Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Hum Cell ; 36(3): 1204-1210, 2023 May.
Artigo em Inglês | MEDLINE | ID: mdl-36737540

RESUMO

Lung cancer remains the most chronic form of cancer and the leading cause of cancer mortality in the world. Despite significant improvements in the treatment of lung cancer, the current therapeutic interventions are only partially effective, necessitating the continued search for better, novel alternative treatments. Angiogenesis and cancer stem cells play a central role in the initiation and propagation of cancers. Tumor angiogenesis is triggered by an angiogenic switch when pro-angiogenic factors exceed anti-angiogenic components. Although many anti-angiogenic agents are used in cancer treatment, there are therapeutic limitations with significant side effects. In recent years, cannabinoids have been investigated extensively for their potential anti-neoplastic effects. Our previous findings showed that cannabidiol (CBD) could impede tumor growth in mouse models of melanoma and glioblastoma. Importantly, CBD has been suggested to possess anti-angiogenic activity. In this study, we tested, for the first time, inhalant CBD in the treatment of heterotopic lung cancer and whether such potential effects could reduce cancer stem cell numbers and inhibit tumor angiogenesis. We implanted NCI H1437 human lung cancer cells in nude mice and treated the mice with inhalant CBD or placebo. The outcomes were measured by tumor size and imaging, as well as by immunohistochemistry and flow cytometric analysis for CD44, VEGF, and P-selectin. Our findings showed that CBD decreased tumor growth rate and suppressed expression of CD44 and the angiogenic factors VEGF and P-selectin. These results suggest, for the first time, that inhalant CBD can impede lung cancer growth by suppressing CD44 and angiogenesis.


Assuntos
Canabidiol , Neoplasias Pulmonares , Humanos , Animais , Camundongos , Canabidiol/farmacologia , Canabidiol/uso terapêutico , Selectina-P , Fator A de Crescimento do Endotélio Vascular , Camundongos Nus , Neoplasias Pulmonares/patologia , Neovascularização Patológica/tratamento farmacológico , Neovascularização Patológica/patologia
2.
Nat Commun ; 12(1): 704, 2021 01 29.
Artigo em Inglês | MEDLINE | ID: mdl-33514736

RESUMO

p53 mutations with single amino acid changes in cancer often lead to dominant oncogenic changes. Here, we have developed a mouse model of gain-of-function (GOF) p53-driven lung cancer utilizing conditionally active LSL p53-R172H and LSL K-Ras-G12D knock-in alleles that can be activated by Cre in lung club cells. Mutation of the p53 transactivation domain (TAD) (p53-L25Q/W26S/R172H) eliminating significant transactivation activity resulted in loss of tumorigenicity, demonstrating that transactivation mediated by or dependent on TAD is required for oncogenicity by GOF p53. GOF p53 TAD mutations significantly reduce phosphorylation of nearby p53 serine 20 (S20), which is a target for PLK3 phosphorylation. Knocking out PLK3 attenuated S20 phosphorylation along with transactivation and oncogenicity by GOF p53, indicating that GOF p53 exploits PLK3 to trigger its transactivation capability and exert oncogenic functions. Our data show a mechanistic involvement of PLK3 in mutant p53 pathway of oncogenesis.


Assuntos
Carcinogênese/genética , Neoplasias Pulmonares/genética , Proteínas Serina-Treonina Quinases/metabolismo , Proteína Supressora de Tumor p53/genética , Animais , Carcinogênese/patologia , Linhagem Celular Tumoral , Modelos Animais de Doenças , Feminino , Mutação com Ganho de Função , Técnicas de Introdução de Genes , Técnicas de Inativação de Genes , Humanos , Pulmão/patologia , Neoplasias Pulmonares/patologia , Camundongos , Camundongos Transgênicos , Fosforilação/genética , Domínios Proteicos/genética , Proteínas Serina-Treonina Quinases/genética , Serina/metabolismo , Esferoides Celulares , Ativação Transcricional , Proteína Supressora de Tumor p53/metabolismo , Proteínas Supressoras de Tumor
3.
Mol Oncol ; 11(6): 696-711, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28423230

RESUMO

Gain-of-function (GOF) mutants of p53 upregulate genes implicated in cell proliferation and oncogenesis. Here, we report that GOF p53 induces tumorigenicity through simultaneous activation of key oncogenic pathways including those controlling putative tumor-initiating cell functions. We determined that in cells expressing p53-R273H, GOF p53 simultaneously upregulates genes from multiple signaling pathways by recognizing promoters containing distinct transcription factor (TF) binding sites. Our analytical data support a model in which GOF p53 complexes with two TFs on the promoter-a mediator protein, Med17, and a histone acetyl transferase, activating histone acetylation-and enhances gene expression to signal cell proliferation and oncogenesis. Thus, therapeutic inhibition of one GOF p53-induced pathway would be insufficient to prevent tumor growth as the oncoprotein activates a multitude of parallel pathways. This discovery suggests enormous selection advantage for cancer cells with GOF p53 to induce oncogenic growth, highlighting the problems of cancer therapy.


Assuntos
Carcinogênese/genética , Mutação com Ganho de Função , Regulação Neoplásica da Expressão Gênica , Neoplasias Pulmonares/genética , Proteína Supressora de Tumor p53/genética , Análise de Variância , Animais , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/genética , Histona Acetiltransferases/genética , Humanos , Neoplasias Pulmonares/patologia , Complexo Mediador/genética , Camundongos , Camundongos Nus , Invasividade Neoplásica , Ativação Transcricional/genética , Proteína Supressora de Tumor p53/metabolismo , Regulação para Cima , Ensaios Antitumorais Modelo de Xenoenxerto
4.
Subcell Biochem ; 85: 71-90, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25201189

RESUMO

p53 is a tumor suppressor protein whose key function is to maintain the integrity of the cell. Mutations in p53 have been found in up to 50 % of all human cancers and cause an increase in oncogenic phenotypes such as proliferation and tumorigenicity. Both wild-type and mutant p53 have been shown to transactivate their target genes, either through directly binding to DNA, or indirectly through protein-protein interactions. This review discusses possible mechanisms behind both wild-type and mutant p53-mediated transactivation and touches on the concept of addiction to mutant p53 of cancer cells and how that may be used for future therapies.


Assuntos
Genes p53 , Mutação , Transcrição Gênica/genética , Animais , Humanos , Camundongos , Modelos Animais , Neoplasias/genética , Oncogenes
5.
Subcell Biochem ; 85: 215-34, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25201197

RESUMO

Frequent overexpression of MDM2 in human cancers suggests that the protein confers a survival advantage to cancer cells. However, overexpression of MDM2 in normal cells seems to restrict cell proliferation. This review discusses the cell growth regulatory functions of MDM2 in normal and genetically defective cells to assess how cancer cells evade the growth-restricting consequence of MDM2 overexpression. Similar to oncoproteins that induce a DNA damage response and oncogene induced senescence in non-transformed cells, MDM2 induces G1-arrest and intra-S phase checkpoint responses that control untimely DNA replication in the face of genetic challenges.


Assuntos
Proliferação de Células , Neoplasias/metabolismo , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Transdução de Sinais , Animais , Ciclo Celular , Ciclinas/metabolismo , Replicação do DNA , Humanos , Camundongos , Camundongos Knockout , Neoplasias/patologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Proto-Oncogênicas c-mdm2/genética
6.
Subcell Biochem ; 85: 359-70, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25201204

RESUMO

Over the past few decades, advances in cancer research have enabled us to understand the different mechanisms that contribute to the aberrant proliferation of normal cells into abnormal cells that result in tumors. In the pursuit to find cures, researchers have primarily focused on various molecular level changes that are unique to cancerous cells. In humans, about 50 % or more cancers have a mutated tumor suppressor p53 gene thereby resulting in accumulation of p53 protein and losing its function to activate the target genes that regulate cell cycle and apoptosis. Extensive research conducted in murine cancer models with activated p53, loss of p53, or p53 missense mutations have facilitated researchers to understand the role of this key protein. Despite the identification of numerous triggers that causes lung cancer specific cure still remain elusive. One of the primary reasons attributed to this is due to the fact that the tumor tissue is heterogeneous and contains numerous sub-populations of cells. Studies have shown that a specific sub-population of cells termed as cancer stem cells (CSCs) drive the recurrence of cancer in response to standard chemotherapy. These CSCs are mutated cells with core properties similar to those of adult stem cells. They reside in a microenvironment within the tumor tissue that supports their growth and make them less susceptible to drug treatment. These cells possess properties of symmetric self-renewal and migration thus driving tumor formation and metastasis. Therefore, research specifically targeting these cells has gained prominence towards developing new therapeutic agents against cancer. This chapter focuses on lung cancer stem cells, p53 mutations noted in these cells, and importance of MDM2 interactions. Further, research approaches for better understanding of molecular mechanisms that drive CSC function and developing appropriate therapies are discussed.


Assuntos
Genes p53 , Neoplasias Pulmonares/patologia , Mutação , Células-Tronco Neoplásicas/patologia , Proteínas Proto-Oncogênicas c-mdm2/genética , Humanos , Neoplasias Pulmonares/genética
7.
Nucleic Acids Res ; 42(2): 926-40, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24163099

RESUMO

Conventional paradigm ascribes the cell proliferative function of the human oncoprotein mouse double minute2 (MDM2) primarily to its ability to degrade p53. Here we report that in the absence of p53, MDM2 induces replication stress eliciting an early S-phase checkpoint response to inhibit further firing of DNA replication origins. Partially synchronized lung cells cultured from p53-/-:MDM2 transgenic mice enter S phase and induce S-phase checkpoint response earlier than lung cells from p53-/- mice and inhibit firing of DNA replication origins. MDM2 activates chk1 phosphorylation, elevates mixed lineage lymphoma histone methyl transferase levels and promotes checkpoint-dependent tri-methylation of histone H3 at lysine 4, known to prevent firing of late replication origins at the early S phase. In the absence of p53, a condition that disables inhibition of cyclin A expression by MDM2, MDM2 increases expression of cyclin D2 and A and hastens S-phase entry of cells. Consistently, inhibition of cyclin-dependent kinases, known to activate DNA replication origins during firing, inhibits MDM2-mediated induction of chk1 phosphorylation indicating the requirement of this activity in MDM2-mediated chk1 phosphorylation. Our data reveal a novel pathway, defended by the intra-S-phase checkpoint, by which MDM2 induces unscheduled origin firing and accelerates S-phase entry of cells in the absence of p53.


Assuntos
Replicação do DNA , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Origem de Replicação , Pontos de Checagem da Fase S do Ciclo Celular/genética , Animais , Cafeína/farmacologia , Linhagem Celular , Quinase 1 do Ponto de Checagem , Ciclina A/metabolismo , Ciclina D2/metabolismo , Genes p53 , Histona-Lisina N-Metiltransferase/metabolismo , Histonas/metabolismo , Humanos , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Inibidores de Proteínas Quinases/farmacologia , Proteínas Quinases/metabolismo , Proteínas Proto-Oncogênicas c-mdm2/genética , Origem de Replicação/efeitos dos fármacos , Estresse Fisiológico/genética
8.
Methods Mol Biol ; 962: 127-33, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23150442

RESUMO

Chemoresistance and increased growth rate are two gain-of-function functions that mutant p53 is thought to possess. Here, we describe two methods for measuring the sensitiveness of cells to chemotherapeutic drugs and the rate of cell growth. Both of which can be used with a wide range of cell types. The clonogenic assay can be used with many different chemotoxic drugs and the growth assay described here presents an alternative to the MTT assay and allows for a long-term measurement of cell growth. These protocols are both easy, flexible, require relatively little effort, and are inexpensive to carry out.


Assuntos
Proliferação de Células/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica , Neoplasias Pulmonares/genética , Proteína Supressora de Tumor p53/metabolismo , Antineoplásicos/farmacologia , Apoptose , Ciclo Celular/efeitos dos fármacos , Ensaio de Unidades Formadoras de Colônias/métodos , Humanos , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/genética
9.
Methods Mol Biol ; 962: 147-55, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23150444

RESUMO

DNA replication involves a coordinated progression through S phase, and disruption of these regulated steps may cause gene abnormalities, which may lead to cancer. Different stages of DNA replication can be detected immunofluorescently that would indicate how replication is progressing in a cell population or under specific conditions. We describe a method for labeling replicating DNA with two nucleotide analogs, and then detecting the sequential patterns of incorporation using fluorescently labeled antibodies on DNA spread onto a glass slide. Quantification of the different types of replication patterns produced by this method reveals how replication is achieved under different conditions by the predominance and lengths of elongating replication forks progressing from single or clustered origins, as well as the sites of termination from two converging forks.


Assuntos
Replicação do DNA , Proteínas Mutantes/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Linhagem Celular Tumoral , DNA/genética , Humanos , Proteínas Mutantes/genética , Fase S , Proteína Supressora de Tumor p53/genética
10.
Methods Mol Biol ; 962: 193-9, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23150448

RESUMO

In order to study the functions of a cell's endogenous mutant p53, the p53 protein levels must be knocked-down. Transient transfection of small interfering RNAs is one way to accomplish this. Another is the stable expression of short hairpin RNAs. This chapter presents a method by which a short hairpin RNA (shRNA) targeting p53 is inserted into the genome of a cell via lentivirus infection. These p53 knock-down cell lines are stable and may be grown long term for use in a wide range of applications.


Assuntos
Técnicas de Silenciamento de Genes/métodos , Proteína Supressora de Tumor p53/genética , DNA Viral/genética , Vetores Genéticos , Células HEK293 , Humanos , Lentivirus , Proteínas Mutantes/genética , RNA Mensageiro/genética , RNA Interferente Pequeno/genética , Transfecção
11.
Carcinogenesis ; 33(2): 442-51, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22114072

RESUMO

The role of dominant transforming p53 in carcinogenesis is poorly understood. Our previous data suggested that aberrant p53 proteins can enhance tumorigenesis and metastasis. Here, we examined potential mechanisms through which gain-of-function (GOF) p53 proteins can induce motility. Cells expressing GOF p53 -R175H, -R273H and -D281G showed enhanced migration, which was reversed by RNA interference (RNAi) or transactivation-deficient mutants. In cells with engineered or endogenous p53 mutants, enhanced migration was reduced by downregulation of nuclear factor-kappaB2, a GOF p53 target. We found that GOF p53 proteins upregulate CXC-chemokine expression, the inflammatory mediators that contribute to multiple aspects of tumorigenesis. Elevated expression of CXCL5, CXCL8 and CXCL12 was found in cells expressing oncogenic p53. Transcription was elevated as CXCL5 and CXCL8 promoter activity was higher in cells expressing GOF p53, whereas wild-type p53 repressed promoter activity. Chromatin immunoprecipitation assays revealed enhanced presence of acetylated histone H3 on the CXCL5 promoter in H1299/R273H cells, in agreement with increased transcriptional activity of the promoter, whereas RNAi-mediated repression of CXCL5 inhibited cell migration. Consistent with this, knockdown of the endogenous mutant p53 in lung cancer or melanoma cells reduced CXCL5 expression and cell migration. Furthermore, short hairpin RNA knockdown of mutant p53 in MDA-MB-231 cells reduced expression of a number of key targets, including several chemokines and other inflammatory mediators. Finally, CXCL5 expression was also elevated in lung tumor samples containing GOF p53, indicating relevance to human cancer. The data suggest a mechanistic link between GOF p53 proteins and chemokines in enhanced cell motility.


Assuntos
Movimento Celular/genética , Quimiocinas CXC/genética , Mutação , Proteína Supressora de Tumor p53/genética , Acetilação , Linhagem Celular Tumoral , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Quimiocinas CXC/metabolismo , Imunoprecipitação da Cromatina/métodos , Regulação para Baixo , Regulação Neoplásica da Expressão Gênica , Técnicas de Silenciamento de Genes/métodos , Histonas/metabolismo , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Melanoma/genética , Melanoma/metabolismo , Subunidade p52 de NF-kappa B/genética , Subunidade p52 de NF-kappa B/metabolismo , Regiões Promotoras Genéticas , Interferência de RNA , RNA Interferente Pequeno/genética , Ativação Transcricional , Proteína Supressora de Tumor p53/metabolismo , Regulação para Cima
12.
Genes Cancer ; 2(10): 943-55, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22701761

RESUMO

The current model predicts that MDM2 is primarily overexpressed in cancers with wild-type (WT) p53 and contributes to oncogenesis by degrading p53. Following a correlated expression of MDM2 and NF-κB2 transcripts in human lung tumors, we have identified a novel transactivation function of MDM2. Here, we report that in human lung tumors, overexpression of MDM2 was found in approximately 30% of cases irrespective of their p53 status, and expression of MDM2 and NF-κB2 transcripts showed a highly significant statistical correlation in tumors with WT p53. We investigated the significance of this correlated expression in terms of mechanism and biological function. Increase in MDM2 expression from its own promoter in transgenic mice remarkably enhanced expression of NF-κB2 compared with its non-transgenic littermates. Knockdown or elimination of endogenous MDM2 expression in cultured non-transformed or lung tumor cells drastically reduced expression of NF-κB2 transcripts, suggesting a normal physiological role of MDM2 in regulating NF-κB2 transcription. MDM2 could up-regulate expression of NF-κB2 transcripts when its p53-interaction domain was blocked with Nutlin-3, indicating that the MDM2-p53 interaction is dispensable for up-regulation of NF-κB2 expression. Consistently, analysis of functional domains of MDM2 indicated that although the p53-interaction domain of MDM2 contributes to the up-regulation of the NFκB2 promoter, MDM2 does not require direct interactions with p53 for this function. Accordingly, MDM2 overexpression in non-transformed or lung cancer cells devoid of p53 also generated a significant increase in the expression of NF-κB2 transcript and its targets CXCL-1 and CXCL-10, whereas elimination of MDM2 expression had the opposite effects. MDM2-mediated increase in p100/NF-κB2 expression reduced cell death mediated by paclitaxel. Furthermore, knockdown of NF-κB2 expression retarded cell proliferation. Based on these data, we propose that MDM2-mediated NF-κB2 up-regulation is a combined effect of p53-dependent and independent mechanisms and that it confers a survival advantage to lung cancer cells.

13.
Mol Cancer Res ; 7(8): 1253-67, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19671680

RESUMO

Overexpression of MDM2 has been related to oncogenesis. In this communication, we present evidence to show that MDM2 controls the cell cycle-dependent expression of cyclin A by using a pathway that ensures its timely expression. MDM2 does not inhibit cyclin D or E expression. Silencing of endogenous MDM2 expression elevates cyclin A expression. The p53-binding domain of MDM2 harbors a SWIB region homologous to a conserved domain of a chromosome remodeling factor BRG1-associated protein. The SWIB domain of MDM2 inhibits cyclin A expression in a p53- and BRG1-dependent fashion, suggesting that MDM2 interferes with p53 binding of the BRG1 complex freeing it to repress cyclin A expression. Silencing of cyclin-dependent kinase (cdk) inhibitor p16 prevents MDM2-mediated inhibition of cyclin A expression, implicating its role in the process. MDM2-mediated repression of cyclin A expression induces G(1)-S arrest, which can be rescued by ectopic expression of cyclin A. Cancer cells lacking p53, p16, or BRG1 escape MDM2-mediated repression of cyclin A expression and growth arrest. Our data propose a novel mechanism by which MDM2 controls the cell cycle in normal cells and how cancer cells may escape this important safety barrier.


Assuntos
Ciclo Celular , Ciclina A/metabolismo , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Linhagem Celular Transformada , Linhagem Celular Tumoral , Ciclina E/metabolismo , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , DNA Helicases/metabolismo , Regulação para Baixo , Citometria de Fluxo , Fase G1 , Humanos , Microscopia Confocal , Modelos Biológicos , Proteínas Nucleares/metabolismo , Estrutura Terciária de Proteína , Proteínas Proto-Oncogênicas c-mdm2/química , Fatores de Tempo , Fatores de Transcrição/metabolismo , Proteína Supressora de Tumor p53/metabolismo
14.
Mol Cell Biol ; 25(22): 10097-110, 2005 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16260623

RESUMO

Overexpression of mutant p53 is a common theme in tumors, suggesting a selective pressure for p53 mutation in cancer development and progression. To determine how mutant p53 expression may lead to survival advantage in human cancer cells, we generated stable cell lines expressing p53 mutants p53-R175H, -R273H, and -D281G by use of p53-null human H1299 (lung carcinoma) cells. Compared to vector-transfected cells, H1299 cells expressing mutant p53 showed a survival advantage when treated with etoposide, a common chemotherapeutic agent; however, cells expressing the transactivation-deficient triple mutant p53-D281G (L22Q/W23S) had significantly lower resistance to etoposide. Gene expression profiling of cells expressing transcriptionally active mutant p53 proteins revealed the striking pattern that all three p53 mutants induced expression of approximately 100 genes involved in cell growth, survival, and adhesion. The gene NF-kappaB2 is a prominent member of this group, whose overexpression in H1299 cells also leads to chemoresistance. Treatment of H1299 cells expressing p53-R175H with small interfering RNA specific for NF-kappaB2 made these cells more sensitive to etoposide. We have also observed activation of the NF-kappaB2 pathway in mutant p53-expressing cells. Thus, one possible pathway through which mutants of p53 may induce loss of drug sensitivity is via the NF-kappaB2 pathway.


Assuntos
Regulação Neoplásica da Expressão Gênica , Mutação , Subunidade p52 de NF-kappa B/biossíntese , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/fisiologia , Adenoviridae/genética , Adenoviridae/metabolismo , Bromodesoxiuridina/farmacologia , Linhagem Celular Tumoral , Núcleo Celular/metabolismo , Clonagem Molecular , DNA Complementar/metabolismo , Progressão da Doença , Etoposídeo/farmacologia , Éxons , Vetores Genéticos , Humanos , NF-kappa B/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Reação em Cadeia da Polimerase , RNA/metabolismo , RNA Interferente Pequeno/metabolismo , Transcrição Gênica , Ativação Transcricional , Transfecção , Proteína Supressora de Tumor p53/metabolismo , Regulação para Cima
15.
Cancer Res ; 64(20): 7447-54, 2004 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-15492269

RESUMO

p53 mutants with a single amino acid substitution are overexpressed in a majority of human cancers containing a p53 mutation. Overexpression of the mutant protein suggests that there is a selection pressure on the cell indicative of an active functional role for mutant p53. Indeed, H1299 cells expressing mutant p53-R175H, p53-R273H or p53-D281G grow at a faster rate compared with a control cell line. Using p53-specific small interfering RNA, we show that the growth rate of mutant p53-expressing cells decreases as mutant p53 level decreases, demonstrating that the increased cellular growth is dependent on p53 expression. Increased growth rate is not observed for H1299 cell clones expressing mutant p53-D281G (L22Q/W23S), which has been shown to be defective in transactivation in transient transcriptional assays. This shows that the increased growth rate imparted by mutant p53 in H1299 cells requires the transactivation function of mutant p53. By performing microarray hybridization analyses, we show that constitutive expression of three common p53 mutants (p53-R175H, p53-R273H, and p53-D281G) in H1299 human lung carcinoma cells evokes regulation of a common set of genes, a significant number of which are involved in cell growth regulation. Predictably, H1299 cells expressing p53-D281G (L22Q/W23S) are defective in up-regulating a number of these genes. The differences in expression profiles induced by individual p53 mutants in the cells may be representative of the p53 mutants and how they can affect gene expression resulting in the observed "gain of function" phenotypes (i.e., increased growth rate, decreased sensitivity to chemotherapeutic agents, and so forth).


Assuntos
Regulação Neoplásica da Expressão Gênica/genética , Neoplasias Pulmonares/genética , Ativação Transcricional/genética , Proteína Supressora de Tumor p53/genética , Processos de Crescimento Celular/genética , Linhagem Celular Tumoral , Humanos , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Análise de Sequência com Séries de Oligonucleotídeos , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/farmacologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transfecção , Proteína Supressora de Tumor p53/biossíntese , Regulação para Cima
16.
Oncogene ; 23(25): 4430-43, 2004 May 27.
Artigo em Inglês | MEDLINE | ID: mdl-15077194

RESUMO

We have studied the mechanism of mutant p53-mediated oncogenesis using several tumor-derived mutants. Using a colony formation assay, we found that the majority of the mutants increased the number of colonies formed compared to the vector. Expression of tumor-derived p53 mutants increases the rate of cell growth, suggesting that the p53 mutants have 'gain of function' properties. We have studied the gene expression profile of cells expressing tumor-derived p53-D281G to identify genes transactivated by mutant p53. We report the transactivation of two genes, asparagine synthetase and human telomerase reverse transcriptase. Quantitative real-time PCR confirms this upregulation. Transient transfection promoter assays verify that tumor-derived p53 mutants transactivate these promoters significantly. An electrophoretic mobility shift assay shows that tumor-derived p53-mutants cannot bind to the wild-type p53 consensus sequence. The results presented here provide some evidence of a possible mechanism for mutant p53-mediated transactivation.


Assuntos
Transformação Celular Neoplásica/genética , Genes p53 , Neoplasias/genética , Ativação Transcricional/genética , Proteína Supressora de Tumor p53/fisiologia , Substituição de Aminoácidos , Aspartato-Amônia Ligase/biossíntese , Aspartato-Amônia Ligase/genética , Divisão Celular , Linhagem Celular Tumoral , Sequência Consenso , Proteínas de Ligação a DNA/metabolismo , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Genes Supressores de Tumor , Humanos , Mutação de Sentido Incorreto , Proteínas Nucleares/metabolismo , Mutação Puntual , Regiões Promotoras Genéticas/genética , Ligação Proteica , Estrutura Terciária de Proteína , Proteínas Recombinantes de Fusão/fisiologia , Relação Estrutura-Atividade , Telomerase/biossíntese , Telomerase/genética , Proteína Tumoral p73 , Ensaio Tumoral de Célula-Tronco , Proteína Supressora de Tumor p53/química , Proteínas Supressoras de Tumor
17.
Methods Mol Biol ; 234: 93-110, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-12824527

RESUMO

The methods outlined in this chapter are designed to facilitate the study of the transactivation and transrepression properties of p53 (as well as p63 and p73). Once a gene of interest is identified, its presumptive promoter region can be cloned upstream of a luciferase gene in a plasmid. The most common reason for transfection experiments is to study gene expression patterns in the presence or absence of a particular gene product (e.g., p53). Three methods of transfection are outlined in this chapter: (i) cationic lipofection; (ii) calcium phosphate precipitation; and (iii) BES precipitation. The first method is ideal for the study of transactivation and transrepression properties of p53 (or other transcription factors). The last two are more suited for experiments where larger numbers of transfected cells are needed. Several examples of transfections and their respective results are provided.


Assuntos
Regulação da Expressão Gênica , Proteínas Repressoras/metabolismo , Ativação Transcricional , Transfecção/métodos , Proteína Supressora de Tumor p53/metabolismo , Linhagem Celular Tumoral , Genes Reporter , Vetores Genéticos , Humanos , Regiões Promotoras Genéticas , Proteínas Repressoras/genética , Proteína Supressora de Tumor p53/genética
18.
Methods Mol Biol ; 234: 257-67, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-12824538

RESUMO

Although MDM2, the product of mouse double minute-2 (mdm2) gene, or its human homologue possesses the potential to confer tumorigenic properties, it induces G1/S arrest in nontransformed cells. Flow cytometry provides a way to determine the effects of MDM2 on the cell cycle by expressing the protein ectopically, immunostaining cells expressing MDM2 and analyzing their DNA content. The DNA histograms of MDM2-transfected and untransfected cells can then be used to visualize the effect of ectopically expressed MDM2 on the cell cycle. Fluorescence-activated cell sorter (FACS) analysis following bromodeoxyuridine (BrdU) incorporation can be used to determine whether MDM2-expressing cells are synthesizing DNA. Incorporation of BrdU during DNA synthesis or repair can be detected in partially denatured DNA with a BrdU-specific fluorescent antibody. Subsequent staining of transfected MDM2 with a different fluorochrome provides information about whether transfected cells make significant progression through S phase. Further analysis of the growth-regulatory properties of MDM2 will elucidate both its normal function and the ways in which its deregulation leads to tumorigenesis.


Assuntos
Ciclo Celular/fisiologia , DNA/análise , Citometria de Fluxo/métodos , Inibidores do Crescimento/metabolismo , Proteínas Nucleares , Proteínas Proto-Oncogênicas/metabolismo , Animais , Antimetabólitos/metabolismo , Bromodesoxiuridina/metabolismo , Separação Celular/métodos , Humanos , Camundongos , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas c-mdm2
19.
Mol Cancer Res ; 1(14): 1009-16, 2003 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-14707284

RESUMO

The protein (MDM2) coded by the mouse double minute-2 (mdm2) gene or its human homologue is well known as an oncoprotein. Malignant human tumors particularly breast tumors and soft tissue sarcomas frequently overexpress MDM2. Artificial amplification of mdm2 gene derived from a transformed murine cell line enhances tumorigenic potential of murine cells. Consistent with its tumorigenic property, mouse or human MDM2 can inactivate several functions of the tumor suppressor p53 and can degrade p53. The protein also interacts with other tumor suppressors, and these interactions may contribute to its tumorigenic property. In spite of its oncogenic role, mouse or human MDM2 induces G(1) arrest in normal human or murine cells. Some cell lines bearing known genetic mutations are insensitive to MDM2-mediated growth arrest. This review is aimed to collect available information on the functions of MDM2 that could potentially regulate cell cycle and to discuss how this information may fit in one model that could explain the two apparently opposite G(1) arrest and oncogenic function of MDM2.


Assuntos
Ciclo Celular/fisiologia , Proteínas Nucleares/fisiologia , Proteínas Proto-Oncogênicas/fisiologia , Animais , Humanos , Proteínas Proto-Oncogênicas c-mdm2
20.
Front Biosci ; 7: d235-43, 2002 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-11779693

RESUMO

The protein MDM2 coded by the human homologue of mouse double minute-2 (mdm2) gene frequently overexpresses in malignant human breast and other tumors. Artificial amplification of mouse mdm2 gene derived from a transformed murine cell line enhances tumorigenic potential of murine cells. These evidences suggest oncogenic properties of human or mouse MDM2. The tumorigenic property of MDM2 is not unexpected as MDM2 can inactivate several functions of the tumor suppressor p53. The protein also interacts with several cell cycle regulatory proteins that may contribute to its tumorigenic ability. Several spliced forms of MDM2 have been detected in cells that overexpress MDM2. The function of the proteins coded by these spliced forms is not well understood. Overexpression of full-length MDM2 from its cDNA arrests G1 to S phase transition of normal human or murine cells. Elimination of the growth inhibitory domains of the oncoprotein induces tumorigenesis. Some cancer-derived cell lines are partially insensitive to MDM2-mediated growth arrest. Normal cells can induce MDM2 in response to oncogenic challenges such as UV irradiation or estrogen treatment. Normal cells may induce full-length MDM2 in response to oncogenic challenges to protect against premature cell cycle progression. If the oncoprotein is defective in growth arrest or if the cells are insensitive to MDM2 mediated growth arrest, premature progression of cell cycle may lead to tumorigenesis. Elucidation of the growth regulatory functions of MDM2 may help develop new drug design for cancer treatment.


Assuntos
Proteínas de Neoplasias/fisiologia , Proteínas Nucleares , Proteínas Proto-Oncogênicas/fisiologia , Animais , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Transformação Celular Neoplásica/patologia , Regulação Neoplásica da Expressão Gênica/fisiologia , Humanos , Proteínas de Neoplasias/biossíntese , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Proteínas Proto-Oncogênicas/biossíntese , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-mdm2
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...