Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Stem Cell Res ; 53: 102311, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33799276

RESUMEN

Heterozygous variants in the KCNQ3 gene cause epileptic and/or developmental disorders of varying severity. Here we describe the generation of induced pluripotent stem cells (iPSCs) from a 9-year-old girl with pharmacodependent neonatal-onset epilepsy and intellectual disability who carry a homozygous single-base duplication in exon 12 of KCNQ3 (NM_004519.3: KCNQ3 c.1599dup; KCNQ3 p.PHE534ILEfs*15), and from a non-carrier brother of the proband. For iPSC generation, non-integrating episomal plasmid vectors were used to transfect fibroblasts isolated from skin biopsies. The obtained iPSC lines had a normal karyotype, showed embryonic stem cell-like morphology, expressed pluripotency markers, and possessed trilineage differentiation potential.


Asunto(s)
Epilepsia , Células Madre Pluripotentes Inducidas , Discapacidad Intelectual , Diferenciación Celular , Niño , Epilepsia/genética , Femenino , Homocigoto , Humanos , Discapacidad Intelectual/genética , Masculino , Hermanos
2.
Epilepsia Open ; 4(3): 464-475, 2019 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-31440727

RESUMEN

OBJECTIVE: Heterozygous variants in KCNQ2 or, more rarely, KCNQ3 genes are responsible for early-onset developmental/epileptic disorders characterized by heterogeneous clinical presentation and course, genetic transmission, and prognosis. While familial forms mostly include benign epilepsies with seizures starting in the neonatal or early-infantile period, de novo variants in KCNQ2 or KCNQ3 have been described in sporadic cases of early-onset encephalopathy (EOEE) with pharmacoresistant seizures, various age-related pathological EEG patterns, and moderate/severe developmental impairment. All pathogenic variants in KCNQ2 or KCNQ3 occur in heterozygosity. The aim of this work was to report the clinical, molecular, and functional properties of a new KCNQ3 variant found in homozygous configuration in a 9-year-old girl with pharmacodependent neonatal-onset epilepsy and non-syndromic intellectual disability. METHODS: Exome sequencing was used for genetic investigation. KCNQ3 transcript and subunit expression in fibroblasts was analyzed with quantitative real-time PCR and Western blotting or immunofluorescence, respectively. Whole-cell patch-clamp electrophysiology was used for functional characterization of mutant subunits. RESULTS: A novel single-base duplication in exon 12 of KCNQ3 (NM_004519.3:c.1599dup) was found in homozygous configuration in the proband born to consanguineous healthy parents; this frameshift variant introduced a premature termination codon (PTC), thus deleting a large part of the C-terminal region. Mutant KCNQ3 transcript and protein abundance was markedly reduced in primary fibroblasts from the proband, consistent with nonsense-mediated mRNA decay. The variant fully abolished the ability of KCNQ3 subunits to assemble into functional homomeric or heteromeric channels with KCNQ2 subunits. SIGNIFICANCE: The present results indicate that a homozygous KCNQ3 loss-of-function variant is responsible for a severe phenotype characterized by neonatal-onset pharmacodependent seizures, with developmental delay and intellectual disability. They also reveal difference in genetic and pathogenetic mechanisms between KCNQ2- and KCNQ3-related epilepsies, a crucial observation for patients affected with EOEE and/or developmental disabilities.

3.
Front Oncol ; 9: 1498, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31993371

RESUMEN

Glioblastoma is among the most common malignant brain tumors and has a dismal prognosis due to the poor response to therapeutic regimens such as ionizing radiation and DNA-alkylating agents. In our study, we investigated the radiosensitizing activity of the N6-isopentenyladenosine (iPA), an naturally modified adenosine harboring an isopenenyl moiety, which shows antiproliferative effects on glioblastoma cell lines. We observed that co-treatment with ionizing radiation and iPA at micromolar concentration inhibited colony formation and viability of glioblastoma cell lines but not of non-malignant human cells. The combined treatment significantly attenuated the repair of radiation-induced DNA damage by inhibiting both the expression and irradiation-induced foci formation of RAD51, a key player in the homologous recombination repair process, leading to persistent DNA damage, as reflected by an increase of γ-H2AX foci. The radiosensitizing effect relied also on the inhibition of STAT5a/b activation, which is crucial for RAD51 expression, suggesting that iPA modulates the STAT5a/b-RAD51 axis following exposure to ionizing radiation. Overall, these data suggest that iPA, by acting through RAD51 inhibition at the mechanistic level, could function as a promising radiosensitizing agent and warrants further evaluation in prospective clinical trials.

4.
Int J Dev Biol ; 62(11-12): 827-836, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30604852

RESUMEN

Prep1 (pKnox1) is a homeodomain transcription factor essential for in utero and post-natal development and an oncosuppressor gene in human and adult mice. We have analyzed its role in the development of the mouse mammary gland. We used Prep1i/i hypomorphic and Prep1F/F-Ker5CRE crosses to analyze the role of Prep1 in vivo in adult mouse mammary gland development. We also cultured mammary gland stem/progenitor cells in mammospheres to perform biochemical studies. Prep1 was expressed in mammary gland progenitors and fully differentiated mammary gland cells. Using different Prep1-deficient mouse models we show that in vivo Prep1 contributes to mammary gland branching since the branching efficiency of the mammary gland in Prep1-deleted or Prep1 hypomorphic mice was largely reduced. In-vitro, Prep1 sustained functions of the mammary stem/progenitor compartment. Prep1-deficient mammary stem/progenitor cells showed reduced ability to form mammospheres; they were not able to branch in a 3D assay, and exhibited reduced expression of Snail1, Snail2 and vimentin. The branching phenotype associated with increased Tp53-dependent apoptosis and inability to properly activate signals involved in branching morphogenesis. Finally, Prep1 formed complexes with Snail2, a transcription factor essential in branching morphogenesis, and its absence destabilizes and promotes Snail2 proteasome-mediated degradation. We conclude that Prep1 is required for normal adult mammary gland development, in particular at its branching morphogenesis step. By binding Snail2, Prep1 protects it from the proteasomal degradation.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/genética , Glándulas Mamarias Animales/embriología , Morfogénesis/genética , Animales , Células Epiteliales/metabolismo , Femenino , Proteínas de Homeodominio/metabolismo , Glándulas Mamarias Animales/metabolismo , Ratones , Ratones Noqueados , Complejo de la Endopetidasa Proteasomal/metabolismo , Factores de Transcripción de la Familia Snail/genética , Factores de Transcripción de la Familia Snail/metabolismo , Vimentina/genética , Vimentina/metabolismo
6.
Proc Natl Acad Sci U S A ; 111(36): E3775-84, 2014 Sep 09.
Artículo en Inglés | MEDLINE | ID: mdl-25157139

RESUMEN

Pre-B-cell leukemia homeobox (Pbx)-regulating protein-1 (Prep1) is a ubiquitous homeoprotein involved in early development, genomic stability, insulin sensitivity, and hematopoiesis. Previously we have shown that Prep1 is a haploinsufficient tumor suppressor that inhibits neoplastic transformation by competing with myeloid ecotropic integration site 1 for binding to the common heterodimeric partner Pbx1. Epithelial-mesenchymal transition (EMT) is controlled by complex networks of proinvasive transcription factors responsive to paracrine factors such as TGF-ß. Here we show that, in addition to inhibiting primary tumor growth, PREP1 is a novel EMT inducer and prometastatic transcription factor. In human non-small cell lung cancer (NSCLC) cells, PREP1 overexpression is sufficient to trigger EMT, whereas PREP1 down-regulation inhibits the induction of EMT in response to TGF-ß. PREP1 modulates the cellular sensitivity to TGF-ß by inducing the small mothers against decapentaplegic homolog 3 (SMAD3) nuclear translocation through mechanisms dependent, at least in part, on PREP1-mediated transactivation of a regulatory element in the SMAD3 first intron. Along with the stabilization and accumulation of PBX1, PREP1 induces the expression of multiple activator protein 1 components including the proinvasive Fos-related antigen 1 (FRA-1) oncoprotein. Both FRA-1 and PBX1 are required for the mesenchymal changes triggered by PREP1 in lung tumor cells. Finally, we show that the PREP1-induced mesenchymal transformation correlates with significantly increased lung colonization by cells overexpressing PREP1. Accordingly, we have detected PREP1 accumulation in a large number of human brain metastases of various solid tumors, including NSCLC. These findings point to a novel role of the PREP1 homeoprotein in the control of the TGF-ß pathway, EMT, and metastasis in NSCLC.


Asunto(s)
Adenocarcinoma/patología , Carcinoma de Pulmón de Células no Pequeñas/patología , Transición Epitelial-Mesenquimal , Proteínas de Homeodominio/metabolismo , Neoplasias Pulmonares/patología , Transducción de Señal , Proteína smad3/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Adenocarcinoma/genética , Adenocarcinoma del Pulmón , Animales , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/secundario , Carcinoma de Pulmón de Células no Pequeñas/genética , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Proliferación Celular/efectos de los fármacos , Proteínas de Unión al ADN/metabolismo , Elementos de Facilitación Genéticos/genética , Transición Epitelial-Mesenquimal/efectos de los fármacos , Transición Epitelial-Mesenquimal/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Intrones/genética , Neoplasias Pulmonares/genética , Ratones , Modelos Biológicos , Metástasis de la Neoplasia , Péptido Hidrolasas/metabolismo , Factor de Transcripción 1 de la Leucemia de Células Pre-B , Unión Proteica/efectos de los fármacos , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-fos/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Proteína smad3/genética , Análisis de Supervivencia , Factor de Transcripción AP-1/metabolismo , Transcripción Genética/efectos de los fármacos , Factor de Crecimiento Transformador beta/farmacología
7.
Proc Natl Acad Sci U S A ; 111(10): E896-905, 2014 Mar 11.
Artículo en Inglés | MEDLINE | ID: mdl-24578510

RESUMEN

Pbx-regulating protein-1 (Prep1) is a tumor suppressor, whereas myeloid ecotropic viral integration site-1 (Meis1) is an oncogene. We show that, to perform these activities in mouse embryonic fibroblasts, both proteins competitively heterodimerize with pre-B-cell leukemia homeobox-1 (Pbx1). Meis1 alone transforms Prep1-deficient fibroblasts, whereas Prep1 overexpression inhibits Meis1 tumorigenicity. Pbx1 can, therefore, alternatively act as an oncogene or tumor suppressor. Prep1 posttranslationally controls the level of Meis1, decreasing its stability by sequestering Pbx1. The different levels of Meis1 and the presence of Prep1 are followed at the transcriptional level by the induction of specific transcriptional signatures. The decrease of Meis1 prevents Meis1 interaction with Ddx3x and Ddx5, which are essential for Meis1 tumorigenesis, and modifies the growth-promoting DNA binding landscape of Meis1 to the growth-controlling landscape of Prep1. Hence, the key feature of Prep1 tumor-inhibiting activity is the control of Meis1 stability.


Asunto(s)
Unión Competitiva/fisiología , Carcinogénesis/metabolismo , Proteínas de Homeodominio/metabolismo , Proteínas de Neoplasias/metabolismo , Estabilidad Proteica , Factores de Transcripción/metabolismo , Animales , Línea Celular , Inmunoprecipitación de Cromatina , ARN Helicasas DEAD-box/metabolismo , Cartilla de ADN/genética , Inmunoprecipitación , Espectrometría de Masas , Ratones , Ratones Desnudos , Proteína 1 del Sitio de Integración Viral Ecotrópica Mieloide , Factor de Transcripción 1 de la Leucemia de Células Pre-B , ARN Helicasas/metabolismo , ARN Interferente Pequeño/genética , Reacción en Cadena en Tiempo Real de la Polimerasa
8.
Proc Natl Acad Sci U S A ; 108(29): E314-22, 2011 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-21715654

RESUMEN

Prep1 is a homeodomain transcription factor that is essential in embryonic development and functions in the adult as a tumor suppressor. We show here that Prep1 is involved in maintaining genomic stability and preventing neoplastic transformation. Hypomorphic homozygous Prep1(i/i) fetal liver cells and mouse embryonic fibroblasts (MEFs) exhibit increased basal DNA damage and normal DNA damage response after γ-irradiation compared with WT. Cytogenetic analysis shows the presence of numerous chromosomal aberrations and aneuploidy in very early-passage Prep1(i/i) MEFs. In human fibroblasts, acute Prep1 down-regulation by siRNA induces DNA damage response, like in Prep1(i/i) MEFs, together with an increase in heterochromatin-associated modifications: rapid increase of histone methylation and decreased transcription of satellite DNA. Ectopic expression of Prep1 rescues DNA damage and heterochromatin methylation. Inhibition of Suv39 activity blocks the chromatin but not the DNA damage phenotype. Finally, Prep1 deficiency facilitates cell immortalization, escape from oncogene-induced senescence, and H-Ras(V12)-dependent transformation. Importantly, the latter can be partially rescued by restoration of Prep1 level. The results show that the tumor suppressor role of Prep1 is associated with the maintenance of genomic stability.


Asunto(s)
Transformación Celular Neoplásica/metabolismo , Regulación de la Expresión Génica/fisiología , Inestabilidad Genómica/fisiología , Proteínas de Homeodominio/fisiología , Factores de Transcripción/fisiología , Animales , Inmunoprecipitación de Cromatina , Ensayo Cometa , Análisis Citogenético , Daño del ADN/genética , Embrión de Mamíferos/citología , Fibroblastos/metabolismo , Rayos gamma , Regulación de la Expresión Génica/genética , Heterocromatina/genética , Heterocromatina/efectos de la radiación , Proteínas de Homeodominio/metabolismo , Humanos , Ratones , Oligonucleótidos/genética , Factores de Transcripción/metabolismo
9.
Diabetes ; 60(1): 138-47, 2011 01.
Artículo en Inglés | MEDLINE | ID: mdl-20864515

RESUMEN

OBJECTIVE: We investigated the function of the Prep1 gene in insulin-dependent glucose homeostasis in liver. RESEARCH DESIGN AND METHODS: Prep1 action on insulin glucoregulatory function has been analyzed in liver of Prep1-hypomorphic mice (Prep1(i/i)), which express 2-3% of Prep1 mRNA. RESULTS: Based on euglycemic hyperinsulinemic clamp studies and measurement of glycogen content, livers from Prep1(i/i) mice feature increased sensitivity to insulin. Tyrosine phosphorylation of both insulin receptor (IR) and insulin receptor substrate (IRS)1/2 was significantly enhanced in Prep1(i/i) livers accompanied by a specific downregulation of the SYP and SHP1 tyrosine phosphatases. Prep1 overexpression in HepG2 liver cells upregulated SYP and SHP1 and inhibited insulin-induced IR and IRS1/2 phosphorylation and was accompanied by reduced glycogen content. Consistently, overexpression of the Prep1 partner Pbx1, but not of p160MBP, mimicked Prep1 effects on tyrosine phosphorylations, glycogen content, and on SYP and SHP1 expression. In Prep1 overexpressing cells, antisense silencing of SHP1, but not that of SYP, rescued insulin-dependent IR phosphorylation and glycogen accumulation. Both Prep1 and Pbx1 bind SHP1 promoter at a site located between nucleotides -2,113 and -1,778. This fragment features enhancer activity and induces luciferase function by 7-, 6-, and 30-fold, respectively, in response to Prep1, Pbx1, or both. CONCLUSIONS: SHP1, a known silencer of insulin signal, is a transcriptional target of Prep1. In liver, transcriptional activation of SHP1 gene by Prep1 attenuates insulin signal transduction and reduces glucose storage.


Asunto(s)
Proteínas de Homeodominio/genética , Insulina/fisiología , Hígado/fisiología , Proteína Tirosina Fosfatasa no Receptora Tipo 6/genética , Animales , Línea Celular , Cartilla de ADN , Grasas de la Dieta/farmacología , Glucosa/metabolismo , Células Hep G2/metabolismo , Humanos , Hígado/enzimología , Hígado/metabolismo , Ratones , Ratones Endogámicos C57BL , Oligonucleótidos/química , Plásmidos/genética , Factor de Transcripción 1 de la Leucemia de Células Pre-B , Proteína Tirosina Fosfatasa no Receptora Tipo 11/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 6/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Factores de Transcripción/genética , Transcripción Genética , Triglicéridos/metabolismo
10.
Development ; 137(20): 3393-403, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20826531

RESUMEN

Disruption of mouse Prep1, which codes for a homeodomain transcription factor, leads to embryonic lethality during post-implantation stages. Prep1(-/-) embryos stop developing after implantation and before anterior visceral endoderm (AVE) formation. In Prep1(-/-) embryos at E6.5 (onset of gastrulation), the AVE is absent and the proliferating extra-embryonic ectoderm and epiblast, marked by Bmp4 and Oct4, respectively, are reduced in size. At E.7.5, Prep1(-/-) embryos are small and very delayed, showing no evidence of primitive streak or of differentiated embryonic lineages. Bmp4 is expressed residually, while the reduced number of Oct4-positive cells is constant up to E8.5. At E6.5, Prep1(-/-) embryos retain a normal mitotic index but show a major increase in cleaved caspase 3 and TUNEL staining, indicating apoptosis. Therefore, the mouse embryo requires Prep1 when undergoing maximal expansion in cell number. Indeed, the phenotype is partially rescued in a p53(-/-), but not in a p16(-/-), background. Apoptosis is probably due to DNA damage as Atm downregulation exacerbates the phenotype. Despite this early lethal phenotype, Prep1 is not essential for ES cell establishment. A differential embryonic expression pattern underscores the unique function of Prep1 within the Meis-Prep family.


Asunto(s)
Apoptosis/fisiología , Desarrollo Embrionario/fisiología , Estratos Germinativos/citología , Proteínas de Homeodominio/metabolismo , Células Madre Pluripotentes/citología , Animales , Southern Blotting , Cartilla de ADN/genética , Ensayo de Cambio de Movilidad Electroforética , Técnica del Anticuerpo Fluorescente , Genotipo , Proteínas de Homeodominio/genética , Etiquetado Corte-Fin in Situ , Ratones , Ratones Noqueados , Células Madre Pluripotentes/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteína p53 Supresora de Tumor/metabolismo
11.
Nucleic Acids Res ; 38(11): 3595-604, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20110257

RESUMEN

PREP1 (PKNOX1) maps in the Down syndrome (DS) critical region of chromosome 21, is overexpressed in some DS tissues and might be involved in the DS phenotype. By using fibroblasts from DS patients and by overexpressing Prep1 in F9 teratocarcinoma and Prep1(i/i) MEF to single out the role of the protein, we report that excess Prep1 increases the sensitivity of cells to genotoxic stress and the extent of the apoptosis directly correlates with the level of Prep1. The apoptotic response of Prep1-overexpressing cells is mediated by the pro-apoptotic p53 protein that we show is a direct target of Prep1, as its depletion reverts the apoptotic phenotype. The induction of p53 overcomes the anti-apoptotic role of Bcl-X(L), previously shown to be also a Prep1 target, the levels of which are increased in Prep1-overexpressing cells as well. Our results provide a rationale for the involvement of PREP1 in the apoptotic phenotype of DS tissues and indicate that differences in Prep1 level can have drastic effects.


Asunto(s)
Apoptosis , Síndrome de Down/metabolismo , Proteínas de Homeodominio/metabolismo , Animales , Células Cultivadas , Síndrome de Down/patología , Células Madre de Carcinoma Embrionario , Etopósido/toxicidad , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Fibroblastos/efectos de la radiación , Vectores Genéticos , Proteínas de Homeodominio/genética , Humanos , Ratones , Fenotipo , Transcripción Genética , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Rayos Ultravioleta , Proteína bcl-X/genética , Proteína bcl-X/metabolismo
12.
Mol Biol Cell ; 20(15): 3543-51, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19477923

RESUMEN

We have analyzed the role of actin polymerization in retinoic acid (RA)-induced HoxB transcription, which is mediated by the HoxB regulator Prep1. RA induction of the HoxB genes can be prevented by the inhibition of actin polymerization. Importantly, inhibition of actin polymerization specifically affects the transcription of inducible Hox genes, but not that of their transcriptional regulators, the RARs, nor of constitutively expressed, nor of actively transcribed Hox genes. RA treatment induces the recruitment to the HoxB2 gene enhancer of a complex composed of "elongating" RNAPII, Prep1, beta-actin, and N-WASP as well as the accessory splicing components p54Nrb and PSF. We show that inhibition of actin polymerization prevents such recruitment. We conclude that inducible Hox genes are selectively sensitive to the inhibition of actin polymerization and that actin polymerization is required for the assembly of a transcription complex on the regulatory region of the Hox genes.


Asunto(s)
Actinas/metabolismo , Proteínas de Homeodominio/genética , Factores de Transcripción/genética , Transcripción Genética/efectos de los fármacos , Tretinoina/farmacología , Actinas/genética , Antineoplásicos/farmacología , Línea Celular Tumoral , Citocalasina D/farmacología , Proteínas de Unión al ADN , Regulación hacia Abajo , Proteínas de Homeodominio/metabolismo , Humanos , Immunoblotting , Mutación , Proteínas Asociadas a Matriz Nuclear/genética , Proteínas Asociadas a Matriz Nuclear/metabolismo , Inhibidores de la Síntesis del Ácido Nucleico/farmacología , Factores de Transcripción de Octámeros/genética , Factores de Transcripción de Octámeros/metabolismo , Factor de Empalme Asociado a PTB , Polímeros/metabolismo , Interferencia de ARN , ARN Polimerasa II/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Receptores de Ácido Retinoico/genética , Receptores de Ácido Retinoico/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteína Neuronal del Síndrome de Wiskott-Aldrich/genética , Proteína Neuronal del Síndrome de Wiskott-Aldrich/metabolismo
13.
Dev Biol ; 311(2): 324-34, 2007 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-17904118

RESUMEN

Most of the hypomorphic Prep1(i/i) embryos (expressing 3-10% of the Prep1 protein), die between E17.5 and P0, with profound anemia, eye malformations and angiogenic anomalies [Ferretti, E., Villaescusa, J.C., Di Rosa, P., Fernandez-Diaz, L.-C., Longobardi, E., Mazzieri, R., Miccio, A., Micali, N., Selleri, L., Ferrari G., Blasi, F. (2006). Hypomorphic mutation of the TALE gene Prep1 (pKnox1) causes a major reduction of Pbx and Meis proteins and a pleiotropic embryonic phenotype. Mol. Cell. Biol. 26, 5650-5662]. We now report on the hematopoietic phenotype of these embryos. Prep1(i/i) fetal livers (FL) are hypoplastic, produce less common myeloid progenitors colonies (CFU-GEMM) in cytokine-supplemented methylcellulose and have an increased number of B-cells precursors that differentiate poorly. Prep1(i/i) FL is able to protect lethally irradiated mice only at high cell doses but the few protected mice show major anomalies in all hematopoietic lineages in both bone marrow (BM) and peripheral organs. Prep1(i/i) FL cells compete inefficiently with wild type bone marrow in competitive repopulation experiments, suggesting that the major defect lies in long-term repopulating hematopoietic stem cells (LTR-HSC). Indeed, wt embryonic expression of Prep1 in the aorta-gonad-mesonephros (AGM) region, fetal liver (FL), cKit(+)Sca1(+)Lin(-)AA4.1(+) (KSLA) cells and B-lymphocytes precursors agrees with the observed phenotype. We therefore conclude that Prep1 is required for a correct and complete hematopoiesis.


Asunto(s)
Hematopoyesis/fisiología , Células Madre Hematopoyéticas/metabolismo , Proteínas de Homeodominio/metabolismo , Células Madre/metabolismo , Factores de Transcripción/metabolismo , Animales , Linfocitos B/citología , Linfocitos B/fisiología , Biomarcadores/metabolismo , Células de la Médula Ósea/citología , Células de la Médula Ósea/fisiología , Diferenciación Celular , Linaje de la Célula , Embrión de Mamíferos/citología , Embrión de Mamíferos/fisiología , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/citología , Proteínas de Homeodominio/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Células Madre/citología , Factores de Transcripción/genética
14.
Mol Cell Biol ; 27(22): 7981-90, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17875935

RESUMEN

Prep1 is known to interact in vivo with Pbx1 to regulate development and organogenesis. We have identified a novel Prep1-interacting protein, p160 c-Myb binding protein (p160). p160 and Pbx1 compete for Prep1 in vitro, and p160 inhibits Prep1-dependent HoxB2 expression in retinoic acid-treated NT2-D1 cells. The N-terminal physiologically truncated form of p160, p67, binds the sequence 63LFPLL67 in the HR1 domain of Prep1. Mutation of both L63 and L66 impairs the binding of Prep1 to both p160/p67 and Pbx1. The sequences required to bind Prep1 are mainly located in residues 51 to 151. Immunofluorescence colocalization and coimmunoprecipitation of endogenous p160 and Prep1 are induced by ActD, which translocates p160 from the nucleolus to the nucleoplasm. These data therefore show that p160 is a novel regulator of Prep1-Pbx1 transcriptional activity.


Asunto(s)
Proteínas Portadoras/metabolismo , Regulación de la Expresión Génica , Proteínas de Homeodominio/metabolismo , Proteínas Nucleares/metabolismo , Transcripción Genética , Secuencia de Aminoácidos , Animales , Proteínas Portadoras/genética , Núcleo Celular/metabolismo , Proteínas de Unión al ADN , Proteínas de Homeodominio/genética , Ratones , Datos de Secuencia Molecular , Células 3T3 NIH , Proteínas Nucleares/genética , Factor de Transcripción 1 de la Leucemia de Células Pre-B , Proteínas de Unión al ARN , Alineación de Secuencia , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
15.
Mol Cell Biol ; 26(15): 5650-62, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16847320

RESUMEN

The interaction of Prep1 and Pbx homeodomain transcription factors regulates their activity, nuclear localization, and likely, function in development. To understand the in vivo role of Prep1, we have analyzed an embryonic lethal hypomorphic mutant mouse (Prep1(i/i)). Prep1(i/i) embryos die at embryonic day 17.5 (E17.5) to birth with an overall organ hypoplasia, severe anemia, impaired angiogenesis, and eye anomalies, particularly in the lens and retina. The anemia correlates with delayed differentiation of erythroid progenitors and may be, at least in part, responsible for intrauterine death. At E14.5, Prep1 is present in fetal liver (FL) cMyb-positive cells, whose deficiency causes a marked hematopoietic phenotype. Prep1 is also localized to FL endothelial progenitors, consistent with the observed angiogenic phenotype. Likewise, at the same gestational day, Prep1 is present in the eye cells that bear Pax6, implicated in eye development. The levels of cMyb and Pax6 in FL and in the retina, respectively, are significantly decreased in Prep1(i/i) embryos, consistent with the hematopoietic and eye phenotypes. Concomitantly, Prep1 deficiency results in the overall decrease of protein levels of its related family member Meis1 and its partners Pbx1 and Pbx2. As both Prep1 and Meis interact with Pbx, the overall Prep1/Meis-Pbx DNA-binding activity is strongly reduced in whole Prep1(i/i) embryos and their organs. Our data indicate that Prep1 is an essential gene that acts upstream of and within a Pbx-Meis network that regulates multiple aspects of embryonic development.


Asunto(s)
Embrión de Mamíferos , Proteínas de Homeodominio/metabolismo , Mutación , Proteínas de Neoplasias/metabolismo , Fenotipo , Factores de Transcripción/metabolismo , Alantoides/citología , Anemia/genética , Animales , Células Cultivadas , Embrión de Mamíferos/anatomía & histología , Embrión de Mamíferos/fisiología , Ojo/anatomía & histología , Ojo/embriología , Ojo/patología , Femenino , Marcación de Gen , Edad Gestacional , Células Madre Hematopoyéticas/fisiología , Proteínas de Homeodominio/genética , Ratones , Ratones Endogámicos C57BL , Proteína 1 del Sitio de Integración Viral Ecotrópica Mieloide , Proteínas de Neoplasias/genética , Neovascularización Fisiológica , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Factor de Transcripción 1 de la Leucemia de Células Pre-B , Embarazo , ARN Mensajero/metabolismo , Factores de Transcripción/genética
16.
Mol Cell Biol ; 25(19): 8541-52, 2005 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16166636

RESUMEN

The Hoxb1 autoregulatory enhancer directs segmental expression in vertebrate hindbrain. Three conserved repeats (R1, R2, and R3) in the enhancer have been described as Pbx-Hoxb1 (PH) binding sites, and one Pbx-Meinox (PM) binding site has also been characterized. We have investigated the importance and relative roles of PH and PM binding sites with respect to protein interactions and in vivo regulatory activity. We have identified a new PM site (PM2) and found that it cooperates with the R3 PH site to form ternary Prep1-Pbx1-Hoxb1 complexes. In vivo, the combination of the R3 and PM2 sites is sufficient to mediate transgenic reporter activity in the developing chick hindbrain. In both chicken and mouse transgenic embryos, mutations of the PM1 and PM2 sites reveal that they cooperate to modulate in vivo regulatory activity of the Hoxb1 enhancer. Furthermore, we have shown that the R2 motif functions as a strong PM site, with a high binding affinity for Prep1-Pbx1 dimers, and renamed this site R2/PM3. In vitro R2/PM3, when combined with the PM1 and R3 motifs, inhibits ternary complex formation mediated by these elements and in vivo reduces and restricts reporter expression in transgenic embryos. These inhibitory effects appear to be a consequence of the high PM binding activity of the R2/PM3 site. Taken together, our results demonstrate that the activity of the Hoxb1 autoregulatory enhancer depends upon multiple Prep1-Pbx1 (PM1, PM2, and PM3) and Pbx1-Hoxb1 (R1 and R3) binding sites that cooperate to modulate and spatially restrict the expression of Hoxb1 in r4 rhombomere.


Asunto(s)
Proteínas de Homeodominio/metabolismo , Factores de Transcripción/metabolismo , Secuencia de Aminoácidos , Animales , Animales Modificados Genéticamente , Secuencia de Bases , Sitios de Unión , Encéfalo/embriología , Encéfalo/metabolismo , Diferenciación Celular , Línea Celular , Núcleo Celular/metabolismo , Embrión de Pollo , Drosophila melanogaster , Electroporación , Embrión de Mamíferos/metabolismo , Embrión no Mamífero , Elementos de Facilitación Genéticos , Regulación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Genes Reporteros , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Ratones Transgénicos , Datos de Secuencia Molecular , Neuronas/metabolismo , Oligonucleótidos/química , Factor de Transcripción 1 de la Leucemia de Células Pre-B , Unión Proteica , Homología de Secuencia de Ácido Nucleico , Transgenes
17.
J Biol Chem ; 278(40): 39235-41, 2003 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-12871956

RESUMEN

To bind DNA and to be retained in the nucleus, PBX proteins must form heterodimeric complexes with members of the MEINOX family. Therefore the balance between PBX and MEINOX must be an important regulatory feature. We show that overexpression of PREP-1 influences the level of PBX-2 protein maintaining the PREP-1-PBX balance. This effect has important functional consequences. F9 teratocarcinoma cells stably transfected with PREP-1 had an increased DNA binding activity to a PREP-PBX-responsive element. Because PREP-1 binds DNA efficiently only when dimerized to PBX, the increased DNA binding activity suggests that the level of PBX might also have increased. Indeed PREP-1-overexpressing cells had a higher level of PBX-2 and PBX-1b proteins. PBX-2 increase did not depend on increased mRNA level or a higher rate of translation but rather because of a protein stabilization process. Indeed, PBX-2 level drastically decreased after 3 h of cycloheximide treatment in control but not in PREP-1-overexpressing cells and the proteasome inhibitor MG132 prevented PBX-2 decay in control cells. Hence, dimerization with PREP-1 appears to decrease proteasomal degradation of PBX-2. Retinoic acid induces differentiation of F9 teratocarcinoma cells with a cascade synthesis of HOX proteins. In PREP-1-overexpressing cells, HOXb1 induction was more sustained (3 days versus 1 day) and the induced level of MEIS-1b, another TALE (three amino acid loop extension) protein involved in embryonal development, was higher. Thus an increase in PREP-1 leads to changes in the fate-determining HOXb1 and has therefore important functional consequences.


Asunto(s)
Proteínas de Homeodominio/biosíntesis , Proteínas de Homeodominio/química , Proteínas de Homeodominio/fisiología , Proteínas Proto-Oncogénicas/biosíntesis , Proteínas Proto-Oncogénicas/química , Teratocarcinoma/metabolismo , Secuencia de Aminoácidos , Northern Blotting , Diferenciación Celular , Núcleo Celular/metabolismo , Cicloheximida/farmacología , Cisteína Endopeptidasas/metabolismo , Citoplasma/metabolismo , ADN/metabolismo , Dimerización , Proteínas de Homeodominio/metabolismo , Immunoblotting , Cinética , Leupeptinas/farmacología , Modelos Biológicos , Datos de Secuencia Molecular , Complejos Multienzimáticos/antagonistas & inhibidores , Complejos Multienzimáticos/metabolismo , Proteína 1 del Sitio de Integración Viral Ecotrópica Mieloide , Proteínas de Neoplasias/metabolismo , Complejo de la Endopetidasa Proteasomal , Unión Proteica , Biosíntesis de Proteínas , Inhibidores de la Síntesis de la Proteína/farmacología , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Tiempo , Transfección , Tretinoina/farmacología , Células Tumorales Cultivadas
18.
Blood ; 100(9): 3325-32, 2002 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-12384434

RESUMEN

Activated transcription of the urokinase-type plasminogen activator (uPA) gene depends on the enhancer, located approximately 2 kb from the start of transcription. The proximal promoter, driving basal transcription, contains a GC-/GA-rich sequence immediately upstream of the TATA box. We have investigated the role played by this element in the transcription of the uPA gene in HeLa and PC3 cells, which do not express or constitutively express the gene, respectively. This region binds either Sp1 or Sp3, as monomers or multimers, but not a combination of the 2 proteins. The more efficient binding of Sp1 to the proximal promoter in PC3 cells is correlated to its phosphorylation state. Polymerase chain reaction (PCR)-coupled, chromatin immunoprecipitation experiments with anti-Sp1 antibodies indeed show an enrichment of proximal promoter sequences in PC3 cells and support the observed difference in transcription levels from proximal promoter constructs in HeLa versus PC3 cells. Furthermore, overexpression of Sp1 increases transcription from the reporter construct in HeLa cells, whereas in PC3 cells, overexpression of Sp3 does not reduce transcription from the same construct, indicating that the Sp1/Sp3 balance cannot be shifted. We conclude that the GC-/GA-rich element of the uPA regulatory region is an independent functional element, regulated by Sp family proteins. Phosphorylation of Sp1 determines the presence in vivo and the functionality of this element in PC3 cells. Thus, the cellular context determines the relevance of the GC-/GA-rich region in uPA gene transcription, which contributes to constitutive gene expression, related, in turn, to the invasive phenotype.


Asunto(s)
Adenocarcinoma/patología , Elementos de Facilitación Genéticos/genética , Regulación Neoplásica de la Expresión Génica/genética , Invasividad Neoplásica/genética , Proteínas de Neoplasias/genética , Regiones Promotoras Genéticas/genética , Neoplasias de la Próstata/patología , Factor de Transcripción Sp1/metabolismo , Activador de Plasminógeno de Tipo Uroquinasa/genética , Adenocarcinoma/genética , Secuencia de Bases , Proteínas de Unión al ADN/metabolismo , Desoxirribonucleasa I/farmacología , Genes Reporteros , Células HeLa/metabolismo , Humanos , Luciferasas/biosíntesis , Luciferasas/genética , Masculino , Datos de Secuencia Molecular , Familia de Multigenes , Proteínas de Neoplasias/biosíntesis , Fosforilación , Reacción en Cadena de la Polimerasa , Neoplasias de la Próstata/genética , Unión Proteica , Procesamiento Proteico-Postraduccional , Proteínas Recombinantes de Fusión/biosíntesis , Factor de Transcripción Sp3 , Factores de Transcripción/metabolismo , Células Tumorales Cultivadas/patología , Activador de Plasminógeno de Tipo Uroquinasa/biosíntesis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...