Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 61
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Sci Rep ; 10(1): 243, 2020 01 14.
Artigo em Inglês | MEDLINE | ID: mdl-31937831

RESUMO

Oropharyngeal squamous cell carcinoma (OPSCC) incidence is increasing at a nearly epidemic rate, largely driven by the human papillomavirus (HPV). Despite the generally favorable clinical outcomes of patients with HPV driven (HPV+) OPSCC, a significant subset of HPV tumors associated with tobacco exposure have diminished treatment response and worse survival. The tumor immune microenvironment (TIME) has been shown to be a critical driver of treatment response and oncologic outcomes in OPSCC generally and HPV+ OPSCC more specifically. However, the impact of tobacco exposure on the TIME in OPSCC patients remains unclear. We analyzed the relationship between TIME, tobacco exposure and clinical outcomes in OPSCC patients (n = 143) with extensive tobacco exposure (median pack-years = 40). P16 overexpression, a surrogate marker of HPV association, was a strong predictor of relapse-free (RFS) and overall survival (OS) (p < 0.001, p < 0.001 respectively) regardless of tobacco exposure and associated strongly with differential infiltration of the tumor by both CD3 and CD8 lymphocytes measured via immunohistochemistry (p < 001, p < 0.001 respectively). CD3 and CD8 infiltration was a strong predictor of RFS and OS and associated strongly with disease stage (AJCC 8th Edition Staging Manual). Tobacco exposure correlated significantly (p < 0.001) with decreased CD8 infiltration in p16+ OPSCC tumors. Our findings demonstrate that the HPV+ OPSCC clinical outcomes are strongly correlated with the TIME, which is potentially modulated by tobacco exposure. Immunomodulatory strategies targeting this disease in smokers must take into consideration the potential modifying effects of tobacco exposure on treatment effectiveness and clinical outcomes.


Assuntos
Antígenos CD8/metabolismo , Nicotiana/efeitos adversos , Neoplasias Orofaríngeas/induzido quimicamente , Neoplasias Orofaríngeas/metabolismo , Carcinoma de Células Escamosas/induzido quimicamente , Carcinoma de Células Escamosas/imunologia , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/virologia , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Neoplasias Orofaríngeas/imunologia , Neoplasias Orofaríngeas/virologia , Papillomaviridae/fisiologia , Estudos Retrospectivos , Risco , Microambiente Tumoral/efeitos dos fármacos , Microambiente Tumoral/imunologia
2.
Science ; 275(5308): 1943-7, 1997 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-9072974

RESUMO

Mapping of homozygous deletions on human chromosome 10q23 has led to the isolation of a candidate tumor suppressor gene, PTEN, that appears to be mutated at considerable frequency in human cancers. In preliminary screens, mutations of PTEN were detected in 31% (13/42) of glioblastoma cell lines and xenografts, 100% (4/4) of prostate cancer cell lines, 6% (4/65) of breast cancer cell lines and xenografts, and 17% (3/18) of primary glioblastomas. The predicted PTEN product has a protein tyrosine phosphatase domain and extensive homology to tensin, a protein that interacts with actin filaments at focal adhesions. These homologies suggest that PTEN may suppress tumor cell growth by antagonizing protein tyrosine kinases and may regulate tumor cell invasion and metastasis through interactions at focal adhesions.


Assuntos
Cromossomos Humanos Par 10 , Genes Supressores de Tumor , Mutação , Neoplasias/genética , Monoéster Fosfórico Hidrolases , Proteínas Tirosina Fosfatases/genética , Proteínas Supressoras de Tumor , Sequência de Aminoácidos , Neoplasias Encefálicas/genética , Neoplasias da Mama/genética , Mapeamento Cromossômico , Feminino , Mutação da Fase de Leitura , Glioblastoma/genética , Humanos , Masculino , Proteínas dos Microfilamentos/química , Dados de Sequência Molecular , Transplante de Neoplasias , PTEN Fosfo-Hidrolase , Fosfotirosina/metabolismo , Neoplasias da Próstata/genética , Proteínas Tirosina Fosfatases/química , Proteínas Tirosina Fosfatases/fisiologia , Proteínas Tirosina Quinases/antagonistas & inibidores , Deleção de Sequência , Homologia de Sequência de Aminoácidos , Tensinas , Transplante Heterólogo , Células Tumorais Cultivadas
3.
Mol Cell Biol ; 7(10): 3386-93, 1987 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-3683386

RESUMO

We have cloned the human genomic DNA and the corresponding cDNA for the gene which complements the mutation of tsBN51, a temperature-sensitive (Ts) cell cycle mutant of BHK cells which is blocked in G1 at the nonpermissive temperature. After transfecting human DNA into TsBN51 cells and selecting for growth at 39.5 degrees C, Ts+ transformants were identified by their content of human AluI repetitive DNA sequences. Following two additional rounds of transfection, a genomic library was constructed from a tertiary Ts+ transformant and a recombinant phage containing the complementing gene isolated by screening for human AluI sequences. A genomic probe from this clone recognized a 2-kilobase mRNA in human and tertiary transformant cell lines, and this probe was used to isolate a biologically active cDNA from the Okayama-Berg cDNA expression library. Sequencing of this cDNA revealed a single open reading frame encoding a polypeptide of 395 amino acids. The deduced BN51 gene product has a high proportion of acidic and basic amino acids which are clustered in four hydrophilic domains spaced at 60- to 80-amino-acid intervals. These domains have strong sequence homology to each other. Thus, the tsBN51 protein consists of periodic repetitive clusters of acidic and basic amino acids.


Assuntos
Ciclo Celular , Genes , Sequência de Aminoácidos , Animais , Sequência de Bases , Clonagem Molecular , Cricetinae , DNA/genética , Regulação da Expressão Gênica , Teste de Complementação Genética , Humanos , Dados de Sequência Molecular , Conformação Proteica , RNA Mensageiro/genética
4.
Mol Cell Biol ; 13(4): 1998-2010, 1993 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-8455594

RESUMO

The K-fgf/hst oncogene encodes a growth factor of the fibroblast growth factor (FGF) family and transforms cells through an autocrine mechanism which requires extracellular activation of its receptor(s). To identify the cell and tissue targets of K-fgf oncogenic potential in vivo, we constructed a recombinant retrovirus carrying the human K-fgf cDNA and injected it, together with helper Moloney murine leukemia virus, into immunocompetent as well as nude mice. The original construct was highly transforming in tissue culture but produced no detectable pathologies in vivo with the exception of a single fibrosarcoma which arose after a long latency. The virus produced by this tumor appears to have undergone a complex series of recombination events involving the helper Moloney murine leukemia virus. It encodes an Env/K-FGF fusion protein whose expression is under the control of a hybrid long terminal repeat. This virus (designated MFS, for meningeal fibrosarcoma) induces tumors in mice with high frequency and short latency. These neoplasms consist of aggressive fibrosarcomas of soft tissue as well as diffuse meningeal tumors originating from the dura mater that surround the whole central nervous system and cause severe hydrocephalus. The Env/K-FGF fusion protein expressed by the MFS virus has retained all of the biological properties of native K-FGF, including secretion, mitogenic activity, heparin binding, and neutralization by anti-K-FGF antibodies. These and other results indicate that the tumors induced by the MFS virus result from the oncogenic potential of K-FGF.


Assuntos
Transformação Celular Neoplásica/genética , Fibrossarcoma/genética , Neoplasias Meníngeas/genética , Animais , Ciclo Celular , Clonagem Molecular , Regulação Neoplásica da Expressão Gênica , Vetores Genéticos , Hibridização In Situ , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos ICR , Camundongos Nus , Neoplasias Experimentais/genética , RNA Mensageiro/genética , Proteínas Recombinantes de Fusão , Recombinação Genética , Retroviridae/genética , Transfecção
5.
Mol Cell Biol ; 15(1): 94-101, 1995 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-7799973

RESUMO

The three eukaryotic nuclear RNA polymerase (Pol) contain common and unique subunits. Cloning of the unique Pol III subunit genes in yeast cells has revealed a potential homolog in the mammalian system, the BN51 gene. The human BN51 gene was originally isolated as a suppressor of a temperature-sensitive cell cycle mutant of BHK cells (tsBN51). Although tsBN51 cells have a marked decrease in RNA Pol III activity at the nonpermissive temperature, direct biochemical evidence for the BN51 protein being a human Pol III subunit was lacking. Using antibodies directed against the BN51 protein, we show the following: (i) the BN51 protein copurifies with Pol III activity, (ii) Pol III activity can be specifically immunoprecipitated from HeLa nuclear extracts, and (iii) the immunopurified BN51 complex is active in restoring both nonspecific and promoter-specific Pol III activity. Our findings provide direct biochemical evidence for BN51 being a Pol III-specific subunit. Despite the fact that BN51 is not a subunit of Pol I, the production of mature Pol I transcripts is inhibited in tsBN51 cells at the nonpermissive temperature. tsBN51 cells appear defective in processing the 32S precursor rRNA into mature 5.8S and 28S rRNA at the nonpermissive temperature. We surmise that ribosome assembly has halted because of the loss of Pol III transcripts. Thus, there is regulation of the synthesis of mature Pol I transcripts by a posttranscriptional mechanism based on the availability of Pol III transcripts.


Assuntos
Proteínas Nucleares/química , RNA Polimerase III/química , RNA Ribossômico/genética , Células HeLa , Humanos , Substâncias Macromoleculares , Processamento Pós-Transcricional do RNA , RNA Ribossômico/metabolismo , Transcrição Gênica
6.
Mol Cell Biol ; 9(6): 2350-9, 1989 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-2569668

RESUMO

The human ts11 gene was isolated on the basis of its ability to complement the mutation of the BHK cell cycle ts11 mutant, which is blocked in G1 at the nonpermissive temperature. This gene has now been identified as the structural gene for asparagine synthetase (AS) on the bases of sequence homology and the ability of exogenous asparagine to bypass the ts11 block. The ts11 (AS) mRNA has a size of about 2 kilobases and is induced in mid-G1 phase in human, mouse, and hamster cell lines. We have studied the organization and regulation of expression of the ts11 gene. The human ts11 gene consists of 13 exons (the first two noncoding) interspersed in a region of about 21 kilobases of DNA. Transient expression assays using the bacterial chloramphenicol acetyltransferase reporter gene identified two separate promoters: one (ts11 P1) contained in a 280-base-pair region upstream of the first exon and the other (ts11 P2) contained in the first intron. ts11 P1 produced about sixfold more chloramphenicol acetyltransferase activity than did ts11 P2 and had features of the promoters of housekeeping genes: high G + C content, multiple transcription start sites, absence of a TATA box, and presence of putative Sp1 binding sites. ts11 P2 contained a TATA sequence and other elements characteristic of a promoter, but so far we have no evidence of its physiological utilization. The ts11 gene was overexpressed in ts11 cells exposed to the nonpermissive temperature. Addition of asparagine to the culture medium led to a drastic decrease in mRNA levels and prevented G1 induction in serum-stimulated cells, which indicated that expression of the AS gene is regulated by a mechanism of end product inhibition.


Assuntos
Aspartato-Amônia Ligase/genética , Ciclo Celular , Genes , Ligases/genética , Asparagina/fisiologia , Bacteriófagos/genética , Sequência de Bases , Células Cultivadas , Cloranfenicol O-Acetiltransferase/genética , Clonagem Molecular , Éxons , Regulação da Expressão Gênica , Humanos , Íntrons , Dados de Sequência Molecular , Plasmídeos , Regiões Promotoras Genéticas , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Mapeamento por Restrição , Homologia de Sequência do Ácido Nucleico , Transfecção
7.
Oncogene ; 36(5): 618-627, 2017 02 02.
Artigo em Inglês | MEDLINE | ID: mdl-27345403

RESUMO

The role of Notch signaling in prostate cancer has not been defined definitively. Several large scale tissue microarray studies have revealed that the expression of some Notch signaling components including the Jagged1 ligand are upregulated in advanced human prostate cancer specimens. Jagged1 expressed by tumor cells may activate Notch signaling in both adjacent tumor cells and cells in tumor microenvironment. However, it remains undetermined whether increased Jagged1 expression reflects a cause for or a consequence of tumor progression in vivo. To address this question, we generated a novel R26-LSL-JAG1 mouse model that enables spatiotemporal Jagged1 expression. Prostate specific upregulation of Jagged1 neither interferes with prostate epithelial homeostasis nor significantly accelerates tumor initiation or progression in the prostate-specific Pten deletion mouse model for prostate cancer. However, Jagged1 upregulation results in increased inflammatory foci in tumors and incidence of intracystic adenocarcinoma. In addition, Jagged1 overexpression upregulates Tgfß signaling in prostate stromal cells and promotes progression of a reactive stromal microenvironment in the Pten null prostate cancer model. Collectively, Jagged1 overexpression does not significantly accelerate prostate cancer initiation and progression in the context of loss-of-function of Pten, but alters tumor histopathology and microenvironment. Our study also highlights an understudied role of Notch signaling in regulating prostatic stromal homeostasis.


Assuntos
Proteína Jagged-1/metabolismo , PTEN Fosfo-Hidrolase/deficiência , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Animais , Linhagem Celular Tumoral , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Humanos , Proteína Jagged-1/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , PTEN Fosfo-Hidrolase/metabolismo , Neoplasias da Próstata/genética , Células Estromais/metabolismo , Células Estromais/patologia , Regulação para Cima
8.
Oncogene ; 36(33): 4767-4777, 2017 08 17.
Artigo em Inglês | MEDLINE | ID: mdl-28414305

RESUMO

The E3 ubiquitin ligase adaptor speckle-type POZ protein (SPOP) is frequently dysregulated in prostate adenocarcinoma (PC), via either somatic mutations or mRNA downregulation, suggesting an important tumour suppressor function. To examine its physiologic role in the prostate epithelium in vivo, we generated mice with prostate-specific biallelic ablation of Spop. These mice exhibited increased prostate mass, prostate epithelial cell proliferation, and expression of c-MYC protein compared to littermate controls, and eventually developed prostatic intraepithelial neoplasia (PIN). We found that SPOPWT can physically interact with c-MYC protein and, upon exogenous expression in vitro, can promote c-MYC ubiquitination and degradation. This effect was attenuated in PC cells by introducing PC-associated SPOP mutants or upon knockdown of SPOP via short-hairpin-RNA, suggesting that SPOP inactivation directly increases c-MYC protein levels. Gene Set Enrichment Analysis revealed enrichment of Myc-induced genes in transcriptomic signatures associated with SPOPMT. Likewise, we observed strong inverse correlation between c-MYC activity and SPOP mRNA levels in two independent PC patient cohorts. The core SPOPMT;MYCHigh transcriptomic response, defined by the overlap between the SPOPMT and c-MYC transcriptomic programmes, was also associated with inferior clinical outcome in human PCs. Finally, the organoid-forming capacity of Spop-null murine prostate cells was more sensitive to c-MYC inhibition than that of Spop-WT cells, suggesting that c-MYC upregulation functionally contributes to the proliferative phenotype of Spop knock-out prostates. Taken together, our data highlight SPOP as an important regulator of luminal epithelial cell proliferation and c-MYC expression in prostate physiology, identify c-MYC as a novel bona fide SPOP substrate, and help explain the frequent inactivation of SPOP in human PC. We propose SPOPMT-induced stabilization of c-MYC protein as a novel mechanism that can increase total c-MYC levels in PC cells, in addition to amplification of c-MYC locus.


Assuntos
Adenocarcinoma/patologia , Proteínas Nucleares/metabolismo , Próstata/patologia , Neoplasias da Próstata/patologia , Proteínas Proto-Oncogênicas c-myc/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-myc/metabolismo , Proteínas Repressoras/metabolismo , Ubiquitinação , Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Animais , Linhagem Celular Tumoral , Proliferação de Células , Estudos de Coortes , Humanos , Masculino , Camundongos , Camundongos Knockout , Mutação , Proteínas Nucleares/genética , Próstata/metabolismo , Neoplasia Prostática Intraepitelial/genética , Neoplasia Prostática Intraepitelial/metabolismo , Neoplasia Prostática Intraepitelial/patologia , Neoplasias da Próstata/genética , Neoplasias da Próstata/metabolismo , Inibidores de Proteassoma/farmacologia , Receptores Androgênicos/metabolismo , Proteínas Repressoras/genética , Complexos Ubiquitina-Proteína Ligase
9.
Neuroscience ; 138(1): 133-48, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16414199

RESUMO

Human neural stem cells have the ability to differentiate into all three major cell types in the CNS including neurons, astrocytes and oligodendrocytes. The multipotency of human neural stem cells shed a light on the possibility of using stem cells as a therapeutic tool for various neurological disorders including neurodegenerative diseases and neurotrauma that involve a loss of functional neurons. We have discovered previously a priming procedure to direct primarily cultured human neural stem cells to differentiate into almost pure neurons when grafted into adult CNS. However, the molecular mechanism underlying this phenomenon is still unknown. To unravel transcriptional changes of human neural stem cells upon priming, cDNA microarray was used to study temporal changes in human neural stem cell gene expression profile during priming and differentiation. As a result, transcriptional levels of 520 annotated genes were detected changed in at least at two time points during the priming process. In addition, transcription levels of more than 3000 hypothetical protein encoding genes and EST genes were modulated during the priming and differentiation processes of human neural stem cells. We further analyzed the named genes and grouped them into 14 functional categories. Of particular interest, key cell signal transduction pathways, including the G-protein-mediated signaling pathways (heterotrimeric and small monomeric GTPase pathways), the Wnt signaling pathway and the TGF-beta pathway, are modulated by the neural stem cell priming, suggesting important roles of these key signaling pathways in priming and differentiation of human neural stem cells.


Assuntos
Neurônios/fisiologia , Transdução de Sinais/fisiologia , Células-Tronco/fisiologia , Proteínas Morfogenéticas Ósseas/fisiologia , Diferenciação Celular/fisiologia , Células , AMP Cíclico/fisiologia , Imunofluorescência , GTP Fosfo-Hidrolases/metabolismo , Perfilação da Expressão Gênica , Humanos , Linfotoxina-alfa/fisiologia , Microcomputadores , Proteínas Quinases Ativadas por Mitógeno/fisiologia , Família Multigênica , Análise de Sequência com Séries de Oligonucleotídeos , RNA/biossíntese , RNA/genética , Receptores Acoplados a Proteínas G/fisiologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Tretinoína/fisiologia , Fosfolipases Tipo C/fisiologia , Proteínas Wnt/fisiologia
10.
J Natl Cancer Inst ; 93(23): 1783-90, 2001 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-11734594

RESUMO

BACKGROUND: Expression of fibroblast growth factors (FGFs) is increased in a substantial fraction of human prostate cancers in vivo and in prostate cancer cell lines. Altered FGF signaling can potentially have a variety of effects, including stimulating cell proliferation and inhibiting cell death. To determine the biologic significance of altered FGF signaling in human prostate cancer, we disrupted signaling by expression of a dominant-negative (DN) FGF receptor in prostate cancer cell lines. METHODS: PC-3, LNCaP, and DU145 prostate cancer cells were stably transfected with DN FGFR constructs, and LNCaP and DU145 cells were infected with a recombinant adenovirus expressing DN FGFR-1. The effect of DN FGFR-1 expression was assessed by colony-formation assays, cell proliferation assays, flow cytometry, and cytogenetic analysis. Key regulators involved in the G(2)-to-M cell cycle transition were assessed by western blotting to examine cyclin B1 expression and by in vitro kinase assay to assess cdc2 kinase activity. RESULTS: Stable transfection of the DN FGFR-1 construct inhibited colony formation by more than 99% in all three cell lines. Infection of LNCaP and DU145 prostate cancer cells with adenovirus expressing DN FGFR-1 led to extensive cell death within 48 hours. Flow cytometry and cytogenetic analysis revealed that the DN FGFR-1 receptor led to arrest in the G(2) phase of the cell cycle before cell death. Cyclin B1 accumulated in DN FGFR-1-infected LNCaP cells, but cdc2 kinase activity was decreased. CONCLUSIONS: These findings reveal an unexpected dependence of prostate cancer cells on FGF receptor signal transduction to traverse the G(2)/M checkpoint. The mechanism for the G(2) arrest is not clear. Our results raise the possibility that FGF-signaling antagonists might enhance the cell death induced by other prostate cancer therapies.


Assuntos
Neoplasias da Próstata/metabolismo , Receptores de Fatores de Crescimento de Fibroblastos/metabolismo , Receptores de Fatores de Crescimento de Fibroblastos/fisiologia , Transdução de Sinais , Adenoviridae/genética , Western Blotting , Proteína Quinase CDC2/metabolismo , Ciclo Celular , Morte Celular , Divisão Celular , Sobrevivência Celular , Cromossomos , Ciclina B/biossíntese , Ciclina B1 , Citogenética , Citometria de Fluxo , Genes Dominantes , Humanos , Óperon Lac , Masculino , Testes de Precipitina , Fatores de Tempo , Transfecção , Células Tumorais Cultivadas
11.
Cancer Res ; 56(9): 2143-7, 1996 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-8616863

RESUMO

A total of 83 prostate adenocarcinomas was evaluated for allelic loss on chromosome 10 by analysis of loss of polymorphic microsatellite repeats. Initially, 64 stage B carcinomas were analyzed at 12 loci on chromosome 10. Nine cases showed loss of chromosome 10 sequences, with a fractional allelic loss of 20%. These nine cases were then analyzed at an additional 19 loci to define better the regions of loss. Four areas of loss were identified, including 10p (2 of 64 cases), 10q23.1 (7 of 64 cases), 10q23.3 (4 of 64 cases), and 10q26 (2 of 64 cases). Three loci in these regions, D10S111, D10S185, and D10S192, were then analyzed in 19 advanced (stage C and D) carcinomas. Seven (37%) of 19 advanced carcinomas showed allelic loss at one or more of these loci. A statistically significant increase in the fractional allelic loss at both D10S111 (10p) and D10S185 (10q23.1) was observed. Thus, a complex pattern of loss is seen on chromosome 10 in prostate carcinoma, with regions of loss on 10p and 10q, and such loss occurs at a higher rate in clinically advanced disease.


Assuntos
Alelos , Carcinoma/genética , Cromossomos Humanos Par 10 , Neoplasias da Próstata/genética , Deleção de Genes , Humanos , Masculino
12.
Cancer Res ; 60(15): 4245-50, 2000 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-10945637

RESUMO

Fibroblast growth factors (FGFs) are known to play an important role in the growth of normal prostatic epithelial cells. In addition to their effects on proliferation, FGFs can promote cell motility, increase tumor angiogenesis, and inhibit apoptosis, all of which play an important role in tumor progression. To determine whether FGFs are overexpressed in human prostate cancers, we analyzed 26 prostate cancer RNAs by reverse transcription-PCR for expression of FGF3, FGF4, and FGF6, which cannot be detected in normal prostate tissue by this technique. Fourteen of 26 prostate cancers expressed FGF6 mRNA. No expression of FGF3 or FGF4 was detected. An ELISA of tissue extracts of normal prostate, high-grade prostatic intraepithelial neoplasia (PIN), and prostate cancer for FGF6 showed that this growth factor was undetectable in normal prostate but was present at elevated levels in 4 of 9 PIN lesions and in 15 of 24 prostate cancers. Immunohistochemical analysis with anti-FGF6 antibody revealed weak staining of prostatic basal cells in normal prostate that was markedly elevated in PIN. In the prostate cancers, the majority of cases revealed expression of FGF6 by the prostate cancer cells themselves. In two cases, expression was present in prostatic stromal cells. Exogenous FGF6 was able to stimulate proliferation of primary prostatic epithelial and stromal cells, immortalized prostatic epithelial cells, and prostate cancer cell lines in tissue culture. FGF receptor 4, which is the most potent FGF receptor for FGF6, is expressed in the human prostate in vivo and in all of the cultured cell lines. Thus, FGF6 is increased in PIN and prostate cancer and can promote the proliferation of the transformed prostatic epithelial cells via paracrine and autocrine mechanisms.


Assuntos
Fatores de Crescimento de Fibroblastos/biossíntese , Neoplasia Prostática Intraepitelial/metabolismo , Neoplasias da Próstata/metabolismo , Proteínas Proto-Oncogênicas/biossíntese , Divisão Celular/efeitos dos fármacos , Divisão Celular/fisiologia , Ensaio de Imunoadsorção Enzimática , Células Epiteliais/citologia , Células Epiteliais/efeitos dos fármacos , Fator 6 de Crescimento de Fibroblastos , Fatores de Crescimento de Fibroblastos/farmacologia , Fatores de Crescimento de Fibroblastos/fisiologia , Humanos , Imuno-Histoquímica , Masculino , Próstata/citologia , Próstata/efeitos dos fármacos , Neoplasias da Próstata/patologia , Proteínas Proto-Oncogênicas/farmacologia , Proteínas Proto-Oncogênicas/fisiologia , Receptor Tipo 4 de Fator de Crescimento de Fibroblastos , Receptores de Fatores de Crescimento de Fibroblastos/biossíntese , Proteínas Recombinantes/farmacologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células Estromais/citologia , Células Estromais/efeitos dos fármacos , Células Tumorais Cultivadas
13.
Cancer Res ; 61(10): 3882-5, 2001 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-11358800

RESUMO

Caveolin-1 is an integral protein of caveolae, known to play important roles in signal transduction and lipid transport. We demonstrate that caveolin-1 expression is significantly increased in primary and metastatic human prostate cancer after androgen ablation therapy. We also show that caveolin-1 is secreted by androgen-insensitive prostate cancer cells, and that this secretion is regulated by steroid hormones. Significantly, caveolin-1 was detected in the MDL(3) fraction of serum specimens from patients with advanced prostate cancer and to a lesser extent in normal subjects. Conditioned media from high passage caveolin-1 secreting, androgen-insensitive, LNCaP cells stimulated increased viability and clonal growth of low passage, caveolin-1-negative, androgen-sensitive, LNCaP cells in vitro, and this effect was blocked by treating the media with caveolin-1 antibody. i.p. injections of caveolin-1 antibody suppressed the orthotopic growth and spontaneous metastasis of highly metastatic, androgen-insensitive caveolin-1-secreting mouse prostate cancer. Overall, our results establish caveolin-1 as an autocrine/paracrine factor that is associated with androgen-insensitive prostate cancer. We demonstrate the potential for caveolin-1 as a therapeutic target for this important malignancy.


Assuntos
Caveolinas/metabolismo , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Antagonistas de Androgênios/farmacologia , Animais , Anticorpos/farmacologia , Antineoplásicos Hormonais/farmacologia , Neoplasias Ósseas/metabolismo , Neoplasias Ósseas/patologia , Neoplasias Ósseas/secundário , Caveolina 1 , Caveolinas/antagonistas & inibidores , Caveolinas/biossíntese , Divisão Celular/fisiologia , Sobrevivência Celular/fisiologia , Meios de Cultivo Condicionados , Humanos , Masculino , Camundongos , Metástase Neoplásica , Estadiamento de Neoplasias , Neoplasias Hormônio-Dependentes/metabolismo , Neoplasias Hormônio-Dependentes/patologia
14.
Cancer Res ; 60(4): 944-9, 2000 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-10706109

RESUMO

We analyzed the frequency and relevance of mutations in the coding region of the androgen receptor (AR) in genomic DNA extracted from 137 specimens of prostate cancer. The specimens were obtained from the primary tumors of patients affected by stage B disease [15 nonmicrodissected (group 1A) and 84 microdissected (group 1B)] and from the metastatic deposits of individuals with stage D1 disease [8 nonmicrodissected (group 2A) and 30 microdissected (group 2B)] who had not undergone androgen ablation therapy. The study was conducted by PCR-single strand conformational polymorphism (SSCP) analysis of exons 2-8 in the four groups and direct sequence analysis of exon 1 in group 1B. As positive and negative controls, we used genomic DNA extracted from genital skin fibroblasts of patients affected by various forms of androgen resistance with known mutations in the AR. To control for genetic instability, PCR-SSCP analysis of exon 2 of the human progesterone receptor was carried out on each specimen. The overall number of mutations detected was 11 (8%). No mutations were detected in any of the 99 patients with stage B disease. Eleven mutations were detected in exons 2-8 in 8 of the 38 patients with stage D1 disease (all in group 2B). Simultaneous analysis of exon 2 of the progesterone receptor was carried out, and no SSCP changes were identified. These data suggest that AR mutations are rare and presumably do not play a role in the initial phase of prostatic carcinogenesis. The presence of a significant number of AR mutations in metastatic disease indicates that mutations of this molecule may play a role in the most advanced phases of the natural history of this disease, either by facilitating growth or acquisition of the metastatic phenotype.


Assuntos
Mutação , Neoplasias da Próstata/genética , Receptores Androgênicos/genética , Humanos , Masculino , Metástase Neoplásica , Estadiamento de Neoplasias , Polimorfismo Conformacional de Fita Simples , Neoplasias da Próstata/patologia
15.
Oncogene ; 35(46): 5963-5976, 2016 11 17.
Artigo em Inglês | MEDLINE | ID: mdl-26804168

RESUMO

Advanced prostate adenocarcinomas enriched in stem-cell features, as well as variant androgen receptor (AR)-negative neuroendocrine (NE)/small-cell prostate cancers are difficult to treat, and account for up to 30% of prostate cancer-related deaths every year. While existing therapies for prostate cancer such as androgen deprivation therapy (ADT), destroy the bulk of the AR-positive cells within the tumor, eradicating this population eventually leads to castration-resistance, owing to the continued survival of AR-/lo stem-like cells. In this study, we identified a critical nexus between p38MAPK signaling, and the transcription factor Forkhead Box Protein C2 (FOXC2) known to promote cancer stem-cells and metastasis. We demonstrate that prostate cancer cells that are insensitive to ADT, as well as high-grade/NE prostate tumors, are characterized by elevated FOXC2, and that targeting FOXC2 using a well-tolerated p38 inhibitor restores epithelial attributes and ADT-sensitivity, and reduces the shedding of circulating tumor cells in vivo with significant shrinkage in the tumor mass. This study thus specifies a tangible mechanism to target the AR-/lo population of prostate cancer cells with stem-cell properties.


Assuntos
Resistencia a Medicamentos Antineoplásicos , Epitélio/metabolismo , Epitélio/patologia , Fatores de Transcrição Forkhead/metabolismo , Células-Tronco Neoplásicas/efeitos dos fármacos , Células-Tronco Neoplásicas/metabolismo , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Androgênios/metabolismo , Animais , Benzamidas , Linhagem Celular Tumoral , Modelos Animais de Doenças , Transição Epitelial-Mesenquimal/genética , Fatores de Transcrição Forkhead/genética , Expressão Gênica , Humanos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Masculino , Camundongos , Modelos Biológicos , Gradação de Tumores , Nitrilas , Fenótipo , Feniltioidantoína/análogos & derivados , Feniltioidantoína/farmacologia , Antígeno Prostático Específico/metabolismo , Neoplasias da Próstata/tratamento farmacológico , Receptores Androgênicos/metabolismo , Recidiva , Ensaios Antitumorais Modelo de Xenoenxerto , Homeobox 1 de Ligação a E-box em Dedo de Zinco/metabolismo
16.
Prostate Cancer Prostatic Dis ; 8(2): 108-18, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-15724144

RESUMO

The PI3-K-Akt pathway plays a central role in the development and progression of prostate cancer and other malignancies. We review original studies and summarize relevant sections of previous reviews concerning the relationships between abnormalities in the PI3-K-Akt pathway and prostate cancer progression. We discuss laboratory and clinical data that indicate gene perturbation and dysregulation of PI3-K-Akt pathway is common in prostate cancer and other malignancies. We further discuss the critical role of the PI3-K-Akt pathway in the oncogenic signaling network and provide examples that establish the PI3-K-Akt pathway as a focal point for the future development of informative biomarkers and effective therapies for prostate cancer.


Assuntos
Regulação Neoplásica da Expressão Gênica , Fosfatidilinositol 3-Quinases/fisiologia , Neoplasias da Próstata/enzimologia , Neoplasias da Próstata/fisiopatologia , Proteínas Serina-Treonina Quinases/fisiologia , Proteínas Proto-Oncogênicas/fisiologia , Biomarcadores Tumorais/análise , Progressão da Doença , Humanos , Masculino , Fosfatidilinositol 3-Quinases/biossíntese , Fosfatidilinositol 3-Quinases/genética , Proteínas Serina-Treonina Quinases/biossíntese , Proteínas Serina-Treonina Quinases/genética , Proteínas Proto-Oncogênicas/biossíntese , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas c-akt , Transdução de Sinais
17.
Clin Cancer Res ; 5(5): 1063-71, 1999 May.
Artigo em Inglês | MEDLINE | ID: mdl-10353739

RESUMO

Fibroblast growth factors (FGFs) play an important role in the growth and maintenance of the normal prostate. There is increasing evidence from both animal models and analysis of human prostate cancer cell lines that alterations of FGFs and/or FGF receptors (FGFRs) may play an important role in prostate cancer progression. To better define the role of FGF2 and FGF7 in human prostate cancer in vivo, we have quantified these two growth factors in clinically localized human prostate cancers and uninvolved prostate by ELISA and Western blotting and determined their localization by immunohistochemistry. The expression of two of the primary receptors for these growth factors, FGFR-1 and FGFR-2, were also analyzed by immunohistochemistry and Western blotting in these same samples. We have found that FGF2 is significantly increased in prostate cancers when compared with uninvolved prostate and that the FGF2 is present in the stromal fibroblasts and endothelial cells but not the cancer cells. In addition, we have observed overexpression of both FGFR-1 and FGFR-2 in the prostate cancer epithelial cells in a subset of prostate cancers and that such overexpression is correlated with poor differentiation. Thus, there is both an increase in FGF2 concentration in prostate cancers and an increased expression of a receptor capable of responding to this growth factor, establishing a potential paracrine stimulation of prostate cancer cells by the surrounding stromal cells, which may play an important role in prostate cancer progression.


Assuntos
Adenocarcinoma/genética , Fator 2 de Crescimento de Fibroblastos/biossíntese , Fatores de Crescimento de Fibroblastos , Regulação Neoplásica da Expressão Gênica , Proteínas de Neoplasias/biossíntese , Neoplasias da Próstata/genética , Receptores Proteína Tirosina Quinases/biossíntese , Receptores de Fatores de Crescimento de Fibroblastos/biossíntese , Adenocarcinoma/metabolismo , Western Blotting , Diferenciação Celular , Divisão Celular , Cromatografia de Afinidade , Progressão da Doença , Ensaio de Imunoadsorção Enzimática , Células Epiteliais/química , Células Epiteliais/patologia , Fator 10 de Crescimento de Fibroblastos , Fator 2 de Crescimento de Fibroblastos/genética , Fator 2 de Crescimento de Fibroblastos/fisiologia , Fator 7 de Crescimento de Fibroblastos , Fibroblastos/química , Fibroblastos/patologia , Substâncias de Crescimento/biossíntese , Substâncias de Crescimento/genética , Substâncias de Crescimento/fisiologia , Humanos , Técnicas Imunoenzimáticas , Masculino , Proteínas de Neoplasias/genética , Neoplasias da Próstata/metabolismo , Receptores Proteína Tirosina Quinases/genética , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos , Receptores de Fatores de Crescimento de Fibroblastos/genética , Células Estromais/química , Células Estromais/patologia , Células Tumorais Cultivadas
18.
Clin Cancer Res ; 3(10): 1867-72, 1997 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-9815575

RESUMO

To clarify the role in prostate tumorigenesis played by loss of the three known or putative tumor suppressor loci on the centromeric portion of chromosome 13q, we examined 80 clinically localized and 15 advanced prostate carcinomas for allelic loss at microsatellite markers mapped to this region, including markers tightly linked to the BRCA-2, retinoblastoma (Rb), and DBM (deleted in B-cell malignancy) loci. Among the 80 clinically localized cases, 24 showed allelic loss at one or more 13q loci. In all cases with loss, the Rb and/or DBM loci were lost. No cases were found with loss of Rb without loss of DBM or loss of DBM without loss of Rb, implying a role for both the Rb and DBM loci in clinically localized prostate cancer. Loss of the BRCA-2 locus was less common (4 of 55 informative cases) and was always associated with loss of Rb and/or DBM loci. Thus, the BRCA-2 locus does not appear to play as important a role in clinically localized prostate cancer as the Rb and/or DBM loci. Allelic loss on 13q was extremely common in the clinically advanced cases; it was present in 14 of the 15 cases. The rate of allelic loss at each of the three tumor suppressor loci was increased significantly in the advanced cases (P < 0.01, Fisher's exact test). Thus, loss of heterozygosity on 13q is very common in prostate cancer and occurs at all three known or putative tumor suppressor loci on the centromeric portion of chromosome 13q.


Assuntos
Carcinoma/genética , Cromossomos Humanos Par 13/genética , Genes Supressores de Tumor , Neoplasias da Próstata/genética , Alelos , Proteína BRCA2 , Carcinoma/patologia , Genes do Retinoblastoma , Humanos , Perda de Heterozigosidade , Masculino , Proteínas de Neoplasias/genética , Neoplasias da Próstata/patologia , Fatores de Transcrição/genética
19.
Clin Cancer Res ; 4(3): 811-5, 1998 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-9533551

RESUMO

A novel tumor suppressor gene, PTEN, which encodes a dual-specificity protein phosphatase, has recently been identified on chromosome 10q23. We have previously shown that both alleles of this gene are inactivated in three of four prostate cancer cell lines tested. To evaluate the role of inactivation of this gene in primary stage B prostate cancers, 60 cases were analyzed using Southern blotting with PTEN probes and microsatellites on 10q23. Eight of 60 cases had homozygous deletions by Southern blotting. In three of these cases, homozygous deletion was confirmed by apparent retention of heterozygosity at PTEN with loss of heterozygosity at telomeric and centromeric loci. In the remaining five cases, microsatellite analysis was consistent with homozygous deletion. Loss of heterozygosity at PTEN was found in only two cases both by microsatellite analysis and quantitative Southern blotting. No small mutations within PTEN exons were found in any tumors exhibiting alterations on 10q23. Thus, inactivation of the PTEN gene by homozygous deletion occurs in approximately 10-15% of primary stage B prostate carcinomas.


Assuntos
Adenocarcinoma/genética , Cromossomos Humanos Par 10 , Deleção de Genes , Genes Supressores de Tumor , Homozigoto , Monoéster Fosfórico Hidrolases , Neoplasias da Próstata/genética , Proteínas Tirosina Fosfatases/genética , Proteínas Supressoras de Tumor , Adenocarcinoma/classificação , Adenocarcinoma/patologia , Southern Blotting , Mapeamento Cromossômico , Humanos , Masculino , Repetições de Microssatélites , Estadiamento de Neoplasias , PTEN Fosfo-Hidrolase , Reação em Cadeia da Polimerase , Neoplasias da Próstata/classificação , Neoplasias da Próstata/patologia
20.
Clin Cancer Res ; 6(9): 3430-3, 2000 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-10999725

RESUMO

Clinical studies suggest that African-American (AA) prostate cancer patients manifest a more aggressive form of the disease compared with white prostate cancer patients. However, the biological underpinnings of this potential difference remain unresolved. To address this issue, we used specific quantitative immunostaining protocols to determine whether a panel of biomarkers related to apoptosis including caveolin-1, bcl-2, p53, and c-myc were differentially expressed in AA versus white prostate cancer patients with similar clinical and pathological characteristics. We further attempted to correlate biomarker positivity with proliferation-related markers including Ki-67 labeling index and apoptotic index. Interestingly, our results indicated that only the incidence of caveolin-1 staining was significantly different between these two ethnic/racial groups of prostate cancer patients. The incidence of caveolin-1 staining in white patients was 17% compared with 39% in AA patients (P = 0.0048; Fisher's exact test). In addition, the percentage of caveolin-1-positive prostate cancer cells was also higher in moderately differentiated (Gleason score 6) prostate cancer patients in AA versus whites. Because caveolin-1 has been shown previously to demonstrate antiapoptotic activities in prostate cancer cells, our results suggest that differences in caveolin-1 expression may in part underlie the apparent differences in the clinical virulence of this disease in AA versus white prostate cancer patients.


Assuntos
Biomarcadores Tumorais/biossíntese , População Negra , Caveolinas/biossíntese , Neoplasias da Próstata/metabolismo , População Branca , Apoptose/fisiologia , Caveolina 1 , Humanos , Antígeno Ki-67/biossíntese , Masculino , Pessoa de Meia-Idade , Neoplasias da Próstata/patologia , Proteínas Proto-Oncogênicas c-bcl-2/biossíntese , Proteínas Proto-Oncogênicas c-myc/biossíntese , Proteína Supressora de Tumor p53/biossíntese
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA