Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 52
Filtrar
1.
J Pathol ; 194(3): 341-8, 2001 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-11439367

RESUMO

Loss of PTEN (phosphatase and tensin homologue deleted from chromosome 10) function has been implicated in the progression of several types of cancer. Allele loss close to the PTEN locus occurs in sporadic colon cancer and germline PTEN mutations cause Cowden disease, an inherited cancer syndrome characterized by an increased incidence of gastrointestinal tract lesions that can progress to colorectal carcinoma. However, although PTEN is a good candidate for involvement in the pathogenesis of sporadic colon cancer, previous analyses have not revealed a high frequency of somatic mutations in colorectal tumours. Alternative mechanisms which could lead to a loss of PTEN expression in colon cancer have not been investigated. This study monitored PTEN mRNA and protein levels in a panel of 50 tumour tissues obtained from 35 patients with sporadic colon cancer. RT-PCR and immunohistochemistry were used to evaluate the expression of mRNA and protein, respectively, in normal, adenoma and adenocarcinoma colorectal tissues as well as in metastatic lesions. To overcome the problem of heterogeneity and normal stromal cell contamination in homogenized tissue specimens, specific cell types were isolated by microdissection prior to PCR analysis. No loss of PTEN expression was evident in any of the colon tissues examined. PTEN protein was localized exclusively in the cytoplasm of normal and tumour cells and no correlation of immunostaining intensity and tumour stage or grade was revealed. As with previous deletion and mutation analyses, the present study suggests that loss of PTEN expression is not prevalent in sporadic colon cancer.


Assuntos
Adenocarcinoma/genética , Adenocarcinoma/secundário , Neoplasias Colorretais/genética , Neoplasias Colorretais/secundário , Monoéster Fosfórico Hidrolases/genética , RNA Mensageiro/análise , Proteínas Supressoras de Tumor , Adenoma/genética , Animais , Western Blotting/métodos , Deleção de Genes , Humanos , Imuno-Histoquímica/métodos , Camundongos , Camundongos Nus , Transplante de Neoplasias , PTEN Fosfo-Hidrolase , Monoéster Fosfórico Hidrolases/análise , Reação em Cadeia da Polimerase Via Transcriptase Reversa
2.
Cancer Gene Ther ; 8(12): 982-9, 2001 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11781661

RESUMO

Progressive growth and metastasis of solid tumors require angiogenesis, or the formation of new blood vessels. Endostatin is a 20-kDa carboxy-terminal fragment of collagen XVIII that has been shown to inhibit endothelial cell proliferation and tumor angiogenesis. Replication-deficient recombinant adenovirus (rAd) vectors were constructed, which encoded secreted forms of human and mouse endostatin (HECB and MECB, respectively), and, as a control, human alkaline phosphatase (APCB). Accumulation of endostatin was demonstrated in supernatants of cultured cells infected with the endostatin rAds. These supernatants disrupted tubule formation, inhibited migration and proliferation, and induced apoptosis in human dermal vascular endothelial cells or human vascular endothelial cells. Endostatin-containing supernatants had no effect on the proliferation of MidT2-1 mouse mammary tumor cells in vitro. A pharmacokinetic study of MECB in immunocompetent FVB mice demonstrated a 10-fold increase of serum endostatin concentrations 3 days after intravenous administration of 1x10(10) particles of this rAd (215-257 ng/mL compared to 12-38 ng/mL in control rAd-treated mice). Intravenous administration of MECB reduced b-FGF stimulated angiogenesis into Matrigel plugs by 38%. Intratumoral MECB inhibited growth of MidT2-1 syngeneic mammary tumors in FVB mice, but had minimal impact on the growth of MDA-MB-231 human breast tumors in SCID mice. Intravenous therapy with MECB also initially inhibited growth of MidT2-1 tumors, but this activity was subsequently blocked by induced anti-rAd antibodies. In summary, endostatin gene therapy effectively suppressed angiogenic processes in vitro and in vivo in several model systems.


Assuntos
Inibidores da Angiogênese/genética , Colágeno/genética , Terapia Genética , Neovascularização Patológica/genética , Neovascularização Patológica/terapia , Fragmentos de Peptídeos/genética , Adenoviridae , Inibidores da Angiogênese/uso terapêutico , Animais , Células Cultivadas , Colágeno/uso terapêutico , Colágeno Tipo XVIII , Endostatinas , Endotélio Vascular/patologia , Endotélio Vascular/fisiologia , Vetores Genéticos , Humanos , Camundongos , Fragmentos de Peptídeos/uso terapêutico , Vírus Reordenados
3.
J Biol Chem ; 275(17): 12889-95, 2000 Apr 28.
Artigo em Inglês | MEDLINE | ID: mdl-10777587

RESUMO

PTEN is a tumor suppressor with sequence homology to protein-tyrosine phosphatases and the cytoskeleton protein tensin. PTEN is capable of dephosphorylating phosphatidylinositol 3,4, 5-trisphosphate in vitro and down-regulating its levels in insulin-stimulated 293 cells. To study the role of PTEN in insulin signaling, we overexpressed PTEN in 3T3-L1 adipocytes approximately 30-fold above uninfected or control virus (green fluorescent protein)-infected cells, using an adenovirus gene transfer system. PTEN overexpression inhibited insulin-induced 2-deoxy-glucose uptake by 36%, GLUT4 translocation by 35%, and membrane ruffling by 50%, all of which are phosphatidylinositol 3-kinase-dependent processes, compared with uninfected cells or cells infected with control virus. Microinjection of an anti-PTEN antibody increased basal and insulin stimulated GLUT4 translocation, suggesting that inhibition of endogenous PTEN function led to an increase in intracellular phosphatidylinositol 3,4,5-trisphosphate levels, which stimulates GLUT4 translocation. Further, insulin-induced phosphorylation of downstream targets Akt and p70S6 kinase were also inhibited significantly by overexpression of PTEN, whereas tyrosine phosphorylation of the insulin receptor and IRS-1 or the phosphorylation of mitogen-activated protein kinase were not affected, suggesting that the Ras/mitogen-activated protein kinase pathway remains fully functional. Thus, we conclude that PTEN may regulate phosphatidylinositol 3-kinase-dependent insulin signaling pathways in 3T3-L1 adipocytes.


Assuntos
Adipócitos/metabolismo , Insulina/metabolismo , Proteínas Musculares , Monoéster Fosfórico Hidrolases/fisiologia , Proteínas Supressoras de Tumor , Células 3T3 , Adenoviridae/genética , Animais , Western Blotting , Linhagem Celular , DNA Complementar/metabolismo , Desoxiglucose/metabolismo , Técnicas de Transferência de Genes , Transportador de Glucose Tipo 4 , Proteínas de Fluorescência Verde , Humanos , Proteínas Luminescentes/metabolismo , Sistema de Sinalização das MAP Quinases , Camundongos , Microscopia de Fluorescência , Proteínas de Transporte de Monossacarídeos/metabolismo , PTEN Fosfo-Hidrolase , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação
4.
Cancer Res ; 59(10): 2318-23, 1999 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-10344736

RESUMO

Genetic alterations in the MMAC1 tumor suppressor gene (also referred to as PTEN or TEP1) occur in several types of human cancers including glioblastoma. Growth suppression induced by overexpression of MMAC1 in cells with mutant MMAC1 alleles is thought to be mediated by the inhibition of signaling through the phosphatidylinositol 3-kinase pathway. However, the exact biochemical mechanisms by which MMAC1 exerts its growth-inhibitory effects are still unknown. Here we report that recombinant adenovirus-mediated overexpression of MMAC1 in three different MMAC1-mutant glioblastoma cell lines blocked progression from G0/G1 to S phase of the cell cycle. Cell cycle arrest correlated with the recruitment of the cyclin-dependent kinase (CDK) inhibitor, p27Kip1, to cyclin E immunocomplexes, which resulted in a reduction in CDK2 kinase activities and a decrease in levels of endogenous phosphorylated retinoblastoma protein. CDK4 kinase activities were unaffected, as were the levels of the CDK inhibitor p21Cip1 present in cyclin E immunocomplexes. Therefore, overexpression of MMAC1 via adenovirus-mediated gene transfer suppresses tumor cell growth through cell cycle inhibitory mechanisms, and as such, represents a potential therapeutic approach to treating glioblastomas.


Assuntos
Quinases relacionadas a CDC2 e CDC28 , Proteínas de Ciclo Celular , Ciclina E/metabolismo , Quinases Ciclina-Dependentes/metabolismo , Glioblastoma/patologia , Proteínas Associadas aos Microtúbulos/metabolismo , Proteínas de Neoplasias/metabolismo , Proteínas de Neoplasias/fisiologia , Monoéster Fosfórico Hidrolases/fisiologia , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas , Fase S/fisiologia , Proteínas Supressoras de Tumor , Adenovírus Humanos/genética , Complexo Antígeno-Anticorpo/metabolismo , Ciclo Celular/fisiologia , Divisão Celular/efeitos dos fármacos , Ciclina E/imunologia , Quinase 2 Dependente de Ciclina , Quinase 4 Dependente de Ciclina , Inibidor de Quinase Dependente de Ciclina p21 , Inibidor de Quinase Dependente de Ciclina p27 , Ciclinas/análise , Vetores Genéticos/genética , Humanos , Substâncias Macromoleculares , PTEN Fosfo-Hidrolase , Monoéster Fosfórico Hidrolases/genética , Fosforilação , Processamento de Proteína Pós-Traducional , Proteínas Recombinantes de Fusão/fisiologia , Proteína do Retinoblastoma/metabolismo , Transdução de Sinais , Transfecção , Células Tumorais Cultivadas
5.
Hum Gene Ther ; 10(1): 77-84, 1999 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-10022532

RESUMO

A replication-deficient adenovirus encoding human interferon alpha2b, driven by the human cytomegalovirus (CMV) promoter, was constructed and characterized. This construct was used to infect human cells derived from different types of cancer. The production of protein and its secretion into the culture medium were tested by Western blotting and immunoassay. Inhibition of cell proliferation and antiviral activity, two of the most important biological activities of interferon, were observed with this construct. PC-3 cells, derived from human prostatic cancer, or Hep3B cells, derived from human hepatocellular carcinoma, were injected subcutaneously to generate and establish in vivo tumors in athymic nude mice. Intratumoral injection with the recombinant adenovirus expressing interferon alpha2b resulted in complete regression of tumor growth. Our results demonstrate that interferon gene delivery using recombinant adenoviral vectors may be a useful approach to treat a variety of cancers.


Assuntos
Terapia Genética/métodos , Interferon-alfa/genética , Adenoviridae/genética , Animais , Western Blotting , Divisão Celular , Citomegalovirus/genética , Feminino , Vetores Genéticos , Humanos , Imunoensaio , Interferon alfa-2 , Interferon-alfa/biossíntese , Camundongos , Camundongos Endogâmicos BALB C , Neoplasias Experimentais/tratamento farmacológico , Proteínas Recombinantes , Fatores de Tempo , Células Tumorais Cultivadas
6.
Oncogene ; 18(6): 1261-6, 1999 Feb 11.
Artigo em Inglês | MEDLINE | ID: mdl-10022807

RESUMO

MMAC1, also known as PTEN or TEP-1, was recently identified as a gene commonly mutated in a variety of human neoplasias. Sequence analysis revealed that MMAC1 harbored sequences similar to those found in several protein phosphatases. Subsequent studies demonstrated that MMAC1 possessed in vitro enzymatic activity similar to that exhibited by dual specificity phosphatases. To characterize the potential cellular functions of MMAC1, we expressed wild-type and several mutant variants of MMAC1 in the human glioma cell line, U373, that lacks endogenous expression. While expression of wild-type MMAC1 in these cells significantly reduced their growth rate and saturation density, expression of enzymatically inactive MMAC1 significantly enhanced growth in soft agar. Our observations indicate that while wild-type MMAC1 exhibits activities compatible with its proposed role as a tumor suppressor, cellular expression of MMAC1 containing mutations in the catalytic domain may yield protein products that enhance transformation characteristics.


Assuntos
Genes Supressores de Tumor , Glioma/genética , Monoéster Fosfórico Hidrolases/biossíntese , Proteínas Supressoras de Tumor , Domínio Catalítico/genética , Adesão Celular , Divisão Celular , Transformação Celular Neoplásica , Glioma/enzimologia , Humanos , Mutação , PTEN Fosfo-Hidrolase , Fenótipo , Monoéster Fosfórico Hidrolases/genética , Proteínas Recombinantes/biossíntese , Células Tumorais Cultivadas
7.
Genes Chromosomes Cancer ; 24(1): 42-7, 1999 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-9892107

RESUMO

Tumor cell-specific homozygous deletions coinciding at a particular genetic location may indicate the inactivation of a nearby tumor suppressor gene. Forty-six human cancer cell lines of prostate, pancreatic, lung, liver, and colon origin were screened for homozygous deletions of 139 expressed sequence tag (EST) and sequence-tagged site (STS) loci spanning the entire short arm of chromosome 8. Only one Southern blot-verified homozygous deletion was detected in this set of cell lines. The deletion, in pancreatic tumor cell line MIA-PaCa-2, encompassed two screening loci, D8S549 and D8S1992, and overlapped another previously described homozygous deletion of band 8p22 in a metastatic prostate cancer specimen. Both deletions entirely removed the candidate tumor suppressor gene N33. These data define a consensus homozygous deletion region in chromosome band 8p22.


Assuntos
Deleção Cromossômica , Cromossomos Humanos Par 8/genética , Homozigoto , Neoplasias/genética , Mapeamento Cromossômico/métodos , DNA de Neoplasias/análise , Marcadores Genéticos , Humanos , Masculino , Reação em Cadeia da Polimerase/métodos , Células Tumorais Cultivadas
8.
Cancer Res ; 58(23): 5285-90, 1998 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-9850049

RESUMO

The MMAC/PTEN tumor suppressor gene encodes for a phosphatase that recently has been shown to have phosphotidylinositol phosphatase activity, implicating its possible involvement in phosphatidylinositol 3'-kinase-mediated signaling. To investigate possible alterations in growth factor-mediated signal transduction, an adenovirus containing MMAC/PTEN, Ad-MMAC, previously shown to inhibit growth and tumorigenicity in glioma cells, was used to acutely express the transgene. Human glioma cells infected with Ad-MMAC but not with control adenoviruses exhibited an inhibition of phosphorylation of both activating residues of Akt, Ser-473, and Thr-308, along with Akt's serine/threonine kinase activity, without significantly altering Akt expression. The effects of functional MMAC/PTEN expression were relatively specific, because members of several other growth factor-mediated signaling pathways showed no altered responses. The presence of MMAC/PTEN also inhibited phosphorylation of BAD, although no evidence of apoptosis in the in situ treated cells was observed. However, U251 glioma cells infected with Ad-MMAC were induced to undergo anoikis at a significantly higher rate than U251 cels treated with control viruses or mock infected with media. These results demonstrate that the acute administration of MMAC/PTEN results in the inhibition of Akt-mediated signaling, growth inhibition, and anoikis, implying that loss of MMAC/PTEN increases cellular proliferation and significantly augments a cell's survival potential during cellular processes that are associated with malignancy.


Assuntos
Apoptose/fisiologia , Glioblastoma/metabolismo , Monoéster Fosfórico Hidrolases/biossíntese , Monoéster Fosfórico Hidrolases/fisiologia , Proteínas Serina-Treonina Quinases , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Supressoras de Tumor , Adenoviridae/genética , Adenoviridae/metabolismo , Ativação Enzimática , Receptores ErbB/biossíntese , Receptores ErbB/fisiologia , Expressão Gênica , Genes Supressores de Tumor , Glioblastoma/enzimologia , Glioblastoma/genética , Humanos , PTEN Fosfo-Hidrolase , Monoéster Fosfórico Hidrolases/genética , Fosforilação , Proteínas Proto-Oncogênicas c-akt , Transdução de Sinais/fisiologia , Transgenes , Células Tumorais Cultivadas
9.
Cancer Res ; 58(11): 2331-4, 1998 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-9622068

RESUMO

Mutated in multiple advanced cancers 1/phosphatase and tensin homologue (MMAC1/PTEN) is a novel tumor suppressor gene candidate located on chromosome 10 that is commonly mutated in human glioblastoma multiforme and several other cancer types. To evaluate the function of this gene as a tumor suppressor, we constructed a replication-defective adenovirus (MMCB) for efficient, transient transduction of MMAC1 into tumor cells. Infection of MMAC1-mutated U87MG glioblastoma cells with MMCB resulted in dose-dependent exogenous MMAC1 protein expression as detected by Western blotting of cell lysates. In vitro proliferation of U87MG cells was inhibited by MMCB in comparison to several control adenoviruses at equal viral doses, implying a specific effect of MMAC1 expression. Anchorage-independent growth in soft agar was also inhibited by MMCB compared to control adenovirus. Tumorigenicity in nude mice of transiently transduced mass cell cultures was then assessed. MMCB-infected U87MG cells were almost completely nontumorigenic compared to untreated and several control adenovirus-treated cells at equal viral doses. These data support an in vivo tumor suppression activity of MMAC1/PTEN and suggest that in vivo gene transfer with this recombinant adenoviral vector has a potential use in cancer gene therapy.


Assuntos
Técnicas de Transferência de Genes , Genes Supressores de Tumor , Terapia Genética , Glioblastoma/terapia , Monoéster Fosfórico Hidrolases , Proteínas Tirosina Fosfatases/genética , Proteínas Supressoras de Tumor , Adenoviridae/genética , Animais , Cromossomos Humanos Par 10 , Citometria de Fluxo , Mutação em Linhagem Germinativa , Glioblastoma/patologia , Humanos , Camundongos , Camundongos Nus , Transplante de Neoplasias , PTEN Fosfo-Hidrolase , Células Tumorais Cultivadas
10.
Oncogene ; 15(9): 1111-4, 1997 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-9285566

RESUMO

Allelic deletions of chromosome 18q are reported to be common in prostate and colorectal cancers, suggesting that one or more tumor suppressor genes on 18q are involved in the genesis of these neoplasms. The DPC4 gene, a recently identified candidate tumor suppressor in 18q21, was examined for evidence of inactivation in prostatic carcinomas, and results compared to those of a parallel analysis of colorectal carcinomas, for which DPC4 mutation has been reported in approximately 10% of cases. In this study, only three (10%) of 29 informative primary prostate cancers showed allelic loss of chromosome 18q21 markers, and no point mutations or deletions of DPC4 were detected in the complete set of 45 primary or metastatic cases. In contrast, five (56%) of nine primary colorectal tumors displayed allelic loss of 18q markers and in one of these a somatically acquired G-->T missense mutation was found in exon 1. Of twelve colorectal tumor cell lines, one showed a G-->C missense mutation in exon 8 and two had partial homozygous deletions that would likely abrogate gene function. These data suggest that DPC4 is rarely if ever mutated during prostatic oncogenesis, whereas inactivation of this gene may contribute to the genesis of a subset of colorectal carcinomas.


Assuntos
Neoplasias Colorretais/genética , Análise Mutacional de DNA , Proteínas de Ligação a DNA , Genes Supressores de Tumor , Neoplasias da Próstata/genética , Transativadores/genética , Alelos , Cromossomos Humanos Par 18 , Deleção de Genes , Homozigoto , Humanos , Masculino , Mutação Puntual , Proteína Smad4 , Células Tumorais Cultivadas
11.
Br J Urol ; 79 Suppl 1: 28-36, 1997 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-9088270

RESUMO

Genetic alterations, such as mutation, methylation and aneuploidy, are thought to underlie the multistep genesis and progression of many human cancers. However, the genetic events occurring in prostatic oncogenesis are still relatively poorly understood. This is especially so in early-stage tumours, in which mutations of known oncogenes or tumour-suppressor genes appear to be quite infrequent. Allelic losses of chromosome arms 7q, 8p, 10, 16q and 18q suggest the involvement of novel suppressor loci on these chromosomes; allelic losses of chromosome arm 8p are especially frequent and may be detected even in early-stage tumours. We have used a positional approach to seek novel genetic targets in prostate cancer, including allelic-loss mapping of chromosome 8p and physical mapping of chromosome band 8p22 around the MSR gene. A homozygous somatic deletion in one prostatic nodal metastasis was mapped in this region and spanned 730-970 kb. This region was then examined in detail for expressed sequences. One novel gene, called N33, was found to be silenced by a methylation mechanism in most colon cancer cell lines and some primary colorectal tumours. Characterization of additional chromosome 8p22 candidates is in progress.


Assuntos
Genes Supressores de Tumor/fisiologia , Mutação , Oncogenes/fisiologia , Neoplasias da Próstata/genética , Sequência de Aminoácidos , Southern Blotting , Mapeamento Cromossômico , Cromossomos Humanos Par 6 , Deleção de Genes , Regulação Neoplásica da Expressão Gênica , Marcadores Genéticos , Humanos , Masculino , Metilação , Dados de Sequência Molecular
12.
Semin Cancer Biol ; 8(1): 11-9, 1997 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-9299577

RESUMO

Inactivation of tumour suppressor gene function is a critical step in the development of human neoplasia. The Rb and CDKN2 tumour suppressor genes are inactivated in many tumour types, including the late stages of prostate cancer, and appear to function in the same suppressor pathway. p53, another major tumour suppressor is also mutated in a subset of advanced-stage prostate carcinomas. E-cadherin and other cell adhesion genes, which have been characterized as suppressors of the metastatic phenotype, are inactivated or downregulated during progression to advanced prostate cancer and have been associated with poor clinical outcome. The early genetic events involved a prostatic neoplasia are poorly understood, but loss of as yet undiscovered tumour suppressor genes may play a role in the initiation of this disease.


Assuntos
Genes Supressores de Tumor , Neoplasias da Próstata/genética , Clonagem Molecular , Humanos , Perda de Heterozigosidade , Masculino , Metástase Neoplásica
13.
Oncogene ; 15(23): 2855-66, 1997 Dec 04.
Artigo em Inglês | MEDLINE | ID: mdl-9419977

RESUMO

We have constructed a panel of substitution mutants which affect one or more of the putative cdk target sites of the RB protein. We have examined the activity of these mutants relative to wild-type RB by both a transcriptional repression assay and by measuring growth suppression in vitro. We find that some phosphorylation site mutants of pRB can repress E2 transcription more strongly than wild-type RB. These mutants are partially resistant to phosphorylation by cdks and can arrest tumor cells in G1 in vitro. Our results indicate a functional correlation between the ability to repress E2F-dependent transcription and the ability to suppress tumor cell growth in vitro. In addition, we describe two classes of RB mutants: N-terminal truncated p56RB and a novel mutant of RB containing multiple substitutions near its nuclear localization signal. Both classes of RB mutants have greater activity than the wild-type protein. Because RB is a key regulator of cell cycle progression, expression of a more potent, phosphorylation resistant RB may have utility in both RB(-/-) and RB(+/+) tumors as well as in hyperproliferative disorders.


Assuntos
Substituição de Aminoácidos/genética , Proteínas de Transporte , Proteínas de Ciclo Celular , Proteínas de Ligação a DNA , Inibidores do Crescimento/genética , Inibidores do Crescimento/fisiologia , Mutagênese Sítio-Dirigida , Proteína do Retinoblastoma/genética , Proteína do Retinoblastoma/fisiologia , Antineoplásicos/antagonistas & inibidores , Antineoplásicos/metabolismo , Sítios de Ligação/genética , Ciclina E/fisiologia , Quinases Ciclina-Dependentes/fisiologia , Fatores de Transcrição E2F , Fase G1/efeitos dos fármacos , Fase G1/genética , Inibidores do Crescimento/antagonistas & inibidores , Inibidores do Crescimento/metabolismo , Humanos , Osteossarcoma , Fosforilação , Proteína do Retinoblastoma/antagonistas & inibidores , Proteína do Retinoblastoma/metabolismo , Proteína 1 de Ligação ao Retinoblastoma , Deleção de Sequência , Fator de Transcrição DP1 , Fatores de Transcrição/fisiologia , Células Tumorais Cultivadas
14.
Genomics ; 35(1): 46-54, 1996 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-8661103

RESUMO

Numerous studies have implicated the short arm of chromosome 8 as the site of one or more tumor suppressor genes inactivated in carcinogenesis of the prostate, colon, lung, and liver. Previously, we identified a homozygous deletion on chromosome 8p22 in a metastatic prostate cancer. To map this homozygous deletion physically, long-range restriction mapping was performed using yeast artificial chromosomes (YACs) spanning approximately 2 Mb of chromosome band 8p22. Subcloned genomic DNA and cDNA probes isolated by hybrid capture from these YACs were mapped in relation to one another, reinforcing map integrity. Mapped single-copy probes from the region were then applied to DNA isolated from a metastatic prostate cancer containing a chromosome 8p22 homozygous deletion and indicated that its deletion spans 730-970 kb. Candidate genes PRLTS (PDGF-receptor beta-like tumor suppressor) and CTSB (cathepsin B) are located outside the region of homozygous deletion. Généthon marker D8S549 is located approximately at the center of this region of homozygous deletion. Two new microsatellite polymorphisms, D8S1991 and D8S1992, also located within the region of homozygous deletion on chromosome 8p22, are described. Physical mapping places cosmid CI8-2644 telomeric to MSR (macrophage scavenger receptor), the reverse of a previously published map, altering the interpretation of published deletion studies. This work should prove helpful in the identification of candidate tumor suppressor genes in this region.


Assuntos
Adenocarcinoma/secundário , Cromossomos Humanos Par 8/genética , Metástase Linfática/genética , Neoplasias da Próstata/genética , Deleção de Sequência , Adenocarcinoma/genética , Sequência de Bases , Cromossomos Artificiais de Levedura/genética , Cosmídeos/genética , Feminino , Humanos , Masculino , Repetições de Microssatélites , Pessoa de Meia-Idade , Dados de Sequência Molecular
15.
Genomics ; 35(1): 55-65, 1996 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-8661104

RESUMO

The structure and expression pattern of a human gene located within a homozygously deleted region of a metastatic prostate cancer have been characterized. Multiple cDNA fragments of this gene were isolated by hybrid capture with yeast artificial chromosome clones covering the deletion region. Eleven coding exons spanned 205-220 kb of the 730- to 970-kb deletion. The predicted amino acid sequence was 43% identical to that of an anonymous Caenorhabditis elegans gene and 20% identical to an accessory or regulatory subunit of the oligosaccharyltransferase enzyme complex in Saccharomyces cerevisiae. Hydrophobicity profiles of all three gene products were similar and showed four putative membrane-spanning domains in the molecules' C-terminal halves, suggesting a general conservation of function. The gene was expressed as an approximately 1.5-kb mRNA in most nonlymphoid human cells/tissues including prostate, lung, liver, and colon. Expression was detected in many epithelial tumor cell lines, but was undetectable by Northern blot or RT-PCR in 14 of 15 colorectal, 1 of 8 lung, and 1 of 4 liver cancer cell lines. Lack of expression in tumor cell lines was highly correlated with hypermethylation of a CpG island located at the gene's 5' end. These findings form a basis for further work on this candidate tumor suppressor gene.


Assuntos
Adenocarcinoma/secundário , Cromossomos Humanos Par 8/genética , Regulação Neoplásica da Expressão Gênica , Genes Supressores de Tumor , Metástase Linfática/genética , Neoplasias da Próstata/genética , Deleção de Sequência , Adenocarcinoma/genética , Sequência de Aminoácidos , Animais , Sequência de Bases , Caenorhabditis elegans/genética , Cromossomos Artificiais de Levedura/genética , Clonagem Molecular , Ilhas de CpG , DNA Complementar/genética , Humanos , Masculino , Metilação , Pessoa de Meia-Idade , Dados de Sequência Molecular , Proteínas de Neoplasias/biossíntese , Proteínas de Neoplasias/genética , Especificidade de Órgãos , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , RNA Neoplásico/biossíntese , RNA Neoplásico/genética , Saccharomyces cerevisiae/genética , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Especificidade da Espécie , Células Tumorais Cultivadas
16.
Semin Oncol ; 23(1): 66-77, 1996 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-8607033

RESUMO

Tumor suppressor genes such as p53 contribute to the oncogenic process via loss-of-function mechanisms such as genetic mutation or complex formation with other cellular or viral proteins. p53 is mutated in approximately 50% of human tumors and has an important role in the genesis or progression of both colorectal and hepatocellular cancers. Colorectal cancer is leading cause of cancer mortality in the United States, whereas hepatocellular cancer is the leading worldwide cause of cancer death; the liver is a primary site of morbidity in both diseases. Because systemic tumor suppressor gene therapy is currently not feasible, we have chosen to develop a regional form of such therapy directed at primary or metastatic liver neoplasms. Gene replacement therapy with p53 is a promising new strategy to treat advanced human cancers.


Assuntos
Carcinoma Hepatocelular/terapia , Neoplasias Colorretais/terapia , Genes p53 , Terapia Genética/métodos , Neoplasias Hepáticas/secundário , Neoplasias Hepáticas/terapia , Adenoviridae/genética , Neoplasias Colorretais/patologia , Técnicas de Transferência de Genes , Vetores Genéticos , Humanos , Retroviridae/genética , Células Tumorais Cultivadas
18.
Genomics ; 24(2): 317-23, 1994 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-7698754

RESUMO

Polymorphic alleles at loci such as LPL (lipoprotein lipase) and MSR (macrophage scavenger receptor) in chromosome band 8p22 are frequently lost during the genesis of several types of human cancer, including colorectal, non-small cell lung, hepatocellular, and prostatic carcinomas. A physical map of 31 published or novel probes and sequence-tagged sites in this genetic region was constructed using a radiation hybrid panel and the CEPH (Centre d'Etude du Polymorphisme Humain) yeast artificial chromosome (YAC) library. Thirty-six overlapping YACs defined a physical order for the following polymorphic markers: tel-D8S26-D8S511-D8S549-MSR-D8S254-D8S233- D8S261-D8S21-LPL-D8S258-cen. These maps unify small consensus regions of allelic loss on chromosome 8p defined by restriction fragment length polymorphisms with more informative PCR-based polymorphisms and widely available YAC mapping resources.


Assuntos
Deleção Cromossômica , Cromossomos Artificiais de Levedura , Cromossomos Humanos Par 8 , Neoplasias/genética , Alelos , Sequência de Bases , Carcinoma Hepatocelular/genética , Carcinoma Pulmonar de Células não Pequenas/genética , Neoplasias Colorretais/genética , Primers do DNA , Genes Supressores de Tumor , Humanos , Células Híbridas/efeitos da radiação , Lipase Lipoproteica/genética , Neoplasias Hepáticas/genética , Neoplasias Pulmonares/genética , Masculino , Dados de Sequência Molecular , Neoplasias da Próstata/genética , Receptores Imunológicos/genética , Receptores Depuradores , Sitios de Sequências Rotuladas
19.
Genes Chromosomes Cancer ; 11(3): 153-62, 1994 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-7530484

RESUMO

Due to problems with primary tumor cell culture, conventional cytogenetics has yielded little insightful information on chromosomal alterations in prostate cancer. The primary aim of this study was to define the ability of comparative genomic hybridization (CGH) to detect and map genetic deletions in prostate tumors. A secondary aim was to apply multiple assays to individual tumors as a means of deciphering the mechanisms of genetic alterations in prostate cancer. CGH results were compared with allelic imbalance measurements at 29 distinct loci on chromosome 8 in 18 specimens (17 malignant and 1 benign). CGH detected no changes in cases where all informative PCR/RFLP loci were retained and detected all p arm deletions consisting of at least two loci. We estimate that in this study, the smallest deletions detected by CGH were approximately 20-30 cM. Physical mapping of subchromosomal arm deletions by CGH correlated well with allelic imbalance mapping by PCR/RFLP: The data agreed at 88% of loci on 8p and 92% of loci on 8q. Fluorescence in situ hybridization (FISH) with multiple centromere probes and DNA content flow cytometry (FCM) also was performed on selected specimens. FISH revealed two cases of chromosome 8 aneusomy. In these two cases and three others, CGH showed simultaneous p arm deletion and q arm gain, suggesting isochromosome 8q formation. Together, these data suggested that, simple chromosomal aberrations were responsible for allelic losses on 8p and allelic gains on 8q in a significant number of prostate tumors. We also used CGH to examine relative DNA sequence copy number throughout the genome. Changes frequently associated with 8p loss include gains of 8q and losses of 13q, 16p, 16q, 17p, 17q, 20q, and Y. Cases with 8p loss exhibited five times the number of alterations as did cases without 8p loss.


Assuntos
Alelos , Cromossomos Humanos Par 8 , Neoplasias da Próstata/genética , Aberrações Cromossômicas , Mapeamento Cromossômico , DNA de Neoplasias/análise , Genoma , Humanos , Hibridização in Situ Fluorescente , Masculino , Hibridização de Ácido Nucleico
20.
Genes Chromosomes Cancer ; 10(3): 151-9, 1994 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-7522037

RESUMO

A previous study of 18 primary or metastatic prostate cancers showed loss of genetic markers on chromosome 8; 10, or 16 in more than 50% of cases [Bergerheim USR et al. (1991) Genes Chromosom Cancer 3:215-220]. The small size and infiltrative nature of primary prostatic tumors have hindered efforts to assess allelic losses by traditional restriction fragment length polymorphism (RFLP)/Southern blotting methods. To improve the sensitivity and specificity of this analysis in early prostate cancer, we have amplified polymorphic microsatellite repeats by polymerase chain reaction (PCR), and have quantitated allelic imbalances with phosphor imaging technology. In this study, 63 primary prostate tumors and matched benign tissues obtained by radical prostatectomy were examined at 28 genetic loci on chromosome 8, all but five of which were located on the short arm. Twenty-nine (46%) of the 63 cases showed loss of at least one locus. Multiple adjacent loci, usually including the LPL and MSR genes in 8p22, were lost in 28 cases. In 10 of these, losses were observed at all informative loci on the p arm. In another 15 tumors, losses were restricted to subregions of the p arm by loci retained either distally toward the p terminus or proximally at the 8p12-8p21 border, or both. In three tumors, two discrete regions of loss were observed within 8p, separated by several retained loci. Allelic loss of 8p loci was associated with higher tumor grade. These data are complementary to previous reports of allelic deletions in colorectal, hepatocellular, and non-small cell lung cancers and suggest the existence of one or more pleotropic tumor suppressor genes on 8p.


Assuntos
Alelos , Deleção Cromossômica , Cromossomos Humanos Par 8 , Neoplasias da Próstata/genética , Sequência de Bases , Southern Blotting , Mapeamento Cromossômico , Primers do DNA , DNA de Neoplasias , DNA Satélite , Humanos , Masculino , Dados de Sequência Molecular , Reação em Cadeia da Polimerase , Polimorfismo de Fragmento de Restrição , Neoplasias da Próstata/fisiopatologia , Sequências Repetitivas de Ácido Nucleico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...