Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 58
Filtrar
1.
Biochem Biophys Res Commun ; 466(1): 89-94, 2015 Oct 09.
Artigo em Inglês | MEDLINE | ID: mdl-26321665

RESUMO

Chemokine receptor CXCR4 is involved in tumor growth, angiogenesis and metastasis. Its function is regulated in many ways and one of them is alternative splicing. We identified two novel coding splice variants (CXCR4-3 and CXCR4-4) of CXCR4 in Ewing sarcoma (EWS) cell lines by whole transcriptome sequencing and validated these with reverse transcriptase- PCR and Sanger sequencing. The novel splice variants were expressed at RNA level in Ewing sarcoma samples and in other tumor cell lines and placenta, but not in lung. Due to inclusion of an additional exon the new isoforms have a 70 and 33 amino acid elongation of the N-terminal end of CXCR4. For validation at protein and functional level, the identified isoforms and normal CXCR4 were cloned into an EYFP tagged vector and ectopically expressed in HEK293T cell line and EWS cell line A673. Of the novel isoforms CXCR4-3 showed cell membrane localization and a functional response after addition of CXCR4 ligand CXCL12a. CXCR4-4 showed strong cytoplasmic accumulation and no response to ligand treatment. The role of the newly discovered isoforms in CXCR4 signaling is likely to be limited. Our data stresses the importance of functional validation of newly identified isoforms.


Assuntos
Neoplasias Ósseas/genética , Receptores CXCR4/genética , Sarcoma de Ewing/genética , Transcriptoma , Neoplasias Ósseas/metabolismo , Neoplasias Ósseas/patologia , Cálcio/metabolismo , Linhagem Celular Tumoral , Perfilação da Expressão Gênica , Células HEK293 , Humanos , Isoformas de Proteínas/análise , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Splicing de RNA , Receptores CXCR4/análise , Receptores CXCR4/metabolismo , Sarcoma de Ewing/metabolismo , Sarcoma de Ewing/patologia
2.
Nat Genet ; 29(1): 92-5, 2001 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-11528400

RESUMO

The p53 protein can inhibit cell cycling or induce apoptosis, and is thus a critical regulator of tumorigenesis. This protein is negatively regulated by a physical interaction with MDM2, an E3 ubiquitin ligase. This interaction is critical for cell viability; loss of Mdm2 causes cell death in vitro and in vivo in a p53-dependent manner. The recently discovered MDM2-related protein MDM4 (also known as MDMX) has some of the same properties as MDM2. MDM4 binds and inhibits p53 transcriptional activity in vitro. Unlike MDM2, however, MDM4 does not cause nuclear export or degradation of p53 (refs. 9,10). To study MDM4 function in vivo, we deleted Mdm4 in mice. Mdm4-null mice died at 7.5-8.5 dpc, owing to loss of cell proliferation and not induction of apoptosis. To assess the importance of p53 in the death of Mdm4-/- embryos, we crossed in the Trp53-null allele. The loss of Trp53 completely rescued the Mdm4-/- embryonic lethality. Thus, MDM2 and MDM4 are nonoverlapping critical regulators of p53 in vivo. These data define a new pathway of p53 regulation and raise the possibility that increased MDM4 levels and the resulting inactivation of p53 contribute to the development of human tumors.


Assuntos
Embrião de Mamíferos/metabolismo , Genes Letais , Genes p53 , Proteínas Nucleares , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Animais , Sequência de Bases , Western Blotting , Primers do DNA , Marcação In Situ das Extremidades Cortadas , Camundongos , Camundongos Knockout , Reação em Cadeia da Polimerase , Testes de Precipitina , Proteínas Proto-Oncogênicas c-mdm2 , Proteína Supressora de Tumor p53/genética
3.
NPJ Breast Cancer ; 7(1): 140, 2021 Oct 27.
Artigo em Inglês | MEDLINE | ID: mdl-34707097

RESUMO

Reactivation of dormant cancer cells can lead to cancer relapse, metastasis, and patient death. Dormancy is a nonproliferative state and is linked to late relapse and death. No targeted therapy is currently available to eliminate dormant cells, highlighting the need for a deeper understanding and reliable models. Here, we thoroughly characterize the dormant D2.OR and ZR-75-1, and proliferative D2A1 breast cancer cell line models in vivo and/or in vitro, and assess if there is overlap between a dormant and a senescent phenotype. We show that D2.OR but not D2A1 cells become dormant in the liver of an immunocompetent model. In vitro, we show that D2.OR and ZR-75-1 cells in response to a 3D environment or serum-free conditions are growth-arrested in G1, of which a subpopulation resides in a 4NG1 state. The dormancy state is reversible and not associated with a senescence phenotype. This will aid future research on breast cancer dormancy.

4.
J Cell Biol ; 151(3): 653-62, 2000 Oct 30.
Artigo em Inglês | MEDLINE | ID: mdl-11062265

RESUMO

Cajal bodies (CBs) are subnuclear organelles that contain components of a number of distinct pathways in RNA transcription and RNA processing. CBs have been linked to other subnuclear organelles such as nucleoli, but the reason for the presence of nucleolar proteins such as fibrillarin in CBs remains uncertain. Here, we use full-length fibrillarin and truncated fibrillarin mutants fused to green fluorescent protein (GFP) to demonstrate that specific structural domains of fibrillarin are required for correct intranuclear localization of fibrillarin to nucleoli and CBs. The second spacer domain and carboxy terminal alpha-helix domain in particular appear to target fibrillarin, respectively, to the nucleolar transcription centers and CBs. The presence of the RNP domain seems to be a prerequisite for correct targeting of fibrillarin. Time-lapse confocal microscopy of human cells that stably express fibrillarin-GFP shows that CBs fuse and split, albeit at low frequencies. Recovered fluorescence of fibrillarin-GFP in nucleoli and CBs after photobleaching indicates that it is highly mobile in both organelles (estimated diffusion constant approximately 0.02 microm(2) s(-1)), and has a significantly larger mobile fraction in CBs than in nucleoli.


Assuntos
Nucléolo Celular/metabolismo , Proteínas Cromossômicas não Histona/metabolismo , Corpos Enovelados/metabolismo , Mutação/genética , Transporte Ativo do Núcleo Celular , Nucléolo Celular/química , Proteínas Cromossômicas não Histona/química , Proteínas Cromossômicas não Histona/genética , Corpos Enovelados/química , Difusão , Imunofluorescência , Humanos , Cinética , Movimento (Física) , Sinais Direcionadores de Proteínas/genética , Sinais Direcionadores de Proteínas/fisiologia , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , RNA Mensageiro/análise , RNA Mensageiro/genética , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/metabolismo , Deleção de Sequência/genética , Transcrição Gênica , Células Tumorais Cultivadas
5.
Cell Death Differ ; 14(12): 2047-57, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17823617

RESUMO

MDMX has been shown to modulate p53 in dividing cells after DNA damage. In this study, we investigated the role of MDMX in primary cultures of neurons undergoing cell death. We found that DNA damage, but also membrane-initiated apoptotic stresses (glutamate receptor; Amyloid beta precursor) or survival factor deprivation downregulated MDMX protein levels. Forced downregulation of murine double minute X (MDMX) by shRNA induced apoptosis suggesting that MDMX is required for survival in neurons. Protease inhibitors prevented the loss of MDMX after neurotoxic treatments, indicating a regulation of protein stability. Some, but not all, neurotoxic stresses induced phosphorylation of MDMX at serine 367, further supporting regulation at the protein level. Interestingly, we found that depending on the stimulus either p53 or E2F1 was induced, but overexpression of MDMX inhibited the transcriptional activity of both proapoptotic factors, and maintained neuronal viability upon neurotoxic stresses. Taken together, our data show that MDMX is an antiapoptotic factor in neurons, whose degradation is induced by various stresses and allows activation of p53 and E2F-1 during neuronal apoptosis.


Assuntos
Apoptose/efeitos dos fármacos , Neurônios/citologia , Neurônios/efeitos dos fármacos , Neurotoxinas/toxicidade , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Precursor de Proteína beta-Amiloide/toxicidade , Animais , Inibidores de Caspase , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Cerebelo/citologia , Cerebelo/efeitos dos fármacos , Fator de Transcrição E2F1/metabolismo , Inibidores Enzimáticos/farmacologia , Inativação Gênica/efeitos dos fármacos , Camundongos , Inibidores de Proteassoma , RNA Interferente Pequeno , Proteína Supressora de Tumor p53/metabolismo
6.
Oncogenesis ; 7(3): 33, 2018 Mar 29.
Artigo em Inglês | MEDLINE | ID: mdl-29593251

RESUMO

Uveal melanoma (UM) is the most frequent ocular cancer in adults, accounting for ~5% of the total melanoma incidence. Although the primary tumor is well treatable, patients frequently develop metastases for which no curative therapy exists. Highly activated protein kinase C (PKC) is a common feature of UM and has shown potential as therapeutic intervention for UM patients. Unfortunately, PKC inhibition as single treatment appears to have only limited clinical benefit. Combining PKC inhibition with activation of p53, which is rarely mutated in UM, by MDM2 inhibitors has shown promising results in vitro and in vivo. However, clinical studies have shown strong adverse effects of MDM2 inhibition. Therefore, we investigated alternative approaches to achieve similar anticancer effects, but with potentially less adverse effects. We studied the potential of targeting MDMX, an essential p53 inhibitor during embryonal development but less universally expressed in adult tissues compared with MDM2. Therefore, targeting MDMX is predicted to have less adverse effects in patients. Depletion of MDMX, like the pharmacological activation of p53, inhibits the survival of UM cells, which is enhanced in combination with PKC inhibition. Also pan-PKC inhibitors elicit adverse effects in patients. As the PKC family consists of 10 different isoforms, it could be hypothesized that targeting a single PKC isoform would have less adverse effects compared with a pan-PKC inhibitor. Here we show that specifically depleting PKCδ inhibits UM cell growth, which can be further enhanced by p53 reactivation. In conclusion, our data show that the synergistic effects of p53 activation by MDM2 inhibition and broad spectrum PKC inhibition on survival of UM cells can also largely be achieved by the presumably less toxic combination of depletion of MDMX and targeting a specific PKC isoform, PKCδ.

7.
Cell Death Differ ; 13(12): 2089-98, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16729027

RESUMO

p53 is a potent inhibitor of cell growth and an inducer of apoptosis. During embryonic development, Mdm2 and Mdm4 inhibit the growth suppressive activities of p53. However, whether tight surveillance of p53 activity is required in quiescent cells is unknown. To test this, conditional inactivation of mdm2 and mdm4 was carried out in smooth muscle cells (SMCs). Upon SMC-specific inactivation of mdm2, and not of mdm4, mice rapidly became ill and died. Necropsy showed small intestinal dilation, and histological analyses indicated a severe reduction in the number of intestinal SMCs. Increased p53 levels and activity were detected in the remaining SMCs, and the phenotype was completely rescued on a p53-null background. Interestingly, intestinal SMCs are caspase-3-negative and therefore did not undergo caspase-3-dependent apoptotic cell death. Together, Mdm2, but not Mdm4, prevents accumulation of active p53 in quiescent SMCs and thereby the induction of p53-mediated caspase-3-independent cell death.


Assuntos
Apoptose/fisiologia , Caspase 3/metabolismo , Miócitos de Músculo Liso/metabolismo , Proteínas Proto-Oncogênicas c-mdm2/fisiologia , Proteínas Proto-Oncogênicas/fisiologia , Proteína Supressora de Tumor p53/metabolismo , Ubiquitina-Proteína Ligases/fisiologia , Animais , Apoptose/genética , Caspase 3/genética , Diferenciação Celular/fisiologia , Regulação da Expressão Gênica/fisiologia , Intestino Delgado/metabolismo , Intestino Delgado/patologia , Camundongos , Camundongos Transgênicos , Miócitos de Músculo Liso/patologia , Proteína Supressora de Tumor p53/genética
8.
Mol Cell Biol ; 19(5): 3885-94, 1999 May.
Artigo em Inglês | MEDLINE | ID: mdl-10207112

RESUMO

Multiple adenovirus (Ad) early proteins have been shown to inhibit transcription activation by p53 and thereby to alter its normal biological functioning. Since these Ad proteins affect the activity of p53 via different mechanisms, we examined whether this inhibition is target gene specific. In addition, we analyzed whether the same Ad early proteins have a comparable effect on transcription activation by the recently identified p53 homologue p73. Our results show that the large E1B proteins very efficiently inhibited the activity of p53 on the Bax, p21(Waf1), cyclin G, and MDM2 reporter constructs but had no effect on the activation of the same reporter constructs by p73, with the exception of some inhibition of the Bax promoter by Ad12 E1B. The repressive effect of the E1A proteins on p53 activity is less than that seen with the large E1B proteins, but the E1A proteins inhibit the activity of both p53 and p73. We could not detect significant inhibition of p53 functions by E4orf6, but a clear repression of the transcription activation by p73 by this Ad early protein was observed. In addition, we found that stable expression of the Ad5 E1A and that of the E1B protein both caused increased p73 protein expression. The large E1B and the E4orf6 proteins together do not target the p73 protein for rapid degradation after adenoviral infection, as has previously been found for the p53 protein, probably because the large E1B protein does not interact with p73. Our results suggest that the p53 and p73 proteins are both inactivated after Ad infection and transformation but via distinct mechanisms.


Assuntos
Proteínas E1A de Adenovirus/metabolismo , Proteínas E1B de Adenovirus/metabolismo , Proteínas E4 de Adenovirus/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas Nucleares/genética , Proteína Supressora de Tumor p53/genética , Inibidor de Quinase Dependente de Ciclina p21 , Ciclinas/genética , Proteínas de Ligação a DNA/metabolismo , Imunofluorescência , Regulação da Expressão Gênica , Genes Reporter/genética , Genes Supressores de Tumor , Humanos , Proteínas Nucleares/metabolismo , Proteínas Repressoras/metabolismo , Ativação Transcricional/genética , Transformação Genética , Células Tumorais Cultivadas , Proteína Tumoral p73 , Proteína Supressora de Tumor p53/metabolismo , Proteínas Supressoras de Tumor
9.
Mol Cell Biol ; 16(5): 2101-9, 1996 May.
Artigo em Inglês | MEDLINE | ID: mdl-8628276

RESUMO

p53 stimulates the transcription of a number of genes, such as MDM2, Waf1, and GADD45. We and others have shown previously that this activity of p53 can be inhibited by adenovirus type 2 or 12 large E1B proteins. Here we show that the adenovirus E1A proteins also can repress the stimulation of transcription by p53, both in transient transfections and in stably transfected cell lines. The inhibition by E1A occurs without a significant effect on the DNA-binding capacity of p53. Furthermore, the activity of a fusion protein containing the N-terminal part of p53 linked to the GAL4 DNA-binding domain can be suppressed by E1A. This indicates that E1A affects the transcription activation domain of p53, although tryptic phosphopeptide mapping revealed that the level of phosphorylation of this domain does not change significantly in E1A-expressing cell lines. Gel filtration studies, however, showed p53 to be present in complexes of increased molecular weight as a result of E1A expression. Apparently, E1A can cause increased homo- or hetero-oligomerization of p53, which might result in the inactivation of the transcription activation domain of p53. Additionally, we found that transfectants stably expressing E1A have lost the ability to arrest in G1 after DNA damage, indicating that E1A can abolish the normal biological function of p53.


Assuntos
Proteínas E1A de Adenovirus/metabolismo , Adenovírus Humanos/genética , Proteínas de Saccharomyces cerevisiae , Fatores de Transcrição , Ativação Transcricional , Proteína Supressora de Tumor p53/metabolismo , Adenovírus Humanos/metabolismo , Sequência de Bases , Ciclo Celular , Linhagem Celular , Núcleo Celular/metabolismo , Proteínas de Ligação a DNA , Proteínas Fúngicas/biossíntese , Proteínas Fúngicas/metabolismo , Humanos , Luciferases/biossíntese , Dados de Sequência Molecular , Sondas de Oligonucleotídeos , Fosforilação , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/metabolismo , Transcrição Gênica , Transfecção , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/antagonistas & inibidores , Proteína Supressora de Tumor p53/biossíntese
10.
Mol Cell Biol ; 14(6): 4126-34, 1994 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-8196650

RESUMO

The human ERCC3 gene, which corrects specifically the nucleotide excision repair defect in human xeroderma pigmentosum group B and cross-complements the repair deficiency in rodent UV-sensitive mutants of group 3, encodes a presumed DNA helicase that is identical to the p89 subunit of the general transcription factor TFIIH/BTF2. To examine the significance of the postulated functional domains in ERCC3, we have introduced mutations in the ERCC3 cDNA by means of site-specific mutagenesis and have determined the repair capacity of each mutant to complement the UV-sensitive phenotype of rodent group 3 cells. A conservative substitution of arginine for the invariant lysine residue in the ATPase motif (helicase domain I), six deletion mutations in the other helicase domains, and a deletion in the potential helix-turn-helix DNA-binding motif fail to complement the ERCC3 excision repair defect of rodent group 3 mutants, which implies that the helicase domains as well as the potential DNA-binding motif are required for the repair function of ERCC3. Analysis of carboxy-terminal deletions suggests that the carboxy-terminal exon may comprise a distinct determinant for the DNA repair function. In addition, we show that a functional epitope-tagged version of ERCC3 accumulates in the nucleus. Deletion of the putative nuclear location signal impairs neither the nuclear location nor the repair function, indicating that other sequences may (also) be involved in translocation of ERCC3 to the nucleus.


Assuntos
Análise Mutacional de DNA , Reparo do DNA , Proteínas de Ligação a DNA/biossíntese , Proteínas de Ligação a DNA/genética , Proteínas de Drosophila , Transcrição Gênica , Xeroderma Pigmentoso/genética , Sequência de Aminoácidos , Animais , Sequência de Bases , Células CHO , Sobrevivência Celular/efeitos da radiação , Cricetinae , Relação Dose-Resposta à Radiação , Drosophila melanogaster/genética , Sequências Hélice-Alça-Hélice/genética , Humanos , Camundongos , Dados de Sequência Molecular , Oligodesoxirribonucleotídeos , Mutação Puntual , Mapeamento por Restrição , Roedores , Deleção de Sequência , Homologia de Sequência de Aminoácidos , Transfecção , Raios Ultravioleta
11.
Cancer Res ; 55(3): 486-9, 1995 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-7834613

RESUMO

Previously, we have shown that the chicken anemia virus-derived VP3 ("apoptin") protein induces apoptosis in chicken mononuclear cells. Here, we report that apoptin also induces apoptosis in human osteosarcoma cells, regardless of whether they expressed wild-type, mutant p53, or no p53 at all. Moreover, the nuclear location of apoptin appears to be important for its optimal induction of apoptosis. The fact that apoptin can induce p53-independent apoptosis in human tumor cells makes apoptin a potential candidate for treatment of frequently occurring types of cancer cells that do not contain functional p53.


Assuntos
Apoptose/fisiologia , Proteínas do Capsídeo , Capsídeo/fisiologia , Proteína Supressora de Tumor p53/fisiologia , Neoplasias Ósseas , Capsídeo/biossíntese , Linhagem Celular , Vírus da Anemia da Galinha , Imunofluorescência , Humanos , Cinética , Osteossarcoma , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/metabolismo , Fatores de Tempo , Transfecção , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/biossíntese
12.
Cancer Res ; 61(5): 1839-42, 2001 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-11280734

RESUMO

It has been shown that the Hdmx gene is amplified in a subset of gliomas, but thus far, no data are available on HDMX protein expression in tumor cells. We now report that a significant fraction of tumor cell lines expresses increased HDMX levels compared with normal cells; in general, HDMX expression in these tumor cell lines correlates with the presence of wild-type p53. Analysis of tumor material showed that high HDMX expression is not a result of cell line establishment. Interestingly, several cell lines express alternative, shorter HDMX proteins. These results suggest that deregulated expression of HDMX plays a role in carcinogenesis as an alternative way to inactivate p53.


Assuntos
Proteínas de Neoplasias/biossíntese , Proteínas Nucleares , Proteínas Proto-Oncogênicas/biossíntese , Proteína Supressora de Tumor p53/genética , Feminino , Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , Melanoma/genética , Melanoma/metabolismo , Proteínas de Neoplasias/genética , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/metabolismo , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas c-mdm2 , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/biossíntese
13.
Cancer Res ; 57(7): 1353-63, 1997 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-9102224

RESUMO

The Wilms' tumor 1 gene (WT1) encodes a transcription factor of the zinc-finger family. As a result of alternative RNA splicing, the gene can be expressed as four polypeptides that differ in the presence or absence of a stretch of 17 amino acids just NH2 terminal of the four zinc fingers and a stretch of three amino acids (+/-KTS) between zinc fingers 3 and 4. In this study, cDNA constructs encoding the four human Wilms' tumor 1 splice variants were transiently transfected into the p53-negative Hep3B and the p53-positive HepG2 hepatoma cell lines. Morphological assessment of the WT1-expressing cells showed that the WT1(-KTS) splice variants induced apoptosis in both cell lines, whereas the WT1(+KTS) isoforms did not. The induction of apoptosis by the WT1(-KTS) isoforms appears to be p53 independent in the hepatoma cell lines. Furthermore, it was found that the WT1(-KTS)-induced apoptosis could not be suppressed by coexpression of either the Mr 21,000 E1B, the Bcl-2, or the BAG-1 protein. Coexpression of either the epidermal growth factor receptor or the insulin receptor, however, partially rescued the cells from apoptosis.


Assuntos
Apoptose , Carcinoma Hepatocelular/genética , Proteínas de Ligação a DNA/genética , Receptores ErbB/metabolismo , Genes p53 , Neoplasias Hepáticas/genética , Receptor de Insulina/metabolismo , Fatores de Transcrição/genética , Western Blotting , Proteínas de Ligação a DNA/metabolismo , Técnica Indireta de Fluorescência para Anticorpo , Regulação Neoplásica da Expressão Gênica , Humanos , Fatores de Transcrição/metabolismo , Transfecção , Células Tumorais Cultivadas , Proteínas WT1
14.
Oncogene ; 20(33): 4576-80, 2001 Jul 27.
Artigo em Inglês | MEDLINE | ID: mdl-11494153

RESUMO

The p53 protein is involved in cell cycle arrest and apoptosis. To ensure that cells under non-stressed conditions are able to grow, p53 sets up a negative feedback loop by inducing Mdm2. Mdm2 is able to both inhibit the transcriptional regulation by p53 and to degrade it, thus maintaining p53 inactive until it is required. The Mdm2 related protein, Hdmx, has also been shown to inhibit the transcriptional activation of p53 but is unable to degrade it. A few years ago, the p53 family member, p63 was identified. Like p53, p63 is able to induce p53 target genes and it was shown to be able to cause cell cycle arrest and apoptosis. In this study we report that, despite the similarities between p53 and p63, neither Hdmx nor Mdm2 are able to interact with p63, to repress p63-induced transcription or to affect its half-life.


Assuntos
Proteínas de Membrana , Proteínas Nucleares , Fosfoproteínas/fisiologia , Proteínas Proto-Oncogênicas/fisiologia , Transativadores/fisiologia , Ativação Transcricional , Proteína Supressora de Tumor p53/antagonistas & inibidores , Apoptose , Neoplasias Ósseas/patologia , Ciclo Celular , Cisteína Endopeptidases/metabolismo , Proteínas de Ligação a DNA , Retroalimentação , Genes Reporter , Genes Supressores de Tumor , Meia-Vida , Humanos , Complexos Multienzimáticos/metabolismo , Osteossarcoma/patologia , Complexo de Endopeptidases do Proteassoma , Isoformas de Proteínas/antagonistas & inibidores , Isoformas de Proteínas/fisiologia , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas c-mdm2 , Proteínas Recombinantes de Fusão/metabolismo , Fatores de Transcrição , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/fisiologia , Proteínas Supressoras de Tumor
15.
Oncogene ; 11(1): 49-57, 1995 Jul 06.
Artigo em Inglês | MEDLINE | ID: mdl-7624131

RESUMO

Loss of function of the tumor-suppressor protein p53 is, in general, either caused by mutation, inducing a conformational change, or by binding to inactivating cellular (e.g. MDM2) or viral (e.g. SV40 large T) proteins. In adenovirus type 12 (Ad12)-transformed cells, p53 is stabilized without detectable binding to the Ad12 E1B/54 kDa protein and still present in a wild-type conformation but contains a mutant-like activity in cellular transformation. In this study we examined whether the changed characteristics of p53 in Ad12-transformed cells are correlated with changes in phosphorylation or complex formation of the protein. By making tryptic phosphopeptide maps we found a significant increase in the phosphorylation of the N-terminus of p53. Furthermore, expression of E1A was found to be essential for the altered phosphorylation, while expression of only Ad12 E1B/54 kDa is sufficient to increase the protein half-life. Additionally, we observed p53 to be present in increased molecular weight complexes in Ad12-transformed cells. We conclude that both the phosphorylation and oligomerization of p53 is changed as a result of Ad12 transformation.


Assuntos
Adenoviridae/fisiologia , Proteína Supressora de Tumor p53/metabolismo , Proteínas E1B de Adenovirus/genética , Biopolímeros , Transformação Celular Viral , Células Cultivadas , Humanos , Fosfatos/metabolismo , Fosforilação
16.
Oncogene ; 9(3): 981-3, 1994 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-8108144

RESUMO

The tumor suppressor gene p53 is thought to be a key factor in the onset of G1 cell-cycle arrest following DNA damage. However, here we describe cells derived from a patient with Bloom's syndrome, lacking any detectable p53 protein, that still shows a functional G1 cell-cycle checkpoint after irradiation with UV-C. Comparison with cells from other Bloom's patients showed that the absence of p53 protein is not a specific characteristic of Bloom's syndrome.


Assuntos
Síndrome de Bloom/patologia , Fase G1 , Proteína Supressora de Tumor p53/metabolismo , Síndrome de Bloom/metabolismo , Linhagem Celular , Fase G1/efeitos da radiação , Humanos , Proteína Supressora de Tumor p53/deficiência , Raios Ultravioleta
17.
Oncogene ; 16(3): 349-57, 1998 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-9467960

RESUMO

It has recently been shown that an adenovirus mutant lacking expression of the large E1B protein (deltaE1B) selectively replicates in p53 deficient cells. However, apart from the large E1B protein the adenovirus early region encodes the E1A and E4orf6 proteins which also have been reported to affect p53 expression as well as its functioning. After infection with wild-type adenovirus we observed a dramatic decrease in wild-type p53 expression while no down-regulation of p53 could be detected after infection with the deltaE1B virus. The different effects of the wild-type and deltaE1B adenovirus on p53 expression were not only found in cells expressing wild-type p53 but were also observed when tumor cells expressing highly stabilized mutant p53 were infected with these two viruses. Infection with different adenovirus mutants indicated the importance of a direct interaction between p53 and the large E1B protein for reduced p53 expression after infection. Moreover, coexpression of the E4orf6 protein was found to be required for this phenomenon, while expression of E1A is dispensable. In addition, we provide evidence that p53 is actively degraded in wild-type adenovirus-infected cells but not in deltaE1B-infected cells.


Assuntos
Adenoviridae/fisiologia , Proteínas E1B de Adenovirus/metabolismo , Proteínas E4 de Adenovirus/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Proteínas E1A de Adenovirus/metabolismo , Proteínas E1B de Adenovirus/genética , Proteínas E4 de Adenovirus/genética , Transformação Celular Viral , Expressão Gênica , Humanos , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/biossíntese , Proteína Supressora de Tumor p53/genética
18.
Oncogene ; 19(6): 791-800, 2000 Feb 10.
Artigo em Inglês | MEDLINE | ID: mdl-10698497

RESUMO

The Wilms' tumor 1 gene (WT1) encodes a transcription factor of the zinc-finger family and is homozygously mutated or deleted in a subset of Wilms' tumors. Through alternative mRNA splicing, the gene is expressed as four main polypeptides that differ by a stretch of 17 amino acids just N-terminal of the four zinc-fingers and three amino acids between zinc fingers 3 and 4. We have previously shown that expression of the WT1(-/-) isoform, lacking both inserts, increases the tumor growth rate of the adenovirus-transformed baby rat kidney (AdBRK) cell line 7C3H2, whereas expression of the WT1(-/+) isoform, lacking the 17aa insert, strongly suppresses the tumorigenic phenotype. In the present study we show that expression of these splice variants does not affect the tumorigenic potential of the similar AdBRK cell line, 7C1T1. In contrast to the 7C3H2 cell line, this AdBRK cell line expresses high endogenous levels of EGR-1 (early growth response-1) protein, a transcription factor structurally related to WT1. Ectopic expression of EGR-1 in the 7C3H2 AdBRK cells significantly increases their in vivo growth rate and nullifies the tumor suppressor activity of the WT1(-/+) protein. Furthermore, we find that EGR-1 levels are elevated in some Wilms' tumors. These data are the first to show that EGR-1 overexpression causes enhanced tumor growth and that WT1 and EGR-1 exert antagonizing effects on growth regulation in baby rat kidney cells, which might reflect the situation in some Wilms' tumors.


Assuntos
Proteínas de Ligação a DNA/fisiologia , Genes do Tumor de Wilms , Proteínas Imediatamente Precoces , Proteínas de Neoplasias/fisiologia , Fatores de Transcrição/fisiologia , Adenoviridae/fisiologia , Animais , Divisão Celular , Linhagem Celular Transformada , Transformação Celular Viral , Proteínas de Ligação a DNA/genética , Proteína 1 de Resposta de Crescimento Precoce , Feminino , Deleção de Genes , Rim , Neoplasias Renais/genética , Neoplasias Renais/patologia , Camundongos , Camundongos Nus , Proteínas de Neoplasias/genética , Transplante de Neoplasias , Splicing de RNA , Ratos , Fatores de Transcrição/genética , Proteínas WT1 , Tumor de Wilms/genética , Tumor de Wilms/patologia , Dedos de Zinco/genética
19.
Oncogene ; 12(3): 537-46, 1996 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-8637710

RESUMO

The Wilms' Tumor 1 gene (WT1) encodes a transcription factor of the zinc-finger family. As a result of alternative RNA splicing, the gene can be expressed as four polypeptides which differ in the presence or absence of two stretches of amino acids: one of 17 residues (17aa) just N-terminal of the four zinc-fingers and of three residues (K-T-S) between zinc finger 3 and 4. In this study, four human cDNA constructs encoding the Wilms' tumor 1 splice variants were stably transfected into adenovirus-transformed baby rat kidney (Ad-BRK) cells. The in vivo produced WT1 proteins that lacked the KTS residues were found to bind efficiently to both the Egr-1 consensus sequence and the recently described WTE DNA sequence, as determined by electrophoretic mobility shift assays. Our studies show distinct effects of the different WT1 isoforms. Expression of the WT1 (-/+) protein, lacking the 17aa insert, strongly suppressed the tumorigenic phenotype of the Ad-BRK cells. Intriguingly, expression of the WT1 (-/-) protein, lacking both inserts, increased the tumor growth rate. In contrast to the growth in vivo, the growth rate of the transfectants in tissue culture is not influenced by any of the WT1 isoforms. However, the suppression of tumorigenicity appears to be correlated with a reduced ability of the cells to grow in serum-free medium.


Assuntos
Processamento Alternativo , Transformação Celular Neoplásica , Proteínas de Ligação a DNA/biossíntese , Genes do Tumor de Wilms , Variação Genética , Neoplasias Renais/patologia , Fatores de Transcrição/biossíntese , Tumor de Wilms/patologia , Animais , Sequência de Bases , Western Blotting , Linhagem Celular , Sequência Consenso , DNA Complementar , Humanos , Rim , Neoplasias Renais/genética , Camundongos , Camundongos Nus , Dados de Sequência Molecular , Oligodesoxirribonucleotídeos , Ratos , Proteínas Recombinantes/biossíntese , Transplante Heterólogo , Proteínas WT1 , Tumor de Wilms/genética , Dedos de Zinco
20.
Oncogene ; 18(1): 189-99, 1999 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-9926934

RESUMO

Mdm2 and MDMX are two structurally related p53-binding proteins which show the highest level of sequence similarity in the N-terminal p53-binding domains. Apart from its ability to inhibit p53 mediated transcription, a feature it shares with mdm2, very little is known about the physiological functions of MDMX. It is clearly distinct from mdm2 since its expression appears not to be regulated by p53 and it cannot compensate for lack of mdm2 in early development. We present data on the structural similarity between the p53 binding pockets of mdm2 and MDMX using p53- and phage-selected peptides. From the results we conclude that our recently devised innovative approach to reverse the mdm2-mediated inhibition of p53's transactivation function in vivo would probably target MDMX as well. Strategies for selectively targeting mdm2 and MDMX are suggested and a possible mechanism for regulating the p53-mdm2/MDMX interactions by protein phosphorylation is discussed.


Assuntos
Proteínas Nucleares , Proteínas Proto-Oncogênicas/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Sequência de Aminoácidos , Expressão Gênica , Humanos , Dados de Sequência Molecular , Peptídeos/metabolismo , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas c-mdm2 , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Solubilidade
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa