Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cells ; 10(2)2021 02 09.
Artículo en Inglés | MEDLINE | ID: mdl-33572475

RESUMEN

Poly-ADP-ribose polymerases (PARPs) are enzymes that catalyze ADP-ribosylation and play critical roles in normal and disease settings. The PARP family member, PARP7, is a mono-ADP-ribosyltransferase that has been suggested to play a tumor suppressive role in breast, ovarian, and colorectal cancer. Here, we have investigated how androgen signaling regulates PARP7 homeostasis in prostate cancer cells, where PARP7 is a direct target gene of AR. We found that the PARP7 protein is extremely short-lived, with a half-life of 4.5 min. We show that in addition to its transcriptional regulation by AR, PARP7 is subject to androgen-dependent post-transcriptional regulation that increases its half-life to 25.6 min. This contrasts with PARP1, PARP2, PARP9, and PARP14, which do not display rapid turnover and are not regulated by androgen signaling. Androgen- and AR-dependent stabilization of PARP7 leads to accumulation in the nucleus, which we suggest is a major site of action. Mutations in the catalytic domain, the Cys3His1 zinc finger, and WWE (tryptophan-tryptophan-glutamate) domains in PARP7 each reduce the degradation rate of PARP7, suggesting the overall structure of the protein is tuned for its rapid turnover. Our finding that PARP7 is regulated by AR signaling both transcriptionally and post-transcriptionally in prostate cancer cells suggests the dosage of PARP7 protein is subject to tight regulation.


Asunto(s)
ADP Ribosa Transferasas/metabolismo , Andrógenos/metabolismo , Regulación de la Expresión Génica , Proteínas de Transporte de Nucleósidos/metabolismo , Neoplasias de la Próstata/enzimología , ADP Ribosa Transferasas/química , Animales , Línea Celular Tumoral , Núcleo Celular/metabolismo , Humanos , Masculino , Ratones , Proteínas de Transporte de Nucleósidos/genética , Neoplasias de la Próstata/patología , Dominios Proteicos , Estabilidad Proteica , Receptores Androgénicos/metabolismo , Transducción de Señal , Transcripción Genética
2.
Genes Dev ; 33(7-8): 388-402, 2019 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-30808659

RESUMEN

Tgif1 (thymine-guanine-interacting factor 1) and Tgif2 repress gene expression by binding directly to DNA or interacting with transforming growth factor (TGF) ß-responsive SMADs. Tgifs are essential for embryogenesis and may function in tumor progression. By analyzing both gain and loss of Tgif function in a well-established mouse model of intestinal cancer, we show that Tgifs promote adenoma growth in the context of mutant Apc (adenomatous polyposis coli). Despite the tumor-suppressive role of TGFß signaling, transcriptome profiling of colon tumors suggests minimal effect of Tgifs on the TGFß pathway. Instead, it appears that Tgifs, which are up-regulated in Apc mutant colon tumors, contribute to reprogramming metabolic gene expression. Integrating gene expression data from colon tumors with other gene expression and chromatin-binding data identifies a set of direct Tgif target genes encoding proteins involved in acetyl CoA and pyruvate metabolism. Analysis of both tumor and nontumor tissues indicates that these genes are targets of Tgif repression in multiple settings, suggesting that this is a core Tgif function. We propose that Tgifs play an important role in regulating basic energy metabolism in normal cells, and that this function of Tgifs is amplified in some cancers.


Asunto(s)
Acetilcoenzima A/genética , Adenoma , Regulación Neoplásica de la Expresión Génica/genética , Proteínas de Homeodominio/metabolismo , Neoplasias Intestinales , Proteínas Represoras/metabolismo , Adenoma/genética , Adenoma/fisiopatología , Poliposis Adenomatosa del Colon/genética , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Metabolismo Energético/genética , Células HCT116 , Humanos , Mucosa Intestinal/fisiopatología , Neoplasias Intestinales/genética , Neoplasias Intestinales/fisiopatología , Ratones , Ratones Endogámicos C57BL
3.
Biochim Biophys Acta Gene Regul Mech ; 1861(11): 983-995, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30312684

RESUMEN

Myelin transcription factor 1 (Myt1) and Myt1l (Myt1-like) are zinc finger transcription factors that regulate neuronal differentiation. Reduced Myt1l expression has been implicated in glioblastoma (GBM), and the related St18 was originally identified as a potential tumor suppressor for breast cancer. We previously analyzed changes in gene expression in a human GBM cell line with re-expression of either Myt1 or Myt1l. This revealed largely overlapping gene expression changes, suggesting similar function in these cells. Here we show that re-expression of Myt1 or Myt1l reduces proliferation in two different GBM cell lines, activates gene expression programs associated with neuronal differentiation, and limits expression of proliferative and epithelial to mesenchymal transition gene-sets. Consistent with this, expression of both MYT1 and MYT1L is lower in more aggressive glioma sub-types. Examination of the gene expression changes in cells expressing Myt1 or Myt1l suggests that both repress expression of the YAP1 transcriptional coactivator, which functions primarily in the Hippo signaling pathway. Expression of YAP1 and its target genes is reduced in Myt-expressing cells, and there is an inverse correlation between YAP1 and MYT1/MYT1L expression in human brain cancer datasets. Proliferation of GBM cell lines is reduced by lowering YAP1 expression and increased with YAP1 over-expression, which overcomes the anti-proliferative effect of Myt1/Myt1l expression. Finally we show that reducing YAP1 expression in a GBM cell line slows the growth of orthotopic tumor xenografts. Together, our data suggest that Myt1 and Myt1l directly repress expression of YAP1, a protein which promotes proliferation and GBM growth.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/fisiología , Neoplasias Encefálicas , Proliferación Celular , Proteínas de Unión al ADN/fisiología , Glioblastoma , Proteínas del Tejido Nervioso/fisiología , Fosfoproteínas/fisiología , Factores de Transcripción/fisiología , Animales , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Línea Celular , Glioblastoma/genética , Glioblastoma/metabolismo , Glioblastoma/patología , Humanos , Ratones Desnudos , Proteínas Señalizadoras YAP
4.
Atherosclerosis ; 275: 246-255, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29980051

RESUMEN

BACKGROUND AND AIMS: Transforming growth factor ß induced factor homeobox 1 (TGIF1) is a transcriptional repressor that limits the response to transforming growth factor ß signaling and also represses transcription independent of this pathway. Recently, we found higher serum cholesterol levels and more hepatic lipid accumulation in mice lacking Tgif1, and showed that TGIF1 can repress the expression of Soat2, the gene encoding the cholesterol esterifying enzyme acyl-Coenzyme A:cholesterol acyltransferase 2. Although there is evidence that TGIF1 plays a role in lipid metabolism, its role in this metabolic pathway is not fully characterized. Here we investigate whether overexpression of TGIF1 affects intestinal cholesterol absorption. METHODS AND RESULTS: TGIF1 was found to repress human and mouse Niemann-Pick C1 like 1 (Npc1l1) promoter activity in intestinal Caco2 cells. We also found TGIF1 to be able to oppose the induction of the promoter activity by sterol regulatory element binding protein 2 and hepatocyte nuclear factor 1α and 4α. To validate these effects of TGIF1 in vivo, we generated transgenic mice specifically overexpressing TGIF1 in the intestine (Villin-Tgif1). We observed lower intestinal expression levels of Npc1l1 that was associated with lower expression of ATP-binding cassette transporter (Abc) a1, Abcg5, and Abcg8. Villin-Tgif1 mice fed regular chow or a high-fat diet had lower levels of markers of intestinal cholesterol absorption than wild types. CONCLUSIONS: We suggest TGIF1 as a new player in intestinal cholesterol metabolism.


Asunto(s)
Colesterol en la Dieta/metabolismo , Proteínas de Homeodominio/metabolismo , Absorción Intestinal , Mucosa Intestinal/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Proteínas Represoras/metabolismo , Transportadoras de Casetes de Unión a ATP/metabolismo , Animales , Células CACO-2 , Colesterol 7-alfa-Hidroxilasa/metabolismo , Regulación hacia Abajo , Factor Nuclear 1-alfa del Hepatocito/genética , Factor Nuclear 1-alfa del Hepatocito/metabolismo , Factor Nuclear 4 del Hepatocito/genética , Factor Nuclear 4 del Hepatocito/metabolismo , Proteínas de Homeodominio/genética , Humanos , Masculino , Proteínas de la Membrana/genética , Proteínas de Transporte de Membrana/genética , Ratones Transgénicos , Proteínas Represoras/genética , Proteína 2 de Unión a Elementos Reguladores de Esteroles/genética , Proteína 2 de Unión a Elementos Reguladores de Esteroles/metabolismo , Regulación hacia Arriba
5.
PLoS Genet ; 14(5): e1007409, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29782499

RESUMEN

Although treatment options for localized prostate cancer (CaP) are initially effective, the five-year survival for metastatic CaP is below 30%. Mutation or deletion of the PTEN tumor suppressor is a frequent event in metastatic CaP, and inactivation of the transforming growth factor (TGF) ß signaling pathway is associated with more advanced disease. We previously demonstrated that mouse models of CaP based on inactivation of Pten and the TGFß type II receptor (Tgfbr2) rapidly become invasive and metastatic. Here we show that mouse prostate tumors lacking Pten and Tgfbr2 have higher expression of stem cell markers and genes indicative of basal epithelial cells, and that basal cell proliferation is increased compared to Pten mutants. To better model the primarily luminal phenotype of human CaP we mutated Pten and Tgfbr2 specifically in luminal cells, and found that these tumors also progress to invasive and metastatic cancer. Accompanying the transition to invasive cancer we observed de-differentiation of luminal tumor cells to an intermediate cell type with both basal and luminal markers, as well as differentiation to basal cells. Proliferation rates in these de-differentiated cells were lower than in either basal or luminal cells. However, de-differentiated cells account for the majority of cells in micro-metastases consistent with a preferential contribution to metastasis. We suggest that active TGFß signaling limits lineage plasticity in prostate luminal cells, and that de-differentiation of luminal tumor cells can drive progression to metastatic disease.


Asunto(s)
Linaje de la Célula/genética , Neoplasias de la Próstata/genética , Transducción de Señal/genética , Factor de Crecimiento Transformador beta/genética , Animales , Proliferación Celular/genética , Progresión de la Enfermedad , Células Epiteliales/metabolismo , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Mutación , Fosfohidrolasa PTEN/genética , Fosfohidrolasa PTEN/metabolismo , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Receptor Tipo II de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/genética , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Análisis de Supervivencia , Factor de Crecimiento Transformador beta/metabolismo
6.
J Cell Biochem ; 119(6): 4644-4655, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29291346

RESUMEN

Myt1 and Myt1l (Myelin transcription factor 1, and Myt1-like) are members of a small family of closely related zinc finger transcription factors, characterized by two clusters of C2HC zinc fingers. Both are widely expressed during early embryogenesis, but are largely restricted to expression within the brain in the adult. Myt1l, as part of a three transcription factor mix, can reprogram fibroblasts to neurons and plays a role in maintaining neuronal identity. Previous analyses have indicated roles in both transcriptional activation and repression and suggested that Myt1 and Myt1l may have opposing functions in gene expression. We show that when targeted to DNA via multiple copies of the consensus Myt1/Myt1l binding site Myt1 represses transcription, whereas Myt1l activates. By targeting via a heterologous DNA binding domain we mapped an activation function in Myt1l to an amino-terminal region that is poorly conserved in Myt1. However, genome wide analyses of the effects of Myt1 and Myt1l expression in a glioblastoma cell line suggest that the two proteins have largely similar effects on endogenous gene expression. Transcriptional repression is likely mediated by binding to DNA via the known consensus site, whereas this site is not associated with the transcriptional start sites of genes with higher expression in the presence of Myt1 or Myt1l. This work suggests that these two proteins function similarly, despite differences observed in analyses based on synthetic reporter constructs.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Regulación Neoplásica de la Expresión Génica , Glioblastoma/metabolismo , Proteínas de Neoplasias/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Elementos de Respuesta , Factores de Transcripción/metabolismo , Transcripción Genética , Células A549 , Proteínas de Unión al ADN/genética , Glioblastoma/genética , Glioblastoma/patología , Células HEK293 , Células HeLa , Humanos , Proteínas de Neoplasias/genética , Proteínas del Tejido Nervioso/genética , Factores de Transcripción/genética
7.
Prostate ; 77(15): 1452-1467, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28875501

RESUMEN

BACKGROUND: Phosphoinositide-3 (PI-3) kinase signaling has a pervasive role in cancer. One of the key effectors of PI-3 kinase signaling is AKT, a kinase that promotes growth and survival in a variety of cancers. Genetically engineered mouse models of prostate cancer have shown that AKT signaling is sufficient to induce prostatic epithelial neoplasia (PIN), but insufficient for progression to adenocarcinoma. This contrasts with the phenotype of mice with prostate-specific deletion of Pten, where excessive PI-3 kinase signaling induces both PIN and locally invasive carcinoma. We reasoned that additional PI-3 kinase effector kinases promote prostate cancer progression via activities that provide biological complementarity to AKT. We focused on the PKN kinase family members, which undergo activation in response to PI-3 kinase signaling, show expression changes in prostate cancer, and contribute to cell motility pathways in cancer cells. METHODS: PKN kinase activity was measured by incorporation of 32 P into protein substrates. Phosphorylation of the turn-motif (TM) in PKN proteins by mTOR was analyzed using the TORC2-specific inhibitor torin and a PKN1 phospho-TM-specific antibody. Amino acid substitutions in the TM of PKN were engineered and assayed for effects on kinase activity. Cell motility-related functions and PKN localization was analyzed by depletion approaches and immunofluorescence microscopy, respectively. The contribution of PKN proteins to prostate tumorigenesis was characterized in several mouse models that express PKN transgenes. The requirement for PKN activity in prostate cancer initiated by loss of phosphatase and tensin homolog deleted on chromosome 10 (Pten), and the potential redundancy between PKN isoforms, was analyzed by prostate-specific deletion of Pkn1, Pkn2, and Pten. RESULTS AND CONCLUSIONS: PKN1 and PKN2 contribute to motility pathways in human prostate cancer cells. PKN1 and PKN2 kinase activity is regulated by TORC2-dependent phosphorylation of the TM, which together with published data indicates that PKN proteins receive multiple PI-3 kinase-dependent inputs. Transgenic expression of active AKT and PKN1 is not sufficient for progression beyond PIN. Moreover, Pkn1 is not required for tumorigenesis initiated by loss of Pten. Triple knockout of Pten, Pkn1, and Pkn2 in mouse prostate results in squamous cell carcinoma, an uncommon but therapy-resistant form of prostate cancer.


Asunto(s)
Neoplasias de la Próstata/enzimología , Neoplasias de la Próstata/patología , Proteína Quinasa C/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Animales , Diferenciación Celular/fisiología , Progresión de la Enfermedad , Células HEK293 , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Fosfohidrolasa PTEN/metabolismo , Neoplasias de la Próstata/genética , Proteína Quinasa C/genética , Serina-Treonina Quinasas TOR/genética
8.
Mol Cell Biol ; 37(5)2017 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-27956704

RESUMEN

Mouse embryos conditionally lacking Tgif1 and Tgif2 have holoprosencephaly and defects in left-right asymmetry. To identify pathways affected by loss of Tgif function during embryogenesis, we performed transcriptome profiling on whole mouse embryos. Among the genes with altered expression in embryos lacking Tgifs were a number with links to cilium function. One of these, Evi5l, encodes a RabGAP that is known to block the formation of cilia when overexpressed. Evi5l expression is increased in Tgif1; Tgif2-null embryos and in double-null mouse embryo fibroblasts (MEFs). Knockdown of Tgifs in a human retinal pigment epithelial cell line also increased EVI5L expression. We show that TGIF1 binds to a conserved consensus TGIF site 5' of the human and mouse Evi5l genes and represses Evi5l expression. In primary MEFs lacking both Tgifs, the number of cells with primary cilia was significantly decreased, and we observed a reduction in the transcriptional response to Shh pathway activation. Reducing Evi5l expression in MEFs lacking Tgifs resulted in a partial restoration of cilium numbers and in the transcriptional response to activation of the Shh pathway. In summary, this work shows that Tgifs regulate ciliogenesis and suggests that Evi5l mediates at least part of this effect.


Asunto(s)
Proteínas Activadoras de GTPasa/genética , Proteínas de Homeodominio/metabolismo , Proteínas Represoras/metabolismo , Factores de Transcripción/genética , Animales , Línea Celular , Cilios/metabolismo , Embrión de Mamíferos/metabolismo , Proteínas Activadoras de GTPasa/metabolismo , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Ontología de Genes , Proteínas Hedgehog/metabolismo , Proteínas de Homeodominio/genética , Humanos , Ratones , Ratones Noqueados , Proteínas Represoras/genética , Transducción de Señal/genética , Factores de Transcripción/metabolismo
9.
Eur J Hum Genet ; 25(2): 208-215, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27924807

RESUMEN

Holoprosencephaly (HPE) is a prevalent craniofacial developmental disorder that has both genetic and environmental causes. The gene encoding TG-interacting factor 1 (TGIF1) is among those that are routinely screened in HPE patients. However, the mechanisms by which TGIF1 variants cause HPE are not fully understood. TGIF1 is a transcriptional repressor that limits the output of the Transforming Growth Factor ß (TGFß)/Nodal signaling pathway, and HPE in patients with TGIF1 variants has been suggested to be due to increased Nodal signaling. Mice lacking both Tgif1 and its paralog, Tgif2, have HPE, and embryos lacking Tgif function do not survive past mid-gestation. Here, we show that in the presence of a Nodal heterozygous mutation, proliferation defects are rescued and a proportion of embryos lacking all Tgif function survive to late gestation. However, these embryos have a classic HPE phenotype, suggesting that this is a Nodal-independent effect of Tgif loss of function. Further, we show that the Gli3 gene is a direct target for repression by Tgifs, independent of TGFß/Nodal signaling, consistent with Tgif mutations causing HPE via Nodal-independent effects on the Sonic Hedgehog (Shh) pathway. Based on this work, we propose a model for distinct functions of Tgifs in the Nodal and Shh/Gli3 pathways during forebrain development.


Asunto(s)
Holoprosencefalia/genética , Proteínas de Homeodominio/genética , Factores de Transcripción de Tipo Kruppel/genética , Proteínas del Tejido Nervioso/genética , Tubo Neural/embriología , Proteína Nodal/genética , Proteínas Represoras/genética , Animales , Línea Celular Tumoral , Células Cultivadas , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Proteínas de Homeodominio/metabolismo , Humanos , Factores de Transcripción de Tipo Kruppel/metabolismo , Ratones , Ratones Endogámicos C57BL , Mutación , Proteínas del Tejido Nervioso/metabolismo , Tubo Neural/metabolismo , Proteína Nodal/metabolismo , Proteínas Represoras/metabolismo , Transducción de Señal , Proteína Gli3 con Dedos de Zinc
10.
PLoS One ; 11(5): e0155837, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27187787

RESUMEN

Tgif1 and Tgif2 are transcriptional repressors that inhibit the transcriptional response to transforming growth factor ß signaling, and can repress gene expression by direct binding to DNA. Loss of function mutations in TGIF1 are associated with holoprosencephaly (HPE) in humans. In mice, embryos lacking both Tgif1 and Tgif2 fail to complete gastrulation, and conditional double null embryos that survive past gastrulation have HPE and do not survive past mid-gestation. Here we show that in mice of a relatively pure C57BL/6 strain background, loss of Tgif1 alone results in defective axial patterning and altered expression of Hoxc6. The primary defects in Tgif1 null embryos are the presence of extra ribs on the C7 vertebra, consistent with a posterior transformation phenotype. In addition we observed defective cervical vertebrae, primarily C1-C5, in both adult mice and embryos that lacked Tgif1. The combination of Tgif1 and Tgif2 mutations increases the severity and penetrance of the posterior transformation phenotype, without altering the type of defects seen. Similarly, exposure of Tgif1 mutant embryos to retinoic acid at E8.5 increased the severity and penetrance of the Tgif1 phenotype. This suggests that Tgif1 and Tgif2 regulate axial patterning and that reduced TGIF function sensitizes embryos to the effects of retinoic acid.


Asunto(s)
Tipificación del Cuerpo/fisiología , Proteínas de Homeodominio/fisiología , Proteínas Represoras/fisiología , Animales , Tipificación del Cuerpo/efectos de los fármacos , Tipificación del Cuerpo/genética , Vértebras Cervicales/anomalías , Vértebras Cervicales/embriología , Proteínas de Homeodominio/biosíntesis , Proteínas de Homeodominio/genética , Ratones , Ratones Endogámicos C57BL , Mutación , Proteínas Represoras/genética , Costillas/anomalías , Costillas/embriología , Esqueleto/anomalías , Tretinoina/farmacología
11.
PLoS One ; 9(3): e92800, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24651496

RESUMEN

Recent work with mouse models of prostate cancer (CaP) has shown that inactivation of TGFß signaling in prostate epithelium can cooperate with deletion of the Pten tumor suppressor to drive locally aggressive cancer and metastatic disease. Here, we show that inactivating the TGFß pathway by deleting the gene encoding the TGFß type II receptor (Tgfbr2) in combination with a deletion of the Apc tumor suppressor gene specifically in mouse prostate epithelium, results in the rapid onset of invasive CaP. Micro-metastases were observed in the lymph nodes and lungs of a proportion of the double mutant mice, whereas no metastases were observed in Apc single mutant mice. Prostate-specific Apc;Tgfbr2 mutants had a lower frequency of metastasis and survived significantly longer than Pten;Tgfbr2 double mutants. However, all Apc;Tgfbr2 mutants developed invasive cancer by 30 weeks of age, whereas invasive cancer was rarely observed in Apc single mutant animals, even by one year of age. Further comparison of the Pten and Apc models of CaP revealed additional differences, including adenosquamous carcinoma in the Apc;Tgfbr2 mutants that was not seen in the Pten model, and a lack of robust induction of the TGFß pathway in Apc null prostate. In addition to causing high-grade prostate intra-epithelial neoplasia (HGPIN), deletion of either Pten or Apc induced senescence in affected prostate ducts, and this restraint was overcome by loss of Tgfbr2. In summary, this work demonstrates that TGFß signaling restrains the progression of CaP induced by different tumor suppressor mutations, suggesting that TGFß signaling exerts a general tumor suppressive effect in prostate.


Asunto(s)
Proteína de la Poliposis Adenomatosa del Colon/deficiencia , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo , Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Adenocarcinoma/patología , Proteína de la Poliposis Adenomatosa del Colon/genética , Animales , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patología , Línea Celular , Senescencia Celular/genética , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Eliminación de Gen , Homocigoto , Queratina-10 , Masculino , Ratones , Ratones Noqueados , Mutación , Invasividad Neoplásica , Fosfohidrolasa PTEN/genética , Fosfohidrolasa PTEN/metabolismo , Fenotipo , Neoplasias de la Próstata/mortalidad , Neoplasias de la Próstata/patología , Proteínas Serina-Treonina Quinasas/genética , Receptor Tipo II de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/genética , Células del Estroma/metabolismo
12.
J Lipid Res ; 55(4): 709-17, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24478032

RESUMEN

Acat2 [gene name: sterol O-acyltransferase 2 (SOAT2)] esterifies cholesterol in enterocytes and hepatocytes. This study aims to identify repressor elements in the human SOAT2 promoter and evaluate their in vivo relevance. We identified TG-interacting factor 1 (Tgif1) to function as an important repressor of SOAT2. Tgif1 could also block the induction of the SOAT2 promoter activity by hepatocyte nuclear factor 1α and 4α. Women have ∼ 30% higher hepatic TGIF1 mRNA compared with men. Depletion of Tgif1 in mice increased the hepatic Soat2 expression and resulted in higher hepatic lipid accumulation and plasma cholesterol levels. Tgif1 is a new player in human cholesterol metabolism.


Asunto(s)
Silenciador del Gen , Proteínas de Homeodominio/fisiología , Proteínas Represoras/fisiología , Esterol O-Aciltransferasa/genética , Animales , Sitios de Unión , Línea Celular Tumoral , Represión Enzimática , Femenino , Cálculos Biliares/enzimología , Factor Nuclear 1-alfa del Hepatocito/fisiología , Factor Nuclear 4 del Hepatocito/fisiología , Proteínas de Homeodominio/metabolismo , Humanos , Lípidos/sangre , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Regiones Promotoras Genéticas , Unión Proteica , Caracteres Sexuales , Esterol O-Aciltransferasa/metabolismo , Factores de Transcripción/metabolismo , Transcripción Genética , Esterol O-Aciltransferasa 2
13.
PLoS One ; 7(4): e35460, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22514746

RESUMEN

Transforming growth factor ß (TGFß) signaling regulates cell cycle progression in several cell types, primarily by inducing a G1 cell cycle arrest. Tgif1 is a transcriptional corepressor that limits TGFß responsive gene expression. Here we demonstrate that primary mouse embryo fibroblasts (MEFs) lacking Tgif1 proliferate slowly, accumulate increased levels of DNA damage, and senesce prematurely. We also provide evidence that the effects of loss of Tgif1 on proliferation and senescence are not limited to primary cells. The increased DNA damage in Tgif1 null MEFs can be partially reversed by culturing cells at physiological oxygen levels, and growth in normoxic conditions also partially rescues the proliferation defect, suggesting that in the absence of Tgif1 primary MEFs are less able to cope with elevated levels of oxidative stress. Additionally, we show that Tgif1 null MEFs are more sensitive to TGFß-mediated growth inhibition, and that treatment with a TGFß receptor kinase inhibitor increases proliferation of Tgif1 null MEFs. Conversely, persistent treatment of wild type cells with low levels of TGFß slows proliferation and induces senescence, suggesting that TGFß signaling also contributes to cellular senescence. We suggest that in the absence of Tgif1, a persistent increase in TGFß responsive transcription and a reduced ability to deal with hyperoxic stress result in premature senescence in primary MEFs.


Asunto(s)
Senescencia Celular/fisiología , Fibroblastos/citología , Fibroblastos/metabolismo , Proteínas de Homeodominio/metabolismo , Proteínas Represoras/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Animales , Benzamidas/farmacología , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Senescencia Celular/genética , Dioxoles/farmacología , Fibroblastos/efectos de los fármacos , Proteínas de Homeodominio/genética , Ratones , Proteínas Represoras/genética , Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Factor de Crecimiento Transformador beta/genética
14.
J Cell Biochem ; 111(2): 380-90, 2010 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-20506222

RESUMEN

TG-interacting factor (Tgif1) represses gene expression by interaction with general corepressors, and can be recruited to target genes by transforming growth factor beta (TGFß) activated Smads, or by the retinoid X receptor (RXR). Here we show that Tgif1 interacts with the LXRα nuclear receptor and can repress transcription from a synthetic reporter activated by LXRα. In cultured cells reducing endogenous Tgif1 levels resulted in increased expression of LXRα target genes. To test the in vivo role of Tgif1, we analyzed LXRα-dependent gene expression in mice lacking Tgif1. In the livers of Tgif1 null mice, we observed significant derepression of the apolipoprotein genes, Apoa4 and Apoc2, suggesting that Tgif1 is an important in vivo regulator of apolipoprotein gene expression. In contrast, we observed relatively minimal effects on expression of other LXR target genes. This work suggests that Tgif1 can regulate nuclear receptor complexes, in addition to those containing retinoic acid receptors, but also indicates that there is some specificity to which NR target genes are repressed by Tgif1.


Asunto(s)
Apolipoproteínas/genética , Regulación de la Expresión Génica , Proteínas de Homeodominio/fisiología , Hígado/metabolismo , Proteínas Represoras/fisiología , Animales , Apolipoproteína C-II/análisis , Apolipoproteína C-II/genética , Apolipoproteínas/análisis , Apolipoproteínas A/análisis , Apolipoproteínas A/genética , Receptores X del Hígado , Ratones , Receptores Nucleares Huérfanos , Receptores Citoplasmáticos y Nucleares/metabolismo
15.
J Mol Biol ; 398(5): 657-71, 2010 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-20361981

RESUMEN

Pc2 (Cbx4) is a member of the chromobox family of polycomb proteins, and is a SUMO E3 ligase for the transcriptional corepressor CtBP1. Here, we show that both CtBP1 and Pc2 are phosphorylated by the kinase Akt1, which is activated by growth factor signaling via the PI3-kinase pathway. In the presence of Pc2, phosphorylation of CtBP1 is increased, and this requires interaction of both CtBP1 and Akt1 with Pc2. Pc2 promotes CtBP1 phosphorylation by recruiting Akt1 and, in part, by preventing de-phosphorylation of activated Akt1. Alteration of the Akt-phosphorylated residue in CtBP1 to a phosphomimetic results in decreased CtBP1 dimerization, but does not prevent interaction with other transcriptional regulators. The phosphomimetic mutant of CtBP1 is expressed at a lower level than the wild type protein, resulting in decreased transcriptional repression. We show that this CtBP1 mutant is targeted for poly-ubiquitylation and is less stable than the wild type protein. Co-expression of Pc2 and Akt1 results in both phosphorylation and ubiquitylation of CtBP1, thereby targeting CtBP1 for degradation. This work suggests that Pc2 might coordinate multiple enzymatic activities to regulate CtBP1 function.


Asunto(s)
Oxidorreductasas de Alcohol/antagonistas & inhibidores , Proteínas de Unión al ADN/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Represoras/metabolismo , Sustitución de Aminoácidos , Línea Celular , Perfilación de la Expresión Génica , Humanos , Ligasas , Mutagénesis Sitio-Dirigida , Fosforilación , Proteínas del Grupo Polycomb , Ubiquitina-Proteína Ligasas , Ubiquitinación
16.
PLoS One ; 5(1): e8794, 2010 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-20098713

RESUMEN

BACKGROUND: Modification of proteins by the small ubiquitin like modifier (SUMO) is an essential process in mammalian cells. SUMO is covalently attached to lysines in target proteins via an enzymatic cascade which consists of E1 and E2, SUMO activating and conjugating enzymes. There is also a variable requirement for non-enzymatic E3 adapter like proteins, which can increase the efficiency and specificity of the sumoylation process. In addition to covalent attachment of SUMO to target proteins, specific non-covalent SUMO interaction motifs (SIMs) that are generally short hydrophobic peptide motifs have been identified. METHODOLOGY/PRINCIPAL FINDINGS: Intriguingly, consensus SIMs are present in most SUMO E3s, including the polycomb protein, Pc2/Cbx4. However, a role for SIMs in SUMO E3 activity remains to be shown. We show that Pc2 contains two functional SIMs, both of which contribute to full E3 activity in mammalian cells, and are also required for sumoylation of Pc2 itself. Pc2 forms distinct sub-nuclear foci, termed polycomb bodies, and can recruit partner proteins, such as the corepressor CtBP. We demonstrate that mutation of the SIMs in Pc2 prevents Pc2-dependent CtBP sumoylation, and decreases enrichment of SUMO1 and SUMO2 at polycomb foci. Furthermore, mutational analysis of both SUMO1 and SUMO2 reveals that the SIM-interacting residues of both SUMO isoforms are required for Pc2-mediated sumoylation and localization to polycomb foci. CONCLUSIONS/SIGNIFICANCE: This work provides the first clear evidence for a role for SIMs in SUMO E3 activity.


Asunto(s)
Proteínas Represoras/metabolismo , Proteína SUMO-1/metabolismo , Secuencia de Aminoácidos , Sitios de Unión , Western Blotting , Cromatografía de Afinidad , Humanos , Inmunoprecipitación , Ligasas , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Proteínas del Grupo Polycomb , Proteínas Represoras/química , Proteínas Represoras/genética , Proteína SUMO-1/química , Proteína SUMO-1/genética , Homología de Secuencia de Aminoácido , Ubiquitina-Proteína Ligasas
17.
Development ; 137(2): 249-59, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20040491

RESUMEN

Tgif1 and Tgif2 are transcriptional co-repressors that limit the response to TGFbeta signaling and play a role in regulating retinoic-acid-mediated gene expression. Mutations in human TGIF1 are associated with holoprosencephaly, but it is unclear whether this is a result of deregulation of TGFbeta/Nodal signaling, or of effects on other pathways. Surprisingly, mutation of Tgif1 in mice results in only relatively mild developmental phenotypes in most strain backgrounds. Here, we show that loss-of-function mutations in both Tgif1 and Tgif2 result in a failure of gastrulation. By conditionally deleting Tgif1 in the epiblast, we demonstrate that a single wild-type allele of Tgif1 in the extra-embryonic tissue allows the double null embryos to gastrulate and begin organogenesis, suggesting that extra-embryonic Tgif function is required for patterning the epiblast. Genetically reducing the dose of Nodal in embryos lacking all Tgif function results in partial rescue of the gastrulation defects. Conditional double null embryos have defects in left-right asymmetry, which are also alleviated by reducing the dose of Nodal. Together, these data show that Tgif function is required for gastrulation, and provide the first clear evidence that Tgifs limit the transcriptional response to Nodal signaling during early embryogenesis.


Asunto(s)
Gastrulación/fisiología , Proteínas de Homeodominio/metabolismo , Transducción de Señal , Animales , Línea Celular Tumoral , Embrión de Mamíferos/metabolismo , Gastrulación/genética , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/genética , Humanos , Hibridación in Situ , Ratones , Ratones Mutantes
18.
Dev Biol ; 319(2): 285-97, 2008 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-18508043

RESUMEN

The mammalian placenta is the site of exchange of nutrients and waste between mother and embryo. In humans, placental insufficiency can result in intrauterine growth retardation, perinatal death and spontaneous abortion. We show that in C57BL/6J mice a null mutation in the gene encoding the transcriptional corepressor, Tgif, causes placental defects. The major defects are decreased vascularization of the placenta, due to a decrease in the fetal blood vessels, and decreased expression of the gap junction protein Gjb2 (Cx26). These defects result in severe growth retardation in a proportion of Tgif null embryos in Tgif heterozygous mothers, and an overall growth delay in Tgif null animals. Placental defects are much more severe if the mother also completely lacks Tgif function, and placentas from heterozygous Tgif embryos are defective in a Tgif null mother. Embryo transfer experiments show that even the placenta from a wild type embryo is compromised in the absence of maternal Tgif. These results demonstrate that Tgif functions in the normal development of the placenta, and suggest a role for maternal factors in regulating the morphogenesis of embryonically-derived placental tissues.


Asunto(s)
Embrión de Mamíferos/fisiología , Proteínas de Homeodominio/fisiología , Placenta/irrigación sanguínea , Proteínas Represoras/fisiología , Animales , Peso Corporal , Supervivencia Celular , Conexina 26 , Conexinas , Transferencia de Embrión , Embrión de Mamíferos/citología , Desarrollo Embrionario/genética , Desarrollo Embrionario/fisiología , Femenino , Proteínas de Homeodominio/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Morfogénesis , Tamaño de los Órganos , Placenta/anatomía & histología , Embarazo , Proteínas Represoras/genética
19.
BMC Mol Biol ; 7: 2, 2006 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-16436215

RESUMEN

BACKGROUND: TGIF and TGIF2 are homeodomain proteins, which act as TGFbeta specific Smad transcriptional corepressors. TGIF recruits general repressors including mSin3 and CtBP. The related TGIF2 protein functions in a similar manner, but does not bind CtBP. In addition to repressing TGFbeta activated gene expression, TGIF and TGIF2 repress gene expression by binding directly to DNA. TGIF and TGIF2 share two major blocks of similarity, encompassing the homeodomain, and a conserved carboxyl terminal repression domain. Here we characterize two splice variants of the Tgif2 gene from mouse and demonstrate that the Tgif2 gene contains a retained intron. RESULTS: By PCR from mouse cDNA, we identified two alternate splice forms of the Tgif2 gene. One splice variant encodes the full length 237 amino acid Tgif2, whereas the shorter form results in the removal of 39 codons from the centre of the coding region. The generation of this alternate splice form occurs with the mouse RNA, but not the human, and both splice forms are present in all mouse tissues analyzed. Human and mouse Tgif2 coding sequences contain a retained intron, which in mouse Tgif2 is removed by splicing from around 25-50% of RNAs, as assessed by RT-PCR. This splicing event is dependent on sequences within the mouse Tgif2 coding sequence. Both splice forms of mouse Tgif2 encode proteins which are active transcriptional repressors, and can repress both TGFbeta dependent and independent transcription. In addition, we show that human and mouse Tgif2 interact with the transcriptional corepressor mSin3. CONCLUSION: These data demonstrate that the Tgif2 gene contains a retained intron, within the second coding exon. This retained intron is not removed from the human mRNA at a detectable level, but is spliced out in a significant proportion of mouse RNAs. This alternate splicing is dependent entirely on sequences within the mouse Tgif2 coding sequence, suggesting the presence of an exonic splicing enhancer. Both splice forms of mouse Tgif2 produce proteins which are functional transcriptional repressors.


Asunto(s)
Proteínas de Homeodominio/genética , Intrones/genética , Ratones/genética , Proteínas Represoras/genética , Empalme Alternativo , Animales , Células COS , Carcinoma Hepatocelular/patología , Chlorocebus aethiops , ADN Complementario/genética , Exones/genética , Humanos , Neoplasias Hepáticas/patología , ARN Mensajero/genética , Proteínas Recombinantes de Fusión/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transfección
20.
Mol Cell Biol ; 26(3): 990-1001, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16428452

RESUMEN

TGIF (TG-interacting factor) represses transforming growth factor beta (TGF-beta)-activated gene expression and can repress transcription via a specific retinoid response element. Mutations in human TGIF are associated with holoprosencephaly, a severe defect of craniofacial development with both genetic and environmental causes. Both TGF-beta and retinoic acid signaling are implicated in craniofacial development. Here, we analyze the role of TGIF in regulating retinoid responsive gene expression. We demonstrate that TGIF interacts with the ligand binding domain of the RXRalpha retinoid receptor and represses transcription from retinoid response elements. TGIF recruits the general corepressor, CtBP, to RXRalpha, and this recruitment is required for full repression by TGIF. Interaction between TGIF and RXRalpha is reduced by the addition of retinoic acid, consistent with a role for TGIF as an RXRalpha transcriptional corepressor. We created a Tgif null mutation in mice and tested the sensitivity of mutant mice to increased levels of retinoic acid. Tgif mutant embryos are more sensitive to retinoic acid-induced teratogenesis, and retinoid target genes are expressed at a higher level in tissues from Tgif null mice. These results demonstrate an important role for TGIF as a transcriptional corepressor, which regulates developmental signaling by retinoic acid, and raises the possibility that TGIF may repress other RXR-dependent transcriptional responses.


Asunto(s)
Regulación de la Expresión Génica , Proteínas de Homeodominio/metabolismo , Proteínas Represoras/metabolismo , Receptor alfa X Retinoide/antagonistas & inhibidores , Receptor alfa X Retinoide/genética , Tretinoina/metabolismo , Animales , Antineoplásicos/farmacología , Dimerización , Resistencia a Antineoplásicos/genética , Embrión de Mamíferos/efectos de los fármacos , Desarrollo Embrionario/genética , Femenino , Proteínas de Homeodominio/genética , Masculino , Ratones , Ratones Mutantes , Proteínas Represoras/genética , Elementos de Respuesta/genética , Receptor alfa X Retinoide/metabolismo , Eliminación de Secuencia , Transcripción Genética/efectos de los fármacos , Tretinoina/farmacología , Tretinoina/toxicidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA