Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
J Immunol ; 193(8): 3914-24, 2014 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-25200950

RESUMEN

Thrombospondin-1 (TSP1) inhibits angiogenesis, in part, by interacting with the ubiquitous cell-surface receptor CD47. In endothelial cells, CD47 interacts directly with vascular endothelial growth factor receptor (VEGFR)-2, and TSP1 inhibits VEGFR2 phosphorylation and signaling by disrupting this association. We show that CD47 similarly associates with and regulates VEGFR2 in T cells. TSP1 inhibits phosphorylation of VEGFR2 and its downstream target Src in wild type but not in CD47-deficient human Jurkat and primary murine T cells. VEGFR2 signaling inhibits proliferation and TCR signaling in wild type T cells. However, ligation of CD47 by TSP1 or loss of CD47 expression reverses some inhibitory effects of VEGF on proliferation and T cell activation. We further found that VEGF and VEGFR2 expression are upregulated in CD47-deficient murine CD4(+) and human Jurkat T cells, and the resulting autocrine VEGFR2 signaling enhances proliferation and some TCR responses in the absence of CD47. Thus, CD47 signaling modulates the ability of VEGF to regulate proliferation and TCR signaling, and autocrine production of VEGF by T cells contributes to this regulation. This provides a mechanism to understand the context-dependent effects of TSP1 and VEGF on T cell activation, and reveals an important role for CD47 signaling in regulating T cell production of the major angiogenic factor VEGF.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Antígeno CD47/inmunología , Tolerancia Inmunológica , Trombospondina 1/inmunología , Factor A de Crecimiento Endotelial Vascular/inmunología , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Animales , Antígeno CD47/biosíntesis , Antígeno CD47/genética , Linfocitos T CD8-positivos/inmunología , Comunicación Celular/inmunología , Línea Celular Tumoral , Proliferación Celular , Humanos , Células Jurkat , Activación de Linfocitos/inmunología , Ratones , Ratones Noqueados , Neovascularización Patológica/inmunología , Fosforilación , Receptores de Antígenos de Linfocitos T/inmunología , Transducción de Señal/inmunología , Regulación hacia Arriba , Factor A de Crecimiento Endotelial Vascular/biosíntesis , Receptor 2 de Factores de Crecimiento Endotelial Vascular/biosíntesis , Familia-src Quinasas/metabolismo
2.
Proc Natl Acad Sci U S A ; 109(52): E3659-67, 2012 Dec 26.
Artículo en Inglés | MEDLINE | ID: mdl-23236152

RESUMEN

Cancers with Ras mutations represent a major therapeutic problem. Recent RNAi screens have uncovered multiple nononcogene addiction pathways that are necessary for the survival of Ras mutant cells. Here, we identify the evolutionarily conserved gene enhancer of rudimentary homolog (ERH), in which depletion causes greater toxicity in cancer cells with mutations in the small GTPase KRAS compared with KRAS WT cells. ERH interacts with the spliceosome protein SNRPD3 and is required for the mRNA splicing of the mitotic motor protein CENP-E. Loss of ERH leads to loss of CENP-E and consequently, chromosome congression defects. Gene expression profiling indicates that ERH is required for the expression of multiple cell cycle genes, and the gene expression signature resulting from ERH down-regulation inversely correlates with KRAS signatures. Clinically, tumor ERH expression is inversely associated with survival of colorectal cancer patients whose tumors harbor KRAS mutations. Together, these findings identify a role of ERH in mRNA splicing and mitosis, and they provide evidence that KRAS mutant cancer cells are dependent on ERH for their survival.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Proteínas Cromosómicas no Histona/genética , Secuencia Conservada , Evolución Molecular , Mutación/genética , Proteínas Proto-Oncogénicas/genética , Empalme del ARN/genética , Factores de Transcripción/metabolismo , Proteínas ras/genética , Ciclo Celular/genética , Línea Celular Tumoral , Supervivencia Celular/genética , Proteínas Cromosómicas no Histona/metabolismo , Cromosomas Humanos/metabolismo , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Regulación Neoplásica de la Expresión Génica , Humanos , Oncogenes , Unión Proteica , Proteínas Proto-Oncogénicas p21(ras) , ARN Mensajero/genética , ARN Mensajero/metabolismo , Análisis de Supervivencia , Proteínas Nucleares snRNP/metabolismo
3.
J Biol Chem ; 288(17): 12253-68, 2013 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-23486470

RESUMEN

Epithelial cell adhesion molecule (EpCAM) (CD326) is a surface glycoprotein expressed by invasive carcinomas and some epithelia. Herein, we report that EpCAM regulates the composition and function of tight junctions (TJ). EpCAM accumulated on the lateral interfaces of human colon carcinoma and normal intestinal epithelial cells but did not co-localize with TJ. Knockdown of EpCAM in T84 and Caco-2 cells using shRNAs led to changes in morphology and adhesiveness. TJ formed readily after EpCAM knockdown; the acquisition of trans-epithelial electroresistance was enhanced, and TJ showed increased resistance to disruption by calcium chelation. Preparative immunoprecipitation demonstrated that EpCAM bound tightly to claudin-7. Co-immunoprecipitation documented associations of EpCAM with claudin-7 and claudin-1 but not claudin-2 or claudin-4. Claudin-1 associated with claudin-7 in co-transfection experiments, and claudin-7 was required for association of claudin-1 with EpCAM. EpCAM knockdown resulted in decreases in claudin-7 and claudin-1 proteins that were reversed with lysosome inhibitors. Immunofluorescence microscopy revealed that claudin-7 and claudin-1 continually trafficked into lysosomes. Although EpCAM knockdown decreased claudin-1 and claudin-7 protein levels overall, accumulations of claudin-1 and claudin-7 in TJ increased. Physical interactions between EpCAM and claudins were required for claudin stabilization. These findings suggest that EpCAM modulates adhesion and TJ function by regulating intracellular localization and degradation of selected claudins.


Asunto(s)
Antígenos de Neoplasias/metabolismo , Moléculas de Adhesión Celular/metabolismo , Claudinas/metabolismo , Proteolisis , Uniones Estrechas/metabolismo , Antígenos de Neoplasias/genética , Células CACO-2 , Adhesión Celular/fisiología , Moléculas de Adhesión Celular/genética , Claudinas/genética , Molécula de Adhesión Celular Epitelial , Técnicas de Silenciamiento del Gen , Humanos , Uniones Estrechas/genética , Transfección
4.
J Biol Chem ; 287(16): 13137-58, 2012 Apr 13.
Artículo en Inglés | MEDLINE | ID: mdl-22351766

RESUMEN

C1 domains, the recognition motif of the second messenger diacylglycerol and of the phorbol esters, are classified as typical (ligand-responsive) or atypical (not ligand-responsive). The C1 domain of Vav1, a guanine nucleotide exchange factor, plays a critical role in regulation of Vav activity through stabilization of the Dbl homology domain, which is responsible for exchange activity of Vav. Although the C1 domain of Vav1 is classified as atypical, it retains a binding pocket geometry homologous to that of the typical C1 domains of PKCs. This study clarifies the basis for its failure to bind ligands. Substituting Vav1-specific residues into the C1b domain of PKCδ, we identified five crucial residues (Glu(9), Glu(10), Thr(11), Thr(24), and Tyr(26)) along the rim of the binding cleft that weaken binding potency in a cumulative fashion. Reciprocally, replacing these incompatible residues in the Vav1 C1 domain with the corresponding residues from PKCδ C1b (δC1b) conferred high potency for phorbol ester binding. Computer modeling predicts that these unique residues in Vav1 increase the hydrophilicity of the rim of the binding pocket, impairing membrane association and thereby preventing formation of the ternary C1-ligand-membrane binding complex. The initial design of diacylglycerol-lactones to exploit these Vav1 unique residues showed enhanced selectivity for C1 domains incorporating these residues, suggesting a strategy for the development of ligands targeting Vav1.


Asunto(s)
Diglicéridos/metabolismo , Ésteres del Forbol/metabolismo , Proteínas Proto-Oncogénicas c-vav/química , Proteínas Proto-Oncogénicas c-vav/metabolismo , Secuencia de Aminoácidos , Línea Celular Tumoral , Humanos , Lactonas/metabolismo , Ligandos , Masculino , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Fosfolípidos/metabolismo , Neoplasias de la Próstata , Unión Proteica/fisiología , Proteína Quinasa C-delta/metabolismo , Estructura Terciaria de Proteína , Proteínas Proto-Oncogénicas c-vav/genética , Transducción de Señal/fisiología
5.
Chembiochem ; 12(8): 1242-51, 2011 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-21542090

RESUMEN

Phorbol 12-myristate 13-acetate (PMA) and bryostatin 1 are both potent protein kinase C (PKC) activators. In LNCaP human prostate cancer cells, PMA induces tumor necrosis factor alpha (TNFα) secretion and inhibits proliferation; bryostatin 1 does not, and indeed blocks the response to PMA. This difference has been attributed to bryostatin 1 not localizing PKCδ to the plasma membrane. Since phorbol ester lipophilicity influences PKCδ localization, we have examined in LNCaP cells a series of phorbol esters and related derivatives spanning some eight logs in lipophilicity (logP) to see if any behave like bryostatin 1. The compounds showed marked differences in their effects on proliferation and TNFα secretion. For example, maximal responses for TNFα secretion relative to PMA ranged from 97 % for octyl-indolactam V to 24 % for phorbol 12,13-dibenzoate. Dose-response curves ranged from monophasic for indolactam V to markedly biphasic for sapintoxin D. The divergent patterns of response, however, correlated neither to lipophilicity, to plasma membrane translocation of PKCδ, nor to the ability to interact with model membranes. In U937 human leukemia cells, a second system in which PMA and bryostatin 1 have divergent effects, viz. PMA but not bryostatin 1 inhibits proliferation and induces attachment, all the compounds acted like PMA for proliferation, but several induced a reduced level or a biphasic dose-response curve for attachment. We conclude that active phorbol esters are not all equivalent. Depending on the system, some might partially resemble bryostatin 1 in their behavior; this encourages the concept that bryostatin-like behavior may be obtained from other structural templates.


Asunto(s)
Antineoplásicos/farmacología , Brioestatinas/farmacología , Ésteres del Forbol/farmacología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Humanos , Concentración 50 Inhibidora , Leucemia/patología , Masculino , Estructura Molecular , Neoplasias de la Próstata/patología
6.
Chembiochem ; 12(15): 2331-40, 2011 Oct 17.
Artículo en Inglés | MEDLINE | ID: mdl-23106081

RESUMEN

N-methyl-substituted diacylglycerol-indololactones (DAG-indololactones) are newly synthesized effectors of protein kinase C (PKC) isoforms and exhibit substantial selectivity between RasGRP3 and PKCα. We present a comprehensive analysis of membrane interactions and biological activities of several DAG-indololactones. Translocation and binding activity assays underline significant variations between the PKC translocation characteristics affected by the ligands as compared to their binding activities. In parallel, the fluorescent properties of the ligands were employed for analysis of their membrane association profiles. Specifically, we found that a slight change in the linkage to the indole ring resulted in significant differences in membrane binding and association of the DAG-indololactones with lipid bilayers. Our analysis shows that seemingly small structural modifications of the hydrophobic regions of these biomimetic PKC effectors contribute to pronounced modulation of membrane interactions of the ligands.


Asunto(s)
Lactonas/química , Lactonas/farmacología , Proteína Quinasa C/antagonistas & inhibidores , Proteína Quinasa C/metabolismo , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/farmacología , Animales , Células CHO , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Cricetinae , Diglicéridos/química , Diglicéridos/farmacología , Factores de Intercambio de Guanina Nucleótido/metabolismo , Humanos , Indoles/química , Indoles/farmacocinética , Indoles/farmacología , Isomerismo , Lactonas/farmacocinética , Isoformas de Proteínas/antagonistas & inhibidores , Isoformas de Proteínas/metabolismo , Proteína Quinasa C-alfa/antagonistas & inhibidores , Proteína Quinasa C-alfa/metabolismo , Inhibidores de Proteínas Quinasas/farmacocinética , Transporte de Proteínas/efectos de los fármacos
7.
Oncotarget ; 7(9): 10133-52, 2016 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-26840086

RESUMEN

CD47 is a signaling receptor for thrombospondin-1 and the counter-receptor for signal-regulatory protein-α (SIRPα). By inducing inhibitory SIRPα signaling, elevated CD47 expression by some cancers prevents macrophage phagocytosis. The anti-human CD47 antibody B6H12 inhibits tumor growth in several xenograft models, presumably by preventing SIRPα engagement. However, CD47 signaling in nontransformed and some malignant cells regulates self-renewal, suggesting that CD47 antibodies may therapeutically target cancer stem cells (CSCs). Treatment of MDA-MB-231 breast CSCs with B6H12 decreased proliferation and asymmetric cell division. Similar effects were observed in T47D CSCs but not in MCF7 breast carcinoma or MCF10A breast epithelial cells. Gene expression analysis in breast CSCs treated with B6H12 showed decreased expression of epidermal growth factor receptor (EGFR) and the stem cell transcription factor KLF4. EGFR and KLF4 mRNAs are known targets of microRNA-7, and B6H12 treatment correspondingly enhanced microRNA-7 expression in breast CSCs. B6H12 treatment also acutely inhibited EGF-induced EGFR tyrosine phosphorylation. Expression of B6H12-responsive genes correlated with CD47 mRNA expression in human breast cancers, suggesting that the CD47 signaling pathways identified in breast CSCs are functional in vivo. These data reveal a novel SIRPα-independent mechanism by which therapeutic CD47 antibodies could control tumor growth by autonomously forcing differentiation of CSC.


Asunto(s)
Anticuerpos Bloqueadores/farmacología , Antígeno CD47/metabolismo , Receptores ErbB/metabolismo , Células Madre Neoplásicas/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Anticuerpos Bloqueadores/inmunología , Antígenos de Diferenciación/genética , Antígenos de Diferenciación/metabolismo , Western Blotting , Antígeno CD47/genética , Antígeno CD47/inmunología , Línea Celular , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Factor de Crecimiento Epidérmico/genética , Factor de Crecimiento Epidérmico/metabolismo , Factor de Crecimiento Epidérmico/farmacología , Receptores ErbB/genética , Femenino , Perfilación de la Expresión Génica/métodos , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Factor 4 Similar a Kruppel , Células MCF-7 , MicroARNs/genética , Microscopía Confocal , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Fosforilación/efectos de los fármacos , Receptores Inmunológicos/genética , Receptores Inmunológicos/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/genética , Trombospondina 1/genética , Trombospondina 1/metabolismo , Neoplasias de la Mama Triple Negativas/genética , Neoplasias de la Mama Triple Negativas/metabolismo , Neoplasias de la Mama Triple Negativas/patología
8.
Stem Cell Reports ; 4(1): 155-169, 2015 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-25497455

RESUMEN

Many tumors are hierarchically organized with a minority cell population that has stem-like properties and enhanced ability to initiate tumorigenesis and drive therapeutic relapse. These cancer stem cells (CSCs) are typically identified by complex combinations of cell-surface markers that differ among tumor types. Here, we developed a flexible lentiviral-based reporter system that allows direct visualization of CSCs based on functional properties. The reporter responds to the core stem cell transcription factors OCT4 and SOX2, with further selectivity and kinetic resolution coming from use of a proteasome-targeting degron. Cancer cells marked by this reporter have the expected properties of self-renewal, generation of heterogeneous offspring, high tumor- and metastasis-initiating activity, and resistance to chemotherapeutics. With this approach, the spatial distribution of CSCs can be assessed in settings that retain microenvironmental and structural cues, and CSC plasticity and response to therapeutics can be monitored in real time.


Asunto(s)
Expresión Génica , Genes Reporteros , Células Madre Neoplásicas/metabolismo , Animales , Antineoplásicos/farmacología , División Celular Asimétrica , Diferenciación Celular , Línea Celular Tumoral , Movimiento Celular/genética , Rastreo Celular , Transformación Celular Neoplásica/genética , Resistencia a Antineoplásicos/genética , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Orden Génico , Vectores Genéticos , Xenoinjertos , Humanos , Inmunofenotipificación , Ratones , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/patología , Fenotipo , Regiones Promotoras Genéticas , Unión Proteica , Elementos de Respuesta , Factores de Transcripción/metabolismo , Células Tumorales Cultivadas
9.
J Clin Endocrinol Metab ; 88(1): 459-63, 2003 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-12519890

RESUMEN

We have previously identified two second hit mechanisms involved in the development of multiple endocrine neoplasia type 2 (MEN 2)-associated tumors: trisomy 10 with duplication of the mutant RET allele and loss of the wild-type RET allele. However, some of the MEN 2-associated tumors investigated did not demonstrate either mechanism. Here, we studied the TT cell line derived from MEN 2-associated medullary thyroid carcinoma with a RET germline mutation in codon 634, for alternative mechanisms of tumorigenesis. Although we observed a 2:1 ratio between mutant and wild-type RET at the genomic DNA level in this cell line, fluorescence in situ hybridization analysis revealed neither trisomy 10 nor loss of the normal chromosome 10. Instead, a tandem duplication event was responsible for amplification of mutant RET. In further studies we could for the first time demonstrate that the genomic chromosome 10 abnormalities in this cell line cause an increased production of mutant RET mRNA. These findings provide evidence for a third second hit mechanism resulting in overrepresentation and overexpression of mutant RET in MEN 2-associated tumors.


Asunto(s)
Carcinoma Medular/genética , Amplificación de Genes , Expresión Génica , Mutación de Línea Germinal/genética , Neoplasia Endocrina Múltiple Tipo 2a/genética , Proteínas Oncogénicas/genética , Proteínas Tirosina Quinasas Receptoras/genética , Neoplasias de la Tiroides/genética , Aberraciones Cromosómicas , Cromosomas Humanos Par 10/genética , Humanos , Proteínas Proto-Oncogénicas c-ret , ARN Mensajero/biosíntesis , Secuencias Repetidas en Tándem , Células Tumorales Cultivadas
10.
ACS Chem Biol ; 8(4): 767-77, 2013 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-23369356

RESUMEN

The bryostatins are a group of 20 macrolides isolated by Pettit and co-workers from the marine organism Bugula neritina. Bryostatin 1, the flagship member of the family, has been the subject of intense chemical and biological investigations due to its remarkably diverse biological activities, including promising indications as therapy for cancer, Alzheimer's disease, and HIV. Other bryostatins, however, have attracted far less attention, most probably due to their relatively low natural abundance and associated scarcity of supply. Among all macrolides in this family, bryostatin 7 is biologically the most potent protein kinase C (PKC) ligand (in terms of binding affinity) and also the first bryostatin to be synthesized in the laboratory. Nonetheless, almost no biological studies have been carried out on this agent. We describe herein the total synthesis of bryostatin 7 based on our pyran annulation technology, which allows for the first detailed biological characterizations of bryostatin 7 with side-by-side comparisons to bryostatin 1. The results suggest that the more easily synthesized and less lipophilic bryostatin 7 may be an effective surrogate for bryostatin 1.


Asunto(s)
Brioestatinas/farmacología , Lípidos/química , Brioestatinas/síntesis química , Brioestatinas/química , Línea Celular Tumoral , Regulación hacia Abajo , Humanos , Isoenzimas/metabolismo , Masculino , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Proteína Quinasa C/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Fracciones Subcelulares/enzimología , Células U937
11.
Biochem Pharmacol ; 81(11): 1296-308, 2011 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-21458422

RESUMEN

Bryostatin 1 has attracted considerable attention both as a cancer chemotherapeutic agent and for its unique activity. Although it functions, like phorbol esters, as a potent protein kinase C (PKC) activator, it paradoxically antagonizes many phorbol ester responses in cells. Because of its complex structure, little is known of its structure-function relations. Merle 23 is a synthetic derivative, differing from bryostatin 1 at only four positions. However, in U-937 human leukemia cells, Merle 23 behaves like a phorbol ester and not like bryostatin 1. Here, we characterize the behavior of Merle 23 in the human prostate cancer cell line LNCaP. In this system, bryostatin 1 and phorbol ester have contrasting activities, with the phorbol ester but not bryostatin 1 blocking cell proliferation or tumor necrosis factor alpha secretion, among other responses. We show that Merle 23 displays a highly complex pattern of activity in this system. Depending on the specific biological response or mechanistic change, it was bryostatin-like, phorbol ester-like, intermediate in its behavior, or more effective than either. The pattern of response, moreover, varied depending on the conditions. We conclude that the newly emerging bryostatin derivatives such as Merle 23 provide powerful tools to dissect subsets of bryostatin mechanism and response.


Asunto(s)
Brioestatinas/farmacología , Apoptosis/efectos de los fármacos , Brioestatinas/química , División Celular/efectos de los fármacos , Línea Celular Tumoral , Regulación hacia Abajo , Humanos , Masculino , Fosforilación , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Proteína Quinasa C/metabolismo , Fracciones Subcelulares/efectos de los fármacos , Fracciones Subcelulares/enzimología , Fracciones Subcelulares/metabolismo , Acetato de Tetradecanoilforbol/farmacología , Factor de Necrosis Tumoral alfa/metabolismo , Células U937
12.
Virology ; 369(1): 131-42, 2007 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-17727912

RESUMEN

Oncolytic adenoviruses as a treatment for cancer have demonstrated limited clinical activity. Contributing to this may be the relevance of preclinical animal models used to study these agents. Syngeneic mouse tumor models are generally non-permissive for adenoviral replication, whereas human tumor xenograft models exhibit attenuated immune responses to the vector. The cotton rat (Sigmodon hispidus) is susceptible to human adenovirus infection, permissive for viral replication and exhibits similar inflammatory pathology to humans with adenovirus replicating in the lungs, respiratory passages and cornea. We evaluated three transplantable tumorigenic cotton rat cell lines, CCRT, LCRT and VCRT as models for the study of oncolytic adenoviruses. All three cells lines were readily infected with adenovirus type-5-based vectors and exhibited high levels of transgene expression. The cell lines supported viral replication demonstrated by the induction of cytopathogenic effect (CPE) in tissue culture, increase in virus particle numbers and assembly of virions seen on transmission electron microscopy. In vivo, LCRT and VCRT tumors demonstrated delayed growth after injection with replicating adenovirus. No in vivo antitumor activity was seen in CCRT tumors despite in vitro oncolysis. Adenovirus was also rapidly cleared from the CCRT tumors compared to LCRT and VCRT tumors. The effect observed with the different cotton rat tumor cell lines mimics the variable results of human clinical trials highlighting the potential relevance of this model for assessing the activity and toxicity of oncolytic adenoviruses.


Asunto(s)
Adenoviridae/crecimiento & desarrollo , Modelos Animales de Enfermedad , Neoplasias/terapia , Neoplasias/virología , Viroterapia Oncolítica/métodos , Animales , Línea Celular Tumoral , Efecto Citopatogénico Viral , Microscopía Electrónica de Transmisión , Ratas , Sigmodontinae , Trasplante Isogénico , Ensayo de Placa Viral , Virión/ultraestructura
13.
Cell Cycle ; 6(7): 836-42, 2007 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-17377495

RESUMEN

Animal models of human diseases are widely used to address questions of tumor development. Selection of a particular animal model depends upon a variety of factors, among them: animal cost, species lifespan and hardiness; availability of biomolecular and genetic tools for that species; and evolutionary distance from humans. In spite of the growth in genomic data in the past several years, many animal models cannot yet be studied extensively due to gaps in genetic mapping, sequencing and functional analyses. Thus, alternative molecular genetic approaches are needed. We have designed an interspecies comparative genomic hybridization approach to analyze genetic changes in radiation-induced brain tumors in the non-human primate, Macaca mulatta. Using homologies between the primate and human genomes, we adapted widely-available CGH techniques to generate cytogenetic profiles of malignant gliomas in four monkey tumors. Losses and gains were projected onto the corresponding homologous chromosomal regions in the human genome, thus directly translating the status of the monkey gliomas into human gene content. This represents a novel method of comparative interspecies cytogenetic mapping that permits simultaneous analysis of genomic imbalances of unknown sequences in disparate species and correlation with potential or known human disease-related genes.


Asunto(s)
Neoplasias Encefálicas/genética , Regulación Neoplásica de la Expresión Génica/genética , Glioma/genética , Macaca mulatta/genética , Biología Molecular/métodos , Hibridación de Ácido Nucleico/genética , Animales , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica/métodos , Genoma Humano/genética , Inestabilidad Genómica/genética , Biblioteca Genómica , Glioma/patología , Humanos , Masculino , Especificidad de la Especie
14.
Genes Chromosomes Cancer ; 43(1): 72-82, 2005 May.
Artículo en Inglés | MEDLINE | ID: mdl-15704128

RESUMEN

Twenty-four pituitary adenomas, both the sporadic type (n = 18) and the type arising in association with either multiple endocrine neoplasia, type 1 (MEN1; n = 2), or Carney complex (CNC, n = 4) were analyzed by comparative genomic hybridization. Twenty-one (88%) tumors displayed chromosomal alterations. The number of chromosomal aberrations in each tumor varied from 2 to greater than 10. Several recurrent chromosomal abnormalities were identified in this study. The most frequently detected losses of chromosomal material involved 1p (14 of 24, 58%), 11p (11 of 24, 46%), 17 (10 of 24, 42%), 16p (9 of 24, 38%), 4 (8 of 24, 33%), 10p (6 of 24, 25%), 12 (6 of 24, 25%), 20 (6 of 24, 25%), 22q (6 of 24, 25%), 13q (5 of 24, 21%), and 9p (4 of 24, 17%). Copy number increases were detected on 4q (7 of 24, 29%), 17 (8 of 24, 33%), 19 (7 of 24, 29%), 1p (6 of 24, 25%), 5 (6 of 24, 25%), 20 (6 of 24, 25%), 6q (5 of 24, 21%), 13q21-qter (5 of 24, 21%), and 16p (5 of 24, 21%). Chromosome 11 loss, which involved 11p in all cases, was the most significant finding and was common to tumors arising sporadically and in association with MEN1 and CNC. In addition, the majority of the tumors (18 of 24, 75% overall and 86% of all tumors with chromosomal abnormalities) showed involvement of chromosome 1. Tumors had either loss (14 of 24, 58%) or gain (6 of 24, 25%) in the 1p32-1pter region. Finally, changes on chromosome 17, either loss or gain, occurred in 71% (17) of all 24 patients. In summary, all the tumors with chromosomal rearrangements (21 of 24, 88%), whether sporadic pituitary adenomas or those associated with MEN1 or CNC, had alteration(s) of 1p32, 11p, or 17.


Asunto(s)
Adenoma/genética , Neoplasias Hipofisarias/genética , Adenoma/cirugía , Adolescente , Adulto , Preescolar , Deleción Cromosómica , Mapeo Cromosómico , ADN de Neoplasias/genética , Femenino , Humanos , Masculino , Persona de Mediana Edad , Hibridación de Ácido Nucleico/genética , Neoplasias Hipofisarias/cirugía
15.
Proc Natl Acad Sci U S A ; 99(10): 6806-11, 2002 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-12011441

RESUMEN

CTCF, a conserved, ubiquitous, and highly versatile 11-zinc-finger factor involved in various aspects of gene regulation, forms methylation-sensitive insulators that regulate X chromosome inactivation and expression of imprinted genes. We document here the existence of a paralogous gene with the same exons encoding the 11-zinc-finger domain as mammalian CTCF genes and thus the same DNA-binding potential, but with distinct amino and carboxy termini. We named this gene BORIS for Brother of the Regulator of Imprinted Sites. BORIS is present only in the testis, and expressed in a mutually exclusive manner with CTCF during male germ cell development. We show here that erasure of methylation marks during male germ-line development is associated with dramatic up-regulation of BORIS and down-regulation of CTCF expression. Because BORIS bears the same DNA-binding domain that CTCF employs for recognition of methylation marks in soma, BORIS is a candidate protein for the elusive epigenetic reprogramming factor acting in the male germ line.


Asunto(s)
Proteínas de Unión al ADN/genética , Impresión Genómica , Proteínas Represoras , Testículo/metabolismo , Factores de Transcripción/genética , Dedos de Zinc , Secuencia de Aminoácidos , Animales , Factor de Unión a CCCTC , Clonación Molecular , Metilación de ADN , Expresión Génica , Marcadores Genéticos , Humanos , Masculino , Ratones , Datos de Secuencia Molecular , Estructura Terciaria de Proteína , Homología de Secuencia de Aminoácido
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA