Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
PLoS Biol ; 7(2): e39, 2009 Feb 24.
Artículo en Inglés | MEDLINE | ID: mdl-19243219

RESUMEN

Colon cancer accounts for more than 10% of all cancer deaths annually. Our genetic evidence from Drosophila and previous in vitro studies of mammalian Atonal homolog 1 (Atoh1, also called Math1 or Hath1) suggest an anti-oncogenic function for the Atonal group of proneural basic helix-loop-helix transcription factors. We asked whether mouse Atoh1 and human ATOH1 act as tumor suppressor genes in vivo. Genetic knockouts in mouse and molecular analyses in the mouse and in human cancer cell lines support a tumor suppressor function for ATOH1. ATOH1 antagonizes tumor formation and growth by regulating proliferation and apoptosis, likely via activation of the Jun N-terminal kinase signaling pathway. Furthermore, colorectal cancer and Merkel cell carcinoma patients show genetic and epigenetic ATOH1 loss-of-function mutations. Our data indicate that ATOH1 may be an early target for oncogenic mutations in tissues where it instructs cellular differentiation.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Carcinoma de Células de Merkel/genética , Neoplasias Colorrectales/genética , Genes Supresores de Tumor/fisiología , Neoplasias Cutáneas/genética , Animales , Apoptosis/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Carcinoma de Células de Merkel/metabolismo , Carcinoma de Células de Merkel/patología , Línea Celular Tumoral , Proliferación Celular , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Análisis Mutacional de ADN , Regulación Neoplásica de la Expresión Génica , Humanos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Proteínas Quinasas JNK Activadas por Mitógenos , Masculino , Ratones , Ratones Noqueados , Mutación , Transducción de Señal , Neoplasias Cutáneas/metabolismo , Neoplasias Cutáneas/patología
2.
Gastroenterology ; 139(3): 918-28, 928.e1-6, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20621629

RESUMEN

BACKGROUND & AIMS: The atonal homolog 1 (Atoh1) transcription factor is required for intestinal secretory (goblet, Paneth, enteroendocrine) cell differentiation. Notch/gamma-secretase inhibitors (GSIs) block proliferation and induce secretory cell differentiation in the intestine. We used genetic analyses of mice to determine whether Atoh1 mediates the effects of GSIs in normal and cancerous intestinal epithelia. METHODS: We studied mice with intestine-specific disruption of Atoh1 (Atoh1(Deltaintestine)), the adenomatosis polyposis coli (APC)(min) mutation, both mutations (Atoh1(Deltaintestine); APC(min)), or littermate controls; mice were given GSI or vehicle. Colorectal cancer (CRC) cell lines were treated with GSI or vehicle and with small hairpin RNAs to reduce ATOH1. Differentiation and homeostasis were assessed by protein, RNA, and histologic analyses. RESULTS: GSIs failed to induce secretory cell differentiation or apoptosis or decrease proliferation of Atoh1-null progenitor cells, compared with wild-type cells. Exposure of APC(min) adenomas to GSIs decreased proliferation and increased secretory cell numbers in an Atoh1-dependent manner. In CRC cells treated with GSI, ATOH1 levels were correlated inversely with proliferation. ATOH1 was required for secretory cell gene expression in cell lines and in mice. CONCLUSIONS: ATOH1 is required for all effects of GSIs in intestinal crypts and adenomas; Notch has no unique function in intestinal progenitors and cancer cells other than to regulate ATOH1 expression. Reducing ATOH1 activity might mitigate intestinal toxicity from systemic GSI therapy for nonintestinal diseases. Among gastrointestinal malignancies, ATOH1 mediates the effects of GSIs, so ATOH1 expression levels might predict responses to these inhibitors. We propose that only the subset of CRCs that retain ATOH1 expression will respond to GSIs.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide/antagonistas & inhibidores , Antineoplásicos/farmacología , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Neoplasias Colorrectales/metabolismo , Células Epiteliales/efectos de los fármacos , Mucosa Intestinal/efectos de los fármacos , Receptores Notch/antagonistas & inhibidores , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Animales , Apoptosis/efectos de los fármacos , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Células Epiteliales/metabolismo , Células Epiteliales/patología , Regulación Neoplásica de la Expresión Génica , Genes APC , Células HCT116 , Células HT29 , Humanos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Ratones , Ratones Transgénicos , Interferencia de ARN , ARN Mensajero/metabolismo , Receptores Notch/metabolismo , Factores de Tiempo
3.
Exp Cell Res ; 316(3): 452-65, 2010 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-19786015

RESUMEN

BACKGROUND AND AIMS: SPDEF (also termed PDEF or PSE) is an ETS family transcription factor that regulates gene expression in the prostate and goblet cell hyperplasia in the lung. Spdef has been reported to be expressed in the intestine. In this paper, we identify an important role for Spdef in regulating intestinal epithelial cell homeostasis and differentiation. METHODS: SPDEF expression was inhibited in colon cancer cells to determine its ability to control goblet cell gene activation. The effects of transgenic expression of Spdef on intestinal differentiation and homeostasis were determined. RESULTS: In LS174T colon cancer cells treated with Notch/gamma-secretase inhibitor to activate goblet cell gene expression, shRNAs that inhibited SPDEF also repressed expression of goblet cell genes AGR2, MUC2, RETLNB, and SPINK4. Transgenic expression of Spdef caused the expansion of intestinal goblet cells and corresponding reduction in Paneth, enteroendocrine, and absorptive enterocytes. Spdef inhibited proliferation of intestinal crypt cells without induction of apoptosis. Prolonged expression of the Spdef transgene caused a progressive reduction in the number of crypts that expressed Spdef, consistent with its inhibitory effects on cell proliferation. CONCLUSIONS: Spdef was sufficient to inhibit proliferation of intestinal progenitors and induce differentiation into goblet cells; SPDEF was required for activation of goblet cell associated genes in vitro. These data support a model in which Spdef promotes terminal differentiation into goblet cells of a common goblet/Paneth progenitor.


Asunto(s)
Diferenciación Celular , Células Caliciformes/citología , Células Caliciformes/metabolismo , Proteínas Proto-Oncogénicas c-ets/metabolismo , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Recuento de Células , Diferenciación Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Neoplasias del Colon/genética , Neoplasias del Colon/patología , Proteínas de Unión al ADN/metabolismo , Doxiciclina/farmacología , Células Epiteliales/citología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Células Caliciformes/efectos de los fármacos , Humanos , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Mucosa Intestinal/ultraestructura , Ratones , Especificidad de Órganos/efectos de los fármacos , Transporte de Proteínas/efectos de los fármacos , Proteínas Proto-Oncogénicas c-ets/genética , Células Madre/citología , Células Madre/efectos de los fármacos , Células Madre/metabolismo , Factores de Transcripción/metabolismo , Transgenes/genética
4.
Sci Rep ; 9(1): 8980, 2019 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-31222072

RESUMEN

Hydrosalpinx, the blockage of fallopian tubes, can result from pelvic inflammatory disease. Hydrosalpinx is a cause of infertility and negatively impacts in vitro fertilization. To better understand the pathobiology of hydrosalpinx, we compared the proteome of lavages from disease vs. healthy fallopian tubes. Results indicate a disruption of redox homeostasis and activation of the complement system, immune cell infiltration, and phagocytosis; pathways that may drive tubal injury. To our surprise among the most prominent proteins with hydrosalpinx was mesothelin (MSLN), which until now has only been associated with epithelial malignancies. Analogous to mesothelioma and ovarian carcinoma, a significant increase of MSLN was detected in plasma from patients with hydrosalpinx. This finding suggests MSLN may provide clinical diagnosis in lieu of the current approaches that require invasive imaging. Importantly, these findings implicate MSLN in a benign disease, indicating that the activation and role of MSLN is not restricted to cancer.


Asunto(s)
Enfermedades de las Trompas Uterinas/metabolismo , Trompas Uterinas/metabolismo , Proteoma , Cromatografía Liquida , Susceptibilidad a Enfermedades , Enfermedades de las Trompas Uterinas/etiología , Enfermedades de las Trompas Uterinas/patología , Femenino , Fertilidad , Proteínas Ligadas a GPI/sangre , Humanos , Inmunohistoquímica , Mesotelina , Proteómica/métodos , Espectrometría de Masas en Tándem , Irrigación Terapéutica
5.
Sci Rep ; 7(1): 17883, 2017 12 20.
Artículo en Inglés | MEDLINE | ID: mdl-29263436

RESUMEN

Preterm infants are at significantly increased risk for lifelong neurodevelopmental disability with male offspring disproportionately affected. Corticosteroids (such as betamethasone) and magnesium sulphate (MgSO4) are administered to women in preterm labor to reduce neurologic morbidity. Despite widespread use of MgSO4 in clinical practice, its effects on adult offspring are not well known nor have sex-specific differences in therapeutic response been explored. The objective of our study was to examine the long-term effects of perinatal neuroinflammation and the effectiveness of prenatal MgSO4/betamethasone treatments between males and females in a murine model via histologic and expression analyses. Our results demonstrate that male but not female offspring exposed to intrauterine inflammation demonstrated impaired performance in neurodevelopmental testing in early life assessed via negative geotaxis, while those exposed to injury plus treatment fared better. Histologic analysis of adult male brains identified a significant reduction in hippocampal neural density in the injured group compared to controls. Evaluation of key neural markers via qRT-PCR demonstrated more profound differences in gene expression in adult males exposed to injury and treatment compared to female offspring, which largely showed resistance to injury. Prenatal treatment with MgSO4/betamethasone confers long-term benefits beyond cerebral palsy prevention with sex-specific differences in response.


Asunto(s)
Betametasona/farmacología , Inflamación/tratamiento farmacológico , Sulfato de Magnesio/farmacología , Animales , Biomarcadores/metabolismo , Parálisis Cerebral/tratamiento farmacológico , Parálisis Cerebral/metabolismo , Modelos Animales de Enfermedad , Femenino , Expresión Génica/efectos de los fármacos , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Humanos , Inflamación/metabolismo , Masculino , Ratones , Embarazo , Atención Prenatal , Caracteres Sexuales
6.
Crit Rev Oncol Hematol ; 59(2): 85-97, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16716599

RESUMEN

The growth factor independence-1 (Gfi1) transcription factor is required for proper development of neuroendocrine cells, sensory neurons, and blood. Patients with mutations in Gfi1 exhibit severe congenital neutropenia (SCN) or non-immune chronic idiopathic neutropenia of adults. Gfi1 was initially described as an oncoprotein that mediates tumor progression in a mouse model of leukemia; however, recent data suggest that Gfi1 may act as either an oncogene or an anti-proliferative tumor suppressor gene depending on the cell type. Here we review the latest literature on Gfi1, and emphasize its role in the hematopoietic, sensory and neuroendocrine systems.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Proteínas Oncogénicas/metabolismo , Factores de Transcripción/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Animales , Proteínas de Unión al ADN/genética , Hematopoyesis/genética , Humanos , Ratones , Neuronas Aferentes/metabolismo , Sistemas Neurosecretores/metabolismo , Neutropenia/genética , Neutropenia/metabolismo , Proteínas Oncogénicas/genética , Especificidad de Órganos , Factores de Transcripción/genética , Proteínas Supresoras de Tumor/genética
7.
Cancer Res ; 64(19): 6874-82, 2004 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-15466176

RESUMEN

Human small cell lung cancers might be derived from pulmonary cells with a neuroendocrine phenotype. They are driven to proliferate by autocrine and paracrine neuropeptide growth factor stimulation. The molecular basis of the neuroendocrine phenotype of lung carcinomas is relatively unknown. The Achaete-Scute Homologue-1 (ASH1) transcription factor is critically required for the formation of pulmonary neuroendocrine cells and is a marker for human small cell lung cancers. The Drosophila orthologues of ASH1 (Achaete and Scute) and the growth factor independence-1 (GFI1) oncoprotein (Senseless) genetically interact to inhibit Notch signaling and specify fly sensory organ development. Here, we show that GFI1, as with ASH1, is expressed in neuroendocrine lung cancer cell lines and that GFI1 in lung cancer cell lines functions as a DNA-binding transcriptional repressor protein. Forced expression of GFI1 potentiates tumor formation of small-cell lung carcinoma cells. In primary human lung cancer specimens, GFI1 expression strongly correlates with expression of ASH1, the neuroendocrine growth factor gastrin-releasing peptide, and neuroendocrine markers synaptophysin and chromogranin A (P < 0.0000001). GFI1 colocalizes with chromogranin A and calcitonin-gene-related peptide in embryonic and adult murine pulmonary neuroendocrine cells. In addition, mice with a mutation in GFI1 display abnormal development of pulmonary neuroendocrine cells, indicating that GFI1 is important for neuroendocrine differentiation.


Asunto(s)
Carcinoma Neuroendocrino/metabolismo , Carcinoma de Células Pequeñas/metabolismo , Proteínas de Unión al ADN/biosíntesis , Neoplasias Pulmonares/metabolismo , Pulmón/citología , Sistemas Neurosecretores/citología , Factores de Transcripción/biosíntesis , Animales , Carcinoma Neuroendocrino/genética , Carcinoma de Células Pequeñas/genética , Diferenciación Celular , Extractos Celulares/farmacología , Línea Celular Tumoral , Proteínas de Unión al ADN/genética , Femenino , Humanos , Pulmón/efectos de los fármacos , Neoplasias Pulmonares/genética , Ratones , Trasplante de Neoplasias , Sistemas Neurosecretores/efectos de los fármacos , Embarazo , Factores de Transcripción/genética , Transfección , Trasplante Heterólogo
8.
Reprod Sci ; 21(2): 204-14, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23757314

RESUMEN

Glycosylation of plasma proteins increases during pregnancy. Our objectives were to investigate an anti-inflammatory role of these proteins in normal pregnancies and determine whether aberrant protein glycosylation promotes monocyte adhesion in preeclampsia. Plasma was prospectively collected from nonpregnant controls and nulliparous patients in all 3 trimesters. Patients were divided into cohorts based on the applicable postpartum diagnosis. U937 monocytes were preconditioned with enzymatically deglycosylated plasma, and monocyte adhesion to endothelial cell monolayers was quantified by spectrophotometry. Plasma from nonpregnant controls, first trimester normotensives, and first trimester patients with mild preeclampsia inhibited monocyte-endothelial cell adhesion (P < .05), but plasma from first trimester patients with severe preeclampsia and second and third trimester normotensives did not. Deglycosylating plasma proteins significantly increased adhesion in all the cohorts. These results support a role of plasma glycoprotein interaction in monocyte-endothelial cell adhesion and could suggest a novel therapeutic target for severe preeclampsia.


Asunto(s)
Proteínas Sanguíneas/metabolismo , Monocitos/metabolismo , Preeclampsia/sangre , Preeclampsia/diagnóstico , Índice de Severidad de la Enfermedad , Adulto , Secuencia de Aminoácidos , Proteínas Sanguíneas/genética , Adhesión Celular/fisiología , Estudios de Cohortes , Femenino , Glicosilación , Células Endoteliales de la Vena Umbilical Humana , Humanos , Datos de Secuencia Molecular , Preeclampsia/genética , Embarazo , Células U937 , Adulto Joven
9.
Fertil Steril ; 99(1): 199-205, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23009827

RESUMEN

OBJECTIVE: To investigate the effect of cigarette smoke exposure on ciliation and ciliogenesis in human oviductal epithelium. DESIGN: Molecular analysis using human tubal segments. SETTING: Academic medical center. PATIENT(S): Twenty women undergoing elective tubal sterilization procedure. INTERVENTION(S): Expression of ciliated cell-specific markers was compared in tubal segments from smokers and nonsmokers using quantitative immunohistochemistry and Western blot analysis. The expression of transcription factors in the motile ciliogenesis program was compared using quantitative polymerase chain reaction and quantitative immunohistochemistry. MAIN OUTCOME MEASURE(S): Oviductal ciliation and expression of transcription factors involved in ciliogenesis. RESULT(S): No significant differences were detected in density of ciliation between groups. Neither number of years of smoking nor pack-year history correlated with density of ciliation. Expression of ciliogenic transcription factors FOXJ1, RFX2, and RFX3 was consistent between groups. CONCLUSION(S): Few studies have evaluated the relationship between smoking and ciliated epithelium in human oviducts. Cigarette smoking does not seem to result in quantitative differences in the density of ciliation nor expression of ciliogenesis factors. Our findings suggest that pathophysiologic mechanisms other than ciliation account for the increased risk of ectopic pregnancy in women who smoke.


Asunto(s)
Cilios/patología , Cilios/fisiología , Trompas Uterinas/fisiología , Fumar/efectos adversos , Adulto , Biopsia , Proteínas de Unión al ADN/fisiología , Epitelio/patología , Epitelio/fisiología , Trompas Uterinas/patología , Femenino , Factores de Transcripción Forkhead/fisiología , Humanos , Incidencia , Embarazo , Embarazo Ectópico/epidemiología , Estudios Prospectivos , Factores de Transcripción del Factor Regulador X , Factores de Transcripción/fisiología
10.
Curr Colorectal Cancer Rep ; 7(2): 121-127, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21980310

RESUMEN

The Notch receptor signaling pathway regulates expression of the basic helix-loop-helix transcription factor ATOH1 (Math1/Hath1) to determine cell fate in the intestine. In differentiating intestinal stem cells, high levels of Notch activity specify absorptive enterocyte/colonocyte differentiation, whereas high ATOH1 activity specifies secretory (goblet, enteroendocrine, and Paneth) cell differentiation. In colorectal cancer, ATOH1 is a tumor suppressor that is silenced in most tumors, while Notch is oncogenic and often highly active in human tumors. In other gastrointestinal malignancies with features of intestinal metaplasia, such as esophageal and gastric cancers, the Notch-ATOH1 pathway becomes activated. In cancers and preneoplastic tissues that retain the ability to activate ATOH1, therapeutic targeting of this pathway can be achieved by inhibiting Notch activity (with Notch-targeting antibodies or small-molecule inhibitors of γ-secretase). Thus, targeting the Notch-ATOH1 pathway represents a novel approach to differentiation therapy in gastrointestinal cancers.

12.
J Biol Chem ; 283(46): 32056-65, 2008 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-18805794

RESUMEN

Growth factor independent-1 (Gfi1) is a zinc finger protein with a SNAG-transcriptional repressor domain. Ajuba is a LIM domain protein that shuttles between the cytoplasm and the nucleus. Ajuba functions as a co-repressor for synthetic Gfi1 SNAG-repressor domain-containing constructs, but a role for Ajuba co-repression of the cognate DNA bound Gfi1 protein has not been defined. Co-immunoprecipitation of synthetic and endogenous proteins and co-elution with gel filtration suggest that an endogenous Ajuba.Gfi1.HDAC multiprotein complex is possible. Active histone deacetylase activity co-immunoprecipitates with Ajuba or Gfi1, and both proteins depend upon histone deacetylases for full transcriptional repression activity. Ajuba LIM domains directly bind to Gfi1, but the association is not SNAG domain-dependent. ChIP analysis and reciprocal knockdown experiments suggest that Ajuba selectively functions as a co-repressor for Gfi1 autoregulation. The data suggest that Ajuba is utilized as a corepressor selectively on Gfi1 target genes.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Regulación hacia Abajo , Histona Desacetilasas/metabolismo , Proteínas de Homeodominio/metabolismo , Factores de Transcripción/metabolismo , Línea Celular , Supervivencia Celular , Proteínas de Unión al ADN/genética , Proteínas de Homeodominio/genética , Humanos , Proteínas con Dominio LIM , Unión Proteica , Factores de Transcripción/genética
13.
Lab Invest ; 87(4): 336-44, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17377622

RESUMEN

Naphthalene exposure kills lung airway epithelial (Clara) cells, but is rapidly followed by Clara cell reconstitution coincident with proliferation of pulmonary neuroendocrine cells (PNEC). Although a role for mature PNEC in the reconstitution process has been excluded, the reconstituting progenitor cells have been suggested to enter a transient neuroendocrine (NE) differentiation phase before differentiating to Clara cells. Furthermore, these progenitors were suggested to be the target population for transformation to a NE tumor; small cell lung cancer (SCLC). Although the NE phenotype is central to SCLC oncogenesis, the relevance of NE differentiation to post naphthalene reconstitution remains to be determined. The Growth factor independent-1 (Gfi1) transcription factor is expressed in SCLC and is required for the NE differentiation of PNEC. Gfi1(-/-) mice display a 70% reduction in airway cells that express NE markers, and cells that stain for NE markers show weak expression of some markers. Therefore, to determine the relevance of the NE phenotype to post-naphthalene reconstitution, we examined post-naphthalene reconstitution in Gfi1(-/-) mice. Our analyses indicate that the post-naphthalene regeneration process includes both airway epithelial proliferation and apoptosis. Gfi1 deletion lowered both airway epithelial proliferation and apoptosis; however, the post-naphthalene rate of increase in growth and apoptosis was not significantly different between Gfi1(-/-) mice and wild-type littermates. Moreover, the timing and extent of CC10+ cell regeneration was unaffected by Gfi1 deletion. These data suggest that neither Gfi1 nor the NE phenotype play a dominant role in the regeneration process. However, the few Gfi1(-/-) cells capable of NE differentiation show a significant reduction in post-naphthalene proliferation. The modest proliferation seen in Gfi1(-/-) NE cells is consistent with the previously proposed role for Gfi1 in controlling neuroendocrine cancer growth.


Asunto(s)
Carcinoma de Células Pequeñas/patología , Proliferación Celular/efectos de los fármacos , Proteínas de Unión al ADN/fisiología , Contaminantes Ambientales/toxicidad , Neoplasias Pulmonares/patología , Pulmón/patología , Naftalenos/toxicidad , Sistemas Neurosecretores/patología , Factores de Transcripción/fisiología , Animales , Apoptosis , Carcinoma de Células Pequeñas/inducido químicamente , Diferenciación Celular , Proteínas de Unión al ADN/genética , Neoplasias Pulmonares/inducido químicamente , Ratones , Ratones Noqueados , Fenotipo , Regeneración , Mucosa Respiratoria/patología , Células Madre/citología , Factores de Transcripción/genética
14.
J Cell Biochem ; 89(5): 1005-18, 2003 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-12874834

RESUMEN

Gfi-1 and Gfi-1B can repress transcription and play important roles in hematopoietic cell survival and differentiation. Although these proteins are known to bind DNA through a C-terminal zinc-finger domain and may require an N-terminal SNAG domain (SNAIL/Gfi-1) to repress transcription, the mechanism by which Gfi-1 and Gfi-1B act is unknown. A first step towards understanding the mechanism by which these proteins repress transcription is to identify interacting proteins that could contribute to transcriptional repression. ETO (also termed MTG8), was first identified through its involvement in the (8;21) translocation associated with acute myelogenous leukemia. It attaches to the nuclear matrix and associates with histone deacetylases and the co-repressors N-CoR, SMRT, and mSin3A, and may act as a co-repressor for site-specific transcriptions factors. In this report we demonstrate that Gfi-1 interacts with ETO and related proteins both in vitro and in vivo and with histone deacetylase proteins in vivo. We observed that a portion of Gfi-1 and Gfi-1B associated with the nuclear matrix, as is the case with ETO. Moreover, Gfi-1 and ETO co-localize to punctate subnuclear structures. When co-expressed in mammalian cells, Gfi-1 associates with histone deacetylse-1 (HDAC-1), HDAC-2, and HDAC-3. These data identify ETO as a partner for Gfi-1 and Gfi-1B, and suggest that Gfi-1 proteins repress transcription through recruitment of histone deacetylase-containing complexes.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Histona Desacetilasas/metabolismo , Ácidos Hidroxámicos/metabolismo , Matriz Nuclear/metabolismo , Proteínas Represoras/metabolismo , Factores de Transcripción/metabolismo , Células Cultivadas , Proteínas de Unión al ADN/química , Inhibidores Enzimáticos/metabolismo , Inhibidores Enzimáticos/farmacología , Genes Reporteros/genética , Inhibidores de Histona Desacetilasas , Humanos , Ácidos Hidroxámicos/farmacología , Filamentos Intermedios/metabolismo , Proteínas Luminiscentes/metabolismo , Microscopía Fluorescente , Proteínas Proto-Oncogénicas/metabolismo , Proteína 1 Compañera de Translocación de RUNX1 , Proteínas Represoras/química , Transcripción Genética/fisiología , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA