Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
Oncogene ; 34(28): 3700-10, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25220414

RESUMO

Androgen receptor splicing variants (ARVs) that lack the ligand-binding domain (LBD) are associated with the development of castration-resistant prostate cancer (CRPC), including resistance to the new generation of high-affinity anti-androgens. However, the mechanism by which ARV expression is regulated is not fully understood. In this study, we show that the activation of classical nuclear factor-kappa B (NF-κB) signaling increases the expression of ARVs in prostate cancer (PCa) cells and converts androgen-sensitive PCa cells to become androgen-insensitive, whereas downregulation of NF-κB signaling inhibits ARV expression and restores responsiveness of CRPC to anti-androgen therapy. In addition, we demonstrated that combination of anti-androgen with NF-κB-targeted therapy inhibits efficiently tumor growth of human CRPC xenografts. These results indicate that induction of ARVs by activated NF-κB signaling in PCa cells is a critical mechanism by which the PCa progresses to CRPC. This has important implications as it can prolong the survival of CRPC patients by restoring the tumors to once again respond to conventional androgen-deprivation therapy (ADT).


Assuntos
Antagonistas de Androgênios/administração & dosagem , Antineoplásicos/administração & dosagem , Neoplasias de Próstata Resistentes à Castração/tratamento farmacológico , Receptores Androgênicos/metabolismo , Transdução de Sinais/efeitos dos fármacos , Antagonistas de Androgênios/farmacologia , Anilidas/administração & dosagem , Anilidas/farmacologia , Antineoplásicos/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Ácidos Borônicos/administração & dosagem , Ácidos Borônicos/farmacologia , Bortezomib , Linhagem Celular Tumoral , Humanos , Masculino , NF-kappa B/antagonistas & inibidores , NF-kappa B/metabolismo , Nitrilas/administração & dosagem , Nitrilas/farmacologia , Neoplasias de Próstata Resistentes à Castração/genética , Neoplasias de Próstata Resistentes à Castração/metabolismo , Pirazinas/administração & dosagem , Pirazinas/farmacologia , Receptores Androgênicos/genética , Compostos de Tosil/administração & dosagem , Compostos de Tosil/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto
2.
Prostate Cancer Prostatic Dis ; 17(4): 301-9, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25091041

RESUMO

BACKGROUND: SOX2 is a member of SOX (SRY-related high mobility group box) family of transcription factors. METHODS: In this study, we examined the expression of SOX2 in murine and human prostatic specimens by immunohistochemistry. RESULTS: We found that SOX2 was expressed in murine prostates during budding morphogenesis and in neuroendocrine (NE) prostate cancer (PCa) murine models. Expression of SOX2 was also examined in human prostatic tissue. We found that SOX2 was expressed in 26 of the 30 BPH specimens. In these BPH samples, expression of SOX2 was limited to basal epithelial cells. In contrast, 24 of the 25 primary PCa specimens were negative for SOX2. The only positive primary PCa was the prostatic NE tumor, which also showed co-expression of synaptophysin. Additionally, the expression of SOX2 was detected in all prostatic NE tumor xenograft lines. Furthermore, we have examined the expression of SOX2 on a set of tissue microarrays consisting of metastatic PCa tissues. Expression of SOX2 was detected in at least one metastatic site in 15 of the 24 patients with metastatic castration-resistant PCa; and the expression of SOX2 was correlated with synaptophysin. CONCLUSIONS: SOX2 was expressed in developing prostates, basal cells of BPH, as well as prostatic NE tumors.


Assuntos
Tumores Neuroendócrinos/metabolismo , Próstata/metabolismo , Neoplasias da Próstata/metabolismo , Fatores de Transcrição SOXB1/biossíntese , Animais , Western Blotting , Xenoenxertos , Humanos , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Transgênicos , Próstata/embriologia , Hiperplasia Prostática/metabolismo , Análise Serial de Tecidos
3.
Cell Death Dis ; 3: e361, 2012 Aug 09.
Artigo em Inglês | MEDLINE | ID: mdl-22874998

RESUMO

Recent observations indicate prostatic diseases are comorbidities of systemic metabolic dysfunction. These discoveries revealed fundamental questions regarding the nature of prostate metabolism. We previously showed that prostate-specific ablation of PPARγ in mice resulted in tumorigenesis and active autophagy. Here, we demonstrate control of overlapping and distinct aspects of prostate epithelial metabolism by ectopic expression of individual PPARγ isoforms in PPARγ knockout prostate epithelial cells. Expression and activation of either PPARγ 1 or 2 reduced de novo lipogenesis and oxidative stress and mediated a switch from glucose to fatty acid oxidation through regulation of genes including Pdk4, Fabp4, Lpl, Acot1 and Cd36. Differential effects of PPARγ isoforms included decreased basal cell differentiation, Scd1 expression and triglyceride fatty acid desaturation and increased tumorigenicity by PPARγ1. In contrast, PPARγ2 expression significantly increased basal cell differentiation, Scd1 expression and AR expression and responsiveness. Finally, in confirmation of in vitro data, a PPARγ agonist versus high-fat diet (HFD) regimen in vivo confirmed that PPARγ agonization increased prostatic differentiation markers, whereas HFD downregulated PPARγ-regulated genes and decreased prostate differentiation. These data provide a rationale for pursuing a fundamental metabolic understanding of changes to glucose and fatty acid metabolism in benign and malignant prostatic diseases associated with systemic metabolic stress.


Assuntos
Células Epiteliais/citologia , Redes e Vias Metabólicas , PPAR gama/metabolismo , Animais , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Transformação Celular Neoplásica , Dieta Hiperlipídica , Regulação para Baixo , Células Epiteliais/metabolismo , Ácidos Graxos/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Técnicas de Inativação de Genes , Glucose/metabolismo , Lipogênese , Masculino , Camundongos , Estresse Oxidativo , PPAR gama/agonistas , PPAR gama/genética , Próstata/citologia , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Estearoil-CoA Dessaturase/metabolismo , Tiazolidinedionas/farmacologia
4.
Oncogene ; 31(9): 1130-42, 2012 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-21804603

RESUMO

The stromal microenvironment has key roles in prostate development and cancer, and cancer-associated fibroblasts (CAFs) stimulate tumourigenesis via several mechanisms including the expression of pro-tumourigenic factors. Mesenchyme (embryonic stroma) controls prostate organogenesis, and in some circumstances can re-differentiate prostate tumours. We have applied next-generation Tag profiling to fetal human prostate, normal human prostate fibroblasts (NPFs) and CAFs to identify molecules expressed in prostatic stroma. Comparison of gene expression profiles of a patient-matched pair of NPFs vs CAFs identified 671 transcripts that were enriched in CAFs and 356 transcripts whose levels were decreased, relative to NPFs. Gene ontology analysis revealed that CAF-enriched transcripts were associated with prostate morphogenesis and CAF-depleted transcripts were associated with cell cycle. We selected mRNAs to follow-up by comparison of our data sets with published prostate cancer fibroblast microarray profiles as well as by focusing on transcripts encoding secreted and peripheral membrane proteins, as well as mesenchymal transcripts identified in a previous study from our group. We confirmed differential transcript expression between CAFs and NPFs using QrtPCR, and defined protein localization using immunohistochemistry in fetal prostate, adult prostate and prostate cancer. We demonstrated that ASPN, CAV1, CFH, CTSK, DCN, FBLN1, FHL1, FN, NKTR, OGN, PARVA, S100A6, SPARC, STC1 and ZEB1 proteins showed specific and varied expression patterns in fetal human prostate and in prostate cancer. Colocalization studies suggested that some stromally expressed molecules were also expressed in subsets of tumour epithelia, indicating that they may be novel markers of EMT. Additionally, two molecules (ASPN and STC1) marked overlapping and distinct subregions of stroma associated with tumour epithelia and may represent new CAF markers.


Assuntos
Fibroblastos/metabolismo , Perfilação da Expressão Gênica , Próstata/metabolismo , Neoplasias da Próstata/genética , Adulto , Biomarcadores Tumorais/metabolismo , Caderinas/metabolismo , Feto , Regulação da Expressão Gênica , Humanos , Masculino , Próstata/citologia , Neoplasias da Próstata/metabolismo , Transporte Proteico , Proteoma/metabolismo , Reprodutibilidade dos Testes , Células Estromais/metabolismo , Antígenos Thy-1/metabolismo , Transcriptoma , Microambiente Tumoral/genética
5.
Br J Cancer ; 106(1): 174-81, 2012 Jan 03.
Artigo em Inglês | MEDLINE | ID: mdl-22134510

RESUMO

BACKGROUND: Tumours are made up of a mixed population of different types of cells that include normal structures as well as ones associated with the malignancy, and there are multiple interactions between the malignant cells and the local microenvironment. These intercellular interactions, modulated by the microenvironment, effect tumour progression and represent a largely under-appreciated therapeutic target. We use observations of primary tumour biology from prostate cancer to extrapolate a mathematical model. Specifically, it has been observed that in prostate cancer three disparate cellular outcomes predominate: (i) the tumour remains well differentiated and clinically indolent--in this case the local stromal cells may act to restrain the growth of the cancer; (ii) early in its genesis the tumour acquires a highly malignant phenotype, growing rapidly and displacing the original stromal population (often referred to as small cell prostate cancer)--these less common aggressive tumours are relatively independent of the local microenvironment and (iii) the tumour co-opts the local stroma--taking on a classic stromagenic phenotype where interactions with the local microenvironment are critical to the cancer growth. METHODS: We present an evolutionary game theoretical construct that models the influence of tumour-stroma interactions in driving these outcomes. We consider three characteristic and distinct cellular populations: stromal cells, tumour cells that are self-reliant in terms of microenvironmental factors and tumour cells that depend on the environment for resources, but can also co-opt stroma. RESULTS: Using evolutionary game theory we explore a number of different scenarios that elucidate the impact of tumour-stromal interactions on the dynamics of prostate cancer growth and progression, and how different treatments in the metastatic setting can affect different types of tumours. CONCLUSION: The tumour microenvironment has a crucial role in selecting the traits of the tumour cells that will determine prostate cancer progression. Equally important treatments like hormone therapy affect the selection of these cancer phenotypes making it very important to understand how they impact prostate cancer's somatic evolution.


Assuntos
Evolução Biológica , Modelos Teóricos , Neoplasias da Próstata/patologia , Células Estromais/patologia , Humanos , Masculino
6.
Curr Mol Med ; 10(1): 95-112, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20205682

RESUMO

From the morphogenetic movements of the three germ layers during development to the reactive stromal microenvironment in cancer, tissue interactions are vital to maintaining healthy organ morphologic architecture and function. The stromal compartment is thought to be complicit in tumor progression and, as such, represents an opportune target for disease therapies. However, recent developments in our understanding of the diversity of the stromal compartment and the lack of appropriate models to study its relevance in human disease have limited our further understanding of the role of tissue interactions in tumor progression. The failure any model to fully recapitulate the complexities of systemic biology continue to create a higher imperative for incorporating various perspectives into a broader understanding for the ultimate goal of designing interventional therapies. Understanding this potential, this review examines the biological models used to study stromal-epithelial interactions and includes an attempt to incorporate behavioral terminology to define and mathematically model ecological relationships in stromal-epithelial interactions. In addition, the current attempt to incorporate these diverse ecological perspectives into in silico mathematical models through cross-disciplinary coordination is reviewed, which will provide a fresh perspective on defining cell group behavior and tissue ecology in disease and hopefully lead to the generation of new hypotheses to be empirically validated.


Assuntos
Comunicação Celular , Células Epiteliais/patologia , Neoplasias/patologia , Células Estromais/patologia , Animais , Células Epiteliais/metabolismo , Humanos , Neoplasias/metabolismo , Células Estromais/metabolismo
7.
Oncogene ; 29(17): 2477-87, 2010 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-20140016

RESUMO

The oncogenic PIM1 kinase has been implicated as a cofactor for c-MYC in prostate carcinogenesis. In this study, we show that in human prostate tumors, coexpression of c-MYC and PIM1 is associated with higher Gleason grades. Using a tissue recombination model coupled with lentiviral-mediated gene transfer we find that Pim1 is weakly oncogenic in naive adult mouse prostatic epithelium. However, it cooperates dramatically with c-MYC to induce prostate cancer within 6-weeks. Importantly, c-MYC/Pim1 synergy is critically dependent on Pim1 kinase activity. c-MYC/Pim1 tumors showed increased levels of the active serine-62 (S62) phosphorylated form of c-MYC. Grafts expressing a phosphomimetic c-MYCS62D mutant had higher rates of proliferation than grafts expressing wild type c-MYC but did not form tumors like c-MYC/Pim1 grafts, indicating that Pim1 cooperativity with c-MYC in vivo involves additional mechanisms other than enhancement of c-MYC activity by S62 phosphorylation. c-MYC/Pim1-induced prostate carcinomas show evidence of neuroendocrine (NE) differentiation. Additional studies, including the identification of tumor cells coexpressing androgen receptor and NE cell markers synaptophysin and Ascl1 suggested that NE tumors arose from adenocarcinoma cells through transdifferentiation. These results directly show functional cooperativity between c-MYC and PIM1 in prostate tumorigenesis in vivo and support efforts for targeting PIM1 in prostate cancer.


Assuntos
Neoplasias da Próstata/etiologia , Proteínas Proto-Oncogênicas c-myc/fisiologia , Proteínas Proto-Oncogênicas c-pim-1/fisiologia , Adenocarcinoma/etiologia , Animais , Apoptose , Diferenciação Celular , Proliferação de Células , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Tumores Neuroendócrinos/patologia , Fosforilação , Neoplasias da Próstata/patologia , Proteínas Proto-Oncogênicas c-myc/análise , Proteínas Proto-Oncogênicas c-pim-1/análise
8.
Cell Death Differ ; 17(3): 469-81, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19834493

RESUMO

Peroxisome proliferator-activated receptor-gamma (PPARgamma) regulates the interface between cellular lipid metabolism, redox status and organelle differentiation. Conditional prostatic epithelial knockout of PPARgamma in mice resulted in focal hyperplasia which developed into mouse prostatic intraepithelial neoplasia (mPIN). The grade of PIN became more severe with time. Electron microscopy (EM) showed accumulated secondary lysosomes containing cellular organelles and debris suggestive of autophagy. Consistent with this analysis the autophagy marker LC-3 was found to be upregulated in areas of PIN in PPARgamma KO tissues. We selectively knocked down PPARgamma2 isoform in wild-type mouse prostatic epithelial cells and examined the consequences of this in a tissue recombination model. Histopathologically grafted tissues resembled the conditional PPARgamma KO mouse prostates. EM studies of PPARgamma- and PPARgamma2-deficient epithelial cells in vitro were suggestive of autophagy, consistent with the prostatic tissue analysis. This was confirmed by examining expression of beclin-1 and LC-3. Gene expression profiling in PPARgamma-/gamma2-deficient cells indicated a major dysregulation of cell cycle control and metabolic signaling networks related to peroxisomal and lysosomal maturation, lipid oxidation and degradation. The putative autophagic phenotypes of PPARgamma-deficient cells could be rescued by re-expression of either gamma1 or gamma2 isoform. We conclude that disruption of PPARgamma signaling results in autophagy and oxidative stress during mPIN pathogenesis.


Assuntos
Autofagia/fisiologia , PPAR gama/metabolismo , Neoplasia Prostática Intraepitelial/metabolismo , Neoplasias da Próstata/metabolismo , Isoformas de Proteínas/metabolismo , Transdução de Sinais/fisiologia , Animais , Linhagem Celular , Células Epiteliais/metabolismo , Células Epiteliais/ultraestrutura , Perfilação da Expressão Gênica , Hipoglicemiantes/metabolismo , Masculino , Camundongos , Camundongos SCID , Camundongos Transgênicos , Dados de Sequência Molecular , PPAR gama/genética , Fenótipo , Próstata/citologia , Próstata/metabolismo , Próstata/patologia , Neoplasia Prostática Intraepitelial/patologia , Neoplasias da Próstata/patologia , Isoformas de Proteínas/genética , Rosiglitazona , Tiazolidinedionas/metabolismo , Transplante Heterólogo
9.
Biochim Biophys Acta ; 1787(12): 1433-43, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19524545

RESUMO

Many cancer cells have an unusual ability to grow in hypoxia, but the origins of this metabolic phenotype remain unclear. We compared the metabolic phenotypes of three common prostate cancer cell models (LNCaP, DU145, PC3), assessing energy metabolism, metabolic gene expression, and the response to various culture contexts (in vitro and xenografts). LNCaP cells had a more oxidative phenotype than PC3 and DU145 cells based upon respiration, lactate production, [ATP], metabolic gene expression, and sensitivity of these parameters to hypoxia. PC3 and DU145 cells possessed similar Complex II and mtDNA levels, but lower Complex III and IV activities, and were unresponsive to dinitrophenol or dichloroacetate, suggesting that their glycolytic phenotype is due to mitochondrial dysfunction rather than regulation. High passage under normoxia converted LNCaP from oxidative to glycolytic cells (based on respiration and lactate production), and altered metabolic gene expression. Though LNCaP-derived cells differed from the parental line in mitochondrial enzyme activities, none differed in mitochondrial content (assessed as cardiolipin levels). When LNCaP-derived cells were grown as xenografts in immunodeficient mice, there were elements of a hypoxic response (e.g., elevated VEGF mRNA) but line-specific changes in expression of select glycolytic, mitochondrial and fatty acid metabolic genes. Low oxygen in vitro did not influence the mRNA levels of SREBP axis, nor did it significantly alter triglyceride production in any of the cell lines suggesting that the pathway of de novo fatty acid synthesis is not directly upregulated by hypoxic conditions. Collectively, these studies demonstrate important differences in the metabolism of these prostate cancer models. Such metabolic differences would have important ramifications for therapeutic strategies involving metabolic targets.


Assuntos
Hipóxia Celular , Neoplasias da Próstata/metabolismo , Animais , Linhagem Celular Tumoral , Expressão Gênica , Glicólise , Humanos , Lipídeos/biossíntese , Masculino , Camundongos , Camundongos SCID , Mitocôndrias/enzimologia , Fenótipo , Neoplasias da Próstata/patologia , Proteína de Ligação a Elemento Regulador de Esterol 2/fisiologia
10.
J Physiol Pharmacol ; 60 Suppl 4: 47-55, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20083851

RESUMO

Structural chromosome aberrations are known hallmarks of many solid tumors. In the papillary form of thyroid cancer (PTC), for example, activation of the receptor tyrosine kinase (RTK) genes, ret or the neurotrophic tyrosine kinase receptor type I (NTRK1) by intra- or interchromosomal rearrangements have been suggested as a cause of the disease. The 1986 accident at the nuclear power plant in Chernobyl, Ukraine, led to the uncontrolled release of high levels of radioisotopes. Ten years later, the incidence of childhood papillary thyroid cancer (chPTC) near Chernobyl had risen by two orders of magnitude. Tumors removed from some of these patients showed aberrant expression of the ret RTK gene due to a ret/PTC1 or ret/PTC3 rearrangement involving chromosome 10. However, many cultured chPTC cells show a normal G-banded karyotype and no ret rearrangement. We hypothesize that the "ret-negative" tumors inappropriately express a different oncogene or have lost function of a tumor suppressor as a result of chromosomal rearrangements, and decided to apply molecular and cytogenetic methods to search for potentially oncogenic chromosomal rearrangements in Chernobyl chPTC cases. Knowledge of the kind of genetic alterations may facilitate the early detection and staging of chPTC as well as provide guidance for therapeutic intervention.


Assuntos
Carcinoma Papilar/enzimologia , Cromossomos/ultraestrutura , Proteínas Quinases/biossíntese , Neoplasias da Glândula Tireoide/enzimologia , Animais , Carcinoma Papilar/ultraestrutura , Linhagem Celular , Transplante de Células , Acidente Nuclear de Chernobyl , Aberrações Cromossômicas , Cromossomos Artificiais Bacterianos/genética , Clonagem Molecular , Sondas de DNA , Citometria de Fluxo , Humanos , Processamento de Imagem Assistida por Computador , Cariotipagem , Camundongos , Receptores Proteína Tirosina Quinases/metabolismo , Receptor trkA/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Neoplasias da Glândula Tireoide/ultraestrutura , Translocação Genética
11.
Oncogene ; 27(56): 7118-30, 2008 Nov 27.
Artigo em Inglês | MEDLINE | ID: mdl-18724388

RESUMO

Transforming growth factor (TGF)-beta is an important paracrine factor in tumorigenesis. Ligand binding of the type I and II TGF-beta receptors initiate downstream signaling. The role of stromal TGF-beta signaling in prostate cancer progression is unknown. In mice, the conditional stromal knockout of the TGF-beta type II receptor expression (Tgfbr2(fspKO)) resulted in the development of prostatic intraepithelial neoplasia and progression to adenocarcinoma within 7 months. Clinically, we observed a loss of TGF-beta receptor type II expression in 69% of human prostate cancer-associated stroma, compared to 15% of stroma associated with benign tissues (n=140, P-value <0.0001). To investigate the mechanism of paracrine TGF-beta signaling in prostate cancer progression, we compared the effect of the prostatic stromal cells from Tgfbr2(fspKO) and floxed TGF-beta type II receptor Tgfbr2(floxE2/floxE2) mice on LNCaP human prostate cancer cells in vitro and tissue recombination xenografts. Induction of LNCaP cell proliferation and tumorigenesis was observed by Tgfbr2(fspKO) prostate stroma as a result of elevated Wnt3a expression. Neutralizing antibodies to Wnt3a reversed LNCaP tumorigenesis. The TGF-beta inhibition of Wnt3a expression was in part through the suppression of Stat3 activity on the Wnt3a promoter. In conclusion, the frequent loss of stromal TGF-beta type II receptor expression in human prostate cancer can relieve the paracrine suppression of Wnt3a expression.


Assuntos
Regulação Neoplásica da Expressão Gênica , Neoplasias da Próstata/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Proteínas Wnt/metabolismo , Adenocarcinoma/metabolismo , Animais , Progressão da Doença , Humanos , Ligantes , Masculino , Camundongos , Camundongos Knockout , Camundongos SCID , Modelos Biológicos , Transplante de Neoplasias , Comunicação Parácrina , Proteínas Serina-Treonina Quinases/metabolismo , Receptor do Fator de Crescimento Transformador beta Tipo II , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Transdução de Sinais , Proteína Wnt3 , Proteína Wnt3A
13.
Cancer Res ; 61(22): 8135-42, 2001 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-11719442

RESUMO

The human prostatic epithelial cell line BPH-1 is normally nontumorigenic in nude mice. The present report demonstrates that this cell line can be permanently transformed by its microenvironment to become tumorigenic. The establishment of a series of tumorigenic sublines based on this parental cell line is described. BPH-1 cells were induced to form tumors either by recombination with human prostatic carcinoma-associated fibroblasts (CAFs) or by exposure to carcinogenic doses of testosterone and estradiol (T+E2) after recombination with rat urogenital sinus mesenchyme. Epithelial cells isolated from these tumors were established as cell strains in culture. When regrafted to nude mouse hosts epithelial cells isolated from CAF- or T+E2-induced tumors were found to be consistently tumorigenic even in the absence of CAF or T+E2. The T+E2-induced cell strains have been designated BPH1(TETD)-A and -B and the CAF-induced strains are designated BPH1(CAFTD)-01 through -08. In vitro, the cells had an epithelial morphology with a less well-defined cobblestone pattern than the parental line. They express SV40 large T antigen, confirming their derivation from the parental BPH-1 line. The BPH1(CAFTD) strains formed colonies in soft agar, whereas the parental BPH-1 cells and the BPH1(TETD) sublines did not. There was no immunocytochemically detectable expression of androgen (AR), alpha-estrogen (ERalpha), or progesterone (PR) receptors by the parental BPH-1 cell line or by any of the tumor-derived cell strains. The cells uniformly coexpressed both basal and luminal cell-type cytokeratins and the basal cell marker p63. When grafted beneath the renal capsule of athymic mouse hosts, all of the tumor-derived cell strains consistently formed tumors. These were predominantly poorly or moderately differentiated squamous or adenosquamous tumors, similar in organization to the primary tumors from which the cell strains were derived. The cell strains continued to express both basal- and luminal-type cytokeratins in vivo. Some of the cell strains also coexpressed vimentin. E-cadherin expression was absent from many of the cells, although patches of cells expressing this marker were seen. The cells continued to express SV40T antigen. These cell strains, which are all derived from a common nontumorigenic progenitor, represent a useful resource for examining genetic and phenotypic changes during carcinogenesis.


Assuntos
Transformação Celular Neoplásica/patologia , Próstata/patologia , Neoplasias da Próstata/etiologia , Animais , Comunicação Celular , Transformação Celular Neoplásica/genética , Células Epiteliais/patologia , Estradiol/toxicidade , Feminino , Fibroblastos/patologia , Humanos , Masculino , Camundongos , Camundongos Nus , Transplante de Neoplasias , Gravidez , Neoplasias da Próstata/induzido quimicamente , Neoplasias da Próstata/patologia , Ratos , Testosterona/toxicidade , Transplante Heterólogo
14.
Cancer Res ; 61(22): 8143-9, 2001 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-11719443

RESUMO

Here we report the genetic characterization of immortalized prostate epithelial cells before and after conversion to tumorigenicity using molecular cytogenetics and microarray technology. We were particularly interested to analyze the consequences of acquired chromosomal aneuploidies with respect to modifications of gene expression profiles. Compared with nontumorigenic but immortalized prostate epithelium, prostate tumor cell lines showed high levels of chromosomal rearrangements that led to gains of 1p, 5, 11q, 12p, 16q, and 20q and losses of 1pter, 11p, 17, 20p, 21, 22, and Y. Of 5700 unique targets on a 6.5K cDNA microarray, approximately 3% were subject to modification in expression levels; these included GRO-1, -2, IAP-1,- 2, MMP-9, and cyclin D1, which showed increased expression, and TRAIL, BRCA1, and CTNNA, which showed decreased expression. Thirty % of expression changes occurred in regions the genomic copy number of which remained balanced. Of the remainder, 42% of down-regulated and 51% of up-regulated genes mapped to regions present in decreased or increased genomic copy numbers, respectively. A relative gain or loss of a chromosome or chromosomal arm usually resulted in a statistically significant increase or decrease, respectively, in the average expression level of all of the genes on the chromosome. However, of these genes, very few (e.g., 5 of 101 genes on chromosome 11q), and in some instances only two genes (MMP-9 and PROCR on chromosome 20q), were overexpressed by > or =1.7-fold when scored individually. Cluster analysis by gene function suggests that prostate tumorigenesis in these cell line models involves alterations in gene expression that may favor invasion, prevent apoptosis, and promote growth.


Assuntos
Aneuploidia , Perfilação da Expressão Gênica , Neoplasias da Próstata/genética , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/patologia , Regulação Neoplásica da Expressão Gênica/genética , Humanos , Cariotipagem , Masculino , Hibridização de Ácido Nucleico , Análise de Sequência com Séries de Oligonucleotídeos , Neoplasias da Próstata/patologia , Translocação Genética , Células Tumorais Cultivadas
15.
Cancer Res ; 61(16): 6064-72, 2001 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-11507055

RESUMO

The effects of stromal and hormonal environment on the immortalized but nontumorigenic human prostatic epithelial cell line BPH-1 were investigated in an in vivo model. BPH-1 cells were recombined with rat urogenital sinus mesenchyme (UGM), and the tissue recombinants were grafted to the renal capsule of adult male athymic mouse hosts. BPH-1 + UGM recombinants formed solid branching epithelial cords with a well-defined basement membrane. The cords canalized to form ductal structures. The mesenchymal cells formed thick sheets of well-differentiated smooth muscle surrounding the epithelium, reinforcing the idea that the epithelium dictates the patterning of prostatic stromal cells. When hosts carrying BPH-1 + UGM tissue recombinants were exposed to testosterone propionate and 17-beta-estradiol (T + E2), the tissue recombinants responded by forming invasive carcinomas, demonstrating mixed, predominantly squamous as well as adenocarcinomatous (small acinar and mucinous) differentiation. When either untreated or T + E2-treated hosts were castrated, epithelial apoptosis was observed in the grafts. When tumors were removed and regrafted to fresh hosts they grew rapidly. Tumors were serially regrafted through six generations. Histologically these tumors consisted largely of focally keratinizing squamous cell carcinoma with high-grade malignant cytological features. BPH-1 cells grown in the absence of UGM survived at the graft site but did not form tumors or organized structures. This behavior was not influenced by the presence or absence of T + E2 stimulation. These data show that an immortalized, nontumorigenic human prostatic epithelial cell line can undergo hormonal carcinogenesis in response to T + E2 stimulation. In addition, the data demonstrate that the stromal environment plays an important role in mediating hormonal carcinogenesis.


Assuntos
Transformação Celular Neoplásica/efeitos dos fármacos , Estradiol/toxicidade , Neoplasias da Próstata/induzido quimicamente , Testosterona/toxicidade , Animais , Antígenos Transformantes de Poliomavirus/fisiologia , Apoptose/fisiologia , Divisão Celular/efeitos dos fármacos , Linhagem Celular , Transformação Celular Neoplásica/patologia , Células Epiteliais/patologia , Feminino , Humanos , Masculino , Mesoderma/citologia , Mesoderma/fisiologia , Camundongos , Camundongos Nus , Orquiectomia , Gravidez , Neoplasias da Próstata/patologia , Ratos , Ensaio de Cápsula Sub-Renal , Sistema Urogenital/citologia , Sistema Urogenital/embriologia , Sistema Urogenital/fisiologia
16.
Reprod Fertil Dev ; 13(4): 285-96, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-11800167

RESUMO

Estrogens, alone or in combination with androgens, can induce aberrant growth and/or malignancy of the prostate gland. Squamous metaplasia is an abnormal form of prostatic epithelial differentiation elicited by exogenous estrogen alone. Estrogens elicit their effects via estrogen receptors (ER) in the prostate. Experiments using ERalpha and ERbeta null mice demonstrated that ERalpha, but not ERbeta is essential in the induction of prostatic squamous metaplasia. To determine the respective roles of epithelial versus stromal ERalpha in this response, the following tissue recombinants were constructed with prostatic epithelium (PRE) and stroma (S) from wild-type (wt) and ERalpha knockout (alphaERKO) mice: wt-S + wt-PRE, alphaERKO-S + alphaERKO-PRE, wt-S + alphaERKO-PRE and alphaERKO-S + wt-PRE. A metaplastic response to diethylstilbestrol (DES) was only observed in wt-S + wt-PRE tissue recombinants. Tissue recombinants containing alphaERKO-PRE and/or alphaERKO-S (alphaERKO-S + alphaERKO-PRE, wt-S + alphaERKO-PRE and alphaERKO-S + wt-PRE) failed to respond to DES. Therefore, full and uniform epithelial squamous metaplasia requires ERalpha in both the epithelium and stroma. Estradiol (E2) in combination with testosterone (T) was shown to be effective in inducing prostatic carcinogenesis in a tissue recombinant model composed of rat urogenital sinus mesenchyme plus mouse prostatic epithelium. A particularly efficient model of prostatic carcinogenesis in mice involves T + E2 treatment of mice bearing grafts of wild-type rat urogenital mesenchyme (rUGM) plus retinoblastoma gene (Rb) knockout (Rb-KO) prostatic epithelium (rUGM + Rb-KO-PRE). Such rUGM + Rb-KO-PRE tissue recombinants developed hyperplasia, atypical hyperplasia and invasive prostatic carcinoma with high efficiency. During carcinogenesis in rUGM + Rb-KO-PRE tissue recombinants, epithelial E-cadherin almost totally disappeared and epithelial PCNA labeling was elevated. These epithelial changes were associated with almost total loss of smooth muscle cells in the stroma. The results of this study demonstrate that the absence of the Rb tumor suppressor gene predisposes prostatic epithelial cells to hormonal carcinogenesis.


Assuntos
Estrogênios/fisiologia , Próstata/patologia , Neoplasias da Próstata/metabolismo , Animais , Diferenciação Celular , Receptor alfa de Estrogênio , Humanos , Masculino , Camundongos , Camundongos Knockout , Próstata/metabolismo , Neoplasias da Próstata/fisiopatologia , Receptores de Estrogênio/genética , Receptores de Estrogênio/metabolismo
17.
Cancer Res ; 60(21): 6008-17, 2000 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-11085521

RESUMO

The retinoblastoma (Rb) gene product is a prototypic tumor suppressor. Mice lacking the Rb gene are not viable and die in utero at approximately 13 days of gestation. In this study, we have rescued Rb-/- prostates by grafting pelvic organ rudiments from Rb-/- mouse embryos under the renal capsule of adult male nude mouse hosts. Grafts of embryonic pelvic organs developed into functional prostatic tissue. Some of the prostatic tissue generated was further used to construct chimeric prostatic tissue recombinants by combining wild-type rat urogenital mesenchyme (rUGM) with Rb-/- and Rb+/+ prostatic epithelium (PRE). The tissue recombinants were grown as subcapsular renal grafts and treated from the time of grafting with Silastic capsules containing 25 mg of testosterone plus 2.5 mg of estradiol. During 5-8 weeks of hormone treatment, rUGM+Rb+/+PRE tissue recombinants developed prostatic hyperplasia, whereas PRE in rUGM+Rb-/-PRE tissue recombinants developed hyperplasia, atypical hyperplasia, and carcinoma. During carcinogenesis in rUGM+Rb-/-PRE tissue recombinants, prostatic epithelial cells of the basal lineage disappeared, whereas the luminal cells underwent carcinogenesis. Epithelial E-cadherin almost totally disappeared. In all cases, epithelial PCNA labeling was elevated in tissue recombinants containing Rb-/- versus Rb+/+ epithelium. These epithelial changes were associated with almost total loss of smooth muscle cells in the stroma. In contrast, in untreated hosts rUGM+Rb+/+PRE tissue recombinants developed normally, and rUGM+Rb-/-PRE tissue recombinants developed mild epithelial hyperplasia. The results of this study demonstrate that Rb-/- prostatic tissue can be rescued from embryonic lethal mice and used to test its susceptibility to hormonal carcinogenesis. Deletion of the Rb gene predisposes prostatic epithelium to hyperplasia and increases proliferative activity Susceptibility to hormonal carcinogenesis in response to exogenous testosterone + estradiol is manifested in the progression from atypica hyperplasia to carcinoma. Thus, these findings demonstrate that the absence of the Rb tumor suppressor gene may predispose prostatic epithelial cells to carcinogenesis. Rescue of organs from Rb-/- embryos not only provides an opportunity to analyze the Rb gene pathway in the development and progression of prostate cancer but also provides an opportunity for specifically evaluating the role of the Rb pathway in development and carcinogenesis in other organs, such as the mammary gland and colon. Because rUGM greatly stimulates prostatic epithelial proliferation, the tissue recombinant model is a particularly useful tool for assessing the functional role of other genes in prostatic carcinogenesis through use of the appropriate transgenic or gene knockout mice.


Assuntos
Cocarcinogênese , Estradiol/toxicidade , Hormônios Esteroides Gonadais/toxicidade , Neoplasias da Próstata/induzido quimicamente , Neoplasias da Próstata/genética , Proteína do Retinoblastoma/deficiência , Testosterona/toxicidade , Animais , Modelos Animais de Doenças , Epitélio/efeitos dos fármacos , Epitélio/metabolismo , Epitélio/patologia , Feminino , Genes do Retinoblastoma/fisiologia , Masculino , Mesoderma/efeitos dos fármacos , Mesoderma/metabolismo , Mesoderma/patologia , Camundongos , Camundongos Knockout , Camundongos Nus , Gravidez , Hiperplasia Prostática/induzido quimicamente , Hiperplasia Prostática/genética , Neoplasias da Próstata/patologia , Ratos , Ratos Sprague-Dawley , Proteína do Retinoblastoma/genética , Ensaio de Cápsula Sub-Renal
18.
Radiol Clin North Am ; 38(1): 1-14, 2000 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-10664663

RESUMO

The development of the prostate is controlled by steroid hormones that in turn induce and maintain a complex and little understood cross talk between the various cell types making up the gland. The result of this intercellular communication can be either new growth or growth quiescence, depending upon the differentiation state of the cell type being stimulated. Secretory function of the prostate is dependent upon direct stimulation of fully differentiated prostatic epithelial cells by androgens. The prostate thus seems to be regulated in a similar manner to other organs of the male and female genital tract with proliferative control mediated by cell-cell interactions, whereas differentiated function is determined by direct steroid action on the parenchymal cells.


Assuntos
Próstata/crescimento & desenvolvimento , Androgênios/fisiologia , Comunicação Celular/fisiologia , Diferenciação Celular , Divisão Celular/fisiologia , Células Epiteliais/metabolismo , Células Epiteliais/fisiologia , Feminino , Hormônios Esteroides Gonadais/fisiologia , Humanos , Masculino , Mesoderma/fisiologia , Próstata/citologia , Próstata/metabolismo , Próstata/fisiologia , Esteroides/fisiologia
20.
Cancer Res ; 59(19): 5002-11, 1999 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-10519415

RESUMO

The present study demonstrates that fibroblasts associated with carcinomas stimulate tumor progression of initiated nontumorigenic epithelial cells both in an in vivo tissue recombination system and in an in vitro coculture system. Human prostatic carcinoma-associated fibroblasts grown with initiated human prostatic epithelial cells dramatically stimulated growth and altered histology of the epithelial population. This effect was not detected when normal prostatic fibroblasts were grown with the initiated epithelial cells under the same experimental conditions. In contrast, carcinoma-associated fibroblasts did not affect growth of normal human prostatic epithelial cells under identical conditions. From these data, we conclude that in this human prostate cancer model, carcinoma-associated fibroblasts stimulate progression of tumorigenesis. Thus, carcinoma-associated fibroblasts can direct tumor progression of an initiated prostate epithelial cell.


Assuntos
Células Epiteliais/patologia , Fibroblastos/patologia , Próstata/patologia , Neoplasias da Próstata/patologia , Neoplasias da Próstata/fisiopatologia , Animais , Linhagem Celular , Sobrevivência Celular , Técnicas de Cocultura , Progressão da Doença , Células Epiteliais/citologia , Fibroblastos/citologia , Humanos , Cariotipagem , Queratinas/análise , Masculino , Camundongos , Camundongos Nus , Próstata/citologia , Ratos , Ratos Nus , Transplante Heterólogo , Vimentina/análise
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA