Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 56
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 183(3): 702-716.e14, 2020 10 29.
Artículo en Inglés | MEDLINE | ID: mdl-33125890

RESUMEN

The cellular complexity and scale of the early liver have constrained analyses examining its emergence during organogenesis. To circumvent these issues, we analyzed 45,334 single-cell transcriptomes from embryonic day (E)7.5, when endoderm progenitors are specified, to E10.5 liver, when liver parenchymal and non-parenchymal cell lineages emerge. Our data detail divergence of vascular and sinusoidal endothelia, including a distinct transcriptional profile for sinusoidal endothelial specification by E8.75. We characterize two distinct mesothelial cell types as well as early hepatic stellate cells and reveal distinct spatiotemporal distributions for these populations. We capture transcriptional profiles for hepatoblast specification and migration, including the emergence of a hepatomesenchymal cell type and evidence for hepatoblast collective cell migration. Further, we identify cell-cell interactions during the organization of the primitive sinusoid. This study provides a comprehensive atlas of liver lineage establishment from the endoderm and mesoderm through to the organization of the primitive sinusoid at single-cell resolution.


Asunto(s)
Linaje de la Célula/genética , Hígado/citología , Hígado/metabolismo , Análisis de la Célula Individual , Transcriptoma/genética , Animales , Movimiento Celular , Embrión de Mamíferos/citología , Endotelio/citología , Mesodermo/citología , Ratones , Transducción de Señal , Células Madre/citología
2.
Nature ; 569(7756): 361-367, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30959515

RESUMEN

Here we delineate the ontogeny of the mammalian endoderm by generating 112,217 single-cell transcriptomes, which represent all endoderm populations within the mouse embryo until midgestation. We use graph-based approaches to model differentiating cells, which provides a spatio-temporal characterization of developmental trajectories and defines the transcriptional architecture that accompanies the emergence of the first (primitive or extra-embryonic) endodermal population and its sister pluripotent (embryonic) epiblast lineage. We uncover a relationship between descendants of these two lineages, in which epiblast cells differentiate into endoderm at two distinct time points-before and during gastrulation. Trajectories of endoderm cells were mapped as they acquired embryonic versus extra-embryonic fates and as they spatially converged within the nascent gut endoderm, which revealed these cells to be globally similar but retain aspects of their lineage history. We observed the regionalized identity of cells along the anterior-posterior axis of the emergent gut tube, which reflects their embryonic or extra-embryonic origin, and the coordinated patterning of these cells into organ-specific territories.


Asunto(s)
Endodermo/citología , Endodermo/embriología , Intestinos/citología , Intestinos/embriología , Análisis de la Célula Individual , Animales , Blastocisto/citología , Tipificación del Cuerpo , Diferenciación Celular , Linaje de la Célula , Femenino , Gastrulación , Masculino , Ratones
3.
Development ; 148(17)2021 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-34478514

RESUMEN

Liver development is controlled by key signals and transcription factors that drive cell proliferation, migration, differentiation and functional maturation. In the adult liver, cell maturity can be perturbed by genetic and environmental factors that disrupt hepatic identity and function. Developmental signals and fetal genetic programmes are often dysregulated or reactivated, leading to dedifferentiation and disease. Here, we highlight signalling pathways and transcriptional regulators that drive liver cell development and primary liver cancers. We also discuss emerging models derived from pluripotent stem cells, 3D organoids and bioengineering for improved studies of signalling pathways in liver cancer and regenerative medicine.


Asunto(s)
Neoplasias Hepáticas/patología , Hígado/crecimiento & desarrollo , Transducción de Señal/fisiología , Factores de Transcripción/metabolismo , Diferenciación Celular , Células Epiteliales/citología , Células Epiteliales/metabolismo , Hepatocitos/citología , Hepatocitos/metabolismo , Humanos , Hígado/citología , Hígado/metabolismo , Neoplasias Hepáticas/metabolismo , Regeneración Hepática , Ingeniería de Tejidos
4.
Nucleic Acids Res ; 50(15): 8547-8565, 2022 08 26.
Artículo en Inglés | MEDLINE | ID: mdl-35904801

RESUMEN

The transcription factor SOX9 is activated at the onset of endothelial-to-mesenchymal transition (EndMT) during embryonic development and in pathological conditions. Its roles in regulating these processes, however, are not clear. Using human umbilical vein endothelial cells (HUVECs) as an EndMT model, we show that SOX9 expression alone is sufficient to activate mesenchymal genes and steer endothelial cells towards a mesenchymal fate. By genome-wide mapping of the chromatin landscape, we show that SOX9 displays features of a pioneer transcription factor, such as opening of chromatin and leading to deposition of active histone modifications at silent chromatin regions, guided by SOX dimer motifs and H2A.Z enrichment. We further observe highly transient and dynamic SOX9 binding, possibly promoted through its eviction by histone phosphorylation. However, while SOX9 binding is dynamic, changes in the chromatin landscape and cell fate induced by SOX9 are persistent. Finally, our analysis of single-cell chromatin accessibility indicates that SOX9 opens chromatin to drive EndMT in atherosclerotic lesions in vivo. This study provides new insight into key molecular functions of SOX9 and mechanisms of EndMT and highlights the crucial developmental role of SOX9 and relevance to human disease.


Asunto(s)
Cromatina , Regulación de la Expresión Génica , Factor de Transcripción SOX9/metabolismo , Cromatina/genética , Cromatina/metabolismo , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Transducción de Señal
5.
Hepatology ; 70(4): 1360-1376, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-30933372

RESUMEN

Cell-fate determination is influenced by interactions between master transcription factors (TFs) and cis-regulatory elements. Hepatocyte nuclear factor 4 alpha (HNF4A), a liver-enriched TF, acts as a master controller in specification of hepatic progenitor cells by regulating a network of TFs to control onset of hepatocyte cell fate. Using analysis of genome-wide histone modifications, DNA methylation, and hydroxymethylation in mouse hepatocytes, we show that HNF4A occupies active enhancers in hepatocytes and is essential for active histone and DNA signatures, especially acetylation of lysine 27 of histone 3 (H3K27ac) and 5-hydroxymethylcytosine (5hmC). In mice lacking HNF4A protein in hepatocytes, we observed a decrease in both H3K27ac and hydroxymethylation at regions bound by HNF4A. Mechanistically, HNF4A-associated hydroxymethylation (5hmC) requires its interaction with ten-eleven translocation methylcytosine dioxygenase 3 (TET3), a protein responsible for oxidation from 5mC to 5hmC. Furthermore, HNF4A regulates TET3 expression in liver by directly binding to an enhancer region. Conclusion: In conclusion, we identified that HNF4A is required for the active epigenetic state at enhancers that amplifies transcription of genes in hepatocytes.


Asunto(s)
Metilación de ADN/genética , Epigenómica , Factor Nuclear 4 del Hepatocito/genética , Hepatocitos/metabolismo , Hígado/patología , Animales , Diferenciación Celular/genética , Células Cultivadas , Femenino , Factor Nuclear 4 del Hepatocito/metabolismo , Hepatocitos/patología , Humanos , Ratones , Ratones Endogámicos C57BL , Modelos Animales , Sensibilidad y Especificidad , Células Madre/citología , Células Madre/metabolismo , Activación Transcripcional/genética
6.
Development ; 142(24): 4340-50, 2015 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-26525672

RESUMEN

Heart valve formation initiates when endothelial cells of the heart transform into mesenchyme and populate the cardiac cushions. The transcription factor SOX9 is highly expressed in the cardiac cushion mesenchyme, and is essential for heart valve development. Loss of Sox9 in mouse cardiac cushion mesenchyme alters cell proliferation, embryonic survival, and valve formation. Despite this important role, little is known about how SOX9 regulates heart valve formation or its transcriptional targets. Therefore, we mapped putative SOX9 binding sites by ChIP-Seq in E12.5 heart valves, a stage at which the valve mesenchyme is actively proliferating and initiating differentiation. Embryonic heart valves have been shown to express a high number of genes that are associated with chondrogenesis, including several extracellular matrix proteins and transcription factors that regulate chondrogenesis. Therefore, we compared regions of putative SOX9 DNA binding between E12.5 heart valves and E12.5 limb buds. We identified context-dependent and context-independent SOX9-interacting regions throughout the genome. Analysis of context-independent SOX9 binding suggests an extensive role for SOX9 across tissues in regulating proliferation-associated genes including key components of the AP-1 complex. Integrative analysis of tissue-specific SOX9-interacting regions and gene expression profiles on Sox9-deficient heart valves demonstrated that SOX9 controls the expression of several transcription factors with previously identified roles in heart valve development, including Twist1, Sox4, Mecom and Pitx2. Together, our data identify SOX9-coordinated transcriptional hierarchies that control cell proliferation and differentiation during valve formation.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Válvulas Cardíacas/embriología , Válvulas Cardíacas/metabolismo , Factor de Transcripción SOX9/metabolismo , Animales , Proliferación Celular , Inmunoprecipitación de Cromatina , ADN/metabolismo , Extremidades/embriología , Redes Reguladoras de Genes , Ratones , Modelos Biológicos , Regiones Promotoras Genéticas/genética , Unión Proteica/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Sitio de Iniciación de la Transcripción
7.
Development ; 141(19): 3772-81, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25209250

RESUMEN

Although many regulatory networks involved in defining definitive endoderm have been identified, the mechanisms through which these networks interact to pattern the endoderm are less well understood. To explore the mechanisms involved in midgut patterning, we dissected the transcriptional regulatory elements of nephrocan (Nepn), the earliest known midgut specific gene in mice. We observed that Nepn expression is dramatically reduced in Sox17(-/-) and Raldh2(-/-) embryos compared with wild-type embryos. We further show that Nepn is directly regulated by Sox17 and the retinoic acid (RA) receptor via two enhancer elements located upstream of the gene. Moreover, Nepn expression is modulated by Activin signaling, with high levels inhibiting and low levels enhancing RA-dependent expression. In Foxh1(-/-) embryos in which Nodal signaling is reduced, the Nepn expression domain is expanded into the anterior gut region, confirming that Nodal signaling can modulate its expression in vivo. Together, Sox17 is required for Nepn expression in the definitive endoderm, while RA signaling restricts expression to the midgut region. A balance of Nodal/Activin signaling regulates the anterior boundary of the midgut expression domain.


Asunto(s)
Tipificación del Cuerpo/fisiología , Endodermo/fisiología , Tracto Gastrointestinal/embriología , Regulación del Desarrollo de la Expresión Génica/fisiología , Redes Reguladoras de Genes/fisiología , Glicoproteínas/metabolismo , Transducción de Señal/fisiología , Activinas/metabolismo , Aldehído Oxidorreductasas/metabolismo , Animales , Ensayo de Cambio de Movilidad Electroforética , Redes Reguladoras de Genes/genética , Vectores Genéticos/genética , Proteínas HMGB/metabolismo , Péptidos y Proteínas de Señalización Intercelular , Luciferasas , Ratones , Ratones Noqueados , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores de Ácido Retinoico/metabolismo , Factores de Transcripción SOXF/metabolismo
8.
Gynecol Oncol ; 147(3): 663-671, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-29079036

RESUMEN

OBJECTIVE: APELA is a small, secreted peptide that can function as a ligand for the G-protein coupled receptor, Apelin Receptor (APLNR, APJ). APELA plays an essential role in endoderm differentiation and cardiac development during embryogenesis. We investigated whether APELA exerts any functions in cancer progression. METHODS: The Cancer Genome Atlas (TCGA) RNA sequencing datasets, microarray from an OCCC mouse model, and RNA isolated from fresh frozen and FFPE patient tissue were used to assess APELA expression. APELA knockout ovarian clear cell carcinoma (OCCC) cell lines were generated using CRISPR/Cas9. RESULTS: APELA was expressed in various ovarian cancer histotypes and was especially elevated in OCCC. Disruption of APELA expression in OCCC cell lines suppressed cell growth and migration, and altered cell-cycle progression. Moreover, addition of human recombinant APELA peptide to the OCCC cell line OVISE promoted cell growth and migration. Interestingly, OVISE cells do not express APLNR, suggesting that APELA can function through an APLNR-independent pathway. Furthermore, APELA affected cell growth and cell cycle progression in a p53-dependent manner. In addition, APELA knockdown induced p53 expression in cancer cell lines. CONCLUSIONS: Our findings uncover a potential oncogenic role for APELA in promoting ovarian tumour progression and provide a possible therapeutic strategy in ovarian cancer by targeting APELA.


Asunto(s)
Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Hormonas Peptídicas/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Animales , Apelina/metabolismo , Receptores de Apelina/metabolismo , Procesos de Crecimiento Celular/fisiología , Línea Celular Tumoral , Movimiento Celular/fisiología , Femenino , Xenoinjertos , Humanos , Ratones , Ratones Endogámicos NOD , Ratones SCID
9.
Dev Biol ; 397(2): 257-66, 2015 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-25478910

RESUMEN

Huntington disease (HD) is an adult-onset neurodegenerative disease characterized by motor, cognitive, and psychiatric symptoms that is caused by a CAG expansion in the HTT gene. Palmitoylation is the addition of saturated fatty acids to proteins by DHHC palmitoylacyl transferases. HTT is palmitoylated by huntingtin interacting proteins 14 and 14-like (HIP14 and HIP14L or ZDHHC17 and 13 respectively). Mutant HTT is less palmitoylated and this reduction of palmitoylation accelerates its aggregation and increases cellular toxicity. Mouse models deficient in either Hip14 (Hip14(-/-)) or Hip14l (Hip14l(-/-)) develop HD-like phenotypes. The biological function of HTT palmitoylation and the role that loss of HTT palmitoylation plays in the pathogenesis of HD are unknown. To address these questions mice deficient for both genes were created. Loss of Hip14 and Hip14l leads to early embryonic lethality at day embryonic day 10-11 due to failed chorioallantoic fusion. The chorion is thickened and disorganized and the allantois does not fuse correctly with the chorion and forms a balloon-like shape compared to Hip14l(-/-); Hip14(+/+) littermate control embryos. Interestingly, the Hip14(-/-) ; Hip14(-/-) embryos share many features with the Htt(-/-) embryos, including folding of the yolk sac, a bulb shaped allantois, and a thickened and disorganized chorion. This may be due to a decrease in HTT palmitoylation. In Hip14(-/-); Hip14l(-/-) mouse embryonic fibroblasts show a 25% decrease in HTT palmitoylation compared to wild type cells. This is the first description of a double PAT deficient mouse model where loss of a PAT or multiple PATs results in embryonic lethality in mammals. These results reinforce the physiological importance of palmitoylation during embryogenesis.


Asunto(s)
Aciltransferasas/metabolismo , Membrana Corioalantoides/embriología , Fusión de Membrana/genética , Placenta/embriología , Proteínas de Transporte de Serotonina en la Membrana Plasmática/metabolismo , Aciltransferasas/genética , Animales , Western Blotting , Femenino , Genotipo , Hibridación in Situ , Lipoilación , Fusión de Membrana/fisiología , Ratones , Ratones Noqueados , Embarazo , Reacción en Cadena en Tiempo Real de la Polimerasa
10.
PLoS Pathog ; 10(4): e1004070, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24722419

RESUMEN

Intercalated disks (ICDs) are substantial connections maintaining cardiac structures and mediating signal communications among cardiomyocytes. Deficiency in ICD components such as desmosomes, fascia adherens and gap junctions leads to heart dysfunction. Coxsackievirus B3 (CVB3) infection induces cardiac failure but its pathogenic effect on ICDs is unclear. Here we show that CVB3-induced miR-21 expression affects ICD structure, i.e., upregulated miR-21 targets YOD1, a deubiquitinating enzyme, to enhance the K48-linked ubiquitination and degradation of desmin, resulting in disruption of desmosomes. Inhibition of miR-21 preserves desmin during CVB3 infection. Treatment with proteasome inhibitors blocks miR-21-mediated desmin degradation. Transfection of miR-21 or knockdown of YOD1 triggers co-localization of desmin with proteasomes. We also identified K108 and K406 as important sites for desmin ubiquintination and degradation. In addition, miR-21 directly targets vinculin, leading to disturbed fascia adherens evidenced by the suppression and disorientation of pan-cadherin and α-E-catenin proteins, two fascia adherens-components. Our findings suggest a new mechanism of miR-21 in modulating cell-cell interactions of cardiomyocytes during CVB3 infection.


Asunto(s)
Comunicación Celular , Enterovirus Humano B/metabolismo , Infecciones por Enterovirus/metabolismo , Regulación de la Expresión Génica , MicroARNs/biosíntesis , Miocitos Cardíacos/metabolismo , Animales , Desmina/genética , Desmina/metabolismo , Enterovirus Humano B/genética , Infecciones por Enterovirus/genética , Infecciones por Enterovirus/patología , Técnicas de Silenciamiento del Gen , Masculino , Ratones , MicroARNs/genética , Miocitos Cardíacos/patología , Miocitos Cardíacos/virología , Proteolisis , Ubiquitinación/genética
11.
PLoS Genet ; 8(9): e1002903, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22969434

RESUMEN

Left-sided congenital heart disease (CHD) encompasses a spectrum of malformations that range from bicuspid aortic valve to hypoplastic left heart syndrome. It contributes significantly to infant mortality and has serious implications in adult cardiology. Although left-sided CHD is known to be highly heritable, the underlying genetic determinants are largely unidentified. In this study, we sought to determine the impact of structural genomic variation on left-sided CHD and compared multiplex families (464 individuals with 174 affecteds (37.5%) in 59 multiplex families and 8 trios) to 1,582 well-phenotyped controls. 73 unique inherited or de novo CNVs in 54 individuals were identified in the left-sided CHD cohort. After stringent filtering, our gene inventory reveals 25 new candidates for LS-CHD pathogenesis, such as SMC1A, MFAP4, and CTHRC1, and overlaps with several known syndromic loci. Conservative estimation examining the overlap of the prioritized gene content with CNVs present only in affected individuals in our cohort implies a strong effect for unique CNVs in at least 10% of left-sided CHD cases. Enrichment testing of gene content in all identified CNVs showed a significant association with angiogenesis. In this first family-based CNV study of left-sided CHD, we found that both co-segregating and de novo events associate with disease in a complex fashion at structural genomic level. Often viewed as an anatomically circumscript disease, a subset of left-sided CHD may in fact reflect more general genetic perturbations of angiogenesis and/or vascular biology.


Asunto(s)
Variaciones en el Número de Copia de ADN , Cardiopatías Congénitas/genética , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Animales , Niño , Preescolar , Familia , Femenino , Corazón/embriología , Humanos , Masculino , Ratones , Persona de Mediana Edad , Miocardio/metabolismo , Neovascularización Fisiológica , Adulto Joven
12.
Dev Dyn ; 243(7): 894-905, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24633789

RESUMEN

BACKGROUND: Valvuloseptal defects are the most common congenital heart defects. Notch signaling-induced endothelial-to-mesenchymal transition (EMT) in the atrioventricular canal (AVC) cushions at murine embryonic day (E)9.5 is a required step during early valve development. Insights to the transcriptional network that is activated in endocardial cells (EC) during EMT and how these pathways direct valve maturation are lacking. RESULTS: We show that at E11.5, AVC-EC retain the ability to undergo Notch-dependent EMT when explanted on collagen. EC-Notch inhibition at E10.5 blocks expression of known mesenchymal genes in E11.5 AVC-EC. To understand the genetic network and AVC development downstream of Notch signaling beyond E9.5, we constructed Tag-Seq libraries corresponding to different cell types of the E11.5 AVC and atrium in wild-type mice and in EC-Notch inhibited mice. We identified 1,400 potential Notch targets in the AVC-EC, of which 124 are transcription factors (TF). From the 124 TFs, we constructed a transcriptional hierarchy and identify 10 upstream TFs within the network. CONCLUSIONS: We validated 4 of the upstream TFs as Notch targets that are enriched in AVC-EC. Functionally, we show these 4 TFs regulate EMT in AVC explant assays. These novel signaling pathways downstream of Notch are potentially relevant to valve development.


Asunto(s)
Transdiferenciación Celular/genética , Cojinetes Endocárdicos/embriología , Cojinetes Endocárdicos/metabolismo , Regulación del Desarrollo de la Expresión Génica/genética , Redes Reguladoras de Genes/genética , Receptores Notch/metabolismo , Animales , Línea Celular , Transdiferenciación Celular/fisiología , Femenino , Regulación del Desarrollo de la Expresión Génica/fisiología , Redes Reguladoras de Genes/fisiología , Humanos , Masculino , Ratones , Embarazo , Receptores Notch/genética
13.
Hepatology ; 57(6): 2491-501, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23315977

RESUMEN

MicroRNAs (miRNAs) are recently discovered small RNA molecules that regulate developmental processes, such as proliferation, differentiation, and apoptosis; however, the identity of miRNAs and their functions during liver development are largely unknown. Here we investigated the miRNA and gene expression profiles for embryonic day (E)8.5 endoderm, E14.5 Dlk1(+) liver cells (hepatoblasts), and adult liver by employing Illumina sequencing. We found that miRNAs were abundantly expressed at all three stages. Using K-means clustering analysis, 13 miRNA clusters with distinct temporal expression patterns were identified. mir302b, an endoderm-enriched miRNA, was identified as an miRNA whose predicted targets are expressed highly in E14.5 hepatoblasts but low in the endoderm. We validated the expression of mir302b in the endoderm by whole-mount in situ hybridization. Interestingly, mir20a, the most highly expressed miRNA in the endoderm library, was also predicted to regulate some of the same targets as mir302b. We found that through targeting Tgfbr2, mir302b and mir20a are able to regulate transforming growth factor beta (TGFß) signal transduction. Moreover, mir302b can repress liver markers in an embryonic stem cell differentiation model. Collectively, we uncovered dynamic patterns of individual miRNAs during liver development, as well as miRNA networks that could be essential for the specification and differentiation of liver progenitors. (HEPATOLOGY 2013).


Asunto(s)
Hígado/embriología , MicroARNs/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Animales , Benzodioxoles/farmacología , Diferenciación Celular , Células Madre Embrionarias/fisiología , Endodermo/metabolismo , Femenino , Tracto Gastrointestinal/metabolismo , Perfilación de la Expresión Génica , Genoma , Imidazoles/farmacología , Hígado/metabolismo , Masculino , Ratones , Organogénesis , Proteínas Serina-Treonina Quinasas/metabolismo , Piridinas/farmacología , ARN Mensajero/metabolismo , Receptor Tipo II de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Transducción de Señal , Factores de Transcripción p300-CBP/metabolismo
14.
Cell Mol Life Sci ; 70(16): 2899-917, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23161060

RESUMEN

Congenital heart defects affect approximately 1-5 % of human newborns each year, and of these cardiac defects 20-30 % are due to heart valve abnormalities. Recent literature indicates that the key factors and pathways that regulate valve development are also implicated in congenital heart defects and valve disease. Currently, there are limited options for treatment of valve disease, and therefore having a better understanding of valve development can contribute critical insight into congenital valve defects and disease. There are three major signaling pathways required for early specification and initiation of endothelial-to-mesenchymal transformation (EMT) in the cardiac cushions: BMP, TGF-ß, and Notch signaling. BMPs secreted from the myocardium set up the environment for the overlying endocardium to become activated; Notch signaling initiates EMT; and both BMP and TGF-ß signaling synergize with Notch to promote the transition of endothelia to mesenchyme and the mesenchymal cell invasiveness. Together, these three essential signaling pathways help form the cardiac cushions and populate them with mesenchyme and, consequently, set off the cascade of events required to develop mature heart valves. Furthermore, integration and cross-talk between these pathways generate highly stratified and delicate valve leaflets and septa of the heart. Here, we discuss BMP, TGF-ß, and Notch signaling pathways during mouse cardiac cushion formation and how they together produce a coordinated EMT response in the developing mouse valves.


Asunto(s)
Proteínas Morfogenéticas Óseas/metabolismo , Válvulas Cardíacas/crecimiento & desarrollo , Válvulas Cardíacas/metabolismo , Receptores Notch/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Animales , Humanos , Transducción de Señal
15.
Commun Biol ; 7(1): 144, 2024 01 31.
Artículo en Inglés | MEDLINE | ID: mdl-38297077

RESUMEN

Hepatocyte nuclear factor 4A (HNF4A/NR2a1), a transcriptional regulator of hepatocyte identity, controls genes that are crucial for liver functions, primarily through binding to enhancers. In mammalian cells, active and primed enhancers are marked by monomethylation of histone 3 (H3) at lysine 4 (K4) (H3K4me1) in a cell type-specific manner. How this modification is established and maintained at enhancers in connection with transcription factors (TFs) remains unknown. Using analysis of genome-wide histone modifications, TF binding, chromatin accessibility and gene expression, we show that HNF4A is essential for an active chromatin state. Using HNF4A loss and gain of function experiments in vivo and in cell lines in vitro, we show that HNF4A affects H3K4me1, H3K27ac and chromatin accessibility, highlighting its contribution to the establishment and maintenance of a transcriptionally permissive epigenetic state. Mechanistically, HNF4A interacts with the mixed-lineage leukaemia 4 (MLL4) complex facilitating recruitment to HNF4A-bound regions. Our findings indicate that HNF4A enriches H3K4me1, H3K27ac and establishes chromatin opening at transcriptional regulatory regions.


Asunto(s)
Elementos de Facilitación Genéticos , Leucemia , Animales , Histonas/genética , Histonas/metabolismo , Cromatina/genética , N-Metiltransferasa de Histona-Lisina/genética , N-Metiltransferasa de Histona-Lisina/metabolismo , Mamíferos/genética
16.
J Biol Chem ; 287(46): 38913-21, 2012 Nov 09.
Artículo en Inglés | MEDLINE | ID: mdl-22995913

RESUMEN

The endothelium contributes to the control of the tissue inflammatory response following stress and in particular after exposure to ionizing radiation. We previously showed that the TG-interacting factor 1 (TGIF1) plays a role in radiation-induced normal tissue injury. In this study we hypothesized that this protein could play a role in inflammation. The role of TGIF1 in the stress-induced proinflammatory phenotype was investigated in human endothelial cells. In HUVECs ionizing radiation induces TGIF1 expression as well as a proinflammatory phenotype associated with up-regulation of IL-6, IL-8, CXCL1, MIP-2, and MCP-1. TGIF1 overexpression enhances the radiation-induced proinflammatory phenotype whereas TGIF1 silencing limits both the TNF-α- and radiation-induced overexpression of proinflammatory cytokines. Interestingly, in vivo, in radiation-induced intestinal inflammation in mice, TGIF1 genetic deficiency is associated with a reduced radiation-induced overexpression of proinflammatory molecules. In HUVECs, TNF-α- and radiation-induced NF-κB pathway activation is not influenced by TGIF1 expression, whereas TGIF1 knockdown inhibits both TNF-α- and radiation-induced p38 MAPK pathway activation. This study demonstrates that TGIF1 plays a role in TNF-α- and radiation-induced inflammation and suggests that it could be a target in limiting this event in the vascular compartment.


Asunto(s)
Células Endoteliales/citología , Proteínas de Homeodominio/metabolismo , Proteínas Represoras/metabolismo , Animales , Citocinas/metabolismo , Endotelio Vascular/citología , Humanos , Inmunohistoquímica/métodos , Inflamación , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo , Radiación Ionizante , Proteínas Smad/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
17.
BMC Genomics ; 14: 550, 2013 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-23941359

RESUMEN

BACKGROUND: Chimeric transcripts, including partial and internal tandem duplications (PTDs, ITDs) and gene fusions, are important in the detection, prognosis, and treatment of human cancers. RESULTS: We describe Barnacle, a production-grade analysis tool that detects such chimeras in de novo assemblies of RNA-seq data, and supports prioritizing them for review and validation by reporting the relative coverage of co-occurring chimeric and wild-type transcripts. We demonstrate applications in large-scale disease studies, by identifying PTDs in MLL, ITDs in FLT3, and reciprocal fusions between PML and RARA, in two deeply sequenced acute myeloid leukemia (AML) RNA-seq datasets. CONCLUSIONS: Our analyses of real and simulated data sets show that, with appropriate filter settings, Barnacle makes highly specific predictions for three types of chimeric transcripts that are important in a range of cancers: PTDs, ITDs, and fusions. High specificity makes manual review and validation efficient, which is necessary in large-scale disease studies. Characterizing an extended range of chimera types will help generate insights into progression, treatment, and outcomes for complex diseases.


Asunto(s)
Duplicación de Gen/genética , Perfilación de la Expresión Génica/métodos , Fusión Génica/genética , Genómica , Neoplasias de la Mama/genética , Exones/genética , Humanos , Leucemia Mieloide Aguda/genética , Anotación de Secuencia Molecular , ARN Mensajero/genética , Estadística como Asunto
18.
Genome Res ; 20(8): 1037-51, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20551221

RESUMEN

The liver and pancreas share a common origin and coexpress several transcription factors. To gain insight into the transcriptional networks regulating the function of these tissues, we globally identify binding sites for FOXA2 in adult mouse islets and liver, PDX1 in islets, and HNF4A in liver. Because most eukaryotic transcription factors bind thousands of loci, many of which are thought to be inactive, methods that can discriminate functionally active binding events are essential for the interpretation of genome-wide transcription factor binding data. To develop such a method, we also generated genome-wide H3K4me1 and H3K4me3 localization data in these tissues. By analyzing our binding and histone methylation data in combination with comprehensive gene expression data, we show that H3K4me1 enrichment profiles discriminate transcription factor occupied loci into three classes: those that are functionally active, those that are poised for activation, and those that reflect pioneer-like transcription factor activity. Furthermore, we demonstrate that the regulated presence of H3K4me1-marked nucleosomes at transcription factor occupied promoters and enhancers controls their activity, implicating both tissue-specific transcription factor binding and nucleosome remodeling complex recruitment in determining tissue-specific gene expression. Finally, we apply these approaches to generate novel insights into how FOXA2, PDX1, and HNF4A cooperate to drive islet- and liver-specific gene expression.


Asunto(s)
Sitios Genéticos , Factor Nuclear 3-beta del Hepatocito/genética , Factor Nuclear 4 del Hepatocito/genética , Histonas/genética , Proteínas de Homeodominio/genética , Islotes Pancreáticos/metabolismo , Hígado/metabolismo , Nucleosomas/genética , Transactivadores/genética , Animales , Secuencia de Bases , Sitios de Unión , Perfilación de la Expresión Génica , Factor Nuclear 3-beta del Hepatocito/metabolismo , Factor Nuclear 4 del Hepatocito/metabolismo , Histonas/metabolismo , Proteínas de Homeodominio/metabolismo , Ratones , Datos de Secuencia Molecular , Nucleosomas/metabolismo , Secuencias Reguladoras de Ácidos Nucleicos , Transactivadores/metabolismo
19.
Nat Methods ; 7(11): 909-12, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20935650

RESUMEN

We describe Trans-ABySS, a de novo short-read transcriptome assembly and analysis pipeline that addresses variation in local read densities by assembling read substrings with varying stringencies and then merging the resulting contigs before analysis. Analyzing 7.4 gigabases of 50-base-pair paired-end Illumina reads from an adult mouse liver poly(A) RNA library, we identified known, new and alternative structures in expressed transcripts, and achieved high sensitivity and specificity relative to reference-based assembly methods.


Asunto(s)
Biología Computacional/métodos , Perfilación de la Expresión Génica , Análisis de Secuencia de ADN/métodos , Animales , Ratones
20.
Nat Rev Gastroenterol Hepatol ; 20(9): 561-581, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37208503

RESUMEN

The metabolic, digestive and homeostatic roles of the liver are dependent on proper crosstalk and organization of hepatic cell lineages. These hepatic cell lineages are derived from their respective progenitors early in organogenesis in a spatiotemporally controlled manner, contributing to the liver's specialized and diverse microarchitecture. Advances in genomics, lineage tracing and microscopy have led to seminal discoveries in the past decade that have elucidated liver cell lineage hierarchies. In particular, single-cell genomics has enabled researchers to explore diversity within the liver, especially early in development when the application of bulk genomics was previously constrained due to the organ's small scale, resulting in low cell numbers. These discoveries have substantially advanced our understanding of cell differentiation trajectories, cell fate decisions, cell lineage plasticity and the signalling microenvironment underlying the formation of the liver. In addition, they have provided insights into the pathogenesis of liver disease and cancer, in which developmental processes participate in disease emergence and regeneration. Future work will focus on the translation of this knowledge to optimize in vitro models of liver development and fine-tune regenerative medicine strategies to treat liver disease. In this Review, we discuss the emergence of hepatic parenchymal and non-parenchymal cells, advances that have been made in in vitro modelling of liver development and draw parallels between developmental and pathological processes.


Asunto(s)
Hepatopatías , Hígado , Humanos , Hepatopatías/etiología , Hepatopatías/metabolismo , Hepatocitos/metabolismo , Diferenciación Celular , Linaje de la Célula
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA