Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Nature ; 580(7803): 402-408, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32296183

RESUMEN

Global insights into cellular organization and genome function require comprehensive understanding of the interactome networks that mediate genotype-phenotype relationships1,2. Here we present a human 'all-by-all' reference interactome map of human binary protein interactions, or 'HuRI'. With approximately 53,000 protein-protein interactions, HuRI has approximately four times as many such interactions as there are high-quality curated interactions from small-scale studies. The integration of HuRI with genome3, transcriptome4 and proteome5 data enables cellular function to be studied within most physiological or pathological cellular contexts. We demonstrate the utility of HuRI in identifying the specific subcellular roles of protein-protein interactions. Inferred tissue-specific networks reveal general principles for the formation of cellular context-specific functions and elucidate potential molecular mechanisms that might underlie tissue-specific phenotypes of Mendelian diseases. HuRI is a systematic proteome-wide reference that links genomic variation to phenotypic outcomes.


Asunto(s)
Proteoma/metabolismo , Espacio Extracelular/metabolismo , Humanos , Especificidad de Órganos , Mapeo de Interacción de Proteínas
2.
Proc Natl Acad Sci U S A ; 118(41)2021 10 12.
Artículo en Inglés | MEDLINE | ID: mdl-34607954

RESUMEN

BRCA1 germline mutations are associated with an increased risk of breast and ovarian cancer. Recent findings of others suggest that BRCA1 mutation carriers also bear an increased risk of esophageal and gastric cancer. Here, we employ a Brca1/Trp53 mouse model to show that unresolved replication stress (RS) in BRCA1 heterozygous cells drives esophageal tumorigenesis in a model of the human equivalent. This model employs 4-nitroquinoline-1-oxide (4NQO) as an RS-inducing agent. Upon drinking 4NQO-containing water, Brca1 heterozygous mice formed squamous cell carcinomas of the distal esophagus and forestomach at a much higher frequency and speed (∼90 to 120 d) than did wild-type (WT) mice, which remained largely tumor free. Their esophageal tissue, but not that of WT control mice, revealed evidence of overt RS as reflected by intracellular CHK1 phosphorylation and 53BP1 staining. These Brca1 mutant tumors also revealed higher genome mutation rates than those of control animals; the mutational signature SBS4, which is associated with tobacco-induced tumorigenesis; and a loss of Brca1 heterozygosity (LOH). This uniquely accelerated Brca1 tumor model is also relevant to human esophageal squamous cell carcinoma, an often lethal tumor.


Asunto(s)
Proteína BRCA1/genética , Neoplasias Esofágicas/genética , Carcinoma de Células Escamosas de Esófago/genética , Pérdida de Heterocigocidad/genética , Proteína p53 Supresora de Tumor/genética , 4-Nitroquinolina-1-Óxido/toxicidad , Animales , Línea Celular Tumoral , Transformación Celular Neoplásica/genética , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1)/metabolismo , Modelos Animales de Enfermedad , Neoplasias Esofágicas/patología , Carcinoma de Células Escamosas de Esófago/inducido químicamente , Carcinoma de Células Escamosas de Esófago/patología , Femenino , Mutación de Línea Germinal/genética , Heterocigoto , Humanos , Pérdida de Heterocigocidad/efectos de los fármacos , Masculino , Ratones , Ratones Noqueados , Proteína 1 de Unión al Supresor Tumoral P53/metabolismo
3.
Mol Cell ; 54(6): 932-945, 2014 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-24857551

RESUMEN

Quiescence (G0) allows cycling cells to reversibly cease proliferation. A decision to enter quiescence is suspected of occurring early in G1, before the restriction point (R). Surprisingly, we have identified G2 as an interval during which inhibition of the protein phosphatase PP2A results in failure to exhibit stable quiescence. This effect is accompanied by shortening of the ensuing G1. The PP2A subcomplex required for stable G0 contains the B56γ B subunit. After PP2A inhibition in G2, aberrant overexpression of cyclin E occurs during mitosis and is responsible for overriding quiescence. Strikingly, suppression of Ras signaling re-establishes normal cyclin E levels during M and restores G0. These data point to PP2A-B56γ-driven Ras signaling modulation in G2 as essential for suppressing aberrant cyclin E expression during mitosis and thereby achieving normal G0 control. Thus, G2 is an interval during which the length and growth factor dependence of the next G1 interval are established.


Asunto(s)
Fase G1/genética , Fase G2/genética , Proteína Oncogénica p21(ras)/genética , Proteína Fosfatasa 2/genética , Fase de Descanso del Ciclo Celular/fisiología , Línea Celular , Ciclina E/biosíntesis , Humanos , Células MCF-7 , Mitosis/genética , Subunidades de Proteína/genética , Interferencia de ARN , ARN Interferente Pequeño , Transducción de Señal/genética
4.
Proc Natl Acad Sci U S A ; 112(1): 232-7, 2015 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-25535366

RESUMEN

High-grade serous ovarian carcinoma (HGSOC) is the most common and aggressive form of epithelial ovarian cancer, for which few targeted therapies exist. To search for new therapeutic target proteins, we performed an in vivo shRNA screen using an established human HGSOC cell line growing either subcutaneously or intraperitoneally in immunocompromised mice. We identified genes previously implicated in ovarian cancer such as AURKA1, ERBB3, CDK2, and mTOR, as well as several novel candidates including BRD4, VRK1, and GALK2. We confirmed, using both genetic and pharmacologic approaches, that the activity of BRD4, an epigenetic transcription modulator, is necessary for proliferation/survival of both an established human ovarian cancer cell line (OVCAR8) and a subset of primary serous ovarian cancer cell strains (DFs). Among the DFs tested, the strains sensitive to BRD4 inhibition revealed elevated expression of either MYCN or c-MYC, with MYCN expression correlating closely with JQ1 sensitivity. Accordingly, primary human xenografts derived from high-MYCN or c-MYC strains exhibited sensitivity to BRD4 inhibition. These data suggest that BRD4 inhibition represents a new therapeutic approach for MYC-overexpressing HGSOCs.


Asunto(s)
Pruebas Genéticas , Terapia Molecular Dirigida , Neoplasias Glandulares y Epiteliales/genética , Neoplasias Glandulares y Epiteliales/terapia , Proteínas Nucleares/metabolismo , Neoplasias Ováricas/genética , Neoplasias Ováricas/terapia , Factores de Transcripción/metabolismo , Animales , Carcinoma Epitelial de Ovario , Proteínas de Ciclo Celular , Línea Celular Tumoral , Proliferación Celular , Femenino , Estudios de Asociación Genética , Humanos , Ratones , Proteínas Proto-Oncogénicas c-myc/metabolismo , ARN Interferente Pequeño/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Proc Natl Acad Sci U S A ; 110(21): 8632-7, 2013 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-23657012

RESUMEN

Germ-line mutations in PALB2 lead to a familial predisposition to breast and pancreatic cancer or to Fanconi Anemia subtype N. PALB2 performs its tumor suppressor role, at least in part, by supporting homologous recombination-type double strand break repair (HR-DSBR) through physical interactions with BRCA1, BRCA2, and RAD51. To further understand the mechanisms underlying PALB2-mediated DNA repair and tumor suppression functions, we targeted Palb2 in the mouse. Palb2-deficient murine ES cells recapitulated DNA damage defects caused by PALB2 depletion in human cells, and germ-line deletion of Palb2 led to early embryonic lethality. Somatic deletion of Palb2 driven by K14-Cre led to mammary tumor formation with long latency. Codeletion of both Palb2 and Tumor protein 53 (Trp53) accelerated mammary tumor formation. Like BRCA1 and BRCA2 mutant breast cancers, these tumors were defective in RAD51 focus formation, reflecting a defect in Palb2 HR-DSBR function, a strongly suspected contributor to Brca1, Brca2, and Palb2 mammary tumor development. However, unlike the case of Brca1-mutant cells, Trp53bp1 deletion failed to rescue the genomic instability of Palb2- or Brca2-mutant primary lymphocytes. Therefore, Palb2-driven DNA damage control is, in part, distinct from that executed by Brca1 and more similar to that of Brca2. The mechanisms underlying Palb2 mammary tumor suppression functions can now be explored genetically in vivo.


Asunto(s)
Neoplasias de la Mama/metabolismo , Neoplasias Mamarias Experimentales/metabolismo , Síndromes Neoplásicos Hereditarios/metabolismo , Proteínas Nucleares/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Animales , Proteína BRCA1/genética , Proteína BRCA1/metabolismo , Proteína BRCA2/genética , Proteína BRCA2/metabolismo , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Modelos Animales de Enfermedad , Proteína del Grupo de Complementación N de la Anemia de Fanconi , Femenino , Eliminación de Gen , Humanos , Neoplasias Mamarias Experimentales/genética , Neoplasias Mamarias Experimentales/patología , Ratones , Ratones Mutantes , Síndromes Neoplásicos Hereditarios/genética , Síndromes Neoplásicos Hereditarios/patología , Proteínas Nucleares/genética , Recombinasa Rad51/genética , Recombinasa Rad51/metabolismo , Proteína p53 Supresora de Tumor/genética , Proteínas Supresoras de Tumor/genética
6.
J Biol Chem ; 289(35): 24617-29, 2014 Aug 29.
Artículo en Inglés | MEDLINE | ID: mdl-25016020

RESUMEN

PALB2 links BRCA1 and BRCA2 in homologous recombinational repair of DNA double strand breaks (DSBs). Mono-allelic mutations in PALB2 increase the risk of breast, pancreatic, and other cancers, and biallelic mutations cause Fanconi anemia (FA). Like Brca1 and Brca2, systemic knock-out of Palb2 in mice results in embryonic lethality. In this study, we generated a hypomorphic Palb2 allele expressing a mutant PALB2 protein unable to bind BRCA1. Consistent with an FA-like phenotype, cells from the mutant mice showed hypersensitivity and chromosomal breakage when treated with mitomycin C, a DNA interstrand crosslinker. Moreover, mutant males showed reduced fertility due to impaired meiosis and increased apoptosis in germ cells. Interestingly, mutant meiocytes showed a significant defect in sex chromosome synapsis, which likely contributed to the germ cell loss and fertility defect. Our results underscore the in vivo importance of the PALB2-BRCA1 complex formation in DSB repair and male meiosis.


Asunto(s)
Proteína BRCA1/metabolismo , Infertilidad Masculina/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Secuencia de Aminoácidos , Animales , Proteína BRCA1/química , Daño del ADN , Reparación del ADN , Proteína del Grupo de Complementación N de la Anemia de Fanconi , Recombinación Homóloga , Humanos , Etiquetado Corte-Fin in Situ , Infertilidad Masculina/genética , Masculino , Ratones , Datos de Secuencia Molecular , Homología de Secuencia de Aminoácido , Proteínas Supresoras de Tumor/química
7.
Proc Natl Acad Sci U S A ; 106(4): 1122-7, 2009 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-19164542

RESUMEN

Dicer is central to the RNA interference (RNAi) pathway, because it is required for processing of double-stranded RNA (dsRNA) precursors into small RNA effector molecules. In principle, any long dsRNA could serve as a substrate for Dicer. The X inactive specific transcript (Xist) is an untranslated RNA that is required for dosage compensation in mammals. It coats and silences 1 of the 2 X chromosomes in female cells and initiates a chromosomewide change in chromatin structure that includes the recruitment of Polycomb proteins, but it is largely unknown how Xist RNA mediates these processes. To investigate a potential link between the RNAi pathway and X inactivation, we generated and analyzed Dicer-deficient embryonic stem (ES) cells. In the absence of Dicer, coating by Xist RNA, initiation of silencing, and recruitment of Polycomb proteins occur normally. Dicer ablation had modest effects on the steady-state levels of spliced Xist RNA. Together our data indicate that the RNAi machinery is not essential for the initiation of X inactivation.


Asunto(s)
ARN Helicasas DEAD-box/deficiencia , Endorribonucleasas/deficiencia , Inactivación del Cromosoma X/genética , Animales , Cruzamientos Genéticos , ARN Helicasas DEAD-box/metabolismo , Doxiciclina/farmacología , Células Madre Embrionarias/citología , Células Madre Embrionarias/efectos de los fármacos , Células Madre Embrionarias/metabolismo , Endorribonucleasas/metabolismo , Femenino , Silenciador del Gen/efectos de los fármacos , Genes Ligados a X , Endogamia , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Genéticos , Proteínas del Grupo Polycomb , ARN Largo no Codificante , ARN no Traducido/metabolismo , Proteínas Represoras/metabolismo , Ribonucleasa III , Tetraciclina/farmacología , Cromosoma X/genética , Inactivación del Cromosoma X/efectos de los fármacos
8.
World J Gastroenterol ; 28(29): 3886-3902, 2022 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-36157534

RESUMEN

BACKGROUND: The high prevalence and persistence of Helicobacter pylori (H. pylori) infection, as well as the diversity of pathologies related to it, suggest that the virulence factors used by this microorganism are varied. Moreover, as its proteome contains 340 hypothetical proteins, it is important to investigate them to completely understand the mechanisms of its virulence and survival. We have previously reported that the hypothetical protein HP0953 is overexpressed during the first hours of adhesion to inert surfaces, under stress conditions, suggesting its role in the environmental survival of this bacterium and perhaps as a virulence factor. AIM: To investigate the expression and localization of HP0953 during adhesion to an inert surface and against gastric (AGS) cells. METHODS: Expression analysis was performed for HP0953 during H. pylori adhesion. HP0953 expression at 0, 3, 12, 24, and 48 h was evaluated and compared using the Kruskal-Wallis equality-of-populations rank test. Recombinant protein was produced and used to obtain polyclonal antibodies for immunolocalization. Immunogold technique was performed on bacterial sections during adherence to inert surfaces and AGS cells, which was analyzed by transmission electron microscopy. HP0953 protein sequence was analyzed to predict the presence of a signal peptide and transmembrane helices, both provided by the ExPASy platform, and using the GLYCOPP platform for glycosylation sites. Different programs, via, I-TASSER, RaptorX, and HHalign-Kbest, were used to perform three-dimensional modeling. RESULTS: HP0953 exhibited its maximum expression at 12 h of infection in gastric epithelium cells. Immunogold technique revealed HP0953 localization in the cytoplasm and accumulation in some peripheral areas of the bacterial body, with greater expression when it is close to AGS cells. Bioinformatics analysis revealed the presence of a signal peptide that interacts with the transmembrane region and then allows the release of the protein to the external environment. The programs also showed a similarity with the Tip-alpha protein of H. pylori. Tip-alpha is an exotoxin that penetrates cells and induces tumor necrosis factor alpha production, and HP0953 could have a similar function as posttranslational modification sites were found; modifications in turn require enzymes located in eukaryotic cells. Thus, to be functional, HP0953 may necessarily need to be translocated inside the cell where it can trigger different mechanisms producing cellular damage. CONCLUSION: The location of HP0953 around infected cells, the probable posttranslational modifications, and its similarity to an exotoxin suggest that this protein is a virulence factor.


Asunto(s)
Infecciones por Helicobacter , Helicobacter pylori , Proteínas Bacterianas/metabolismo , Células Epiteliales/metabolismo , Epitelio/metabolismo , Exotoxinas/metabolismo , Mucosa Gástrica/patología , Infecciones por Helicobacter/microbiología , Humanos , Señales de Clasificación de Proteína , Proteoma/metabolismo , Proteínas Recombinantes/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Factores de Virulencia/metabolismo
10.
Sci Rep ; 9(1): 3952, 2019 03 08.
Artículo en Inglés | MEDLINE | ID: mdl-30850678

RESUMEN

Chromatin remodeler proteins exert an important function in promoting dynamic modifications in the chromatin architecture, performing a central role in regulating gene transcription. Deregulation of these molecular machines may lead to striking perturbations in normal cell function. The CHD7 gene is a member of the chromodomain helicase DNA-binding family and, when mutated, has been shown to be the cause of the CHARGE syndrome, a severe developmental human disorder. Moreover, CHD7 has been described to be essential for neural stem cells and it is also highly expressed or mutated in a number of human cancers. However, its potential role in glioblastoma has not yet been tested. Here, we show that CHD7 is up-regulated in human glioma tissues and we demonstrate that CHD7 knockout (KO) in LN-229 glioblastoma cells suppresses anchorage-independent growth and spheroid invasion in vitro. Additionally, CHD7 KO impairs tumor growth and increases overall survival in an orthotopic mouse xenograft model. Conversely, ectopic overexpression of CHD7 in LN-428 and A172 glioblastoma cell lines increases cell motility and invasiveness in vitro and promotes LN-428 tumor growth in vivo. Finally, RNA-seq analysis revealed that CHD7 modulates a specific transcriptional signature of invasion-related target genes. Further studies should explore clinical-translational implications for glioblastoma treatment.


Asunto(s)
Movimiento Celular , ADN Helicasas/fisiología , Proteínas de Unión al ADN/fisiología , Glioblastoma/patología , Animales , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Glioblastoma/metabolismo , Humanos , Ratones , Ratones Desnudos , Invasividad Neoplásica , Trasplante de Neoplasias
11.
Cancer Discov ; 9(4): 526-545, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30709805

RESUMEN

Although the majority of BRAF-mutant melanomas respond to BRAF/MEK inhibitors, these agents are not typically curative. Moreover, they are largely ineffective in NRAS- and NF1-mutant tumors. Here we report that genetic and chemical suppression of HDAC3 potently cooperates with MAPK pathway inhibitors in all three RAS pathway-driven tumors. Specifically, we show that entinostat dramatically enhances tumor regression when combined with BRAF/MEK inhibitors, in both models that are sensitive or relatively resistant to these agents. Interestingly, MGMT expression predicts responsiveness and marks tumors with latent defects in DNA repair. BRAF/MEK inhibitors enhance these defects by suppressing homologous recombination genes, inducing a BRCA-like state; however, addition of entinostat triggers the concomitant suppression of nonhomologous end-joining genes, resulting in a chemical synthetic lethality caused by excessive DNA damage. Together, these studies identify melanomas with latent DNA repair defects, describe a promising drug combination that capitalizes on these defects, and reveal a tractable therapeutic biomarker. SIGNIFICANCE: BRAF/MEK inhibitors are not typically curative in BRAF-mutant melanomas and are ineffective in NRAS- and NF1-mutant tumors. We show that HDAC inhibitors dramatically enhance the efficacy of BRAF/MEK inhibitors in sensitive and insensitive RAS pathway-driven melanomas by coordinately suppressing two DNA repair pathways, and identify a clinical biomarker that predicts responsiveness.See related commentary by Lombard et al., p. 469.This article is highlighted in the In This Issue feature, p. 453.


Asunto(s)
Reparación del ADN/genética , Genes ras/genética , Quinasas Quinasa Quinasa PAM/genética , Melanoma/genética , Humanos , Proteínas Proto-Oncogénicas B-raf
12.
Cancer Res ; 65(21): 9651-8, 2005 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-16266984

RESUMEN

Pheochromocytomas are catecholamine-secreting tumors that result from mutations of at least six different genes as components of distinct autosomal dominant disorders. However, there remain familial occurrences of pheochromocytoma without a known genetic defect. We describe here a familial pheochromocytoma syndrome consistent with digenic inheritance identified through a combination of global genomics strategies. Multipoint parametric linkage analysis revealed identical LOD scores of 2.97 for chromosome 2cen and 16p13 loci. A two-locus parametric linkage analysis produced maximum LOD score of 5.16 under a double recessive multiplicative model, suggesting that both loci are required to develop the disease. Allele-specific loss of heterozygosity (LOH) was detected only at the chromosome 2 locus in all tumors from this family, consistent with a tumor suppressor gene. Four additional pheochromocytomas with a similar genetic pattern were identified through transcription profiling and helped refine the chromosome 2 locus. High-density LOH mapping with single nucleotide polymorphism-based array identified a total of 18 of 62 pheochromocytomas with LOH within the chromosome 2 region, which further narrowed down the locus to <2 cM. This finding provides evidence for two novel susceptibility loci for pheochromocytoma and adds a recessive digenic trait to the increasingly broad genetic heterogeneity of these tumors. Similarly, complex traits may also be involved in other familial cancer syndromes.


Asunto(s)
Neoplasias de las Glándulas Suprarrenales/genética , Feocromocitoma/genética , Adulto , Cromosomas Humanos Par 16 , Cromosomas Humanos Par 2 , Femenino , Perfilación de la Expresión Génica , Predisposición Genética a la Enfermedad , Genoma Humano , Genómica/métodos , Humanos , Pérdida de Heterocigocidad , Masculino , Persona de Mediana Edad , Linaje
13.
Cancer Res ; 65(5): 1693-9, 2005 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-15753364

RESUMEN

A detailed genome mapping analysis of 213,636 expressed sequence tags (EST) derived from nontumor and tumor tissues of the oral cavity, larynx, pharynx, and thyroid was done. Transcripts matching known human genes were identified; potential new splice variants were flagged and subjected to manual curation, pointing to 788 putatively new alternative splicing isoforms, the majority (75%) being insertion events. A subset of 34 new splicing isoforms (5% of 788 events) was selected and 23 (68%) were confirmed by reverse transcription-PCR and DNA sequencing. Putative new genes were revealed, including six transcripts mapped to well-studied chromosomes such as 22, as well as transcripts that mapped to 253 intergenic regions. In addition, 2,251 noncoding intronic RNAs, eventually involved in transcriptional regulation, were found. A set of 250 candidate markers for loss of heterozygosis or gene amplification was selected by identifying transcripts that mapped to genomic regions previously known to be frequently amplified or deleted in head, neck, and thyroid tumors. Three of these markers were evaluated by quantitative reverse transcription-PCR in an independent set of individual samples. Along with detailed clinical data about tumor origin, the information reported here is now publicly available on a dedicated Web site as a resource for further biological investigation. This first in silico reconstruction of the head, neck, and thyroid transcriptomes points to a wealth of new candidate markers that can be used for future studies on the molecular basis of these tumors. Similar analysis is warranted for a number of other tumors for which large EST data sets are available.


Asunto(s)
Perfilación de la Expresión Génica , Marcadores Genéticos , Neoplasias de Cabeza y Cuello/genética , ARN Mensajero/genética , Neoplasias de la Tiroides/genética , Transcripción Genética , Empalme Alternativo , Etiquetas de Secuencia Expresada , Neoplasias de Cabeza y Cuello/metabolismo , Humanos , Laringe/metabolismo , Boca/metabolismo , Faringe/metabolismo , Reacción en Cadena de la Polimerasa , Isoformas de Proteínas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Análisis de Secuencia de ADN , Glándula Tiroides/metabolismo , Neoplasias de la Tiroides/metabolismo
14.
Mol Biotechnol ; 58(6): 404-14, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-27126696

RESUMEN

Recombinant human factor VIII (rFVIII) is used in replacement therapy for hemophilia A. Current research efforts are focused on bioengineering rFVIII molecules to improve its secretion efficiency and stability, limiting factors for its efficient production. However, high expression yield in mammalian cells of these rFVIII variants is generally associated with limited proteolytic processing. Non-processed single-chain polypeptides constitute non-natural FVIII molecule configurations with unpredictable toxicity and/or antigenicity. Our main objective was to demonstrate the feasibility of promoting full-proteolytic processing of an rFVIII variant retaining a portion of the B-domain, converting it into the smallest natural activatable form of rFVIII, while keeping its main advantage, i.e., improved secretion efficiency. We generated and employed a CHO-DG44 cell clone producing an rFVIII variant retaining a portion of the B-domain and the FVIII native cleavage site between Arg(1648) and Glu(1649). By bioengineering CHO-DG44 cells to express stably the recombinant human endoproteases PACE, PACE-SOL, PCSK5, PCSK6, or PCKS7, we were able to achieve complete intra- or extracellular proteolytic processing of this rFVIII variant. Additionally, our quantitative data indicated that removal of the B-domain segment by intracellular proteolytic processing does not interfere with this rFVIII variant secretion efficiency. This work also provides the first direct evidence of (1) intracellular cleavage at the Arg(1648) FVIII processing site promoted by wild-type PACE and PCSK7 and (2) proteolytic processing at the Arg(1648) FVIII processing site by PCSK6.


Asunto(s)
Factor VIII/química , Factor VIII/metabolismo , Furina/metabolismo , Animales , Células CHO , Cricetulus , Factor VIII/genética , Humanos , Proproteína Convertasas/metabolismo , Proteolisis , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Serina Endopeptidasas/metabolismo , Subtilisinas/metabolismo
15.
Brain Res Mol Brain Res ; 140(1-2): 25-33, 2005 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-16084624

RESUMEN

Diffuse infiltrating gliomas are the most common tumors of the central nervous system (CNS), naturally progressing from a lower-grade to a higher-grade malignancy. Several genetic alterations have been correlated with astrocytic tumors; however, a number of as yet unknown genes may also be involved. Therefore, we set out to search for genes that are differentially expressed in anaplastic astrocytoma and normal CNS tissue by applying a PCR-based subtractive hybridization approach, namely, representational difference analysis (RDA). The results of DNA sequencing of a sample (96 cDNA clones) from the subtracted library allowed the identification of 18 different genes, some of which were represented by several cDNA clones, coding for the Np95, LMO1, FCGBP, DSCAM, and taxilin proteins. Quantitative real-time PCR analysis for five of these genes was performed using samples of astrocytic tumors of different grades, confirming their higher expression when compared to non-tumoral CNS tissue. Identification of differentially expressed genes present in gliomas but not in normal CNS tissue is important not only to better understand the molecular basis of these cancers, but also to generate diagnostic DNA chips, which may be useful in future therapeutic intervention.


Asunto(s)
Neoplasias del Sistema Nervioso Central/genética , ADN Complementario/genética , Regulación Neoplásica de la Expresión Génica , Glioma/genética , Secuencia de Bases , Cartilla de ADN , ADN de Neoplasias/genética , Humanos , Técnicas de Sonda Molecular , Proteínas de Neoplasias/genética , Reacción en Cadena de la Polimerasa/métodos , ARN Neoplásico/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transcripción Genética
16.
Neurosci Lett ; 380(3): 260-4, 2005 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-15862898

RESUMEN

The ADAMs comprises a family of cell surface proteins with putative roles in cell-cell and/or cell-matrix interactions and in protease activities. In this work, we have examined the expression level and the methylation status of the 5' upstream region of the adhesion molecule ADAM23 in two brain tumor cell lines (A172 and T98G) as well as in three primary brain tumors (one grade II astrocytoma and two meningiomas) and 15 glioblastoma xenografts. Using bisulfite sequencing we verified that the percentage of methylated dinucleotides is higher in T98G when compared to A172 and that methylation significantly correlates with ADAM23 mRNA and protein expression. However, we were unable to detect methylation and down-regulation of the ADAM23 gene in brain tumors. Together, these results indicate that ADAM23 down-regulation by methylation in brain tumors is a rare event and it may help explain why brain tumor metastases are rarely found elsewhere in the body.


Asunto(s)
Astrocitoma/genética , Neoplasias Encefálicas/genética , Metilación de ADN , Regulación Neoplásica de la Expresión Génica/genética , Glioblastoma/genética , Glicoproteínas de Membrana/genética , Meningioma/genética , Metaloendopeptidasas/genética , Proteínas ADAM , Animales , Astrocitoma/metabolismo , Neoplasias Encefálicas/metabolismo , Línea Celular Tumoral , Regulación hacia Abajo/genética , Glioblastoma/metabolismo , Humanos , Glicoproteínas de Membrana/metabolismo , Meningioma/metabolismo , Metaloendopeptidasas/metabolismo , Ratones , Ratones Desnudos , Nucleótidos/metabolismo , ARN Mensajero/metabolismo
17.
Oncotarget ; 6(32): 33120-33, 2015 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-26431549

RESUMEN

Glioblastoma multiforme is the most common and lethal of the central nervous system glial-derived tumors. RECK suppresses tumor invasion by negatively regulating at least three members of the matrix metalloproteinase family: MMP-9, MMP-2, and MT1-MMP. A positive correlation has been observed between the abundance of RECK expression in tumor samples and a more favorable prognosis for patients with several types of tumors. In the present study, novel alternatively spliced variants of the RECK gene: RECK-B and RECK-I were isolated by RT-PCR and sequenced. The expression levels and profiles of these alternative RECK transcripts, as well as canonical RECK were determined in tissue samples of malignant astrocytomas of different grades and in a normal tissue RNA panel by qRT-PCR. Our results show that higher canonical RECK expression, accompanied by a higher canonical to alternative transcript expression ratio, positively correlates with higher overall survival rate after chemotherapeutic treatment of GBM patients. U87MG and T98G cells over-expressing the RECK-B alternative variant display higher anchorage-independent clonal growth and do not display modulation of, respectively, MMP-2 and MMP-9 expression. Our findings suggest that RECK transcript variants might have opposite roles in GBM biology and the ratio of their expression levels may be informative for the prognostic outcome of GBM patients.


Asunto(s)
Neoplasias Encefálicas/genética , Proteínas Ligadas a GPI/genética , Glioblastoma/genética , Adulto , Neoplasias Encefálicas/metabolismo , Línea Celular Tumoral , Femenino , Proteínas Ligadas a GPI/metabolismo , Genes Supresores de Tumor , Glioblastoma/metabolismo , Humanos , Masculino , Metaloproteinasas de la Matriz/genética , Metaloproteinasas de la Matriz/metabolismo , Persona de Mediana Edad , Isoformas de Proteínas , Empalme del ARN
18.
J Endocrinol ; 183(3): 455-67, 2004 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15590972

RESUMEN

Strategies to differentiate progenitor cells into beta cells in vitro have been considered as an alternative to increase beta cell availability prior to transplantation. It has recently been suggested that nestin-positive cells could be multipotential stem cells capable of expressing endocrine markers upon specific stimulation; however, this issue still remains controversial. Here, we characterized short- and long-term islet cell cultures derived from three different human islet preparations, with respect to expression of nestin and islet cell markers, using confocal microscopy and semi-quantitative RT-PCR. The number of nestin-positive cells was found to be strikingly high in long-term cultures. In addition, a large proportion (49.7%) of these nestin-positive cells, present in long-term culture, are shown to be proliferative, as judged by BrdU incorporation. The proportion of insulin-positive cells was found to be high in short-term (up to 28 days) cultures and declined thereafter, when cells were maintained in the presence of 10% serum, concomitantly with the decrease in insulin and PDX-1 expression. Interestingly, insulin and nestin co-expression was observed as a rare event in a small proportion of cells present in freshly isolated human islets as well as in purified islet cells cultured in vitro for long periods of time. In addition, upon long-term subculturing of nestin-positive cells in 10% serum, we observed reappearance of insulin expression at the mRNA level; when these cultures were shifted to 1% serum for a month, expression of insulin, glucagon and somatostatin was also detected, indicating that manipulating the culture conditions can be used to modulate the nestin-positive cell's fate. Attempts to induce cell differentiation by plating nestin-positive cells onto Matrigel revealed that these cells tend to aggregate to form islet-like clusters, but this is not sufficient to increase insulin expression upon short-term culture. Our data corroborate previous findings indicating that, at least in vitro, nestin-positive cells may undergo the early stages of differentiation to an islet cell phenotype and that long-term cultures of nestin-positive human islet cells may be considered as a potential source of precursor cells to generate fully differentiated/ functional beta cells.


Asunto(s)
Insulina/análisis , Proteínas de Filamentos Intermediarios/análisis , Islotes Pancreáticos/química , Proteínas del Tejido Nervioso/análisis , Biomarcadores/análisis , Diferenciación Celular , Células Cultivadas , Colágeno , Medios de Cultivo , Combinación de Medicamentos , Proteínas de Homeodominio/análisis , Humanos , Inmunohistoquímica/métodos , Insulina/genética , Laminina , Microscopía Confocal , Nestina , Proteoglicanos , ARN Mensajero/análisis , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Madre/química , Factores de Tiempo , Transactivadores/análisis
19.
Nat Commun ; 5: 5496, 2014 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-25400221

RESUMEN

BRCA1-a breast and ovarian cancer suppressor gene-promotes genome integrity. To study the functionality of BRCA1 in the heterozygous state, we established a collection of primary human BRCA1(+/+) and BRCA1(mut/+) mammary epithelial cells and fibroblasts. Here we report that all BRCA1(mut/+) cells exhibited multiple normal BRCA1 functions, including the support of homologous recombination- type double-strand break repair (HR-DSBR), checkpoint functions, centrosome number control, spindle pole formation, Slug expression and satellite RNA suppression. In contrast, the same cells were defective in stalled replication fork repair and/or suppression of fork collapse, that is, replication stress. These defects were rescued by reconstituting BRCA1(mut/+) cells with wt BRCA1. In addition, we observed 'conditional' haploinsufficiency for HR-DSBR in BRCA1(mut/+) cells in the face of replication stress. Given the importance of replication stress in epithelial cancer development and of an HR defect in breast cancer pathogenesis, both defects are candidate contributors to tumorigenesis in BRCA1-deficient mammary tissue.


Asunto(s)
Replicación del ADN/fisiología , Genes BRCA1/fisiología , Haploinsuficiencia/fisiología , Animales , Mama/citología , Células Cultivadas , Centrosoma/fisiología , Replicación del ADN/genética , Femenino , Haploinsuficiencia/genética , Heterocigoto , Humanos , Ratones , Satélite de ARN/genética , Satélite de ARN/fisiología , Recombinasa Rad51/genética , Recombinasa Rad51/fisiología , Reparación del ADN por Recombinación/genética , Reparación del ADN por Recombinación/fisiología , Polos del Huso/genética , Polos del Huso/fisiología
20.
Cancer Discov ; 3(8): 894-907, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23650262

RESUMEN

Hereditary breast cancers stem from germline mutations in susceptibility genes such as BRCA1, BRCA2, and PALB2, whose products function in the DNA damage response and redox regulation. Autophagy is an intracellular waste disposal and stress mitigation mechanism important for alleviating oxidative stress and DNA damage response activation; it can either suppress or promote cancer, but its role in breast cancer is unknown. Here, we show that similar to Brca1 and Brca2, ablation of Palb2 in the mouse mammary gland resulted in tumor development with long latency, and the tumors harbored mutations in Trp53. Interestingly, impaired autophagy, due to monoallelic loss of the essential autophagy gene Becn1, reduced Palb2-associated mammary tumorigenesis in a Trp53-wild-type but not conditionally null background. These results indicate that, in the face of DNA damage and oxidative stress elicited by PALB2 loss, p53 is a barrier to cancer development, whereas autophagy facilitates cell survival and tumorigenesis.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/genética , Autofagia , Carcinogénesis , Neoplasias Mamarias Experimentales/patología , Proteína p53 Supresora de Tumor/metabolismo , Proteínas Supresoras de Tumor/genética , Animales , Apoptosis , Proteína BRCA1/genética , Proteína BRCA1/metabolismo , Proteína BRCA2/genética , Proteína BRCA2/metabolismo , Beclina-1 , Senescencia Celular , Daño del ADN , Modelos Animales de Enfermedad , Proteína del Grupo de Complementación N de la Anemia de Fanconi , Femenino , Heterocigoto , Humanos , Neoplasias Mamarias Experimentales/genética , Ratones , Ratones Noqueados , Estrés Oxidativo/genética , Estrés Oxidativo/fisiología , Proteína p53 Supresora de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA