Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
1.
Proc Natl Acad Sci U S A ; 115(13): 3452-3457, 2018 03 27.
Artigo em Inglês | MEDLINE | ID: mdl-29540569

RESUMO

Renewable tissues exhibit heightened sensitivity to DNA damage, which is thought to result from a high level of p53. However, cell proliferation in renewable tissues requires p53 down-regulation, creating an apparent discrepancy between the p53 level and elevated sensitivity to DNA damage. Using a combination of genetic mouse models and pharmacologic inhibitors, we demonstrate that it is p53-regulated MDM2 that functions together with MDMX to regulate DNA damage sensitivity by targeting EZH2 (enhancer of zeste homolog 2) for ubiquitination/degradation. As a methyltransferase, EZH2 promotes H3K27me3, and therefore chromatin compaction, to determine sensitivity to DNA damage. We demonstrate that genetic and pharmacologic interference of the association between MDM2 and MDMX stabilizes EZH2, resulting in protection of renewable tissues from radio-/chemotherapy-induced acute injury. In cells with p53 mutation, there are diminished MDM2 levels, and thus accumulation of EZH2, underpinning the resistant phenotype. Our work uncovers an epigenetic mechanism behind tissue sensitivity to DNA damage, carrying important translation implications.


Assuntos
Cromatina/metabolismo , Dano ao DNA , Proteína Potenciadora do Homólogo 2 de Zeste/genética , Epigênese Genética , Proteínas Proto-Oncogênicas c-mdm2/genética , Proteínas Proto-Oncogênicas/genética , Proteína Supressora de Tumor p53/genética , Animais , Apoptose , Cromatina/genética , Proteína Potenciadora do Homólogo 2 de Zeste/metabolismo , Camundongos , Camundongos Transgênicos , Ligação Proteica , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Ubiquitinação
2.
J Biol Chem ; 291(50): 25937-25949, 2016 Dec 09.
Artigo em Inglês | MEDLINE | ID: mdl-27777309

RESUMO

Deregulated receptor tyrosine kinase (RTK) signaling is frequently associated with tumorigenesis and therapy resistance, but its underlying mechanisms still need to be elucidated. In this study, we have shown that the RTK human epidermal growth factor receptor 4 (Her4, also known as Erbb4) can inhibit the tumor suppressor p53 by regulating MDMX-mouse double minute 2 homolog (MDM2) complex stability. Upon activation by either overexpression of a constitutively active vector or ligand binding (Neuregulin-1), Her4 was able to stabilize the MDMX-MDM2 complex, resulting in suppression of p53 transcriptional activity, as shown by p53-responsive element-driven luciferase assay and mRNA levels of p53 target genes. Using a phospho-proteomics approach, we functionally identified a novel Her4-induced posttranslational modification on MDMX at Ser-314, a putative phosphorylation site for the CDK4/6 kinase. Remarkably, inhibition of Ser-314 phosphorylation either with Ser-to-Ala substitution or with a specific inhibitor of CDK4/6 kinase blocked Her4-induced stabilization of MDMX-MDM2 and rescued p53 activity. Our study offers insights into the mechanisms of deregulated RTK-induced carcinogenesis and provides the basis for the use of inhibitors targeting RTK-mediated signals for p53 restoration.


Assuntos
Proteínas Nucleares/metabolismo , Processamento de Proteína Pós-Traducional , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Receptor ErbB-4/metabolismo , Transcrição Gênica , Proteína Supressora de Tumor p53/metabolismo , Animais , Proteínas de Ciclo Celular , Quinase 4 Dependente de Ciclina/genética , Quinase 4 Dependente de Ciclina/metabolismo , Quinase 6 Dependente de Ciclina/genética , Quinase 6 Dependente de Ciclina/metabolismo , Células HEK293 , Humanos , Células MCF-7 , Camundongos , Fosforilação , Proteínas Proto-Oncogênicas c-mdm2/genética , Receptor ErbB-4/genética , Proteína Supressora de Tumor p53/genética
3.
J Biol Chem ; 289(8): 5340-7, 2014 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-24391088

RESUMO

Radiotherapy is the current frontline cancer treatment, but the resulting severe side effects often pose a significant threat to cancer patients, raising a pressing need for the development of effective strategies for radiotherapy protection. We exploited the distinct metabolic characteristics between normal and malignant cells for a metabolic mechanism of normal tissue protection. We showed that low doses of arsenic induce HIF-1α, which activates a metabolic shift from oxidative phosphorylation to glycolysis, resulting in increased cellular resistance to radiation. Of importance is that low-dose arsenic-induced HIF-1α requires functional p53, limiting the glycolytic shift to normal cells. Using tumor-bearing mice, we provide proof of principle for selective normal tissue protection against radiation injury.


Assuntos
Arsênio/farmacologia , Metabolismo/efeitos dos fármacos , Protetores contra Radiação/farmacologia , Proteína Supressora de Tumor p53/metabolismo , Animais , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Glicólise/efeitos dos fármacos , Glicólise/efeitos da radiação , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Metabolismo/efeitos da radiação , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , NF-kappa B/metabolismo , Tolerância a Radiação/efeitos dos fármacos , Tolerância a Radiação/efeitos da radiação , Irradiação Corporal Total
4.
Mutat Res ; 701(1): 12-22, 2010 Aug 14.
Artigo em Inglês | MEDLINE | ID: mdl-20298803

RESUMO

New data and historical evidence from our own and other laboratories are summarized and discussed bearing on several issues relating to mechanisms and processes involved in the formation of chromosomal aberrations following exposure to ionizing radiations. Specifically addressed are: (1) the lesions and processes affecting the appearance of chromatid-type and/or chromosome-type aberrations after radiation, (2) DNA double strand break rejoining processes and the restitution of breaks vs. the formation of exchanges, (3) the role of homologous recombinational repair in protecting cells from induction of chromatid-type aberrations after irradiation of late S/G2 cells, (4) the role of interphase chromatin structure and nuclear organization in aberration induction, (5) cellular responses for aberration induction in relation to their tissue context, and (6) approaches to the detection of aberrations previously known as "cryptic".


Assuntos
Aberrações Cromossômicas , Reparo do DNA , Radiação Ionizante , Recombinação Genética , Animais , Linhagem Celular , Células Cultivadas , Cromatina/química , Humanos , Interfase , Radiogenética , Técnicas de Cultura de Tecidos
5.
Cell Death Differ ; 27(5): 1618-1632, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-31659281

RESUMO

Apart from its well-known prodeath activity, p53 is also implicated in promoting cell survival. How p53 can mediate such seemingly opposing effects is largely unclear. We report here a novel mechanism in which p53-mediated proapoptosis is switched to antiapoptosis via its interaction with a p53 isoform, Δ133p53. We show that the expression of Δ133p53 is induced by mild or a moderate level of stress via an HIF1-dependent mechanism. Increased Δ133p53 levels contribute to the adaptive response by shifting the p53 binding at the Bcl2 promoter from suppressive responsive elements (RE) to activating REs, resulting in induction of Bcl2. In accordance with this mode of action, pretreatment of mice with mild stress induces Δ133p53 and Bcl2, which is associated with protection of animals from toxicity caused by high doses of DNA damage agents. Collectively, our work uncovers a novel functional interplay between p53 and Δ133p53 determining cell fate; survival or death in response to stress.


Assuntos
Mutação/genética , Estresse Fisiológico , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Animais , Apoptose , Sequência de Bases , Sítios de Ligação , Linhagem Celular , Relação Dose-Resposta à Radiação , Glicólise , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Camundongos Endogâmicos C57BL , Regiões Promotoras Genéticas/genética , Isoformas de Proteínas/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais , Regulação para Cima
6.
Transl Cancer Res ; 9(3): 1993-1997, 2020 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-35117546

RESUMO

Via regulation of cellular stress responses, p53 contributes to the maintenance of homeostasis. Contrary to its well-established pro-death function, p53 is also implicated in promoting cell survival by mediating the adaptive stress response. Emerging data reveal that the adaptive stress response is coupled with p53 decline that is a prerequisite for the induction of pro-survival pathways augmenting cell fitness. However, if the adaptive stress responses persist or become chronic, the sustained p53 downregulation would result in a permanent loss of p53 function and p53-dependent homeostasis. The available information suggests a model in which cells respond to different levels of stress by governing the activity and abundance of p53 that, in turn, determines the cell fate dependent on not only the intensity but also the duration of stress.

7.
J Mol Cell Biol ; 12(9): 713-722, 2020 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-32706867

RESUMO

A role of tumor-suppressive activity of p53 in the tumor microenvironment (TME) has been implicated but remains fairly understudied. To address this knowledge gap, we leveraged our MdmxS314A mice as recipients to investigate how implanted tumor cells incapacitate host p53 creating a conducive TME for tumor progression. We found that tumor cell-associated stress induced p53 downregulation in peritumor cells via an MDMX-Ser314 phosphorylation-dependent manner. As a result, an immunosuppressive TME was developed, as reflected by diminished immune cell infiltration into tumors and compromised macrophage M1 polarization. Remarkably, ablation of MDMX-Ser314 phosphorylation attenuated p53 decline in peritumor cells, which was associated with mitigation of immunosuppression and significant tumor growth delay. Our data collectively uncover a novel role of p53 in regulating the tumor immune microenvironment, suggesting that p53 restoration in the TME can be exploited as a potential strategy of anticancer therapy.


Assuntos
Regulação para Baixo , Terapia de Imunossupressão , Proteínas Proto-Oncogênicas/metabolismo , Microambiente Tumoral/imunologia , Proteína Supressora de Tumor p53/metabolismo , Animais , Linhagem Celular Tumoral , Polaridade Celular , Técnicas de Introdução de Genes , Humanos , Macrófagos/metabolismo , Camundongos Endogâmicos C57BL , Transplante de Neoplasias , Fosforilação , Fosfosserina/metabolismo
8.
DNA Repair (Amst) ; 7(3): 515-22, 2008 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-18182331

RESUMO

We reported previously that the homologous recombinational repair (HRR)-deficient Chinese hamster mutant cell line irs3 (deficient in the Rad51 paralog Rad51C) showed only a 50% spontaneous frequency of sister chromatid exchange (SCE) as compared to parental wild-type V79 cells. Furthermore, when irradiated with very low doses of alpha particles, SCEs were not induced in irs3 cells, as compared to a prominent bystander effect observed in V79 cells [H. Nagasawa, Y. Peng, P.F. Wilson, Y.C. Lio, D.J. Chen, J.S. Bedford, J.B. Little, Role of homologous recombination in the alpha-particle-induced bystander effect for sister chromatid exchanges and chromosomal aberrations, Radiat. Res. 164 (2005) 141-147]. In the present study, we examined additional Chinese hamster cell lines deficient in the Rad51 paralogs Rad51C, Rad51D, Xrcc2, and Xrcc3 as well as another essential HRR protein, Brca2. Spontaneous SCE frequencies in non-irradiated wild-type cell lines CHO, AA8 and V79 were 0.33SCE/chromosome, whereas two Rad51C-deficient cell lines showed only 0.16SCE/chromosome. Spontaneous SCE frequencies in cell lines defective in Rad51D, Xrcc2, Xrcc3, and Brca2 ranged from 0.23 to 0.33SCE/chromosome, 0-30% lower than wild-type cells. SCEs were induced significantly 20-50% above spontaneous levels in wild-type cells exposed to a mean dose of 1.3mGy of alpha particles (<1% of nuclei traversed by an alpha particle). However, induction of SCEs above spontaneous levels was minimal or absent after alpha-particle irradiation in all of the HRR-deficient cell lines. These data suggest that Brca2 and the Rad51 paralogs contribute to DNA damage repair processes induced in bystander cells (presumably oxidative damage repair in S-phase cells) following irradiation with very low doses of alpha particles.


Assuntos
Partículas alfa , Efeito Espectador , Reparo do DNA , Recombinação Genética/efeitos da radiação , Troca de Cromátide Irmã/efeitos da radiação , Animais , Proteína BRCA2/fisiologia , Células CHO , Cricetinae , Cricetulus , Proteínas de Ligação a DNA/fisiologia , Relação Dose-Resposta à Radiação , Rad51 Recombinase/fisiologia , Fase S/fisiologia
9.
Radiat Res ; 172(3): 306-13, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19708779

RESUMO

In addition to cell cycle arrest, DNA repair or/and apoptosis, ionizing radiation can also induce premature senescence, which could lead to very different biological consequences depending on the cell type. We show in this report that low-dose radiation-induced senescent stromal fibroblasts stimulate proliferation of cocultured breast carcinoma cells. Such effects of senescent fibroblasts appear to result from their ability to induce the expression in carcinoma cells of mitotic genes and subsequent mitotic division. The elevated proliferation of breast carcinoma cells correlates with resistance to radiation as well as to adriamycin. Of interest is the observation that exposure to lower doses (<20 cGy) augments the ability of senescent fibroblasts to promote the survival of cocultured breast carcinoma cells. The resistance appears to be mediated partially by the Akt pathway, because expression of a dominant negative Akt mutant in breast carcinoma cells results in a partial reversal of the radioresistance. The ability of fibroblasts to modulate the radiosensitivity of nearby carcinoma cells implicates the importance of targeting the stroma during therapy.


Assuntos
Neoplasias da Mama/patologia , Neoplasias da Mama/fisiopatologia , Efeito Espectador/efeitos da radiação , Senescência Celular/efeitos da radiação , Fibroblastos/efeitos da radiação , Tolerância a Radiação/efeitos da radiação , Linhagem Celular Tumoral , Relação Dose-Resposta à Radiação , Humanos , Doses de Radiação , Células Estromais/patologia , Células Estromais/efeitos da radiação
10.
DNA Repair (Amst) ; 6(6): 818-29, 2007 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-17339135

RESUMO

Cells from unaffected parents of retinoblastoma (RB) patients were previously shown to be hypersensitive to radiation induced G(1) arrest and cell killing [1]. The hypersensitivity was similar to that reported for cells from ATM heterozygotes. The latter was consistent with a mild DNA DSB rejoining defect which we demonstrated using a gamma-H2AX focus assay after low dose-rate (LDR) irradiation of non-cycling G(0) cells [2,3]. Since neither parent carried the mutant RB allele of the RB heterozygous probands, these results suggested the possibility of an enhanced germline mutation rate, perhaps resulting from some mild defect in genome maintenance. We therefore examined levels of gamma-H2AX foci for cells from these RB parents in this G(0) LDR assay, which reflects the non-homologous end joining (NHEJ) capacity of cells and in a G(2)/M assay, which reflects additional contributions from other G(2)-related damage processing systems. For several of the cell strains parallel radiosensitivity comparisons were made for cell killing and for G(2) chromosomal radiosensitivities. G(0) cells from the RB parents were clearly hypersensitive both in the LDR gamma-H2AX assay, and for cell killing. In addition, cultured fibroblasts from 6 of 15 apparently normal individuals in this study (and one of six in a previous study) were also hypersensitive in the same assays. In the G(2)/M gamma-H2AX assay, the relative sensitivities were similar to those seen in the low dose-rate G(0) assay and tracked with chromosomal radiosensitivity, but some differences were observed.


Assuntos
Dano ao DNA , Retinoblastoma/genética , Ciclo Celular , Divisão Celular , Quebras de DNA de Cadeia Dupla , Reparo do DNA , Saúde da Família , Fase G2 , Mutação em Linhagem Germinativa , Histonas/metabolismo , Humanos , Imuno-Histoquímica , Recombinação Genética , Fase de Repouso do Ciclo Celular , Fatores de Tempo
11.
Radiat Res ; 169(5): 483-94, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18439048

RESUMO

We previously described an enhanced sensitivity for cell killing and G(1)-phase cell cycle arrest after acute gamma irradiation in primary fibroblast strains derived from 14 hereditary-type retinoblastoma family members (both affected RB1(+/-) probands and unaffected RB1(+/+) parents) as well as distinctive gene expression profiles in unirradiated cultures by microarray analyses. In the present study, we measured the colony formation ability of these cells after exposure to continuous low-dose-rate (0.5-8.4 cGy/h) (137)Cs gamma radiation for a 2-week growth period. Fibroblasts from all RB family members (irrespective of RB1 genotype) and from 5 of 18 apparently normal Coriell cell bank controls were significantly more radiosensitive than the remaining apparently normal controls. The average dose rates required to reduce relative survival to 10% and 1% were approximately 3.1 and 4.7 cGy/h for the Coriell control strains with normal radiosensitivity and approximately 1.4 and 2.5 cGy/h for the radiosensitive RB family member and remaining apparently normal Coriell control strains. The finding that a significant proportion of fibroblast strains derived from apparently normal individuals are sensitive to chronic low-dose-rate irradiation indicates such individuals may harbor hypomorphic genetic variants in genomic maintenance and/or DNA repair genes that may likewise predispose them or their children to cancer.


Assuntos
Saúde , Tolerância a Radiação , Retinoblastoma/patologia , Adulto , Proliferação de Células/efeitos da radiação , Células Cultivadas , Criança , Pré-Escolar , Feminino , Fibroblastos , Humanos , Lactente , Recém-Nascido , Masculino , Pessoa de Meia-Idade
12.
J Biomed Sci ; 15(2): 163-8, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18038118

RESUMO

In cancer, gene silencing via hypermethylation is as common as genetic mutations in p53. Understanding the relationship between mutant p53 and hypermethylation of other tumor suppressor genes is essential when elucidate mechanisms of tumor development. In this study, two isogenic human B lymphoblast cell lines with different p53 status include TK6 containing wild-type p53 and WTK1 with mutant p53 were used and contrasted. Lower levels of p16(ink4A) protein were detected in WTK1 cells than in TK6 cells, which were accompanied by increased DNA (cytosine-5)-methyltransferase 1 (DNMT1) gene expression as well as hypermethylation of the p16 ( ink4A ) promoter. siRNA experiments to transiently knock down wild-type p53 in TK6 cells resulted in increase of DNMT1 expression as well as decrease of p16(ink4A) protein. Conversely, siRNA knockdown of mutant p53 in WTK1 cells did not alter either DNMT1 or p16(ink4A) protein levels. Furthermore, loss of suppression function of mutant p53 to DNMT1 in WTK1 was caused by the attenuation of its binding ability to the DNMT1 promoter. In summary, we provide evidences to elucidate the relationship between mutant p53 and DNMT1. Our results indicate that mutant p53 loses its ability to suppress DNMT1 expression, and thus enhances methylation levels of the p16 ( ink4A ) promoter and subsequently down-regulates p16(ink4A )protein.


Assuntos
Linfócitos B/metabolismo , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , DNA (Citosina-5-)-Metiltransferases/metabolismo , Regulação para Baixo , Neoplasias/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Linhagem Celular Tumoral , Inibidor p16 de Quinase Dependente de Ciclina/genética , DNA (Citosina-5-)-Metiltransferase 1 , DNA (Citosina-5-)-Metiltransferases/genética , Metilação de DNA , Humanos , Mutação , Neoplasias/genética , Regiões Promotoras Genéticas/genética , RNA Interferente Pequeno/genética , Proteína Supressora de Tumor p53/antagonistas & inibidores , Proteína Supressora de Tumor p53/genética
13.
Int J Radiat Biol ; 84(2): 151-64, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18246483

RESUMO

PURPOSE: We describe variations of three radiation-induced endpoints on the basis of cell genotype: Clonogenic survival, expression of apoptosis and cell-cycle redistribution. METHODS: Clonogenic survival, apoptosis and cell-cycle redistribution are measured in multiple cell lines after exposure to radiation between 2 and 16 Gy. Cell lines varied in clonogenic radiosensitivity and expression of specific genes. RESULTS: Clonal radiosensitivity is genotype-dependent, associating with four specific genes: A mutated form of Ataxia telangiectasia mutated (mutATM); with two forms of TP53, the gene that is template for tumor protein p53, wildtype TP53 (wtTP53) and mutated TP53 (mutTP53); and an unidentified gene in radioresistant glioblastoma cells. Apoptosis is also genotype-dependent showing elevated levels in cells that express mutATM and abrogated 14-3-3sigma (an isoform of the 14-3-3 gene) but less variation for different forms of TP53. Cell-cycle redistribution varied in mutATM cells. Kinetics of apoptosis are biphasic for both time and dose; cell lines did not express apoptosis at doses below 5 Gy or times before 24 hours. Kinetics of cell-cycle redistribution changed dynamically in the first 24 hours but showed little change after that time. CONCLUSIONS: Clonogenic survival, radiation-induced apoptosis and radiation-induced redistribution in the cell-cycle vary with cell genotype, but not the same genotypes. There is temporal, not quantitative, correlation between apoptosis and clonal radiosensitivity with apoptosis suppressed by lower, less toxic doses of radiation (<5 Gy) but enabled after larger, more toxic doses. Kinetic patterns for apoptosis and redistribution show a common change at approximately 24 hours.


Assuntos
Apoptose/efeitos da radiação , Ciclo Celular/efeitos da radiação , Glioblastoma/radioterapia , Tolerância a Radiação/genética , Proteínas 14-3-3/genética , Proteínas 14-3-3/metabolismo , Proteínas 14-3-3/efeitos da radiação , Apoptose/genética , Apoptose/fisiologia , Proteínas Mutadas de Ataxia Telangiectasia , Ciclo Celular/genética , Ciclo Celular/fisiologia , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Proteínas de Ciclo Celular/efeitos da radiação , Sobrevivência Celular/genética , Sobrevivência Celular/fisiologia , Sobrevivência Celular/efeitos da radiação , Clonagem Molecular , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Proteínas de Ligação a DNA/efeitos da radiação , Relação Dose-Resposta à Radiação , Genes p53/genética , Genótipo , Glioblastoma/genética , Glioblastoma/patologia , Humanos , Cinética , Mutação , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Serina-Treonina Quinases/efeitos da radiação , Tolerância a Radiação/fisiologia , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Proteína Supressora de Tumor p53/efeitos da radiação , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo , Proteínas Supressoras de Tumor/efeitos da radiação
14.
Int J Radiat Biol ; 84(4): 253-64, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18386191

RESUMO

PURPOSE: We have previously shown in a limited number of tumor cell lines derived from only two histological types that clonogenic survival patterns fall into radiosensitivity groups, each group associating with a specific genotype. We now establish a global, quantitative description of human tumor cells based on genotype-dependent radiosensitivity across histological types. METHODS: We measure clonogenic radiosensitivity in 39 human tumor cell lines that vary in histological type (colorectal, glioblastoma, prostate, bladder, teratoma, breast, melanoma and liver) and expression of several genes purported to influence radiosensitivity: ATM (ataxia telangiectasia mutated), TP53 (tumor protein 53), CDKN1A (cyclin-dependent kinase N1A), 14-3-3sigma (an isoform of the 14-3-3 gene) and DNA mismatch repair genes . For each survival curve we use the linear-quadratic model and a linear-linear model to extract multiple coefficients and seek correlation across histological types. RESULTS: Under one-parameter analysis, survival rate at circa 2 Gy, cell lines segregate into two major, statistically-significant groups that correlate with TP53 status (wildtype versus mutant). Under two-parameter analysis, cell lines segregate into four radiosensitivity groups based on correlations between response at lower doses (ca. 2 Gy) and a component of response to higher doses (>4 Gy). CONCLUSIONS: Intrinsic radiosensitivity of 39 human tumor cell lines segregate into distinct genotype-dependent radiosensitivity groups that associate with mutATM, wtTP53, mutTP53, and an unidentified factor in some glioblastoma cells. Genotype-dependent radiosensitivity underlies histology-dependent variation in radiosensitivity. Our analysis establishes a quantitative overview of radiosensitivity that can predict possible response of human tumors to radiotherapy protocols.


Assuntos
Linhagem Celular Tumoral/fisiologia , Linhagem Celular Tumoral/efeitos da radiação , Sobrevivência Celular/efeitos da radiação , Tolerância a Radiação/genética , Proteína Supressora de Tumor p53/genética , Linhagem Celular Tumoral/classificação , Relação Dose-Resposta à Radiação , Humanos , Doses de Radiação
15.
Cancer Res ; 66(7): 3428-33, 2006 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-16585164

RESUMO

The hereditary form of retinoblastoma (Rb) is associated with a germ line mutation in one RB allele and is characterized by the occurrence of multiple, bilateral Rb tumors and a predisposition to the development of second cancers. In an earlier study, we observed an unexpected hypersensitivity to ionizing radiation in skin fibroblasts derived from unaffected parents of children with hereditary Rb. In at least four of these five families, there was no family history of Rb, indicating a new germ line mutation. We hypothesize that the increased parental cell sensitivity to radiation may reflect the presence of an as yet unrecognized genetic abnormality occurring in one or both parents of children with Rb. In the present study, we use DNA microarray technology to determine whether differences in gene expression profiles occurred in the unaffected parents of patients with hereditary Rb relative to normal individuals. Microarray analyses were validated by quantitative reverse transcription-PCR measurements. A distinct difference was observed in the patterns of gene expression between unaffected Rb parents and normal controls. By use of the prediction analysis for microarrays and principal component analysis methodologies, significant differences between the two groups were identified when as few as nine genes were analyzed. Further study of this phenomenon may offer a new insight into the genetic mechanisms of Rb and perhaps more broadly in cancer biology.


Assuntos
Pais , Neoplasias da Retina/genética , Retinoblastoma/genética , Adulto , Criança , Fibroblastos/efeitos da radiação , Perfilação da Expressão Gênica , Mutação em Linhagem Germinativa , Humanos , Análise de Sequência com Séries de Oligonucleotídeos , RNA/genética , Tolerância a Radiação , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Pele/citologia
16.
Oncogene ; 37(16): 2150-2164, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29371679

RESUMO

Although ΔNp63 is known to promote cancer cell proliferation, the underlying mechanism behind its oncogenic function remains elusive. We report here a functional interplay between ΔNp63 and Δ133p53. These two proteins are co-overexpressed in a subset of human cancers and cooperate to promote cell proliferation. Mechanistically, Δ133p53 binds to ΔNp63 and utilizes its transactivation domain to upregulate GLUT1, GLUT4, and PGM expression driving glycolysis. While increased glycolysis provides cancer cells with anabolic metabolism critical for proliferation and survival, it can be harnessed for selective cancer cell killing. Indeed, we show that tumors overexpressing both ΔNp63 and Δ133p53 exhibit heightened sensitivity to vitamin C that accumulate to a lethal level due to accelerated uptake via overexpressed GLUT1. These observations offer a new therapeutic avenue that could be exploited for clinical applications.


Assuntos
Proliferação de Células/genética , Glicólise/genética , Neoplasias/patologia , Fatores de Transcrição/fisiologia , Proteína Supressora de Tumor p53/fisiologia , Proteínas Supressoras de Tumor/fisiologia , Células A549 , Animais , Metabolismo dos Carboidratos/genética , Células Cultivadas , Códon sem Sentido , Feminino , Células HEK293 , Células HeLa , Células Hep G2 , Humanos , Células MCF-7 , Camundongos , Camundongos Nus , Proteínas Mutantes/genética , Proteínas Mutantes/fisiologia , Neoplasias/genética , Neoplasias/metabolismo , Isoformas de Proteínas/fisiologia , Fatores de Transcrição/genética , Proteína Supressora de Tumor p53/genética , Proteínas Supressoras de Tumor/genética
17.
J Mol Cell Biol ; 10(4): 273-284, 2018 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-30265334

RESUMO

ZBTB7A, a member of the POZ/BTB and Krüppel (POK) family of transcription factors, has been shown to have a context-dependent role in cancer development and progression. The role of ZBTB7A in estrogen receptor alpha (ERα)-positive breast cancer is largely unknown. Approximately 70% of breast cancers are classified as ERα-positive. ERα carries out the biological effects of estrogen and its expression level dictates response to endocrine therapies and prognosis for breast cancer patients. In this study, we find that ZBTB7A transcriptionally regulates ERα expression in ERα-positive breast cancer cell lines by binding to the ESR1 promoter leading to increased transcription of ERα. Inhibition of ZBTB7A in ERα-positive cells results in decreased estrogen responsiveness as demonstrated by diminished estrogen-response element-driven luciferase reporter activity, induction of estrogen target genes, and estrogen-stimulated growth. We also report that ERα potentiates ZBTB7A expression via a post-translational mechanism, suggesting the presence of a positive feedback loop between ZBTB7A and ERα, conferring sensitivity to estrogen in breast cancer. Clinically, we find that ZBTB7A and ERα are often co-expressed in breast cancers and that high ZBTB7A expression correlates with improved overall and relapse-free survival for breast cancer patients. Importantly, high ZBTB7A expression predicts a more favorable outcome for patients treated with endocrine therapies. Together, these findings demonstrate that ZBTB7A contributes to the transcriptional program maintaining ERα expression and potentially an endocrine therapy-responsive phenotype in breast cancer.


Assuntos
Neoplasias da Mama/genética , Proteínas de Ligação a DNA/genética , Receptor alfa de Estrogênio/genética , Regulação Neoplásica da Expressão Gênica , Fatores de Transcrição/genética , Animais , Mama/metabolismo , Mama/patologia , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Feminino , Humanos , Camundongos , Regiões Promotoras Genéticas , Ativação Transcricional
18.
Cancer Res ; 65(15): 6734-44, 2005 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-16061655

RESUMO

Radiation exposure is an important form of environmental carcinogen and has been associated with increased risk of breast cancer. Epigenetic events, especially those involving alterations in the breast stromal microenvironment, may play an important role in radiation-induced carcinogenesis but remain not well understood. We here show that human mammary stromal fibroblasts respond to protracted low-dose ionizing radiation exposures by displaying a senescence-like phenotype. Using a three-dimensional coculture system to model the interactions of different mammary cell types with their neighbors and with their environment, we provide a direct experimental proof that ionizing radiation-induced senescence-like fibroblasts significantly perturb the mammary stromal microenvironment, which is highlighted by impaired formation of pseudopodia networks due to marked cytoskeletal alterations in senescence-like fibroblasts and increased extracellular matrix degradation because of the up-regulation of multiple secreted matrix metalloproteinases. Within such a perturbed environment, mammary ductal morphogenesis is completely disrupted and epithelial cells instead grow into enlarged cystic structures, which further develop and become disorganized cell masses on inactivation of cellular death pathways. Breast carcinoma cells growing in such an environment are enabled to fully express their malignant potential as evidenced by the alpha6beta4 integrin/phosphatidylinositol 3-kinase/Akt/mammalian target of rapamycin pathway-dependent invasive growth. Our results suggest that ionizing radiation, in addition to causing gene mutations in epithelial cells, can contribute to breast carcinogenesis by perturbing the tissue microenvironment that leads to dysregulated cell-cell and cell-matrix interactions.


Assuntos
Neoplasias da Mama/etiologia , Mama/efeitos da radiação , Neoplasias Induzidas por Radiação/etiologia , Animais , Mama/citologia , Mama/enzimologia , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Senescência Celular/efeitos da radiação , Técnicas de Cocultura , Relação Dose-Resposta a Droga , Células Epiteliais/citologia , Células Epiteliais/efeitos da radiação , Matriz Extracelular/enzimologia , Feminino , Fibroblastos/citologia , Fibroblastos/efeitos da radiação , Humanos , Metaloproteinases da Matriz/metabolismo , Neoplasias Induzidas por Radiação/patologia , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Quinases/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-akt , Ratos , Células Estromais/citologia , Células Estromais/efeitos da radiação , Serina-Treonina Quinases TOR
19.
J Mol Cell Biol ; 9(2): 154-165, 2017 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-27927748

RESUMO

Deregulation of the tyrosine kinase signalling is often associated with tumour progression and drug resistance, but its underlying mechanisms are only partly understood. In this study, we investigated the effects of the receptor tyrosine kinase AXL on the stability of the MDMX-MDM2 heterocomplex and the activity of p53 in melanoma cells. Our data demonstrated that AXL overexpression or activation through growth arrest-specific 6 (Gas6) ligand stimulation increases MDMX and MDM2 protein levels and decreases p53 activity. Upon activation, AXL stabilizes MDMX through a post-translational modification that involves phosphorylation of MDMX on the phosphosite Ser314, leading to increased affinity between MDMX and MDM2 and favouring MDMX nuclear translocation. Ser314 phosphorylation can also protect MDMX from MDM2-mediated degradation, leading to stabilization of the MDMX-MDM2 complex. We identified CDK4/6 and p38 MAPK as the two kinases mediating AXL-induced modulation of the MDMX-MDM2 complex, and demonstrated that suppression of AXL, either through siRNA silencing or pharmacological inhibition, increases expression levels of p53 target genes P21, MDM2, and PUMA, improves p53 pathway response to chemotherapy, and sensitizes cells to both Cisplatin and Vemurafenib. Our findings offer an insight into a novel signalling axis linking AXL to p53 and provide a potentially druggable pathway to restore p53 function in melanoma.


Assuntos
Melanoma/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , Transdução de Sinais , Proteína Supressora de Tumor p53/metabolismo , Proteínas de Ciclo Celular , Linhagem Celular Tumoral , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , Cisplatino/farmacologia , Quinases Ciclina-Dependentes/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Humanos , Indóis/farmacologia , Melanoma/patologia , Fosforilação/efeitos dos fármacos , Fosfosserina/metabolismo , Estabilidade Proteica/efeitos dos fármacos , Transporte Proteico/efeitos dos fármacos , Proteólise/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Sulfonamidas/farmacologia , Transcrição Gênica/efeitos dos fármacos , Proteína Supressora de Tumor p53/genética , Vemurafenib , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Receptor Tirosina Quinase Axl
20.
Radiat Res ; 187(2): 259-267, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-28118114

RESUMO

The catalytic subunit of DNA dependent protein kinase (DNA-PKcs) and its kinase activity are critical for mediation of non-homologous end-joining (NHEJ) of DNA double-strand breaks (DSB) in mammalian cells after gamma-ray irradiation. Additionally, DNA-PKcs phosphorylations at the T2609 cluster and the S2056 cluster also affect DSB repair and cellular sensitivity to gamma radiation. Previously we reported that phosphorylations within these two regions affect not only NHEJ but also homologous recombination repair (HRR) dependent DSB repair. In this study, we further examine phenotypic effects on cells bearing various combinations of mutations within either or both regions. Effects studied included cell killing as well as chromosomal aberration induction after 0.5-8 Gy gamma-ray irradiation delivered to synchronized cells during the G0/G1 phase of the cell cycle. Blocking phosphorylation within the T2609 cluster was most critical regarding sensitization and depended on the number of available phosphorylation sites. It was also especially interesting that only one substitution of alanine in each of the two clusters separately abolished the restoration of wild-type sensitivity by DNA-PKcs. Similar patterns were seen for induction of chromosomal aberrations, reflecting their connection to cell killing. To study possible change in coordination between HRR and NHEJ directed repair in these DNA-PKcs mutant cell lines, we compared the induction of sister chromatid exchanges (SCEs) by very low fluencies of alpha particles with mutant cells defective in the HRR pathway that is required for induction of SCEs. Levels of true SCEs induced by very low fluence of alpha-particle irradiation normally seen in wild-type cells were only slightly decreased in the S2056 cluster mutants, but were completely abolished in the T2609 cluster mutants and were indistinguishable from levels seen in HRR deficient cells. Again, a single substitution in the S2056 together with a single substitution in the T2609 cluster abolished SCE formation and thus also effectively interferes with HRR.


Assuntos
Partículas alfa/efeitos adversos , Proteína Quinase Ativada por DNA/metabolismo , Fase G1/efeitos da radiação , Raios gama/efeitos adversos , Fase de Repouso do Ciclo Celular/efeitos da radiação , Serina/metabolismo , Treonina/metabolismo , Animais , Células CHO , Aberrações Cromossômicas/efeitos da radiação , Cricetinae , Cricetulus , Proteína Quinase Ativada por DNA/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA