Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
1.
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.

2.
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.

3.
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
4.
Oncogene ; 34(22): 2943-8, 2015 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-25088193

RESUMO

Mdm4, a protein related to the ubiquitin-ligase Mdm2, is an essential inhibitor of tumor suppressor protein p53. In both human and mouse cells, the Mdm4 gene encodes two major transcripts: one encodes the full-length oncoprotein (designated below as Mdm4-FL), whereas the other, resulting from a variant splicing that skips exon 6, encodes the shorter isoform Mdm4-S. Importantly, increased Mdm4-S mRNA levels were observed in several human cancers, and correlated with poor survival. However, the role of Mdm4-S in cancer progression remains controversial, because the Mdm4-S protein appeared to be a potent p53 inhibitor when overexpressed, but the splice variant also leads to a decrease in Mdm4-FL expression. To unambiguously determine the physiological impact of the Mdm4-S splice variant, we generated a mouse model with a targeted deletion of the Mdm4 exon 6, thereby creating an obligatory exon skipping. The mutant allele (Mdm4(ΔE6)) prevented the expression of Mdm4-FL, but also led to increased Mdm4-S mRNA levels. Mice homozygous for this allele died during embryonic development, but were rescued by a concomitant p53 deficiency. Furthermore in a hypomorphic p53(ΔP/ΔP) context, the Mdm4(ΔE6) allele led to p53 activation and delayed the growth of oncogene-induced tumors. We next determined the effect of Mdm4(+/ΔE6) heterozygosity in a hypermorphic p53(+/Δ31) genetic background, recently shown to be extremely sensitive to Mdm4 activity. Mdm4(+/ΔE6) p53(+/Δ31) pups were born, but suffered from aplastic anemia and died before weaning, again indicating an increased p53 activity. Our results demonstrate that the main effect of a skipping of Mdm4 exon 6 is not the synthesis of the Mdm4-S protein, but rather a decrease in Mdm4-FL expression. These and other data suggest that increased Mdm4-S mRNA levels might correlate with more aggressive cancers without encoding significant amounts of a potential oncoprotein. Hypotheses that may account for this apparent paradox are discussed.


Assuntos
Proteínas Proto-Oncogênicas/genética , Sítios de Splice de RNA/genética , Proteína Supressora de Tumor p53/metabolismo , Ubiquitina-Proteína Ligases/genética , Animais , Células Cultivadas , Embrião de Mamíferos , Éxons/genética , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Nus , Camundongos Transgênicos , Invasividade Neoplásica , Neoplasias/genética , Neoplasias/patologia , Gravidez , Isoformas de Proteínas/genética
5.
Cell Death Differ ; 20(3): 490-502, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23197296

RESUMO

Unrepaired DNA double-strand breaks (DSBs) cause genetic instability that leads to malignant transformation or cell death. Cells respond to DSBs with the ordered recruitment of signaling and repair proteins to the sites of DNA lesions. Coordinated protein SUMOylation and ubiquitylation have crucial roles in regulating the dynamic assembly of protein complexes at these sites. However, how SUMOylation influences protein ubiquitylation at DSBs is poorly understood. We show herein that Rnf4, an E3 ubiquitin ligase that targets SUMO-modified proteins, accumulates in DSB repair foci and is required for both homologous recombination (HR) and non-homologous end joining repair. To establish a link between Rnf4 and the DNA damage response (DDR) in vivo, we generated an Rnf4 allelic series in mice. We show that Rnf4-deficiency causes persistent ionizing radiation-induced DNA damage and signaling, and that Rnf4-deficient cells and mice exhibit increased sensitivity to genotoxic stress. Mechanistically, we show that Rnf4 targets SUMOylated MDC1 and SUMOylated BRCA1, and is required for the loading of Rad51, an enzyme required for HR repair, onto sites of DNA damage. Similarly to inactivating mutations in other key regulators of HR repair, Rnf4 deficiency leads to age-dependent impairment in spermatogenesis. These findings identify Rnf4 as a critical component of the DDR in vivo and support the possibility that Rnf4 controls protein localization at DNA damage sites by integrating SUMOylation and ubiquitylation events.


Assuntos
Reparo do DNA , Proteínas Nucleares/metabolismo , Fatores de Transcrição/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Alelos , Animais , Proteína BRCA1/metabolismo , Proteínas de Ciclo Celular , Linhagem Celular , Quebras de DNA de Cadeia Dupla , Genótipo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Camundongos , Camundongos Transgênicos , Proteínas Nucleares/genética , Rad51 Recombinase/metabolismo , Radiação Ionizante , Sumoilação , Fatores de Transcrição/genética , Ubiquitina-Proteína Ligases , Ubiquitinação
6.
Cell Death Differ ; 19(6): 980-9, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22158418

RESUMO

Reactivation of the p53 tumor-suppressor protein by small molecules like Nutlin-3 and RITA (reactivation of p53 and induction of tumor cell apoptosis) is a promising strategy for cancer therapy. The molecular mechanisms involved in the responses to RITA remain enigmatic. Several groups reported the induction of a p53-dependent DNA damage response. Furthermore, the existence of a p53-dependent S-phase checkpoint has been suggested, involving the checkpoint kinase Chk1. We have recently shown synergistic induction of apoptosis by RITA in combination with Nutlin-3, and we observed concomitant Chk2 phosphorylation. Therefore, we investigated whether Chk2 contributes to the cellular responses to RITA. Strikingly, the induction of apoptosis seemed entirely Chk2 dependent. Transcriptional activity of p53 in response to RITA required the presence of Chk2. A partial rescue of apoptosis observed in Noxa knockdown cells emphasized the relevance of p53 transcriptional activity for RITA-induced apoptosis. In addition, we observed an early p53- and Chk2-dependent block of DNA replication upon RITA treatment. Replicating cells seemed more prone to entering RITA-induced apoptosis. Furthermore, the RITA-induced DNA damage response, which was not a secondary effect of apoptosis induction, was strongly attenuated in cells lacking p53 or Chk2. In conclusion, we identified Chk2 as an essential mediator of the cellular responses to RITA.


Assuntos
Apoptose/efeitos dos fármacos , Furanos/farmacologia , Proteínas Serina-Treonina Quinases/metabolismo , Linhagem Celular Tumoral , Quinase do Ponto de Checagem 2 , Pontos de Checagem da Fase G2 do Ciclo Celular , Células HCT116 , Humanos , Imidazóis/farmacologia , Fosforilação , Piperazinas/farmacologia , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/genética , Proteínas Proto-Oncogênicas c-bcl-2/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Proteína Supressora de Tumor p53/antagonistas & inibidores , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo
7.
Oncogene ; 31(9): 1105-16, 2012 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-21765463

RESUMO

The prognosis of patients with uveal melanoma is poor. Because of the limited efficacy of current treatments, new therapeutic strategies need to be developed. Because p53 mutations are uncommon in uveal melanoma, reactivation of p53 may be used to achieve tumor regression. We investigated the use of combination therapies for intraocular melanoma, based on the p53 activators Nutlin-3 and reactivation of p53 and induction of tumor cell apoptosis (RITA) and the topoisomerase I inhibitor Topotecan. Nutlin-3 treatment induced p53-dependent growth inhibition in human uveal melanoma cell lines. The sensitivity to Nutlin-3 of the investigated cell lines did not correlate with basal Hdm2 or Hdmx levels. Nutlin-3 synergized with RITA and Topotecan to induce apoptosis in uveal melanoma cell lines and short-term cultures. Drug synergy correlated with enhanced induction of p53-Ser46 phosphorylation, which was attenuated by ATM inhibition. Nutlin-3 and Topotecan also significantly delayed tumor growth in vivo in a murine B16F10 model for ocular melanoma. Combination treatment appeared to inhibit tumor growth slightly more efficient than either drug alone. Nutlin-3, RITA and Topotecan lead to comparable p53 activation and growth inhibition under normoxia and hypoxia. Treatment with Nutlin-3 or RITA had no effect on HIF-1α induction by hypoxia, whereas the combination of these two drugs did inhibit hypoxia-induced HIF-1α. Also Topotecan, alone or in combination with Nutlin-3, reduced HIF-1α protein levels, suggesting that a certain level of DNA damage response is required for p53-mediated downregulation of HIF-1α. In conclusion, combination treatments based on small-molecule-induced p53 activation may have clinical potential for uveal melanoma.


Assuntos
Antineoplásicos/uso terapêutico , Melanoma/tratamento farmacológico , Proteína Supressora de Tumor p53/agonistas , Neoplasias Uveais/tratamento farmacológico , Animais , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Apoptose/genética , Proteínas Mutadas de Ataxia Telangiectasia , Proteínas de Ciclo Celular/antagonistas & inibidores , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Proteínas de Ligação a DNA/antagonistas & inibidores , Sinergismo Farmacológico , Furanos/farmacologia , Furanos/uso terapêutico , Humanos , Hipóxia , Imidazóis/farmacologia , Imidazóis/uso terapêutico , Melanoma/genética , Melanoma Experimental/tratamento farmacológico , Melanoma Experimental/genética , Camundongos , Fosforilação/efeitos dos fármacos , Piperazinas/farmacologia , Piperazinas/uso terapêutico , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Topotecan/farmacologia , Topotecan/uso terapêutico , Proteína Supressora de Tumor p53/genética , Proteínas Supressoras de Tumor/antagonistas & inibidores , Neoplasias Uveais/genética
8.
Cell Death Differ ; 18(11): 1736-45, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21546907

RESUMO

Inactivation of the p53 tumour suppressor, either by mutation or by overexpression of its inhibitors Hdm2 and HdmX is the most frequent event in cancer. Reactivation of p53 by targeting Hdm2 and HdmX is therefore a promising strategy for therapy. However, Hdm2 inhibitors do not prevent inhibition of p53 by HdmX, which impedes p53-mediated apoptosis. Here, we show that p53 reactivation by the small molecule RITA leads to efficient HdmX degradation in tumour cell lines of different origin and in xenograft tumours in vivo. Notably, HdmX degradation occurs selectively in cancer cells, but not in non-transformed cells. We identified the inhibition of the wild-type p53-induced phosphatase 1 (Wip1) as the major mechanism important for full engagement of p53 activity accomplished by restoration of the ataxia telangiectasia mutated (ATM) kinase-signalling cascade, which leads to HdmX degradation. In contrast to previously reported transactivation of Wip1 by p53, we observed p53-dependent repression of Wip1 expression, which disrupts the negative feedback loop conferred by Wip1. Our study reveals that the depletion of both HdmX and Wip1 potentiates cell death due to sustained activation of p53. Thus, RITA is an example of a p53-reactivating drug that not only blocks Hdm2, but also inhibits two important negative regulators of p53 - HdmX and Wip1, leading to efficient elimination of tumour cells.


Assuntos
Apoptose/efeitos dos fármacos , Proteínas de Ciclo Celular/metabolismo , Proteínas de Ligação a DNA/metabolismo , Furanos/farmacologia , Fosfoproteínas Fosfatases/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Proteínas Mutadas de Ataxia Telangiectasia , Linhagem Celular Tumoral , Regulação para Baixo , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Proteínas Nucleares/metabolismo , Fosfoproteínas Fosfatases/genética , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteína Fosfatase 2C , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Transdução de Sinais , Proteína Supressora de Tumor p53/antagonistas & inibidores
9.
Oncogene ; 29(16): 2415-26, 2010 Apr 22.
Artigo em Inglês | MEDLINE | ID: mdl-20140020

RESUMO

The p53 tumor suppressor protein is frequently mutated in human tumors. It is thought that the p53 pathway is indirectly impaired in the remaining tumors, for example by overexpression of its important regulators Mdm2 and Mdm4, making them attractive targets for the development of anti-cancer agents. Recent studies have suggested that Mdm4 levels determine the sensitivity of tumor cells for anti-cancer therapy. To investigate this possibility, we studied the drug sensitivity of several breast cancer cell lines containing wild-type p53, but expressing different Mdm4 levels. We show that endogenous Mdm4 levels can affect the sensitivity of breast cancer cells to anti-cancer agents, but in a cell line-dependent manner and depending on an intact apoptotic response. Furthermore, treatment with the non-genotoxic agent Nutlin-3 sensitizes cells for doxorubicin, showing that activation of p53 by targeting its regulators is an efficient strategy to decrease cell viability of breast cancer cells. These results confirm a function of Mdm4 in determining the efficacy of chemotherapeutic agents to induce apoptosis of cancer cells in a p53-dependent manner, although additional undetermined factors also influence the drug response. Targeting Mdm4 to sensitize tumor cells for chemotherapeutic drugs might be a strategy to effectively treat tumors harboring wild-type p53.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Proteínas Nucleares/fisiologia , Proteínas Proto-Oncogênicas/fisiologia , Apoptose/efeitos dos fármacos , Neoplasias da Mama/patologia , Proteínas de Ciclo Celular , Linhagem Celular Tumoral , Doxorrubicina/farmacologia , Resistencia a Medicamentos Antineoplásicos , Feminino , Humanos , Imidazóis/farmacologia , Proteínas Nucleares/análise , Piperazinas/farmacologia , Proteínas Proto-Oncogênicas/análise , Proteína Supressora de Tumor p53/fisiologia
10.
PLoS One ; 3(7): e2819, 2008 Jul 30.
Artigo em Inglês | MEDLINE | ID: mdl-18665269

RESUMO

BACKGROUND: The Forkhead box O (FOXO) class of transcription factors are involved in the regulation of several cellular responses including cell cycle progression and apoptosis. Furthermore, in model organisms FOXOs act as tumor suppressors and affect aging. Previously, we noted that FOXOs and p53 are remarkably similar within their spectrum of regulatory proteins. For example, the de-ubiquitinating enzyme USP7 removes ubiquitin from both FOXO and p53. However, Skp2 has been identified as E3 ligase for FOXO1, whereas Mdm2 is the prime E3 ligase for p53. PRINCIPAL FINDINGS/METHODOLOGY: Here we provide evidence that Mdm2 acts as an E3 ligase for FOXO as well. In vitro incubation of Mdm2 and FOXO results in ATP-dependent (multi)mono-ubiquitination of FOXO similar to p53. Furthermore, in vivo co-expression of Mdm2 and FOXO induces FOXO mono-ubiquitination and consistent with this result, siRNA-mediated depletion of Mdm2 inhibits mono-ubiquitination of FOXO induced by hydrogen peroxide. Regulation of FOXO ubiquitination by Mdm2 is likely to be direct since Mdm2 and FOXO co-immunoprecipitate. In addition, Mdm2-mediated ubiquitination regulates FOXO transcriptional activity. CONCLUSIONS/SIGNIFICANCE: These data identify Mdm2 as a novel E3 ligase for FOXOs and extend the analogous mode of regulation between FOXO and p53.


Assuntos
Fatores de Transcrição Forkhead/metabolismo , Regulação da Expressão Gênica , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Fatores de Transcrição/metabolismo , Células 3T3 , Trifosfato de Adenosina/metabolismo , Animais , Apoptose , Proteínas de Ciclo Celular , Linhagem Celular , Linhagem Celular Tumoral , Humanos , Camundongos , Modelos Biológicos , Estresse Oxidativo
11.
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
12.
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
13.
Gene ; 273(2): 141-61, 2001 Aug 08.
Artigo em Inglês | MEDLINE | ID: mdl-11595161

RESUMO

The Wilms' tumor 1 gene (WT1) has been identified as a tumor suppressor gene involved in the etiology of Wilms' tumor. Approximately 10% of all Wilms' tumors carry mutations in the WT1 gene. Alterations in the WT1 gene have also been observed in other tumor types, such as leukemia, mesothelioma and desmoplastic small round cell tumor. Dependent on the tumor type, WT1 proteins might either function as tumor suppressor proteins or as survival factors. Mutations in the WT1 gene can also result in congenital abnormalities as observed in Denys-Drash and Frasier syndrome patients. Mouse models have proven the critical importance of WT1 expression for the development of several organs, including the kidneys, the gonads and the spleen. The WT1 proteins seem to perform two main functions. They regulate the transcription of a variety of target genes and may be involved in post-transcriptional processing of RNA. The WT1 gene encodes at least 24 protein forms. These isoforms have partially distinct biological functions and effects, which in many cases are also specific for the model system in which WT1 is studied. This review discusses the molecular mechanisms by which the various WT1 isoforms exert their functions in normal development and how alterations in WT1 may lead to developmental abnormalities and tumor growth.


Assuntos
Proteínas de Ligação a DNA/genética , Fatores de Transcrição/genética , Animais , Proteínas de Ligação a DNA/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , Regulação Neoplásica da Expressão Gênica , Humanos , Fatores de Transcrição/fisiologia , Proteínas WT1
14.
EMBO Rep ; 2(11): 1029-34, 2001 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11606419

RESUMO

The p53 protein maintains genomic integrity through its ability to induce cell cycle arrest or apoptosis in response to various forms of stress. Substantial regulation of p53 activity occurs at the level of protein stability, largely determined by the activity of the Mdm2 protein. Mdm2 targets both p53 and itself for ubiquitylation and subsequent proteasomal degradation by acting as an ubiquitin ligase, a function that needs an intact Mdm2 RING finger. For efficient degradation of p53 nuclear export appears to be required. The Mdmx protein, structurally homologous to Mdm2, does not target p53 for degradation, but even stabilizes both p53 and Mdm2, an activity most likely mediated by heterodimerization of the RING fingers of Mdm2 and Mdmx. Here we show that Mdmx expression leads to accumulation of ubiquitylated, nuclear p53 but does not significantly affect the Mdm2-mediated ubiquitylation of p53. In contrast, Mdmx stabilizes Mdm2 by inhibiting its self-ubiquitylation.


Assuntos
Proteínas Nucleares , Proteínas Proto-Oncogênicas/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Transporte Ativo do Núcleo Celular , Animais , Apoptose , Ciclo Celular , Linhagem Celular , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Dimerização , Humanos , Ligases/metabolismo , Camundongos , Camundongos Knockout , Microscopia de Fluorescência , Mutação , Plasmídeos/metabolismo , Ligação Proteica , Estrutura Terciária de Proteína , Proteínas Proto-Oncogênicas c-mdm2 , Transfecção , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligases
15.
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
16.
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
17.
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
18.
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
19.
J Biol Chem ; 275(36): 28039-44, 2000 Sep 08.
Artigo em Inglês | MEDLINE | ID: mdl-10827196

RESUMO

The Mdm2 protein is a key regulator of p53 activity and stability. Upon binding, Mdm2 inhibits the transcription regulatory activity of p53 and promotes its rapid degradation. In this study we investigated the effect of the human Mdm2 homologue Hdmx on p53 stability. We found that Hdmx does not target p53 for degradation, although, like Mdm2, it inhibits p53-mediated transcription activation. On the contrary, Hdmx was found to counteract the degradation of p53 by Mdm2, and to stabilize both p53 and Mdm2. The RING finger of Hdmx was found to be necessary and sufficient for this stabilization, and it probably involves hetero-oligomerization with the RING finger of Mdm2, which may lead to inhibition of Mdm2's ubiquitin ligase activity. However, Hdmx does not relieve the inhibition by Mdm2 of transcription activation by p53, probably due to the formation of a trimeric complex consisting of Hdmx, Mdm2, and p53. We propose a model in which Hdmx secures a pool of largely inactive p53, which, upon the induction of stress, can be quickly activated.


Assuntos
Proteínas Proto-Oncogênicas/química , Proteínas Proto-Oncogênicas/metabolismo , Proteína Supressora de Tumor p53/química , Proteína Supressora de Tumor p53/metabolismo , Humanos , Proteínas Nucleares/metabolismo , Proteínas Proto-Oncogênicas c-mdm2 , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Transcrição Gênica , Transfecção , Células Tumorais Cultivadas , Dedos de Zinco
20.
J Biol Chem ; 275(14): 10202-11, 2000 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-10744705

RESUMO

The WT1 gene, which is heterozygously mutated or deleted in congenital anomaly syndromes and homozygously mutated in about 15% of all Wilms tumors, encodes tissue-specific developmental regulators. Through alternative mRNA splicing, four main WT1 protein isoforms are synthesized. All isoforms can bind to DNA via their zinc fingers, albeit with different affinities and specificities, and thereby modulate the transcriptional activity of their target genes. Several proteins bind to and alter the transcription regulatory properties of the WT1 proteins, including the product of the tumor suppressor gene p53. Interaction between WT1 and p53 was shown to modulate their ability to regulate the transcription of their respective target genes. Here, we report that all four isoforms of WT1 bind to p73, a recently cloned homologue of p53. p73 binds to the zinc finger region of WT1 and thereby inhibits DNA binding and transcription activation by WT1. Similarly, WT1 inhibits p73-induced transcription activation in reporter assays and counteracts p73-induced expression of endogenous Mdm2. This, taken together with our finding that WT1 also interacts with p63/KET, another p53 homologue, suggests that association between WT1 and the members of the p53 family of proteins may be an important determinant of their functions in cell growth and differentiation.


Assuntos
Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/metabolismo , Proteínas Nucleares/química , Proteínas Nucleares/metabolismo , Fatores de Transcrição/química , Fatores de Transcrição/metabolismo , Transcrição Gênica , Processamento Alternativo , Proteínas de Ligação a DNA/genética , Genes Reporter , Genes Supressores de Tumor , Genes do Tumor de Wilms , Glutationa Transferase/genética , Humanos , Luciferases/genética , Proteínas Nucleares/genética , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/metabolismo , Fatores de Transcrição/genética , Transfecção , Células Tumorais Cultivadas , Proteína Tumoral p73 , Proteínas Supressoras de Tumor , Proteínas WT1
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA