Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Genes Dev ; 29(20): 2108-22, 2015 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-26494787

RESUMEN

Gene regulatory networks controlling functional activities of spatially and temporally distinct endodermal cell populations in the early mouse embryo remain ill defined. The T-box transcription factor Eomes, acting downstream from Nodal/Smad signals, directly activates the LIM domain homeobox transcription factor Lhx1 in the visceral endoderm. Here we demonstrate Smad4/Eomes-dependent Lhx1 expression in the epiblast marks the entire definitive endoderm lineage, the anterior mesendoderm, and midline progenitors. Conditional inactivation of Lhx1 disrupts anterior definitive endoderm development and impedes node and midline morphogenesis in part due to severe disturbances in visceral endoderm displacement. Transcriptional profiling and ChIP-seq (chromatin immunoprecipitation [ChIP] followed by high-throughput sequencing) experiments identified Lhx1 target genes, including numerous anterior definitive endoderm markers and components of the Wnt signaling pathway. Interestingly, Lhx1-binding sites were enriched at enhancers, including the Nodal-proximal epiblast enhancer element and enhancer regions controlling Otx2 and Foxa2 expression. Moreover, in proteomic experiments, we characterized a complex comprised of Lhx1, Otx2, and Foxa2 as well as the chromatin-looping protein Ldb1. These partnerships cooperatively regulate development of the anterior mesendoderm, node, and midline cell populations responsible for establishment of the left-right body axis and head formation.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Desarrollo Embrionario/genética , Regulación del Desarrollo de la Expresión Génica , Estratos Germinativos/embriología , Proteínas de Unión al ADN/genética , Embrión de Mamíferos , Elementos de Facilitación Genéticos/fisiología , Eliminación de Gen , Perfilación de la Expresión Génica , Estratos Germinativos/metabolismo , Factor Nuclear 3-beta del Hepatocito/metabolismo , Proteínas con Dominio LIM/metabolismo , Proteínas con Homeodominio LIM/genética , Proteínas con Homeodominio LIM/metabolismo , Complejos Multiproteicos/genética , Complejos Multiproteicos/metabolismo , Factores de Transcripción Otx/metabolismo , Unión Proteica , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Vía de Señalización Wnt
2.
Development ; 143(10): 1663-73, 2016 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-27190036

RESUMEN

Mammary gland morphogenesis depends on a tight balance between cell proliferation, differentiation and apoptosis, to create a defined functional hierarchy within the epithelia. The limited availability of stem cell/progenitor markers has made it challenging to decipher lineage relationships. Here, we identify a rare subset of luminal progenitors that express the zinc finger transcriptional repressor Blimp1, and demonstrate that this subset of highly clonogenic luminal progenitors is required for mammary gland development. Conditional inactivation experiments using K14-Cre and WAPi-Cre deleter strains revealed essential functions at multiple developmental stages. Thus, Blimp1 regulates proliferation, apoptosis and alveolar cell maturation during puberty and pregnancy. Loss of Blimp1 disrupts epithelial architecture and lumen formation both in vivo and in three-dimensional (3D) primary cell cultures. Collectively, these results demonstrate that Blimp1 is required to maintain a highly proliferative luminal subset necessary for mammary gland development and homeostasis.


Asunto(s)
Glándulas Mamarias Animales/embriología , Glándulas Mamarias Animales/metabolismo , Proteínas Represoras/metabolismo , Células Madre/citología , Células Madre/metabolismo , Factores de Transcripción/metabolismo , Animales , Compartimento Celular/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Polaridad Celular/efectos de los fármacos , Células Cultivadas , Células Clonales , Células Epiteliales/citología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Células Epiteliales/ultraestructura , Femenino , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Hormonas/farmacología , Lactancia/efectos de los fármacos , Glándulas Mamarias Animales/citología , Ratones Endogámicos C57BL , Morfogénesis/efectos de los fármacos , Factor 1 de Unión al Dominio 1 de Regulación Positiva , Embarazo , Células Madre/efectos de los fármacos , Esteroides/farmacología , Regulación hacia Arriba/efectos de los fármacos
3.
PLoS Genet ; 11(7): e1005375, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26158850

RESUMEN

The neonatal intestine is a very complex and dynamic organ that must rapidly adapt and remodel in response to a barrage of environmental stimuli during the first few postnatal weeks. Recent studies demonstrate that the zinc finger transcriptional repressor Blimp1/Prdm1 plays an essential role governing postnatal reprogramming of intestinal enterocytes during this period. Functional loss results in global changes in gene expression patterns, particularly in genes associated with metabolic function. Here we engineered a knock-in allele expressing an eGFP-tagged fusion protein under control of the endogenous regulatory elements and performed genome wide ChIP-seq analysis to identify direct Blimp1 targets and further elucidate the function of Blimp1 in intestinal development. Comparison with published human and mouse datasets revealed a highly conserved core set of genes including interferon-inducible promoters. Here we show that the interferon-inducible transcriptional activator Irf1 is constitutively expressed throughout fetal and postnatal intestinal epithelium development. ChIP-seq demonstrates closely overlapping Blimp1 and Irf1 peaks at key components of the MHC class I pathway in fetal enterocytes. The onset of MHC class I expression coincides with down-regulated Blimp1 expression during the suckling to weaning transition. Collectively, these experiments strongly suggest that in addition to regulating the enterocyte metabolic switch, Blimp1 functions as a gatekeeper in opposition to Irf1 to prevent premature activation of the MHC class I pathway in villus epithelium to maintain tolerance in the neonatal intestine.


Asunto(s)
Antígenos de Histocompatibilidad Clase I/inmunología , Factor 1 Regulador del Interferón/metabolismo , Mucosa Intestinal/metabolismo , Placenta/metabolismo , Factores de Transcripción/metabolismo , Animales , Diferenciación Celular/genética , Línea Celular Tumoral , Enterocitos/citología , Femenino , Regulación del Desarrollo de la Expresión Génica , Técnicas de Sustitución del Gen , Proteínas Fluorescentes Verdes/genética , Humanos , Factor 1 Regulador del Interferón/genética , Mucosa Intestinal/crecimiento & desarrollo , Ratones , Placenta/citología , Factor 1 de Unión al Dominio 1 de Regulación Positiva , Embarazo , Regiones Promotoras Genéticas/genética , Elementos Reguladores de la Transcripción/genética , Factores de Transcripción/genética
4.
BMC Cancer ; 12: 252, 2012 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-22708734

RESUMEN

BACKGROUND: While it is now more than a decade since the first description of the gene mutation underlying the tumour predisposition syndrome multiple endocrine neoplasia type 1 (MEN1), the mechanism by which its protein product menin acts to prevent development of tumours is still poorly understood. METHODS: We undertook a genetic experiment to assess whether menin synergises with p53. Mice carrying various combinations of Men1 and Trp53 mutations were generated then survival and pathology assessed. RESULTS: While homozygous loss of Trp53 in mice resulted in early onset, aggressive tumours and profoundly reduced lifespan, heterozygous loss of either Trp53 or Men1 caused later onset disease, with a spectrum of tumours characteristic of each tumour suppressor gene. Loss of one copy of Men1 in animals also lacking both alleles of Trp53 did not exacerbate phenotype, based on survival, animal weight or sites of pathology, compared to Trp53 deletion alone. Dual heterozygous deletion of Men1 and Trp53 resulted in a small reduction in lifespan compared to the individual mutations, without new tumour sites. In the adrenal, we observed development of cortical tumours in dual heterozygous animals, as we have previously seen in Men1+/- animals, and there was loss of heterozygosity at the Men1 allele in these tumours. Median number of pathology observations per animal was increased in dual heterozygous animals compared with heterozygous loss of Trp53 alone. CONCLUSIONS: Simultaneous heterozygous deletion of Men1 in animals with either heterozygous or homozygous deletion of Trp53 did not result in formation of tumours at any new sites, implying additive rather than synergistic effects of these pathways. Mice that were Men1+/- in addition to Trp53+/- had tumours in endocrine as well as other sites, implying that increase in total tumour burden, at sites typically associated with either Men1 or Trp53 loss, contributed to the slight decrease in survival in Men1+/-: Trp53+/- animals in comparison with their littermates.


Asunto(s)
Transformación Celular Neoplásica/genética , Neoplasia Endocrina Múltiple Tipo 1/genética , Proteína p53 Supresora de Tumor/genética , Glándulas Suprarrenales/metabolismo , Glándulas Suprarrenales/patología , Animales , Peso Corporal , Transformación Celular Neoplásica/metabolismo , Genotipo , Ratones , Ratones Noqueados , Neoplasia Endocrina Múltiple Tipo 1/metabolismo , Mutación , Neoplasias/genética , Neoplasias/mortalidad , Neoplasias/patología , Páncreas/metabolismo , Páncreas/patología , Proteína p53 Supresora de Tumor/metabolismo
5.
CNS Drugs ; 36(1): 1-16, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34928485

RESUMEN

The cognitive dysfunction experienced by patients with schizophrenia represents a major unmet clinical need. We believe that enhancing synaptic function and plasticity by targeting kalirin may provide a novel means to remediate these symptoms. Karilin (a protein encoded by the KALRN gene) has multiple functional domains, including two Dbl homology (DH) guanine exchange factor (GEF) domains, which act to enhance the activity of the Rho family guanosine triphosphate (GTP)-ases. Here, we provide an overview of kalirin's roles in brain function and its therapeutic potential in schizophrenia. We outline how it mediates diverse effects via a suite of distinct isoforms that couple to members of the Rho GTPase family to regulate synapse formation and stabilisation, and how genomic and post-mortem data implicate it in schizophrenia. We then review the current state of knowledge about the influence of kalirin on brain function at a systems level, based largely on evidence from transgenic mouse models, which support its proposed role in regulating dendritic spine function and plasticity. We demonstrate that, whilst the GTPases are classically considered to be 'undruggable', targeting kalirin and other Rho GEFs provides a means to indirectly modulate their activity. Finally, we integrate across the information presented to assess the therapeutic potential of kalirin for schizophrenia and highlight the key outstanding questions required to advance it in this capacity; namely, the need for more information about the diversity and function of its isoforms, how these change across neurodevelopment, and how they affect brain function in vivo.


Asunto(s)
Disfunción Cognitiva/tratamiento farmacológico , Factores de Intercambio de Guanina Nucleótido/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Esquizofrenia/tratamiento farmacológico , Disfunción Cognitiva/complicaciones , Humanos , Plasticidad Neuronal , Esquizofrenia/complicaciones
6.
Trends Mol Med ; 27(11): 1022-1032, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34419330

RESUMEN

Patients with schizophrenia experience cognitive dysfunction and negative symptoms that do not respond to current drug treatments. Historical evidence is consistent with the hypothesis that these deficits are due, at least in part, to altered cortical synaptic plasticity (the ability of synapses to strengthen or weaken their activity), making this an attractive pathway for therapeutic intervention. However, while synaptic transmission and plasticity is well understood in model systems, it has been challenging to identify specific therapeutic targets for schizophrenia. New information is emerging from genomic findings, which converge on synaptic plasticity and provide a new window on the neurobiology of schizophrenia. Translating this information into therapeutic advances will require a multidisciplinary and collaborative approach.


Asunto(s)
Esquizofrenia , Genómica , Humanos , Plasticidad Neuronal/genética , Esquizofrenia/tratamiento farmacológico , Esquizofrenia/genética , Sinapsis/metabolismo , Transmisión Sináptica
7.
Nat Commun ; 11(1): 2782, 2020 06 03.
Artículo en Inglés | MEDLINE | ID: mdl-32493987

RESUMEN

The transcriptional repressor Blimp1 controls cell fate decisions in the developing embryo and adult tissues. Here we describe Blimp1 expression and functional requirements within maternal uterine tissues during pregnancy. Expression is robustly up-regulated at early post-implantation stages in the primary decidual zone (PDZ) surrounding the embryo. Conditional inactivation results in defective formation of the PDZ barrier and abnormal trophectoderm invasion. RNA-Seq analysis demonstrates down-regulated expression of genes involved in cell adhesion and markers of decidualisation. In contrast, genes controlling immune responses including IFNγ are up-regulated. ChIP-Seq experiments identify candidate targets unique to the decidua as well as those shared across diverse cell types including a highly conserved peak at the Csf-1 gene promoter. Interestingly Blimp1 inactivation results in up-regulated Csf1 expression and macrophage recruitment into maternal decidual tissues. These results identify Blimp1 as a critical regulator of tissue remodelling and maternal tolerance during early stages of pregnancy.


Asunto(s)
Decidua/metabolismo , Factor 1 de Unión al Dominio 1 de Regulación Positiva/metabolismo , Transcripción Genética , Animales , Decidua/ultraestructura , Ectodermo/metabolismo , Ectodermo/ultraestructura , Implantación del Embrión/genética , Femenino , Regulación del Desarrollo de la Expresión Génica , Factor Estimulante de Colonias de Macrófagos/genética , Factor Estimulante de Colonias de Macrófagos/metabolismo , Macrófagos/metabolismo , Masculino , Ratones Endogámicos C57BL , Mutación/genética , Embarazo , Regiones Promotoras Genéticas , Trofoblastos/metabolismo , Trofoblastos/ultraestructura , Regulación hacia Arriba/genética
8.
Int J Cancer ; 124(5): 1122-32, 2009 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-19058182

RESUMEN

Heterozygous disruption of the Men1 gene predisposes mice to the development of multiple endocrine tumors, accurately mimicking the human MEN1 cancer predisposition syndrome. Additionally, Men1(+/-) mice frequently develop sex cord adenomas. The mechanism underlying the susceptibility of these mice to sex cord tumor development has not been fully determined, but data suggest it may involve transcriptional regulation of key growth promoting/repressing genes. To identify potential menin-regulated genes that may be important for tumor suppression in sex cord cells, we compared the global gene expression profiles of testis and ovary adenomas with other endocrine tumors of the pancreas and pituitary from Men1 heterozygous mice and with control tissues. Gonadal tumors clustered separately from pancreas and pituitary tumors with only a few genes (e.g., Cdkn2c) commonly dysregulated in all tumor types. Testis and ovary tumors displayed a higher level of transcriptional similarity to each other than they did to their respective control tissues. Among genes that had decreased expression in tumors was significant over-representation of genes associated with the TGF-beta, hedgehog and Wnt signaling, indicating that loss of menin function affects these pathways at the level of transcription. Aberrant protein expression in Leydig and granulosa cells of 2 transcriptionally dysregulated gene products, Gata6 and Csf1r were confirmed by immunohistochemistry. We propose that sex cord tumor susceptibility in Men1(+/-) mice involves deregulated cell proliferation due to dysregulation of multiple cell growth regulating genes including: reduced Cdkn2c transcription, loss of TGF-beta pathway tumor suppressor function (e.g., Gata6) and transcriptional activation of Csf1r.


Asunto(s)
Factor de Transcripción GATA6/análisis , Perfilación de la Expresión Génica , Proteínas Proto-Oncogénicas/fisiología , Receptor de Factor Estimulante de Colonias de Macrófagos/análisis , Tumores de los Cordones Sexuales y Estroma de las Gónadas/etiología , Transducción de Señal/fisiología , Factor de Crecimiento Transformador beta/fisiología , Animales , Análisis por Conglomerados , Inmunohistoquímica , Ratones , Ratones Endogámicos C57BL , Proteínas Proto-Oncogénicas/genética
10.
Sci Rep ; 8(1): 237, 2018 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-29321612

RESUMEN

The transcriptional repressor Blimp-1 originally cloned as a silencer of type I interferon (IFN)-ß gene expression controls cell fate decisions in multiple tissue contexts. Conditional inactivation in the mammary gland was recently shown to disrupt epithelial cell architecture. Here we report that Blimp-1 regulates expression of viral defense, IFN signaling and MHC class I pathways, and directly targets the transcriptional activator Stat1. Blimp-1 functional loss in 3D cultures of mammary epithelial cells (MECs) results in accumulation of dsRNA and expression of type III IFN-λ. Cultures treated with IFN lambda similarly display defective lumen formation. These results demonstrate that type III IFN-λ profoundly influences the behavior of MECs and identify Blimp-1 as a critical regulator of IFN signaling cascades.


Asunto(s)
Células Epiteliales/metabolismo , Interferones/metabolismo , Factor 1 de Unión al Dominio 1 de Regulación Positiva/metabolismo , Animales , Células Epiteliales/efectos de los fármacos , Femenino , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Silenciador del Gen , Interferones/farmacología , Ratones , Ratones Noqueados , Factor 1 de Unión al Dominio 1 de Regulación Positiva/genética , Unión Proteica , ARN Bicatenario/genética , ARN Bicatenario/metabolismo , Factor de Transcripción STAT1/metabolismo , Transducción de Señal
11.
Cell Rep ; 24(8): 1977-1985.e7, 2018 08 21.
Artículo en Inglés | MEDLINE | ID: mdl-30134160

RESUMEN

Epiblast cells in the early post-implantation stage mammalian embryo undergo a transition described as lineage priming before cell fate allocation, but signaling pathways acting upstream remain ill defined. Genetic studies demonstrate that Smad2/3 double-mutant mouse embryos die shortly after implantation. To learn more about the molecular disturbances underlying this abrupt failure, here we characterized Smad2/3-deficient embryonic stem cells (ESCs). We found that Smad2/3 double-knockout ESCs induced to form epiblast-like cells (EpiLCs) display changes in naive and primed pluripotency marker gene expression, associated with the disruption of Oct4-bound distal regulatory elements. In the absence of Smad2/3, we observed enhanced Bmp target gene expression and de-repression of extra-embryonic gene expression. Cell fate allocation into all three embryonic germ layers is disrupted. Collectively, these experiments demonstrate that combinatorial Smad2/3 functional activities are required to maintain distinct embryonic and/or extra-embryonic cell identity during lineage priming in the epiblast before gastrulation.


Asunto(s)
Células Madre Embrionarias/metabolismo , Proteína Nodal/metabolismo , Animales , Diferenciación Celular , Humanos , Ratones , Transducción de Señal , Proteína Smad2
12.
Nat Struct Mol Biol ; 25(9): 766-777, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30127357

RESUMEN

The regulation of higher-order chromatin structure is complex and dynamic, and a full understanding of the suite of mechanisms governing this architecture is lacking. Here, we reveal the noncanonical SMC protein Smchd1 to be a novel regulator of long-range chromatin interactions in mice, and we add Smchd1 to the canon of epigenetic proteins required for Hox-gene regulation. The effect of losing Smchd1-dependent chromatin interactions has varying outcomes that depend on chromatin context. At autosomal targets transcriptionally sensitive to Smchd1 deletion, we found increased short-range interactions and ectopic enhancer activation. In contrast, the inactive X chromosome was transcriptionally refractive to Smchd1 ablation, despite chromosome-wide increases in short-range interactions. In the inactive X, we observed spreading of trimethylated histone H3 K27 (H3K27me3) domains into regions not normally decorated by this mark. Together, these data suggest that Smchd1 is able to insulate chromatin, thereby limiting access to other chromatin-modifying proteins.


Asunto(s)
Cromatina/metabolismo , Proteínas Cromosómicas no Histona/fisiología , Genes Homeobox , Familia de Multigenes , Cromosoma X , Animales , Proteínas Cromosómicas no Histona/genética , Elementos de Facilitación Genéticos , Eliminación de Gen , Silenciador del Gen , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
13.
Circ Res ; 97(6): e60-70, 2005 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-16109918

RESUMEN

Vascular endothelial growth factors (VEGFs) play significant roles in endothelial growth, survival, and function, and their potential use as therapeutic agents to promote the revascularization of ischemic tissues in being avidly explored. VEGF-A has received most attention, as it is a potent stimulator of vascular growth. Results in clinical trials of VEGF-A as a therapeutic agent have fallen short of high expectations because of serious edematous side effects caused by its activity in promoting vascular permeability. VEGF-B, a related factor, binds some of the VEGF-A receptors but not to VEGF receptor 2, which is implicated in the vascular permeability promoting activity of VEGF-A. Despite little in vitro evidence to date for the ability of Vegf-B to directly promote angiogenesis, recent data indicate that it may promote postnatal vascular growth in mice, suggesting that it may have potential therapeutic application. We have specifically studied the effects of VEGF-B on vascular growth in vivo and on angiogenesis in vitro by analyzing transgenic mice in which individual isoforms (VEGFB167Tg and VEGFB186Tg) of VEGF-B are overexpressed in endothelial cells. VEGFB167Tg and VEGFB186Tg mice displayed enhanced vascular growth in the Matrigel assay in vivo and during cutaneous wound healing. In the aortic explant assay, explants from VEGFB167Tg and VEGFB186Tg mice displayed elevated vascular growth, suggesting a direct effect of VEGF-B isoforms in potentiating angiogenesis. These data support the use of VEGF-B as a therapeutic agent to promote vascular growth, in part, by potentiating angiogenesis. Furthermore, the lack of vascular permeability activity associated with either transgenic overexpression of the VEGF-B gene in endothelial cells or application of VEGF-B protein to the skin of mice in the Miles assay indicates that use of VEGF-B as a therapy should not be associated with edematous side effects.


Asunto(s)
Células Endoteliales/fisiología , Neovascularización Fisiológica , Factor B de Crecimiento Endotelial Vascular/fisiología , Animales , Anticuerpos Monoclonales/inmunología , Aorta/fisiología , Permeabilidad Capilar , Humanos , Ratones , Ratones Transgénicos , Células Madre/fisiología , Factor B de Crecimiento Endotelial Vascular/genética , Cicatrización de Heridas
14.
Sci Rep ; 7(1): 6793, 2017 07 28.
Artículo en Inglés | MEDLINE | ID: mdl-28754907

RESUMEN

Trophoblast stem cells (TSCs) give rise to specialized cell types within the placenta. However, the regulatory mechanisms that guide trophoblast cell fate decisions during placenta development remain ill defined. Here we exploited ATAC-seq and transcriptional profiling strategies to describe dynamic changes in gene expression and chromatin accessibility during TSC differentiation. We detect significantly increased chromatin accessibility at key genes upregulated as TSCs exit from the stem cell state. However, downregulated gene expression is not simply due to the loss of chromatin accessibility in proximal regions. Additionally, transcriptional targets recognized by the zinc finger transcriptional repressor Prdm1/Blimp1, an essential regulator of placenta development, were identified in ChIP-seq experiments. Comparisons with previously reported ChIP-seq datasets for primordial germ cell-like cells and E18.5 small intestine, combined with functional annotation analysis revealed that Blimp1 has broadly shared as well as cell type-specific functional activities unique to the trophoblast lineage. Importantly, Blimp1 not only silences TSC gene expression but also prevents aberrant activation of divergent developmental programmes. Overall the present study provides new insights into the chromatin landscape and Blimp1-dependent regulatory networks governing trophoblast gene expression.


Asunto(s)
Diferenciación Celular , Cromatina/genética , Factor 1 de Unión al Dominio 1 de Regulación Positiva/genética , Trofoblastos/metabolismo , Animales , Células Cultivadas , Cromatina/metabolismo , Ensamble y Desensamble de Cromatina , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Regulación del Desarrollo de la Expresión Génica , Ratones , Factor 1 de Unión al Dominio 1 de Regulación Positiva/metabolismo , Trofoblastos/citología
15.
Nat Commun ; 7: 11414, 2016 04 25.
Artículo en Inglés | MEDLINE | ID: mdl-27108815

RESUMEN

Growth and survival of the mammalian embryo within the uterine environment depends on the placenta, a highly complex vascularized organ comprised of both maternal and foetal tissues. Recent experiments demonstrate that the zinc finger transcriptional repressor Prdm1/Blimp1 is essential for specification of spiral artery trophoblast giant cells (SpA-TGCs) that invade and remodel maternal blood vessels. To learn more about functional contributions made by Blimp1+ cell lineages here we perform the first single-cell RNA-seq analysis of the placenta. Cell types of both foetal and maternal origin are profiled. Comparisons with microarray datasets from mutant placenta and in vitro differentiated trophoblast stem cells allow us to identify Blimp1-dependent transcripts enriched in SpA-TGCs. Our experiments provide new insights into the functionally distinct cell types present at the maternal-foetal interface and advance our knowledge of dynamic gene expression patterns controlling placental morphogenesis and vascular mimicry.


Asunto(s)
Células Gigantes/metabolismo , Ratones/embriología , Factor 1 de Unión al Dominio 1 de Regulación Positiva/metabolismo , ARN/genética , Trofoblastos/metabolismo , Animales , Diferenciación Celular , Femenino , Regulación del Desarrollo de la Expresión Génica , Células Gigantes/citología , Intercambio Materno-Fetal , Ratones/genética , Ratones/metabolismo , Placenta/citología , Placenta/metabolismo , Factor 1 de Unión al Dominio 1 de Regulación Positiva/genética , Embarazo , ARN/metabolismo , Análisis de Secuencia de ARN , Especificidad de la Especie , Transcripción Genética , Trofoblastos/citología
16.
Epigenetics Chromatin ; 6(1): 19, 2013 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-23819640

RESUMEN

BACKGROUND: Smchd1 is an epigenetic modifier essential for X chromosome inactivation: female embryos lacking Smchd1 fail during midgestational development. Male mice are less affected by Smchd1-loss, with some (but not all) surviving to become fertile adults on the FVB/n genetic background. On other genetic backgrounds, all males lacking Smchd1 die perinatally. This suggests that, in addition to being critical for X inactivation, Smchd1 functions to control the expression of essential autosomal genes. RESULTS: Using genome-wide microarray expression profiling and RNA-seq, we have identified additional genes that fail X inactivation in female Smchd1 mutants and have identified autosomal genes in male mice where the normal expression pattern depends upon Smchd1. A subset of genes in the Snrpn imprinted gene cluster show an epigenetic signature and biallelic expression consistent with loss of imprinting in the absence of Smchd1. In addition, single nucleotide polymorphism analysis of expressed genes in the placenta shows that the Igf2r imprinted gene cluster is also disrupted, with Slc22a3 showing biallelic expression in the absence of Smchd1. In both cases, the disruption was not due to loss of the differential methylation that marks the imprint control region, but affected genes remote from this primary imprint controlling element. The clustered protocadherins (Pcdhα, Pcdhß, and Pcdhγ) also show altered expression levels, suggesting that their unique pattern of random combinatorial monoallelic expression might also be disrupted. CONCLUSIONS: Smchd1 has a role in the expression of several autosomal gene clusters that are subject to monoallelic expression, rather than being restricted to functioning uniquely in X inactivation. Our findings, combined with the recent report implicating heterozygous mutations of SMCHD1 as a causal factor in the digenically inherited muscular weakness syndrome facioscapulohumeral muscular dystrophy-2, highlight the potential importance of Smchd1 in the etiology of diverse human diseases.

17.
Mol Cell Biol ; 32(17): 3403-13, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22733990

RESUMEN

Prdm1/Blimp-1 is a master regulator of gene expression in diverse tissues of the developing embryo and adult organism. Its C-terminal zinc finger domain mediates nuclear import, DNA binding, and recruitment of the corepressors G9a and HDAC1/2. Alternatively spliced transcripts lacking exon 7 sequences encode a structurally divergent isoform (Blimp-1Δexon7) predicted to have distinct functions. Here we demonstrate that the short Blimp-1Δexon7 isoform lacks DNA binding activity and fails to bind G9a or HDAC1/2 but retains the ability to interact with PRMT5. To investigate functional roles of alternative splicing in vivo, we engineered novel mouse strains via embryonic stem (ES) cell technology. Like null mutants, embryos carrying a targeted deletion of exon 7 and exclusively expressing Blimp-1Δexon7 die at around embryonic day 10.5 (E10.5) due to placental defects. In heterozygous Δexon7 mice, there is no evidence of dominant-negative effects. Mice carrying a knock-in allele with an exon 6-exon 7 fusion express full-length Blimp-1 only, develop normally, are healthy and fertile as adults, and efficiently generate mature plasma cells. These findings strongly suggest that the short Blimp-1Δexon7 isoform is dispensable. We propose that developmentally regulated alternative splicing is influenced by chromatin structure at the locus and fine-tunes Blimp-1's functional capabilities.


Asunto(s)
Empalme Alternativo , Proteínas Co-Represoras/metabolismo , ADN/metabolismo , Regulación del Desarrollo de la Expresión Génica , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Diferenciación Celular , Células Cultivadas , Células Dendríticas/citología , Células Dendríticas/metabolismo , Embrión de Mamíferos/embriología , Embrión de Mamíferos/metabolismo , Exones , Femenino , Eliminación de Gen , Marcación de Gen , Ratones , Datos de Secuencia Molecular , Células Plasmáticas/citología , Células Plasmáticas/metabolismo , Factor 1 de Unión al Dominio 1 de Regulación Positiva , Unión Proteica , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Factores de Transcripción/química
18.
Pancreas ; 39(8): 1140-6, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20531244

RESUMEN

OBJECTIVES: To identify gene expression alterations associated with insulinoma formation and progression in 2 mouse models of multiple endocrine neoplasia type 1. METHODS: Mice were killed at 12 or 16 months, and pancreatic islets were isolated by enzymatic and physical disruption. Islets were separated by size representing control, normal, hyperplastic, and adenomous islets. RNA was isolated from these islets and profiled on Sentrix Mouse-6 Expression version 1 BeadChips. Array data were analyzed in GeneSpring. RESULTS: One hundred and one genes that were significantly (P ≤ 0.05) altered in hyperplastic islets and insulinomas compared with normal islets were identified. Of these, 64 gene elements showed reduced messenger RNA levels and 37 gene elements had increased gene expression compared with control islets. Altered expression of 3 genes, namely, Gata6, Tspan8, and s100a8, was confirmed by quantitative reverse transcription-polymerase chain reaction, and aberrant levels of Tspan8 and Lmo2 protein measured by Western blot correlated with the changes in messenger RNA levels. CONCLUSIONS: These results suggest that alterations in gene expression of Gata6, Tspan8, S100a8, and Lmo2 may act via novel pathways that play functionally important roles in Men1-associated tumor progression.


Asunto(s)
Perfilación de la Expresión Génica , Insulinoma/genética , Neoplasia Endocrina Múltiple Tipo 1/genética , Neoplasias Pancreáticas/genética , Proteínas Adaptadoras Transductoras de Señales , Animales , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/metabolismo , Western Blotting , Calgranulina A/genética , Calgranulina A/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Progresión de la Enfermedad , Femenino , Factor de Transcripción GATA6/genética , Factor de Transcripción GATA6/metabolismo , Humanos , Insulinoma/etiología , Insulinoma/metabolismo , Islotes Pancreáticos/metabolismo , Islotes Pancreáticos/patología , Proteínas con Dominio LIM , Masculino , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Metaloproteínas/genética , Metaloproteínas/metabolismo , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Neoplasia Endocrina Múltiple Tipo 1/complicaciones , Neoplasia Endocrina Múltiple Tipo 1/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Neoplasias Pancreáticas/etiología , Neoplasias Pancreáticas/metabolismo , Proteínas Proto-Oncogénicas/deficiencia , Proteínas Proto-Oncogénicas/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Tetraspaninas
19.
Neoplasia ; 12(3): 235-43, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20234817

RESUMEN

Using a Cre/loxP system, we have determined the phenotypic consequences attributable to in vivo deletion of both Rb1 and Trp53 in the mouse adrenal medulla. The coablation of these two tumor suppressor genes during embryogenesis did not disrupt adrenal gland development but resulted in the neoplastic transformation of the neural crest-derived adrenal medulla, yielding pheochromocytomas (PCCs) that developed with complete penetrance and were inevitably bilateral. Despite their typically benign status, these PCCs had profound ramifications on mouse vitality, with effected mice having a median survival of only 121 days. Evaluation of these PCCs by both immunohistochemistry and electron microscopy revealed that most Rb1(-/-):Trp53(-/-) chromaffin cells possessed atypical chromagenic vesicles that did not seem capable of appropriately storing synthesized catecholamines. The structural remodeling of the heart in mice harboring Rb1(-/-):Trp53(-/-) PCCs suggests that the mortality of these mice may be attributable to the inappropriate release of catecholamines from the mutated adrenal chromaffin cells. On the basis of the collective data from Rb1 and Trp53 knockout mouse models, it seems that the conversion of Rb1 loss-driven adrenal medulla hyperplasia to PCC can be greatly enhanced by the compound loss of Trp53, whereas the loss of Trp53 alone is generally ineffectual on adrenal chromaffin cell homeostasis. Consequently, the Trp53 tumor suppressor gene is an efficient genetic modifier of Rb1 loss in the development of PCC, and their compound loss in the adrenal medulla has a profound impact on both cellular homeostasis and animal vitality.


Asunto(s)
Neoplasias de las Glándulas Suprarrenales/patología , Genes p53/fisiología , Feocromocitoma/patología , Proteína de Retinoblastoma/fisiología , Neoplasias de las Glándulas Suprarrenales/genética , Neoplasias de las Glándulas Suprarrenales/metabolismo , Animales , Catecolaminas/metabolismo , Femenino , Técnicas para Inmunoenzimas , Integrasas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Feocromocitoma/genética , Feocromocitoma/metabolismo , Transgenes/fisiología
20.
Pigment Cell Melanoma Res ; 23(4): 564-70, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20518858

RESUMEN

There has been uncertainty regarding the precise role that the pocket protein Rb1 plays in murine melanocyte homeostasis. It has been reported that the TAT-Cre mediated loss of exon 19 from a floxed Rb1 allele causes melanocyte apoptosis in vivo and in vitro. This is at variance with other findings showing, either directly or indirectly, that Rb1 loss in melanocytes has no noticeable effect in vivo, but in vitro leads to a semi-transformed phenotype. In this study, we show that Rb1-null melanocytes lacking exon 19 do not undergo apoptosis and survive both in vitro and in vivo, irrespective of the developmental stage at which Cre-mediated ablation of the exon occurs. Further, Rb1 loss has no serious long-term ramifications on melanocyte homeostasis in vivo, with Rb1-null melanocytes being detected in the skin after numerous hair cycles, inferring that the melanocyte stem cell population carrying the Cre-mediated deletion is maintained. Consequently, whilst Rb1 loss in the melanocyte is able to alter cellular behaviour in vitro, it appears inconsequential with respect to melanocyte homeostasis in the mouse skin.


Asunto(s)
Cabello/metabolismo , Homeostasis , Melanocitos/metabolismo , Proteína de Retinoblastoma/deficiencia , Piel/metabolismo , Animales , Cabello/patología , Ratones , Ratones Noqueados , Proteína de Retinoblastoma/genética , Proteína de Retinoblastoma/metabolismo , Piel/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA