Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
PLoS One ; 12(1): e0169338, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28046085

RESUMEN

The mier family consists of three related genes encoding ELM2-SANT containing proteins. MIER1 has been well characterized and is known to function in transcriptional repression through its ability to recruit HDAC1 and 2. Little is known about MIER2 or MIER3 function and no study characterizing these two proteins has been published. In this report, we investigate MIER2 and MIER3 localization and function. Confocal analysis revealed that, while MIER2 and MIER3 are mainly nuclear proteins, a substantial proportion (32%) of MIER2 is localized in the cytoplasm. Co-immunoprecipitation experiments demonstrated that the MIER proteins do not dimerize; that MIER2, but not MIER3, can recruit HDACs; and that recruitment is cell line-dependent. MIER2 was associated with HDAC1 and HDAC2 in HEK293 cells, but only with HDAC1 in MCF7 and HeLa cells. Little or no MIER3 co-immunoprecipitated with either HDAC1 or 2 in any of the three cell lines tested. By contrast, HDAC1 and 2 were readily detected in MIER1α complexes in all three cell lines. Histone deacetylase assays confirmed that MIER2, but not MIER3 complexes, have associated deacetylase activity, leading to the conclusion that MIER3 does not function in HDAC recruitment in these cell lines. In contrast to what has been reported for other ELM2-SANT associated HDACs, addition of D-myo-inositol-1,4,5,6-tetrakisphosphate led to only a small increase in MIER1α associated deacetylase activity and no effect on that associated with MIER2. Deletion analysis revealed that HDAC recruitment occurs through the ELM2 domain. Finally, using site-directed mutagenesis, we show that, like MIER1, 228W in the ELM2 domain is a critical residue for HDAC recruitment by MIER2.


Asunto(s)
Histona Desacetilasa 1/metabolismo , Histona Desacetilasa 2/metabolismo , Proteínas Nucleares/metabolismo , Factores de Transcripción/metabolismo , Secuencia de Aminoácidos , Núcleo Celular/metabolismo , Proteínas de Unión al ADN , Células HEK293 , Células HeLa , Humanos , Inmunoprecipitación , Fosfatos de Inositol/metabolismo , Células MCF-7 , Complejos Multiproteicos/metabolismo , Proteínas Nucleares/química , Unión Proteica , Dominios Proteicos , Transporte de Proteínas , Alineación de Secuencia , Eliminación de Secuencia , Homología de Secuencia de Aminoácido , Fracciones Subcelulares/metabolismo , Factores de Transcripción/química
2.
BMC Res Notes ; 8: 356, 2015 Aug 18.
Artículo en Inglés | MEDLINE | ID: mdl-26281834

RESUMEN

BACKGROUND: MIER1α is a transcriptional regulator that interacts with estrogen receptor α and inhibits estrogen-stimulated growth of breast carcinoma cells. Interestingly, analysis of MIER1α subcellular localization in breast samples revealed a stepwise shift from the nucleus to the cytoplasm during progression to invasive carcinoma. Previously, we demonstrated that MIER1α is nuclear in MCF7 cells yet it does not contain a nuclear localization signal. Instead MIER1α is targeted to the nucleus through interaction and co-transport with HDAC 1 and 2. RESULTS: In this study, we demonstrate that treatment of MCF7 breast carcinoma cells with either insulin or insulin-like growth factor affects the subcellular localization of MIER1α. Both factors reduce the percentage of cells with nuclear MIER1α from 81 and 89 to 41 and 56%, respectively. Treatment with 17ß-estradiol, on the other hand, had no effect and MIER1α remained nuclear. CONCLUSIONS: Our data demonstrate that insulin and IGF-1 can contribute to loss of nuclear MIER1α in the MCF7 breast carcinoma cell line.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Factor I del Crecimiento Similar a la Insulina/farmacología , Insulina/farmacología , Proteínas Nucleares/genética , Factores de Transcripción/genética , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Citosol/efectos de los fármacos , Citosol/metabolismo , Proteínas de Unión al ADN , Estradiol/farmacología , Femenino , Histona Desacetilasa 1/genética , Histona Desacetilasa 1/metabolismo , Histona Desacetilasa 2/genética , Histona Desacetilasa 2/metabolismo , Humanos , Células MCF-7 , Proteínas Nucleares/metabolismo , Transporte de Proteínas , Transducción de Señal , Factores de Transcripción/metabolismo
3.
PLoS One ; 8(12): e84046, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24376786

RESUMEN

MIER1α is a transcriptional regulator that functions in gene repression through its ability to interact with various chromatin modifiers and transcription factors. We have also shown that MIER1α interacts with ERα and inhibits estrogen-stimulated growth. While MIER1α is localized in the nucleus of MCF7 cells, previous studies have shown that it does not contain a nuclear localization signal. In this report, we investigate the mechanism involved in transporting MIER1α into the nucleus. We explored the possibility that MIER1α is transported into the nucleus through a 'piggyback' mechanism. One obvious choice is via interaction with ERα, however we demonstrate that nuclear targeting of MIER1α does not require ERα. Knockdown of ERα reduced protein expression to 22% of control, but did not alter the percentage of cells with nuclear MIER1α (98% nuclear with scrambled shRNA vs. 95% with ERα shRNA). Further evidence was obtained using two stable transfectants derived from the ER-negative MDA231 cell line: MC2 (ERα+) and VC5 (ERα-). Confocal analysis showed no difference in MIER1α localization (86% nuclear in MC2 vs. 89% in VC5). These data demonstrate that ERα is not involved in nuclear localization of MIER1α. To identify the critical MIER1α sequence, we performed a deletion analysis and determined that the ELM2 domain was necessary and sufficient for nuclear localization. This domain binds HDAC1 & 2, therefore we investigated their role. Confocal analysis of an MIER1α containing an ELM2 point mutation previously shown to abolish HDAC binding revealed that this mutation results in almost complete loss of nuclear targeting: 10% nuclear vs. 97% with WT-MIER1α. Moreover, double knockdown of HDAC1 and 2 caused a reduction in percent nuclear from 86% to 44%. The results of this study demonstrate that nuclear targeting of MIER1α requires an intact ELM2 domain and is dependent on interaction with HDAC1/2.


Asunto(s)
Neoplasias de la Mama/patología , Núcleo Celular/metabolismo , Histona Desacetilasa 1/metabolismo , Histona Desacetilasa 2/metabolismo , Proteínas Nucleares/metabolismo , Factores de Transcripción/metabolismo , Transporte Activo de Núcleo Celular , Proteínas de Unión al ADN , Humanos , Células MCF-7 , Proteínas Nucleares/química , Estructura Terciaria de Proteína , Factores de Transcripción/química
4.
BMC Endocr Disord ; 13: 35, 2013 Sep 23.
Artículo en Inglés | MEDLINE | ID: mdl-24053729

RESUMEN

BACKGROUND: Ghrelin and peptide YY (PYY) are appetite regulating hormones secreted from the gastrointestinal tract (gut). Aside from their known effect on energy homeostasis, accumulating data indicates that these gut hormones also affect bone metabolism. However, data regarding the influence of ghrelin and PYY on bone density in humans is very limited, and the results are inconclusive. Therefore, this study was designed to investigate the potential association between circulating ghrelin and PYY with bone density indices in the general population. METHODS: A total of 2257 adult subjects from the CODING (Complex Diseases in the Newfoundland Population: Environment and Genetics) study participated in this investigation. Acylated ghrelin and total PYY were measured in serum after a 12-hour fasting, with the Enzyme- Linked Immunosorbent Assay (ELISA) method. Bone mineral density was measured by dual-energy X-ray absorptiometry at the spine, femoral neck, and total hip. Multiple regression analyses adjusting for age, BMI, physical activity, smoking, and alcohol consumption were employed to analyze the association between serum ghrelin and PYY with bone mineral density parameters. RESULTS: Significant positive associations of ghrelin concentration with L2-L4 BMD, L2-L4 Z-score, femoral neck BMD, femoral neck Z-score, total hip BMD, and total hip Z-score were found in women. No significant correlations between ghrelin and bone density indices were present in men. After dividing the female group into pre-menopausal and post-menopausal, ghrelin was positively correlated with femoral neck Z-score, and total hip Z-score in pre-menopausal women and L2-L4 BMD, and Z-score in post-menopausal group. Moreover, no significant association was discovered between serum PYY and bone density at any site. CONCLUSION: Our results suggest a beneficial association of circulating ghrelin concentration with bone density in women at the population level. This association is independent of major confounding factors including BMI, physical activity, age, alcohol consumption, and smoking. Effect of menopause on this association seemed to be site specific. However, PYY does not seem to be associated with bone density parameters.

5.
J Mol Histol ; 44(4): 469-79, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23277184

RESUMEN

MIER1 is a transcriptional regulator that exists as several isoforms. Of particular interest is the MIER1α isoform, which contains in its unique C-terminus an LXXLL motif for interaction with nuclear hormone receptors. Indeed, MIER1α has been shown to interact with ERα and inhibit estrogen-stimulated growth of breast carcinoma cells. Moreover, the subcellular localization of MIER1α changes dramatically, from nuclear to cytoplasmic, during progression to invasive breast carcinoma. While human MIER1 RNA and protein expression pattern data have been posted on several websites, none of these studies use probes or antibodies that distinguish between the α and ß isoforms. We report here the first immunohistochemical study of the MIER1α protein expression pattern in human tissues. Our analysis revealed intense staining of specific cell types within virtually every endocrine and reproductive tissue except for the thyroid gland. In particular, we detected intense staining of ovarian follicles and germinal epithelium, ductal epithelial cells of the breast, pancreatic islet cells, all areas of the anterior pituitary and all zones of the adrenal cortex; moderate staining of germ cells and Leydig cells within the testis, patches of chromaffin cells in the adrenal medulla and weak staining of the fibromuscular stroma within the prostate. Immunoreactivity was limited to the cytoplasm in all positive cells except for oocytes and germinal epithelial cells in which the nucleus was also stained and in ductal epithelial cells of the breast in which staining was exclusively nuclear. In general, non-endocrine tissues were negative, however a few exceptions were noted. These included hepatocytes, myocardial fibers and neurons in all regions of the brain examined, with the exception of the thalamus. Neuronal staining was restricted to the cell bodies and dendrites, as most axons were negative. These data suggest that human MIER1α functions specifically in endocrine tissues and in a limited number of non-endocrine organs.


Asunto(s)
Receptor alfa de Estrógeno/metabolismo , Proteínas Nucleares/metabolismo , Factores de Transcripción/metabolismo , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Niño , Preescolar , Proteínas de Unión al ADN , Femenino , Humanos , Masculino , Persona de Mediana Edad , Especificidad de Órganos , Unión Proteica , Adulto Joven
6.
PLoS One ; 7(2): e32499, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22384264

RESUMEN

MIER1 was originally identified in a screen for novel fibroblast growth factor activated early response genes. The mier1 gene gives rise to multiple transcripts encoding protein isoforms that differ in their amino (N-) and carboxy (C-) termini. Much of the work to date has focused on the two C-terminal variants, MIER1α and ß, both of which have been shown to function as transcriptional repressors. Our previous work revealed a dramatic shift in MIER1α subcellular localization from nuclear in normal breast tissue to cytoplasmic in invasive breast carcinoma, suggesting that loss of nuclear MIER1α may play a role in breast cancer development. In the present study, we investigated whether alternative splicing to include a cassette exon and produce an N-terminal variant of MIER1α affects its subcellular localization in MCF7 breast carcinoma cells. We demonstrate that this cassette exon, exon 3A, encodes a consensus leucine-rich nuclear export signal (NES). Inclusion of this exon in MIER1α to produce the MIER1-3Aα isoform altered its subcellular distribution in MCF7 cells from 81% nuclear to 2% nuclear and this change in localization was abrogated by mutation of critical leucines within the NES. Treatment with leptomycin B (LMB), an inhibitor of the nuclear export receptor CRM1, resulted in a significant increase in the percentage of cells with nuclear MIER1-3Aα, from 4% to 53%, demonstrating that cytoplasmic localization of this isoform was due to CRM1-dependent nuclear export. Inclusion of exon 3A in MIER1ß to produce the N-terminal variant MIER1-3Aß however had little effect on the nuclear targeting of this isoform. Our results demonstrate that alternative splicing to include exon 3A specifically affects the localization pattern of the α isoform.


Asunto(s)
Empalme Alternativo , Neoplasias de la Mama/metabolismo , Regulación Neoplásica de la Expresión Génica , Proteínas Nucleares/biosíntesis , Proteínas Nucleares/genética , Factores de Transcripción/biosíntesis , Factores de Transcripción/genética , Transporte Activo de Núcleo Celular/genética , Línea Celular Tumoral , Codón , Proteínas de Unión al ADN , Exones , Femenino , Humanos , Mutagénesis Sitio-Dirigida , Proteínas Nucleares/química , Plásmidos/metabolismo , Isoformas de Proteínas , Estructura Terciaria de Proteína , Empalme del ARN , Factores de Transcripción/química
8.
BMC Res Notes ; 1: 68, 2008 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-18721470

RESUMEN

BACKGROUND: Mier1 encodes a novel transcriptional regulator and was originally isolated as a fibroblast growth factor early response gene. Two major protein isoforms have been identified, MIER1alpha and beta, which differ in their C-terminal sequence. Previously, we demonstrated that both isoforms recruit histone deacetylase 1 (HDAC1) to repress transcription. To further explore the role of MIER1 in chromatin remodeling, we investigated the functional interaction of MIER1 with the histone acetyltransferase (HAT), Creb-binding protein (CBP). FINDINGS: Using GST pull-down assays, we demonstrate that MIER1 interacts with CBP and that this interaction involves the N-terminal half (amino acids 1-283) of MIER1, which includes the acidic activation and ELM2 domains and the C-terminal half (amino acids 1094-2441) of CBP, which includes the bromo-, HAT, C/H3 and glutamine-rich domains. Functional analysis, using HEK293 cells, shows that the CBP bound to MIER1 in vivo has no detectable HAT activity. Histone 4 peptide binding assays demonstrate that this inhibition of HAT activity is not the result of interference with histone binding. CONCLUSION: Our data indicate that an additional mechanism by which MIER1 could repress transcription involves the inhibition of histone acetyltransferase activity.

9.
J Mol Histol ; 39(1): 15-24, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17622490

RESUMEN

MI-ER1 is a novel transcriptional regulator that plays a critical role in embryonic development and is differentially expressed in breast carcinoma. The MI-ER1 protein sequence is highly conserved among species, with 95% identity between mouse and humans and 72% between Xenopus and mouse. There are two major protein isoforms, MI-ER1alpha and MI-ER1beta, which differ in the sequence of their C-terminus. MI-ER1alpha is of particular interest because it contains a consensus LXXLL nuclear receptor interaction motif and the current study was undertaken to determine the expression pattern of MI-ER1alpha protein in adult mouse tissues. Immunohistochemical analysis of paraffin-embedded tissue using an MI-ER1alpha-specific antibody revealed that the majority of mouse adult tissues examined showed very weak or no immunoreactivity; these included tissues of the lung, liver, intestine, uterus, spleen, lymph node, bladder as well as skeletal muscle. Interestingly, a subset of endocrine tissues displayed intense staining for MI-ER1alpha. Specifically, the islets of Langerhans, the zona glomerulosa and medulla of the adrenal gland, the ovary and the hypothalamus were intensely stained. In addition, both anterior and posterior pituitary showed moderate immunoreactivity, as did the parafollicular cells of the thyroid gland and Leydig cells and spermatids in the testes. Negative endocrine tissues included follicular cells of the thyroid gland and the X zone of the adrenal cortex. A few non-endocrine tissues displayed moderate immunoreactivity; these included all tubules and collecting ducts in the kidney, myocardial and endocardial layers of the heart, the hippocampal formation, pyramidal neurons in the cortex and the ductal epithelium of the mammary gland. In all cases, MI-ER1alpha immunoreactivity was cytoplasmic. This study represents the first immunohistochemical analysis of MI-ER1alpha expression in mammals and our data suggest that this transcriptional regulator plays a role in specific endocrine pathways.


Asunto(s)
Proteínas Inmediatas-Precoces/metabolismo , Factores de Transcripción/metabolismo , Animales , Encéfalo/citología , Encéfalo/metabolismo , Sistema Endocrino/citología , Sistema Endocrino/metabolismo , Femenino , Genitales/citología , Genitales/metabolismo , Masculino , Ratones , Especificidad de Órganos
10.
Dev Biol ; 311(2): 383-95, 2007 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-17920056

RESUMEN

The Xenopus egg has a yolk-laden vegetal hemisphere juxtaposed to a darkly pigmented animal hemisphere. Mesoderm is derived from the marginal zone, located at the interface between the two hemispheres. The vegetal-most cells become endoderm and release TGF-beta-related factors, including the Xenopus Nodal related (Xnr) proteins, which diffuse to induce the marginal zone to form mesoderm. The remaining animal cells become ectoderm, but our understanding of the mechanisms that limit the response to induction is incomplete. In this study, we provide evidence to suggest that Xrel3, a member of the Rel/NF-kappaB family, plays a role in defining the boundary separating induced from uninduced cells by regulating Xnr-responsive gene transcription. Ectopic Xrel3 expressed in prospective mesoderm caused repression of mesoderm-specific genes resulting in loss-of-function phenotypes that were rescued by co-expression of Xnr2. Depletion of Xrel3 from embryos with antisense morpholinos increased Xnr-dependent transcription, broadened expression of the pan-mesoderm marker Xbra and sensitized animal cells to mesoderm induction by Xnr2. We propose that an additional component to the mechanism that differentiates the ectoderm from the mesoderm involves regulation of nodal-dependent gene transcription by Xrel3.


Asunto(s)
Inducción Embrionaria , Mesodermo/fisiología , Factores de Transcripción/metabolismo , Proteínas de Xenopus/metabolismo , Xenopus laevis/embriología , Xenopus laevis/metabolismo , Activinas/genética , Activinas/metabolismo , Animales , Biomarcadores/metabolismo , Factor 2 de Crecimiento de Fibroblastos/genética , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Regulación del Desarrollo de la Expresión Génica , Genes Reporteros , Humanos , Mesodermo/citología , FN-kappa B/genética , FN-kappa B/metabolismo , Proteína Nodal , Oligonucleótidos Antisentido/genética , Oligonucleótidos Antisentido/metabolismo , Oocitos/citología , Oocitos/fisiología , Proteínas de Dominio T Box/genética , Proteínas de Dominio T Box/metabolismo , Factores de Transcripción/genética , Transcripción Genética , Factor de Crecimiento Transformador beta , Proteínas de Xenopus/genética , Xenopus laevis/anatomía & histología
11.
DNA Seq ; 16(3): 237-40, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16147882

RESUMEN

Mi-er1 is a fibroblast growth factor immediate-early gene whose expression is differentially regulated in breast tumours. MI-ER1 functions as a transcriptional repressor of a number of genes, including Sp1 target genes. The Xenopus and human orthologs have been described and here we report the characterization of the mouse gene and its products. Mouse mi-er1 is a single copy gene located on chromosome 4. It has the same intron-exon structure as the human gene with the exception of exon 3A, in which an upstream 3' splice acceptor is utilized. As described in humans, multiple transcripts are produced in the mouse, and we have isolated the mouse orthologs of the human N1-beta, N2-beta and N3-beta, all of, which differ in their N-terminal sequence. Furthermore, we have isolated a novel isoform, N4-beta, containing sequence from an additional exon located between exon 4 and exon 5 that produces an fourth alternate N-terminus. The human mi-er1 transcripts also include isoforms encoding two alternate C-termini, alpha and beta. Like Xenopus, only isoforms containing the beta C-terminus have been detected in the mouse. Expression analysis, using a panel of mouse tissues and embryos, revealed that the N1-beta and N3-beta are ubiquitously expressed while N4-beta is only expressed in testis. N2-beta is expressed in most tissues but was not detected in heart, brain, eye or skeletal muscle. Sequence comparison revealed 95% identity between mouse and human MI-ER1 isoforms and 72% identity between mouse and Xenopus. The most conserved region in the MI-ER1 protein is the SANT domain, which is identical in all three species. Further analysis of the SANT domain using sequences retrieved from the genome databases for rat, cow, chicken, zebrafish, trout and Xenopustropicalis revealed that this domain is highly conserved, with 88% identity among the 9 species. Moreover, an additional 10 residues C-terminal to the published end of this domain are 100% conserved, suggesting that in MI-ER1, the functional domain includes this extended sequence.


Asunto(s)
Proteínas Inmediatas-Precoces/biosíntesis , Proteínas Inmediatas-Precoces/genética , Proteínas Nucleares/biosíntesis , Proteínas Nucleares/genética , Secuencia de Aminoácidos , Animales , Clonación Molecular , Proteínas de Unión al ADN , Exones , Etiquetas de Secuencia Expresada , Factores de Crecimiento de Fibroblastos/metabolismo , Humanos , Intrones , Ratones , Datos de Secuencia Molecular , Músculo Esquelético/metabolismo , Reacción en Cadena de la Polimerasa , Isoformas de Proteínas , Estructura Terciaria de Proteína , Homología de Secuencia de Aminoácido , Especificidad de la Especie , Distribución Tisular , Factores de Transcripción/metabolismo , Xenopus
12.
Int J Biochem Cell Biol ; 37(2): 463-77, 2005 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15474990

RESUMEN

Xmi-er1 is a fibroblast growth factor regulated immediate-early gene that is activated during mesoderm induction in Xenopus embryonic explants. This gene encodes a nuclear protein with potent transcriptional regulator activity and overexpression of XMI-ER1 in Xenopus embryos inhibits mesoderm induction and leads to truncations along the anteroposterior axis. We showed previously that XMI-ER1 is retained in the cytoplasm during cleavage stages and only begins to appear in the nucleus at mid-blastula. Such developmentally regulated nuclear translocation may represent an important mechanism for regulating XMI-ER1 activity in the early embryo. Here, we investigate different mechanisms that might control nuclear translocation of XMI-ER1. Using alpha-amanitin to inhibit transcription, we show that nuclear localization is not dependent on zygotic transcription. Nor is it the result of a developmentally regulated import pathway, as the XMI-ER1 nuclear localization signal (NLS) fused to beta-galactosidase (betagal) was able to direct nuclear translocation prior to mid-blastula. Fusion of an additional, heterologous NLS to the N-terminus of XMI-ER1 was not sufficient to overcome cytoplasmic retention, indicating that retention does not involve NLS masking, but rather binding to a cytoplasmic anchor. The anchoring molecule is not an RNA, as microinjection of RNase A did not affect the timing of nuclear translocation. Western blot analysis using antibodies that recognize phosphorylated residues revealed that, while XMI-ER1 is not itself phosphorylated, it is associated with two differentially phosphorylated proteins, suggesting that the anchoring mechanism may involve interaction with a cytoplasmic protein(s). A series of XMI-ER1 deletion mutants was utilized to map the putative retention domain. Our analysis revealed that amino acids 144-175, containing the fourth acidic stretch of the acidic activation domain, are required for retention. These results suggest that XMI-ER1 is retained in the cytoplasm of the early embryo by interaction of the region containing amino acids 144-175 with a cytoplasmic anchor.


Asunto(s)
Blastómeros/metabolismo , Núcleo Celular/metabolismo , Fase de Segmentación del Huevo/metabolismo , Citoplasma/metabolismo , Factores de Transcripción/metabolismo , Proteínas de Xenopus/metabolismo , Cigoto/metabolismo , Animales , Blastómeros/citología , Fase de Segmentación del Huevo/citología , Mesodermo/metabolismo , Unión Proteica/genética , Estructura Terciaria de Proteína/genética , Transporte de Proteínas/genética , Factores de Transcripción/genética , Proteínas de Xenopus/genética , Xenopus laevis , Cigoto/citología
13.
J Biol Chem ; 279(27): 28009-16, 2004 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-15117948

RESUMEN

To gain insight into the regulation of hmi-er1 expression, we cloned a human genomic DNA fragment containing one of the two hmi-er1 promoters and consisting of 1460 bp upstream of the translation initiation codon of hMI-ER1. Computer-assisted sequence analysis revealed that the hmi-er1 promoter region contains a CpG island but lacks an identifiable TATA element, initiator sequence and downstream promoter element. This genomic DNA was able to direct transcription of a luciferase reporter gene in a variety of human cell lines, and the minimal promoter was shown to be located within-68/+144 bp. Several putative Sp1 binding sites were identified, and we show that Sp1 can bind to the hmi-er1 minimal promoter and increase transcription, suggesting that the level of hmi-er1 expression may depend on the availability of Sp1 protein. Functional analysis revealed that hMI-ER1 represses Sp1-activated transcription from the minimal promoter by a histone deacetylase-independent mechanism. Chromatin immunoprecipitation analysis demonstrated that both Sp1 and hMI-ER1 are associated with the chromatin of the hmi-er1 promoter and that overexpression of hMI-ER1 in cell lines that allow Tet-On-inducible expression resulted in loss of detectable Sp1 from the endogenous hmi-er1 promoter. The mechanism by which this occurs does not involve binding of hMI-ER1 to cis-acting elements. Instead, we show that hMI-ER1 physically associates with Sp1 and that endogenous complexes containing the two proteins could be detected in vivo. Furthermore, hMI-ER1 specifically interferes with binding of Sp1 to the hmi-er1 minimal promoter as well as to an Sp1 consensus oligonucleotide. Deletion analysis revealed that this interaction occurs through a region containing the SANT domain of hMI-ER1. Together, these data reveal a functional role for the SANT domain in the action of co-repressor regulatory factors and suggest that the association of hMI-ER1 with Sp1 represents a novel mechanism for the negative regulation of Sp1 target promoters.


Asunto(s)
Proteínas Inmediatas-Precoces/química , Proteínas Nucleares/química , Regiones Promotoras Genéticas , Factor de Transcripción Sp1/metabolismo , Secuencia de Bases , Sitios de Unión , Línea Celular , Cromatina/metabolismo , Clonación Molecular , Islas de CpG , ADN/genética , Proteínas de Unión al ADN , Relación Dosis-Respuesta a Droga , Regulación hacia Abajo , Genes Reporteros , Glutatión Transferasa/metabolismo , Células HeLa , Histona Desacetilasas/metabolismo , Humanos , Proteínas Inmediatas-Precoces/metabolismo , Datos de Secuencia Molecular , Proteínas Nucleares/metabolismo , Plásmidos/metabolismo , Pruebas de Precipitina , Unión Proteica , Estructura Terciaria de Proteína , ARN Mensajero/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Análisis de Secuencia de ADN , Factores de Transcripción , Transcripción Genética , Transfección
14.
Biochem Biophys Res Commun ; 308(4): 679-83, 2003 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-12927772

RESUMEN

Xmi-er1 is an immediate-early gene encoding a transcriptional regulator whose expression is activated by fibroblast growth factor (FGF) during mesoderm induction in Xenopus. In this study, we examined the role of xmi-er1 in embryonic development and mesoderm induction and investigated the importance of various functional domains in the protein sequence. Overexpression of xmi-er1 in embryos resulted in truncations of the anteroposterior axis, with most of the abnormal embryos exhibiting deficiencies in both anterior and posterior structures. Whole mount in situ hybridization for the early mesodermal marker brachyury (Xbra) revealed a dramatic reduction of Xbra expression in xmi-er1-injected embryos, while mesoderm induction assays showed that overexpression of xmi-er1 significantly reduced the percentage of explants induced by FGF-2. Site-directed mutagenesis of several functional domains, including the ELM2 domain, the SANT domain, a putative MEK phosphorylation site, and a proline-rich region showed that only proline 365 in the proline-rich region is required for the effect on embryonic development and mesoderm induction. These data demonstrate that XMI-ER1 is a negative regulator of FGF, perhaps serving to limit the extent of mesoderm formation in vivo, and that this activity is mediated by proline 365.


Asunto(s)
Proteínas Fetales , Regulación del Desarrollo de la Expresión Génica , Proteínas Inmediatas-Precoces/química , Proteínas Inmediatas-Precoces/fisiología , Prolina/química , Proteínas de Xenopus/química , Proteínas de Xenopus/fisiología , Xenopus/embriología , Animales , Western Blotting , ADN Complementario/metabolismo , Relación Dosis-Respuesta a Droga , Factores de Crecimiento de Fibroblastos/metabolismo , Hibridación in Situ , Modelos Genéticos , Mutagénesis Sitio-Dirigida , Mutación , Fosforilación , Plásmidos/metabolismo , Estructura Terciaria de Proteína , ARN Complementario/metabolismo , Proteínas Recombinantes/metabolismo , Transducción de Señal , Proteínas de Dominio T Box/metabolismo , Factores de Tiempo , Agua/química
15.
Mol Cell Biol ; 23(1): 250-8, 2003 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-12482978

RESUMEN

mi-er1 (previously called er1) was first isolated from Xenopus laevis embryonic cells as a novel fibroblast growth factor-regulated immediate-early gene. Xmi-er1 was shown to encode a nuclear protein with an N-terminal acidic transcription activation domain. The human orthologue of mi-er1 (hmi-er1) displays 91% similarity to the Xenopus sequence at the amino acid level and was shown to be upregulated in breast carcinoma cell lines and tumors. Alternative splicing at the 3' end of hmi-er1 produces two major isoforms, hMI-ER1alpha and hMI-ER1beta, which contain distinct C-terminal domains. In this study, we investigated the role of hMI-ER1alpha and hMI-ER1beta in the regulation of transcription. Using fusion proteins of hMI-ER1alpha or hMI-ER1beta tethered to the GAL4 DNA binding domain, we show that both isoforms, when recruited to the G5tkCAT minimal promoter, function to repress transcription. We demonstrate that this repressor activity is due to interaction and recruitment of a trichostatin A-sensitive histone deacetylase 1 (HDAC1). Furthermore, deletion analysis revealed that recruitment of HDAC1 to hMI-ER1alpha and hMI-ER1beta occurs through their common ELM2 domain. The ELM2 domain was first described in the Caenorhabditis elegans Egl-27 protein and is present in a number of SANT domain-containing transcription factors. This is the first report of a function for the ELM2 domain, highlighting its role in the regulation of transcription.


Asunto(s)
Proteínas de Caenorhabditis elegans , Proteínas de Unión al ADN , Histona Desacetilasas/metabolismo , Proteínas Represoras/metabolismo , Factores de Transcripción , Empalme Alternativo , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Carcinoma/genética , Carcinoma/metabolismo , Secuencia Conservada , Inhibidores Enzimáticos/farmacología , Femenino , Proteínas del Helminto/química , Histona Desacetilasa 1 , Inhibidores de Histona Desacetilasas , Histona Desacetilasas/genética , Humanos , Ácidos Hidroxámicos/farmacología , Ratones , Datos de Secuencia Molecular , Isoformas de Proteínas , Estructura Terciaria de Proteína , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Represoras/genética , Transcripción Genética , Células Tumorales Cultivadas , Neoplasias del Cuello Uterino/genética , Neoplasias del Cuello Uterino/metabolismo
16.
Gene ; 295(1): 79-88, 2002 Jul 24.
Artículo en Inglés | MEDLINE | ID: mdl-12242014

RESUMEN

mi-er1 (previously called er1) is a fibroblast growth factor-inducible early response gene activated during mesoderm induction in Xenopus embryos and encoding a nuclear protein that functions as a transcriptional activator. The human orthologue of mi-er1 was shown to be upregulated in breast carcinoma cell lines and breast tumours when compared to normal breast cells. In this report, we investigate the structure of the human mi-er1 (hmi-er1) gene and characterize the alternatively spliced transcripts and protein isoforms. hmi-er1 is a single copy gene located at 1p31.2 and spanning 63 kb. It contains 17 exons and includes one skipped exon, a facultative intron and three polyadenylation signals to produce 12 transcripts encoding six distinct proteins. hmi-er1 transcripts were expressed at very low levels in most human adult tissues and the mRNA isoform pattern varied with the tissue. The 12 transcripts encode proteins containing a common internal sequence with variable N- and C-termini. Three distinct N- and two distinct C-termini were identified, giving rise to six protein isoforms. The two C-termini differ significantly in size and sequence and arise from alternate use of a facultative intron to produce hMI-ER1alpha and hMI-ER1beta. In all tissues except testis, transcripts encoding the beta isoform were predominant. hMI-ER1alpha lacks the predicted nuclear localization signal and transfection assays revealed that, unlike hMI-ER1beta, it is not a nuclear protein, but remains in the cytoplasm. Our results demonstrate that alternate use of a facultative intron regulates the subcellular localization of hMI-ER1 proteins and this may have important implications for hMI-ER1 function.


Asunto(s)
Empalme Alternativo , Proteínas Inmediatas-Precoces/genética , Proteínas Nucleares/genética , Células 3T3 , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Proteínas de Unión al ADN , Femenino , Expresión Génica , Genes/genética , Humanos , Intrones/genética , Masculino , Ratones , Datos de Secuencia Molecular , Isoformas de Proteínas/genética , Homología de Secuencia de Aminoácido , Factores de Transcripción , Transcripción Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA