Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Clin Cancer Res ; 24(1): 197-208, 2018 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-29051323

RESUMEN

Purpose: Epithelial-to-mesenchymal transition (EMT) confers resistance to a number of targeted therapies and chemotherapies. However, it has been unclear why EMT promotes resistance, thereby impairing progress to overcome it.Experimental Design: We have developed several models of EMT-mediated resistance to EGFR inhibitors (EGFRi) in EGFR-mutant lung cancers to evaluate a novel mechanism of EMT-mediated resistance.Results: We observed that mesenchymal EGFR-mutant lung cancers are resistant to EGFRi-induced apoptosis via insufficient expression of BIM, preventing cell death despite potent suppression of oncogenic signaling following EGFRi treatment. Mechanistically, we observed that the EMT transcription factor ZEB1 inhibits BIM expression by binding directly to the BIM promoter and repressing transcription. Derepression of BIM expression by depletion of ZEB1 or treatment with the BH3 mimetic ABT-263 to enhance "free" cellular BIM levels both led to resensitization of mesenchymal EGFR-mutant cancers to EGFRi. This relationship between EMT and loss of BIM is not restricted to EGFR-mutant lung cancers, as it was also observed in KRAS-mutant lung cancers and large datasets, including different cancer subtypes.Conclusions: Altogether, these data reveal a novel mechanistic link between EMT and resistance to lung cancer targeted therapies. Clin Cancer Res; 24(1); 197-208. ©2017 AACR.


Asunto(s)
Proteína 11 Similar a Bcl2/genética , Transición Epitelial-Mesenquimal/efectos de los fármacos , Transición Epitelial-Mesenquimal/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética , Terapia Molecular Dirigida , Compuestos de Anilina/farmacología , Animales , Apoptosis/genética , Ciclo Celular/genética , Modelos Animales de Enfermedad , Resistencia a Antineoplásicos , Receptores ErbB/genética , Humanos , Ratones , Mutación , Regiones Promotoras Genéticas , Inhibidores de Proteínas Quinasas/farmacología , ARN Interferente Pequeño/genética , Sulfonamidas/farmacología
2.
Cancer Cell ; 29(2): 159-72, 2016 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-26859456

RESUMEN

Fewer than half of children with high-risk neuroblastoma survive. Many of these tumors harbor high-level amplification of MYCN, which correlates with poor disease outcome. Using data from our large drug screen we predicted, and subsequently demonstrated, that MYCN-amplified neuroblastomas are sensitive to the BCL-2 inhibitor ABT-199. This sensitivity occurs in part through low anti-apoptotic BCL-xL expression, high pro-apoptotic NOXA expression, and paradoxical, MYCN-driven upregulation of NOXA. Screening for enhancers of ABT-199 sensitivity in MYCN-amplified neuroblastomas, we demonstrate that the Aurora Kinase A inhibitor MLN8237 combines with ABT-199 to induce widespread apoptosis. In diverse models of MYCN-amplified neuroblastoma, including a patient-derived xenograft model, this combination uniformly induced tumor shrinkage, and in multiple instances led to complete tumor regression.


Asunto(s)
Apoptosis/genética , Neuroblastoma/tratamiento farmacológico , Compuestos de Anilina/uso terapéutico , Antineoplásicos/uso terapéutico , Línea Celular Tumoral , Humanos , Proteína Proto-Oncogénica N-Myc , Neuroblastoma/genética , Neuroblastoma/patología , Proteínas Nucleares , Proteínas Oncogénicas , Sulfonamidas/uso terapéutico
3.
Mol Cancer Res ; 14(1): 66-77, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26385560

RESUMEN

UNLABELLED: The activity of mammalian target of rapamycin complex 1 (mTORC1) is frequently enhanced in carcinomas, an effect thought to contribute to the malignant phenotype. Here, it is demonstrated that either deletion or mutation of TP53 in colon or lung carcinoma cells substantially enhances mTORC1 kinase activity by an effect downstream of and independent of AMPK. Mechanistically, it was determined that loss or mutation of p53 decreased expression of TSC2 and Sestrin2 (SESN2). Complementation of p53 null cells with TSC2 or Sestrin2 reduced mTORC1 activity to levels found in p53 wild-type (wt) cells, whereas their genetic depletion enhanced mTORC1 activity in p53 wt cells. However, the primary causal event in enhanced mTORC1 activity upon loss of p53 appeared to be a diminished distribution of TSC2 to lysosomal membranes containing mTOR. Subsequently, there was increased Rheb in the lysosomal compartment, and a higher mTOR association with Raptor. Transfection of TSC2 into p53 null cells replaced TSC2 and diminished Rheb at the lysosome, recapitulating cells with wt p53. In contrast, transfection of Sestrin2 decreased mTOR in lysosomes, but the lower levels of Sestrin2 in p53 null cells did not change lysosomal mTOR. In summary, loss of the transcriptional activity of p53, either by deletion or by key mutations in the DNA-binding domain, diminishes expression of TSC2 and Sestrin2, thus, shifting membrane-bound TSC2 out of lysosomal membranes, increasing lysosomal Rheb and increasing the kinase activity of mTORC1. IMPLICATIONS: This study establishes that loss of p53 function decreases lysosomal TSC2 and increases lysosomal Rheb resulting in hyperactive mTORC1, findings that are consistent with a more malignant phenotype.


Asunto(s)
Lisosomas/metabolismo , Proteínas de Unión al GTP Monoméricas/metabolismo , Complejos Multiproteicos/metabolismo , Neuropéptidos/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Proteína p53 Supresora de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo , Eliminación de Gen , Regulación Neoplásica de la Expresión Génica , Células HCT116 , Humanos , Lisosomas/genética , Diana Mecanicista del Complejo 1 de la Rapamicina , Proteínas de Unión al GTP Monoméricas/genética , Complejos Multiproteicos/genética , Mutación , Neuropéptidos/genética , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteína Homóloga de Ras Enriquecida en el Cerebro , Serina-Treonina Quinasas TOR/genética , Proteína 2 del Complejo de la Esclerosis Tuberosa , Proteínas Supresoras de Tumor/genética
4.
PLoS One ; 10(6): e0129647, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26053857

RESUMEN

The transcriptional coactivator, PGC-1α, is known for its role in mitochondrial biogenesis. Although originally thought to exist as a single protein isoform, recent studies have identified additional promoters which produce multiple mRNA transcripts. One of these promoters (promoter B), approximately 13.7 kb upstream of the canonical PGC-1α promoter (promoter A), yields alternative transcripts present at levels much lower than the canonical PGC-1α mRNA transcript. In skeletal muscle, exercise resulted in a substantial, rapid increase of mRNA of these alternative PGC-1α transcripts. Although the ß2-adrenergic receptor was identified as a signaling pathway that activates transcription from PGC-1α promoter B, it is not yet known what molecular changes occur to facilitate PGC-1α promoter B activation following exercise. We sought to determine whether epigenetic modifications were involved in this exercise response in mouse skeletal muscle. We found that DNA hydroxymethylation correlated to increased basal mRNA levels from PGC-1α promoter A, but that DNA methylation appeared to play no role in the exercise-induced activation of PGC-1α promoter B. The level of the activating histone mark H3K4me3 increased with exercise 2-4 fold across PGC-1α promoter B, but remained unaltered past the canonical PGC-1α transcriptional start site. Together, these data show that epigenetic modifications partially explain exercise-induced changes in the skeletal muscle mRNA levels of PGC-1α isoforms.


Asunto(s)
Epigénesis Genética , Regulación de la Expresión Génica , Condicionamiento Físico Animal , Regiones Promotoras Genéticas , Factores de Transcripción/genética , Empalme Alternativo , Animales , Metilación de ADN , Exones , Femenino , Histonas/metabolismo , Ratones , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , ARN Mensajero/genética , Sitio de Iniciación de la Transcripción
5.
J Biol Chem ; 289(42): 29386-96, 2014 Oct 17.
Artículo en Inglés | MEDLINE | ID: mdl-25164808

RESUMEN

Folylpoly-γ-glutamate synthetase (FPGS) catalyze the addition of multiple glutamates to tetrahydrofolate derivatives. Two mRNAs for the fpgs gene direct isoforms of FPGS to the cytosol and to mitochondria in mouse and human tissues. We sought to clarify the functions of these two compartmentalized isoforms. Stable cell lines were created that express cDNAs for the mitochondrial and cytosolic isoforms of human FPGS under control of a doxycycline-inducible promoter in the AUXB1 cell line. AUXB1 are devoid of endogenous FPGS activity due to a premature translational stop at codon 432 in the fpgs gene. Loss of folates was not measurable from these doxycycline-induced cells or from parental CHO cells over the course of three CHO cell generations. Likewise, there was no detectable transfer of folate polyglutamates either from the cytosol to mitochondria, or from mitochondria to the cytosol. The cell line expressing cytosolic FPGS required exogenous glycine but not thymidine or purine, whereas cells expressing the mitochondrial isoform required exogenous thymidine and purine but not glycine for optimal growth and survival. We concluded that mitochondrial FPGS is required because folate polyglutamates are not substrates for transport across the mitochondrial membrane in either direction and that polyglutamation not only traps folates in the cytosol, but also in the mitochondrial matrix.


Asunto(s)
Citosol/enzimología , Ácido Fólico/química , Mitocondrias/enzimología , Péptido Sintasas/metabolismo , Secuencia de Aminoácidos , Animales , Transporte Biológico , Células CHO , Cricetinae , Cricetulus , ADN Complementario/metabolismo , Humanos , Membranas Mitocondriales/metabolismo , Datos de Secuencia Molecular , Mutación , Isoformas de Proteínas/metabolismo , Homología de Secuencia de Aminoácido
6.
FASEB J ; 28(5): 1998-2008, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24532667

RESUMEN

The mouse is extensively used to model human folate metabolism and therapeutic outcomes with antifolates. However, the folylpoly-γ-glutamate synthetase (fpgs) gene, whose product determines folate/antifolate intracellular retention and antifolate antitumor activity, displays a pronounced species difference. The human gene uses only a single promoter, whereas the mouse uses two: P2, akin to the human promoter, at low levels in most tissues; and P1, an upstream promoter used extensively in liver and kidney. We deleted the mouse P1 promoter through homologous recombination to study the dual-promoter mouse system and to create a mouse with a humanized fpgs gene structure. Despite the loss of the predominant fpgs mRNA species in liver and kidney (representing 95 and 75% of fpgs transcripts in these tissues, respectively), P1-knockout mice developed and reproduced normally. The survival of these mice was explained by increased P2 transcription due to relief of transcriptional interference, by a 3-fold more efficient translation of P2-derived than P1-derived transcripts, and by 2-fold higher stability of P2-derived FPGS. In combination, all 3 effects reinstated FPGS function, even in liver. By eliminating mouse P1, we created a mouse model that mimicked the human housekeeping pattern of fpgs gene expression.


Asunto(s)
Ácido Fólico/metabolismo , Péptido Sintasas/genética , Regiones Promotoras Genéticas , Alelos , Secuencia de Aminoácidos , Animales , Antineoplásicos/farmacología , Secuencia de Bases , Células Madre Embrionarias/citología , Exones , Antagonistas del Ácido Fólico/farmacología , Eliminación de Gen , Perfilación de la Expresión Génica , Vectores Genéticos , Homocigoto , Humanos , Hígado/metabolismo , Ratones , Ratones Noqueados , Datos de Secuencia Molecular , Biosíntesis de Proteínas , Transporte de Proteínas , Recombinación Genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transcripción Genética
7.
Crit Care Nurse ; 31(4): 30-43, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21807682

RESUMEN

BACKGROUND: Hospital-acquired pressure ulcers are a common problem. Although a number of risk factors have been identified, relationships among risk profile characteristics and pressure ulcer outcomes have not been described in hospitalized patients. OBJECTIVES: To describe patients' characteristics and risk factors associated with pressure ulcer outcome. METHODS: A retrospective chart review was used to collect data on 87 patients in whom hospital-acquired pressure ulcers developed from May 2007 to November 2008. All pressure ulcers were staged by a certified wound nurse. Relationships among risk profile characteristics and pressure ulcer outcomes were determined via bivariate analysis and multivariate logistic regression. RESULTS: High severity of illness was present in patients with hospital-acquired pressure ulcers; 89% were intensive care patients. Vasopressor infusion, spinal cord injury, and age 40 or greater conferred risk for nonhealing pressure ulcers. Among pressure ulcer stages, suspected deep tissue injury ulcers were less likely to heal. CONCLUSIONS: Identification of characteristics and risk factors associated with development of nonhealing hospital-acquired pressure ulcers will allow nurses to recognize patients at risk for nonhealing and to take aggressive preventative measures.


Asunto(s)
Hospitalización , Evaluación de Resultado en la Atención de Salud , Úlcera por Presión/epidemiología , Cicatrización de Heridas/fisiología , Adulto , Anciano , Anciano de 80 o más Años , Cuidados Críticos , Femenino , Humanos , Masculino , Persona de Mediana Edad , Úlcera por Presión/enfermería , Estudios Retrospectivos , Factores de Riesgo , Índice de Severidad de la Enfermedad , Adulto Joven
8.
Proc Natl Acad Sci U S A ; 108(9): 3630-5, 2011 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-21321201

RESUMEN

Mitochondrial DNA (mtDNA) has been reported to contain 5-methylcytosine (5mC) at CpG dinucleotides, as in the nuclear genome, but neither the mechanism generating mtDNA methylation nor its functional significance is known. We now report the presence of 5-hydroxymethylcytosine (5hmC) as well as 5mC in mammalian mtDNA, suggesting that previous studies underestimated the level of cytosine modification in this genome. DNA methyltransferase 1 (DNMT1) translocates to the mitochondria, driven by a mitochondrial targeting sequence located immediately upstream of the commonly accepted translational start site. This targeting sequence is conserved across mammals, and the encoded peptide directs a heterologous protein to the mitochondria. DNMT1 is the only member of the three known catalytically active DNA methyltransferases targeted to the mitochondrion. Mitochondrial DNMT1 (mtDNMT1) binds to mtDNA, proving the presence of mtDNMT1 in the mitochondrial matrix. mtDNMT1 expression is up-regulated by NRF1 and PGC1α, transcription factors that activate expression of nuclear-encoded mitochondrial genes in response to hypoxia, and by loss of p53, a tumor suppressor known to regulate mitochondrial metabolism. Altered mtDNMT1 expression asymmetrically affects expression of transcripts from the heavy and light strands of mtDNA. Hence, mtDNMT1 appears to be responsible for mtDNA cytosine methylation, from which 5hmC is presumed to be derived, and its expression is controlled by factors that regulate mitochondrial function.


Asunto(s)
Citosina/análogos & derivados , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Metilación de ADN , Mitocondrias/enzimología , 5-Metilcitosina/análogos & derivados , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Compartimento Celular , Citosina/metabolismo , ADN (Citosina-5-)-Metiltransferasa 1 , ADN (Citosina-5-)-Metiltransferasas/química , ADN (Citosina-5-)-Metiltransferasas/genética , ADN Mitocondrial/metabolismo , Genes Mitocondriales/genética , Células HCT116 , Humanos , Ratones , Mitocondrias/genética , Datos de Secuencia Molecular , Estrés Oxidativo , Unión Proteica , Señales de Clasificación de Proteína , Transcripción Genética
9.
DNA Repair (Amst) ; 8(5): 654-63, 2009 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-19211312

RESUMEN

A homozygous H493R mutation in the active site of tyrosyl-DNA phosphodiesterase (TDP1) has been implicated in hereditary spinocerebellar ataxia with axonal neuropathy (SCAN1), an autosomal recessive neurodegenerative disease. However, it is uncertain how the H493R mutation elicits the specific pathologies of SCAN1. To address this question, and to further elucidate the role of TDP1 in repair of DNA end modifications and general physiology, we generated a Tdp1 knockout mouse and carried out detailed behavioral analyses as well as characterization of repair deficiencies in extracts of embryo fibroblasts from these animals. While Tdp1(-/-) mice appear phenotypically normal, extracts from Tdp1(-/-) fibroblasts exhibited deficiencies in processing 3'-phosphotyrosyl single-strand breaks and 3'-phosphoglycolate double-strand breaks (DSBs), but not 3'-phosphoglycolate single-strand breaks. Supplementing Tdp1(-/-) extracts with H493R TDP1 partially restored processing of 3'-phosphotyrosyl single-strand breaks, but with evidence of persistent covalent adducts between TDP1 and DNA, consistent with a proposed intermediate-stabilization effect of the SCAN1 mutation. However, H493R TDP1 supplementation had no effect on phosphoglycolate (PG) termini on 3' overhangs of double-strand breaks; these remained completely unprocessed. Altogether, these results suggest that for 3'-phosphoglycolate overhang lesions, the SCAN1 mutation confers loss of function, while for 3'-phosphotyrosyl lesions, the mutation uniquely stabilizes a reaction intermediate.


Asunto(s)
Aductos de ADN/química , Glicolatos/metabolismo , Mutación/genética , Hidrolasas Diéster Fosfóricas/fisiología , Fosfotirosina/metabolismo , Ataxias Espinocerebelosas/genética , Animales , Southern Blotting , Western Blotting , Catálisis , Aductos de ADN/genética , Aductos de ADN/metabolismo , Roturas del ADN de Doble Cadena , Roturas del ADN de Cadena Simple , Embrión de Mamíferos/citología , Embrión de Mamíferos/metabolismo , Femenino , Fibroblastos/citología , Fibroblastos/metabolismo , Prueba de Complementación Genética , Masculino , Ratones , Ratones Noqueados , Mutagénesis Sitio-Dirigida , Reacción en Cadena de la Polimerasa , Ataxias Espinocerebelosas/metabolismo
10.
Mol Cell Biol ; 28(2): 836-48, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17998333

RESUMEN

The mouse fpgs gene uses two distantly placed promoters to produce functionally distinct isozymes in a tissue-specific pattern. We queried how the P1 and P2 promoters were differentially controlled. DNA methylation of the CpG-sparse P1 promoter occurred only in tissues not initiating transcription at this site. The P2 promoter, which was embedded in a CpG island, appeared open to transcription in all tissues by several criteria, including lack of DNA methylation, yet was used only in dividing tissues. The patterns of histone modifications over the two promoters were very different: over P1, histone activation marks (acetylated histones H3 and H4 and H3 trimethylated at K4) reflected transcriptional activity and apparently reinforced the effects of hypomethylated CpGs; over P2, these marks were present in tissues whether P2 was active, inactive, or engaged in assembly of futile initiation complexes. Since P1 transcriptional activity coexisted with silencing of P2, we sought the mechanism of this transcriptional interference. We found RNA polymerase II, phosphorylated in a pattern consistent with transcriptional elongation, and only minimal levels of initiation factors over P2 in liver. We concluded that mouse fpgs uses DNA methylation to control tissue-specific expression from a CpG-sparse promoter, which is dominant over a downstream promoter masked by promoter occlusion.


Asunto(s)
Epigénesis Genética/genética , Transcripción Genética/genética , Acetilación , Animales , Cromatina/genética , Citosina/metabolismo , Metilación de ADN , Histonas/metabolismo , Hígado/enzimología , Ratones , Ratones Transgénicos , Especificidad de Órganos , Fosforilación , Regiones Promotoras Genéticas/genética , ARN Polimerasa II/metabolismo , Serina/genética , Serina/metabolismo
11.
Biochemistry ; 46(6): 1557-67, 2007 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-17279620

RESUMEN

The mitochondrial folate transporter (MFT) was previously identified in human and hamster cells. Sequence homology of this protein with the inner mitochondrial membrane transporters suggested a domain structure in which the N- and C-termini of the protein are located on the mitochondrial intermembrane-facing surface, with six membrane-spanning regions interspersed by two intermembrane loops and three matrix-facing loops. We now report the functional significance of insertion of the c-myc epitope into the intermembrane loops and of a series of site-directed mutations at hamster MFT residues highly conserved in orthologues. Insertional mutagenesis in the first predicted intermembrane loop eliminated MFT function, but the introduction of a c-myc peptide into the second loop was without effect. Most of the hamster MFT residues studied by site-directed mutagenesis were remarkably resilient to these mutations, except for R249A and G192E, both of which eliminated folate transport activity. Homology modeling, using the crystal structure of the bovine ADP/ATP carrier (AAC) as a scaffold, suggested a similar three-dimensional structure for the MFT and the AAC. An ion-pair interaction in the AAC thought to be central to the mechanism of membrane penetration by ADP is predicted by this homology model to be replaced by a pi-cation interaction in MFT orthologues and probably also in other members of the family bearing the P(I/L)W motif. This model suggests that the MFT R249A and G192E mutations both modify the base of a basket-shaped structure that appears to constitute a trap door for the flux of folates into the mitochondrial matrix.


Asunto(s)
Ácido Fólico/metabolismo , Proteínas de Transporte de Membrana Mitocondrial/metabolismo , Secuencia de Aminoácidos , Animales , Células CHO , Cricetinae , Cricetulus , Translocasas Mitocondriales de ADP y ATP/química , Translocasas Mitocondriales de ADP y ATP/metabolismo , Proteínas de Transporte de Membrana Mitocondrial/genética , Modelos Moleculares , Mutagénesis Sitio-Dirigida , Proteínas Proto-Oncogénicas c-myc/química , Proteínas Proto-Oncogénicas c-myc/metabolismo , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/metabolismo , Homología de Secuencia de Aminoácido
12.
Curr Mol Med ; 6(6): 645-50, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17022734

RESUMEN

Astrocytomas represent the most common form of glial tumors. The most malignant grade of these tumors, glioblastoma multiforme, may arise as a malignant progression from low-grade astrocytoma through anaplastic astrocytoma to secondary GBM, or else it may arise "de novo" as primary GBM. Both types of glioblastoma are usually histologically indistinguishable. However, distinct molecular alterations have been described between them that potentially allow differentiation between the two mechanisms of origin. Since malignant transformation is a multistep process, we summarize in this review the earliest genetic changes that seem to be involved in the appearance and development of low-grade astrocytic tumors, where early detection and treatment could be possible.


Asunto(s)
Astrocitoma/genética , Neoplasias Encefálicas/genética , Glioblastoma/genética , Modelos Genéticos , Proteínas Supresoras de Tumor/genética , Transformación Celular Neoplásica , Cromosomas Humanos Par 17 , Amplificación de Genes , Humanos , Mutación , Células Madre/patología
13.
J Biol Chem ; 279(32): 33829-36, 2004 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-15140890

RESUMEN

A mutant Chinese hamster ovary cell line, glyB, that required exogenous glycine for survival and growth was reported previously (Kao, F., Chasin, L., and Puck, T. T. (1969) Proc. Natl. Acad. Sci. U. S. A. 64, 1284-1291). We now report that the defect in glyB cells causative of this phenotype is a point mutation in an inner mitochondrial membrane protein required for transport of folates into mitochondria. The CHO mitochondrial folate transporter (mft) was sequenced and compared with that from glyB cells. The hamster sequence was nearly identical to that of the recently reported human mitochondrial folate transporter. The corresponding cDNA from glyB cells contained a single nucleotide change that introduced a glutamate in place of the glycine in wild-type hamster MFT at codon 192 in a predicted transmembrane domain. Transfection of the wild-type hamster cDNA into glyB cells allowed cell survival in the absence of glycine and the accumulation of folates in mitochondria, whereas transfection of the Glu-192 cDNA did not. Genomic sequence analysis and fluorescence in situ hybridization demonstrated a single mutated allele of the mft gene in glyB cells, whereas there were two alleles in CHO cells. We conclude that we have defined the cause of the glyB auxotrophy and that the glyB mft mutation identified a region of this mitochondrial folate carrier vital to its transport function.


Asunto(s)
Supervivencia Celular/fisiología , Ácido Fólico/metabolismo , Glicina/fisiología , Proteínas de Transporte de Membrana Mitocondrial/genética , Mutación Puntual , Alelos , Secuencia de Aminoácidos , Animales , Transporte Biológico , Células CHO , Codón , Cricetinae , ADN Complementario/genética , Expresión Génica , Ácido Glutámico , Humanos , Hibridación Fluorescente in Situ , Proteínas de Transporte de Membrana/química , Proteínas de Transporte de Membrana Mitocondrial/química , Proteínas de Transporte de Membrana Mitocondrial/fisiología , Datos de Secuencia Molecular , Estructura Molecular , Homología de Secuencia , Transfección
14.
Cancer Res ; 63(20): 6579-82, 2003 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-14583449

RESUMEN

Cytosine methylation patterns in genomic DNA are significantly altered in cancer, and de novo CpG island methylation has been implicated in tumor suppressor gene silencing. Here we demonstrate a mechanistic link between the p53 tumor suppressor gene and control of epigenetic regulation by genomic methylation. Deletion of p53in HCT116 human colon carcinoma cells and primary mouse astrocytes resulted in a 6-fold increase of DNA cytosine methyltransferase 1 (Dnmt1) mRNA and protein, suggesting relief of p53-mediated Dnmt1repression. A p53 consensus binding site in exon 1 of the human Dnmt1gene bound recombinant p53 in vitro and endogenous p53 in vivo in the absence of stimuli that activate p53, implying that p53 controls Dnmt1transcription through direct DNA binding. Interestingly, ionizing radiation or etoposide, both of which stabilize and activate p53, diminished p53 binding in chromatin immunoprecipitation assays, concomitant with a 5-fold increase in Dnmt1 levels. Our findings suggest that activation of p53 reduces binding and relieves transcriptional repression of the Dnmt1gene, whereas loss of p53, a frequent, early event in tumorigenesis, may significantly contribute to aberrant genomic methylation.


Asunto(s)
ADN (Citosina-5-)-Metiltransferasas/antagonistas & inhibidores , ADN (Citosina-5-)-Metiltransferasas/genética , ADN de Neoplasias/metabolismo , Proteína p53 Supresora de Tumor/fisiología , Animales , Sitios de Unión , ADN (Citosina-5-)-Metiltransferasa 1 , ADN (Citosina-5-)-Metiltransferasas/biosíntesis , ADN de Neoplasias/genética , Exones , Regulación Enzimológica de la Expresión Génica/fisiología , Regulación Neoplásica de la Expresión Génica/fisiología , Células HCT116 , Humanos , Ratones , Unión Proteica , Proteína p53 Supresora de Tumor/deficiencia , Proteína p53 Supresora de Tumor/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...