Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 65
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Blood ; 139(9): 1359-1373, 2022 03 03.
Artículo en Inglés | MEDLINE | ID: mdl-34852174

RESUMEN

RNA processing is increasingly recognized as a critical control point in the regulation of different hematopoietic lineages including megakaryocytes responsible for the production of platelets. Platelets are anucleate cytoplasts that contain a rich repertoire of RNAs encoding proteins with essential platelet functions derived from the parent megakaryocyte. It is largely unknown how RNA binding proteins contribute to the development and functions of megakaryocytes and platelets. We show that serine-arginine-rich splicing factor 3 (SRSF3) is essential for megakaryocyte maturation and generation of functional platelets. Megakaryocyte-specific deletion of Srsf3 in mice led to macrothrombocytopenia characterized by megakaryocyte maturation arrest, dramatically reduced platelet counts, and abnormally large functionally compromised platelets. SRSF3 deficient megakaryocytes failed to reprogram their transcriptome during maturation and to load platelets with RNAs required for normal platelet function. SRSF3 depletion led to nuclear accumulation of megakaryocyte mRNAs, demonstrating that SRSF3 deploys similar RNA regulatory mechanisms in megakaryocytes as in other cell types. Our study further suggests that SRSF3 plays a role in sorting cytoplasmic megakaryocyte RNAs into platelets and demonstrates how SRSF3-mediated RNA processing forms a central part of megakaryocyte gene regulation. Understanding SRSF3 functions in megakaryocytes and platelets provides key insights into normal thrombopoiesis and platelet pathologies as SRSF3 RNA targets in megakaryocytes are associated with platelet diseases.


Asunto(s)
Plaquetas/metabolismo , Megacariocitos/metabolismo , ARN Mensajero , Factores de Empalme Serina-Arginina , Trombocitopenia , Trombopoyesis/genética , Animales , Ratones , Ratones Noqueados , ARN Mensajero/genética , ARN Mensajero/metabolismo , Factores de Empalme Serina-Arginina/genética , Factores de Empalme Serina-Arginina/metabolismo , Trombocitopenia/genética , Trombocitopenia/metabolismo
2.
Nature ; 557(7705): 439-445, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29743679

RESUMEN

In vertebrate hearts, the ventricular trabecular myocardium develops as a sponge-like network of cardiomyocytes that is critical for contraction and conduction, ventricular septation, papillary muscle formation and wall thickening through the process of compaction 1 . Defective trabeculation leads to embryonic lethality2-4 or non-compaction cardiomyopathy (NCC) 5 . There are divergent views on when and how trabeculation is initiated in different species. In zebrafish, trabecular cardiomyocytes extrude from compact myocardium 6 , whereas in chicks, chamber wall thickening occurs before overt trabeculation 7 . In mice, the onset of trabeculation has not been described, but is proposed to begin at embryonic day 9.0, when cardiomyocytes form radially oriented ribs 2 . Endocardium-myocardium communication is essential for trabeculation, and numerous signalling pathways have been identified, including Notch2,8 and Neuregulin (NRG) 4 . Late disruption of the Notch pathway causes NCC 5 . Whereas it has been shown that mutations in the extracellular matrix (ECM) genes Has2 and Vcan prevent the formation of trabeculae in mice9,10 and the matrix metalloprotease ADAMTS1 promotes trabecular termination 3 , the pathways involved in ECM dynamics and the molecular regulation of trabeculation during its early phases remain unexplored. Here we present a model of trabeculation in mice that integrates dynamic endocardial and myocardial cell behaviours and ECM remodelling, and reveal new epistatic relationships between the involved signalling pathways. NOTCH1 signalling promotes ECM degradation during the formation of endocardial projections that are critical for individualization of trabecular units, whereas NRG1 promotes myocardial ECM synthesis, which is necessary for trabecular rearrangement and growth. These systems interconnect through NRG1 control of Vegfa, but act antagonistically to establish trabecular architecture. These insights enabled the prediction of persistent ECM and cardiomyocyte growth in a mouse NCC model, providing new insights into the pathophysiology of congenital heart disease.


Asunto(s)
Corazón/embriología , Miocardio/citología , Miocardio/metabolismo , Neurregulina-1/metabolismo , Organogénesis , Receptor Notch1/metabolismo , Animales , Modelos Animales de Enfermedad , Endocardio/citología , Endocardio/metabolismo , Matriz Extracelular/metabolismo , Cardiopatías/congénito , Cardiopatías/metabolismo , Ratones , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Neurregulina-1/genética , Receptor Notch1/genética , Transducción de Señal , Factor A de Crecimiento Endotelial Vascular/metabolismo
3.
J Am Soc Nephrol ; 34(1): 88-109, 2023 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-36167728

RESUMEN

BACKGROUND: NPHS2 variants are the most common cause of steroid-resistant nephrotic syndrome in children >1 month old. Missense NPHS2 variants were reported to cause mistrafficking of the encoded protein, PODOCIN, but this conclusion was on the basis of overexpression in some nonpodocyte cell lines. METHODS: We generated a series of human induced pluripotent stem cell (iPSC) lines bearing pathogenic missense variants of NPHS2 , encoding the protein changes p.G92C, p.P118L, p.R138Q, p.R168H, and p.R291W, and control lines. iPSC lines were also generated from a patient with steroid-resistant nephrotic syndrome (p.R168H homozygote) and a healthy heterozygous parent. All lines were differentiated into kidney organoids. Immunofluorescence assessed PODOCIN expression and subcellular localization. Podocytes were transcriptionally profiled and PODOCIN-NEPHRIN interaction interrogated. RESULTS: All variant lines revealed reduced levels of PODOCIN protein in the absence of reduced transcription. Although wild-type PODOCIN localized to the membrane, distinct variant proteins displayed unique patterns of subcellular protein trafficking, some unreported. P118L and R138Q were preferentially retained in the endoplasmic reticulum (ER); R168H and R291W accumulated in the Golgi. Podocyte profiling demonstrated minimal disease-associated transcriptional change. All variants displayed podocyte-specific apoptosis, which was not linked to ER stress. NEPHRIN-PODOCIN colocalization elucidated the variant-specific effect on NEPHRIN association and hence NEPHRIN trafficking. CONCLUSIONS: Specific variants of endogenous NPHS2 result in distinct subcellular PODOCIN localization within organoid podocytes. Understanding the effect of each variant on protein levels and localization and the effect on NEPHRIN provides additional insight into the pathobiology of NPHS2 variants. PODCAST: This article contains a podcast at https://dts.podtrac.com/redirect.mp3/www.asn-online.org/media/podcast/JASN/2023_01_05_JASN2022060707.mp3.


Asunto(s)
Células Madre Pluripotentes Inducidas , Síndrome Nefrótico , Niño , Humanos , Lactante , Síndrome Nefrótico/genética , Síndrome Nefrótico/metabolismo , Riñón/metabolismo , Mutación
4.
Development ; 147(22)2020 11 30.
Artículo en Inglés | MEDLINE | ID: mdl-33144401

RESUMEN

The inability of the adult mammalian heart to regenerate represents a fundamental barrier in heart failure management. By contrast, the neonatal heart retains a transient regenerative capacity, but the underlying mechanisms for the developmental loss of cardiac regenerative capacity in mammals are not fully understood. Wnt/ß-catenin signalling has been proposed as a key cardioregenerative pathway driving cardiomyocyte proliferation. Here, we show that Wnt/ß-catenin signalling potentiates neonatal mouse cardiomyocyte proliferation in vivo and immature human pluripotent stem cell-derived cardiomyocyte (hPSC-CM) proliferation in vitro By contrast, Wnt/ß-catenin signalling in adult mice is cardioprotective but fails to induce cardiomyocyte proliferation. Transcriptional profiling and chromatin immunoprecipitation sequencing of neonatal mouse and hPSC-CMs revealed a core Wnt/ß-catenin-dependent transcriptional network governing cardiomyocyte proliferation. By contrast, ß-catenin failed to re-engage this neonatal proliferative gene network in the adult heart despite partial transcriptional re-activation of a neonatal glycolytic gene programme. These findings suggest that ß-catenin might be repurposed from regenerative to protective functions in the adult heart in a developmental process dependent on the metabolic status of cardiomyocytes.


Asunto(s)
Proliferación Celular , Redes Reguladoras de Genes , Miocitos Cardíacos/metabolismo , Transcripción Genética , Vía de Señalización Wnt , beta Catenina/metabolismo , Animales , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Ratones , Miocitos Cardíacos/citología , beta Catenina/genética
5.
J Mol Cell Cardiol ; 163: 20-32, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34624332

RESUMEN

Understanding the spatial gene expression and regulation in the heart is key to uncovering its developmental and physiological processes, during homeostasis and disease. Numerous techniques exist to gain gene expression and regulation information in organs such as the heart, but few utilize intuitive true-to-life three-dimensional representations to analyze and visualise results. Here we combined transcriptomics with 3D-modelling to interrogate spatial gene expression in the mammalian heart. For this, we microdissected and sequenced transcriptome-wide 18 anatomical sections of the adult mouse heart. Our study has unveiled known and novel genes that display complex spatial expression in the heart sub-compartments. We have also created 3D-cardiomics, an interface for spatial transcriptome analysis and visualization that allows the easy exploration of these data in a 3D model of the heart. 3D-cardiomics is accessible from http://3d-cardiomics.erc.monash.edu/.


Asunto(s)
Corazón , Transcriptoma , Animales , Perfilación de la Expresión Génica/métodos , Mamíferos , Ratones
6.
BMC Bioinformatics ; 23(1): 69, 2022 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-35164667

RESUMEN

BACKGROUND: Gene ontology (GO) enrichment analysis is frequently undertaken during exploration of various -omics data sets. Despite the wide array of tools available to biologists to perform this analysis, meaningful visualisation of the overrepresented GO in a manner which is easy to interpret is still lacking. RESULTS: Monash Gene Ontology (MonaGO) is a novel web-based visualisation system that provides an intuitive, interactive and responsive interface for performing GO enrichment analysis and visualising the results. MonaGO supports gene lists as well as GO terms as inputs. Visualisation results can be exported as high-resolution images or restored in new sessions, allowing reproducibility of the analysis. An extensive comparison between MonaGO and 11 state-of-the-art GO enrichment visualisation tools based on 9 features revealed that MonaGO is a unique platform that simultaneously allows interactive visualisation within one single output page, directly accessible through a web browser with customisable display options. CONCLUSION: MonaGO combines dynamic clustering and interactive visualisation as well as customisation options to assist biologists in obtaining meaningful representation of overrepresented GO terms, producing simplified outputs in an unbiased manner. MonaGO will facilitate the interpretation of GO analysis and will assist the biologists into the representation of the results.


Asunto(s)
Programas Informáticos , Análisis por Conglomerados , Ontología de Genes , Probabilidad , Reproducibilidad de los Resultados
7.
BMC Genomics ; 23(1): 78, 2022 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-35078412

RESUMEN

BACKGROUND: Transcriptional regulation is primarily mediated by the binding of factors to non-coding regions in DNA. Identification of these binding regions enhances understanding of tissue formation and potentially facilitates the development of gene therapies. However, successful identification of binding regions is made difficult by the lack of a universal biological code for their characterisation. RESULTS: We extend an alignment-based method, changept, and identify clusters of biological significance, through ontology and de novo motif analysis. Further, we apply a Bayesian method to estimate and combine binary classifiers on the clusters we identify to produce a better performing composite. CONCLUSIONS: The analysis we describe provides a computational method for identification of conserved binding sites in the human genome and facilitates an alternative interrogation of combinations of existing data sets with alignment data.


Asunto(s)
Algoritmos , Secuencias Reguladoras de Ácidos Nucleicos , Teorema de Bayes , Sitios de Unión , Genoma Humano , Humanos , Secuencias Reguladoras de Ácidos Nucleicos/genética
8.
Nature ; 512(7514): 314-8, 2014 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-25119043

RESUMEN

Haematopoietic stem cells (HSCs) are self-renewing stem cells capable of replenishing all blood lineages. In all vertebrate embryos that have been studied, definitive HSCs are generated initially within the dorsal aorta (DA) of the embryonic vasculature by a series of poorly understood inductive events. Previous studies have identified that signalling relayed from adjacent somites coordinates HSC induction, but the nature of this signal has remained elusive. Here we reveal that somite specification of HSCs occurs via the deployment of a specific endothelial precursor population, which arises within a sub-compartment of the zebrafish somite that we have defined as the endotome. Endothelial cells of the endotome are specified within the nascent somite by the activity of the homeobox gene meox1. Specified endotomal cells consequently migrate and colonize the DA, where they induce HSC formation through the deployment of chemokine signalling activated in these cells during endotome formation. Loss of meox1 activity expands the endotome at the expense of a second somitic cell type, the muscle precursors of the dermomyotomal equivalent in zebrafish, the external cell layer. The resulting increase in endotome-derived cells that migrate to colonize the DA generates a dramatic increase in chemokine-dependent HSC induction. This study reveals the molecular basis for a novel somite lineage restriction mechanism and defines a new paradigm in induction of definitive HSCs.


Asunto(s)
Células Endoteliales/citología , Células Madre Hematopoyéticas/citología , Proteínas de Homeodominio/metabolismo , Somitos/citología , Factores de Transcripción/metabolismo , Proteínas de Pez Cebra/metabolismo , Animales , Aorta/citología , Aorta/embriología , Biomarcadores/análisis , Movimiento Celular , Quimiocina CXCL12/análisis , Quimiocina CXCL12/metabolismo , Embrión de Pollo , Células Endoteliales/metabolismo , Células Madre Hematopoyéticas/metabolismo , Proteínas de Homeodominio/análisis , Proteínas de Homeodominio/genética , Humanos , Ratones , Músculos/citología , Músculos/metabolismo , Mutación/genética , Somitos/metabolismo , Factores de Transcripción/análisis , Factores de Transcripción/genética , Proteínas Wnt/análisis , Proteínas Wnt/metabolismo , Pez Cebra/embriología , Pez Cebra/metabolismo , Proteínas de Pez Cebra/análisis , Proteínas de Pez Cebra/genética
9.
Differentiation ; 107: 11-23, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31102825

RESUMEN

Congenital Heart Disease (CHD) is characterised by a wide range of cardiac defects, from mild to life-threatening, which occur in babies worldwide. To date, there is no cure to CHD, however, progress in surgery has reduced its mortality allowing children affected by CHD to reach adulthood. In an effort to understand its genetic basis, several studies involving whole-genome sequencing (WGS) of patients with CHD have been undertaken and generated a great wealth of information. The majority of putative causative mutations identified in WGS studies fall into the non-coding part of the genome. Unfortunately, due to the lack of understanding of the function of these non-coding mutations, it is challenging to establish a causal link between the non-coding mutation and the disease. Thus, here we review the state-of-the-art approaches to interpret non-coding mutations in the context of CHD and address the following questions: What are the non-coding sequences important for cardiac function? Which technologies are used to identify them? Which resources are available to analyse them? What mutations are expected in these non-coding sequences? Learning from developmental process, what is their expected role in CHD?


Asunto(s)
Cardiopatías Congénitas/genética , Corazón , Corazón/embriología , Corazón/crecimiento & desarrollo , Humanos , Mutación Silenciosa , Regiones no Traducidas , Secuenciación Completa del Genoma
10.
Genesis ; 56(9): e23246, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30114334

RESUMEN

Development of the embryonic head is driven by the activity of gene regulatory networks of transcription factors. LHX1 is a homeobox transcription factor that plays an essential role in the formation of the embryonic head. The loss of LHX1 function results in anterior truncation of the embryo caused by the disruption of morphogenetic movement of tissue precursors and the dysregulation of WNT signaling activity. Profiling the gene expression pattern in the Lhx1 mutant embryo revealed that tissues in anterior germ layers acquire posterior tissue characteristics, suggesting LHX1 activity is required for the allocation and patterning of head precursor tissues. Here, we used LHX1 as an entry point to delineate its transcriptional targets and interactors and construct a LHX1-anchored gene regulatory network. Using a gain-of-function approach, we identified genes that immediately respond to Lhx1 activation. Meta-analysis of the datasets of LHX1-responsive genes and genes expressed in the anterior tissues of mouse embryos at head-fold stage, in conjunction with published Xenopus embryonic LHX1 (Xlim1) ChIP-seq data, has pinpointed the putative transcriptional targets of LHX1 and an array of genetic determinants functioning together in the formation of the mouse embryonic head.


Asunto(s)
Redes Reguladoras de Genes , Genes Homeobox , Cabeza/embriología , Proteínas con Homeodominio LIM/metabolismo , Factores de Transcripción/metabolismo , Animales , Células Madre Embrionarias/metabolismo , Perfilación de la Expresión Génica , Células Germinativas/fisiología , Transcripción Genética , Xenopus laevis/embriología
11.
Circulation ; 136(12): 1123-1139, 2017 Sep 19.
Artículo en Inglés | MEDLINE | ID: mdl-28733351

RESUMEN

BACKGROUND: The inability of the adult mammalian heart to regenerate following injury represents a major barrier in cardiovascular medicine. In contrast, the neonatal mammalian heart retains a transient capacity for regeneration, which is lost shortly after birth. Defining the molecular mechanisms that govern regenerative capacity in the neonatal period remains a central goal in cardiac biology. Here, we assemble a transcriptomic framework of multiple cardiac cell populations during postnatal development and following injury, which enables comparative analyses of the regenerative (neonatal) versus nonregenerative (adult) state for the first time. METHODS: Cardiomyocytes, fibroblasts, leukocytes, and endothelial cells from infarcted and noninfarcted neonatal (P1) and adult (P56) mouse hearts were isolated by enzymatic dissociation and fluorescence-activated cell sorting at day 3 following surgery. RNA sequencing was performed on these cell populations to generate the transcriptome of the major cardiac cell populations during cardiac development, repair, and regeneration. To complement our transcriptomic data, we also surveyed the epigenetic landscape of cardiomyocytes during postnatal maturation by performing deep sequencing of accessible chromatin regions by using the Assay for Transposase-Accessible Chromatin from purified mouse cardiomyocyte nuclei (P1, P14, and P56). RESULTS: Profiling of cardiomyocyte and nonmyocyte transcriptional programs uncovered several injury-responsive genes across regenerative and nonregenerative time points. However, the majority of transcriptional changes in all cardiac cell types resulted from developmental maturation from neonatal stages to adulthood rather than activation of a distinct regeneration-specific gene program. Furthermore, adult leukocytes and fibroblasts were characterized by the expression of a proliferative gene expression network following infarction, which mirrored the neonatal state. In contrast, cardiomyocytes failed to reactivate the neonatal proliferative network following infarction, which was associated with loss of chromatin accessibility around cell cycle genes during postnatal maturation. CONCLUSIONS: This work provides a comprehensive framework and transcriptional resource of multiple cardiac cell populations during cardiac development, repair, and regeneration. Our findings define a regulatory program underpinning the neonatal regenerative state and identify alterations in the chromatin landscape that could limit reinduction of the regenerative program in adult cardiomyocytes.


Asunto(s)
Perfilación de la Expresión Génica , Corazón/fisiología , Transcriptoma , Animales , Animales Recién Nacidos , Fibroblastos/citología , Fibroblastos/metabolismo , Regulación del Desarrollo de la Expresión Génica/genética , Redes Reguladoras de Genes , Leucocitos/citología , Leucocitos/metabolismo , Ratones , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , ARN/aislamiento & purificación , ARN/metabolismo , Regeneración/fisiología , Análisis de Secuencia de ARN , Transducción de Señal/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
12.
BMC Genomics ; 19(1): 238, 2018 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-29621972

RESUMEN

BACKGROUND: A strong focus of the post-genomic era is mining of the non-coding regulatory genome in order to unravel the function of regulatory elements that coordinate gene expression (Nat 489:57-74, 2012; Nat 507:462-70, 2014; Nat 507:455-61, 2014; Nat 518:317-30, 2015). Whole-genome approaches based on next-generation sequencing (NGS) have provided insight into the genomic location of regulatory elements throughout different cell types, organs and organisms. These technologies are now widespread and commonly used in laboratories from various fields of research. This highlights the need for fast and user-friendly software tools dedicated to extracting cis-regulatory information contained in these regulatory regions; for instance transcription factor binding site (TFBS) composition. Ideally, such tools should not require prior programming knowledge to ensure they are accessible for all users. RESULTS: We present TrawlerWeb, a web-based version of the Trawler_standalone tool (Nat Methods 4:563-5, 2007; Nat Protoc 5:323-34, 2010), to allow for the identification of enriched motifs in DNA sequences obtained from next-generation sequencing experiments in order to predict their TFBS composition. TrawlerWeb is designed for online queries with standard options common to web-based motif discovery tools. In addition, TrawlerWeb provides three unique new features: 1) TrawlerWeb allows the input of BED files directly generated from NGS experiments, 2) it automatically generates an input-matched biologically relevant background, and 3) it displays resulting conservation scores for each instance of the motif found in the input sequences, which assists the researcher in prioritising the motifs to validate experimentally. Finally, to date, this web-based version of Trawler_standalone remains the fastest online de novo motif discovery tool compared to other popular web-based software, while generating predictions with high accuracy. CONCLUSIONS: TrawlerWeb provides users with a fast, simple and easy-to-use web interface for de novo motif discovery. This will assist in rapidly analysing NGS datasets that are now being routinely generated. TrawlerWeb is freely available and accessible at: http://trawler.erc.monash.edu.au .


Asunto(s)
Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Análisis de Secuencia de ADN/métodos , Programas Informáticos , Animales , Secuencia de Bases , Sitios de Unión , Secuencia Conservada , ADN/química , ADN/metabolismo , Humanos , Internet , Mesotelina , Ratones , Motivos de Nucleótidos , Ratas , Factores de Transcripción/metabolismo
13.
Development ; 142(5): 832-9, 2015 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-25655700

RESUMEN

Divisions that generate one neuronal lineage-committed and one self-renewing cell maintain the balance of proliferation and differentiation for the generation of neuronal diversity. The asymmetric inheritance of apical domains and components of the cell division machinery has been implicated in this process, and might involve interactions with cell fate determinants in regulatory feedback loops of an as yet unknown nature. Here, we report the dynamics of Anillin - an essential F-actin regulator and furrow component - and its contribution to progenitor cell divisions in the developing zebrafish retina. We find that asymmetrically dividing retinal ganglion cell progenitors position the Anillin-rich midbody at the apical domain of the differentiating daughter. anillin hypomorphic conditions disrupt asymmetric apical domain inheritance and affect daughter cell fate. Consequently, the retinal cell type composition is profoundly affected, such that the ganglion cell layer is dramatically expanded. This study provides the first in vivo evidence for the requirement of Anillin during asymmetric neurogenic divisions. It also provides insights into a reciprocal regulation between Anillin and the ganglion cell fate determinant Ath5, suggesting a mechanism whereby the balance of proliferation and differentiation is accomplished during progenitor cell divisions in vivo.


Asunto(s)
Proteínas Contráctiles/metabolismo , Células Ganglionares de la Retina/citología , Células Ganglionares de la Retina/metabolismo , Células Madre/citología , Células Madre/metabolismo , Animales , Proteínas Contráctiles/genética , Técnica del Anticuerpo Fluorescente , Hibridación in Situ , Microscopía Confocal , Pez Cebra , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
14.
Genomics ; 109(2): 75-82, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28189763

RESUMEN

DNA adenine methyltransferase identification (DamID) is an enzymatic technology for detecting DNA regions targeted by chromatin-associated proteins. Proteins are fused to bacterial DNA adenine methyltransferase (Dam) and expressed in cultured cells or whole organisms. Here, we used DamID to detect DNA regions bound by the cardiac-restricted transcription factors (TFs) NKX2-5 and SRF, and ubiquitously-expressed co-factors ELK1 and ELK4. We compared targets bound by these TFs as N- and C-terminal fusions with Dam, for both wild type (WT) NKX2-5 and mutant proteins mimicking those found in congenital heart disease. Overall, DamID is highly robust: while the orientation of WT Dam fusions can affect the size of the target sets, their signatures remained largely reproducible. Furthermore, a severe NKX2-5 mutant lacking the homeodomain showed strong steric effects negatively impacting target discovery. The extent of steric effect is likely to be dependent on the protein in question and the orientation of Dam fusion.


Asunto(s)
Cromatina/metabolismo , Regulación de la Expresión Génica , Técnicas Genéticas , Cardiopatías Congénitas/metabolismo , Metiltransferasa de ADN de Sitio Específico (Adenina Especifica) , Animales , ADN/metabolismo , Cardiopatías Congénitas/genética , Proteína Homeótica Nkx-2.5/metabolismo , Humanos , Ratones , Factor de Respuesta Sérica/metabolismo , Proteína Elk-1 con Dominio ets/metabolismo , Proteína Elk-4 del Dominio ets/metabolismo
15.
Differentiation ; 91(1-3): 29-41, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26897459

RESUMEN

Nkx2-5 is one of the master regulators of cardiac development, homeostasis and disease. This transcription factor has been previously associated with a suite of cardiac congenital malformations and impairment of electrical activity. When disease causative mutations in transcription factors are considered, NKX2-5 gene dysfunction is the most common abnormality found in patients. Here we describe a novel mouse model and subsequent implications of Nkx2-5 loss for aspects of myocardial electrical activity. In this work we have engineered a new Nkx2-5 conditional knockout mouse in which flox sites flank the entire Nkx2-5 locus, and validated this line for the study of heart development, differentiation and disease using a full deletion strategy. While our homozygous knockout mice show typical embryonic malformations previously described for the lack of the Nkx2-5 gene, hearts of heterozygous adult mice show moderate morphological and functional abnormalities that are sufficient to sustain blood supply demands under homeostatic conditions. This study further reveals intriguing aspects of Nkx2-5 function in the control of cardiac electrical activity. Using a combination of mouse genetics, biochemistry, molecular and cell biology, we demonstrate that Nkx2-5 regulates the gene encoding Kcnh2 channel and others, shedding light on potential mechanisms generating electrical abnormalities observed in patients bearing NKX2-5 dysfunction and opening opportunities to the study of novel therapeutic targets for anti-arrhythmogenic therapies.


Asunto(s)
Canal de Potasio ERG1/genética , Cardiopatías Congénitas/genética , Corazón/crecimiento & desarrollo , Proteína Homeótica Nkx-2.5/genética , Animales , Modelos Animales de Enfermedad , Canal de Potasio ERG1/metabolismo , Regulación del Desarrollo de la Expresión Génica , Corazón/embriología , Corazón/fisiopatología , Cardiopatías Congénitas/fisiopatología , Humanos , Canales Iónicos/genética , Canales Iónicos/metabolismo , Ratones , Ratones Noqueados , Mutación
16.
EMBO Rep ; 15(12): 1254-67, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25391905

RESUMEN

Low-density lipoprotein receptor-related proteins 5 and 6 (LRP5/6) function as transmembrane receptors to transduce Wnt signals. A key mechanism for signalling is Wnt-induced serine/threonine phosphorylation at conserved PPPSPxS motifs in the LRP6 cytoplasmic domain, which promotes pathway activation. Conserved tyrosine residues are positioned close to all PPPSPxS motifs, which suggests they have a functional significance. Using a cell culture-based cDNA expression screen, we identified the non-receptor tyrosine kinases Src and Fer as novel LRP6 modifiers. Both Src and Fer associate with LRP6 and phosphorylate LRP6 directly. In contrast to the known PPPSPxS Ser/Thr kinases, tyrosine phosphorylation by Src and Fer negatively regulates LRP6-Wnt signalling. Epistatically, they function upstream of ß-catenin to inhibit signalling and in agreement with a negative role in regulating LRP6, MEF cells lacking these kinases show enhanced Wnt signalling. Wnt3a treatment of cells enhances tyrosine phosphorylation of endogenous LRP6 and, mechanistically, Src reduces cell surface LRP6 levels and disrupts LRP6 signalosome formation. Interestingly, CK1γ inhibits Fer-induced LRP6 phosphorylation, suggesting a mechanism whereby CK1γ acts to de-represses inhibitory LRP6 tyrosine phosphorylation. We propose that LRP6 tyrosine phosphorylation by Src and Fer serves a negative regulatory function to prevent over-activation of Wnt signalling at the level of the Wnt receptor, LRP6.


Asunto(s)
Proteína-6 Relacionada a Receptor de Lipoproteína de Baja Densidad/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Tirosina/metabolismo , Proteínas Wnt/metabolismo , beta Catenina/metabolismo , Familia-src Quinasas/metabolismo , Línea Celular , Humanos , Hibridación in Situ , Proteína-6 Relacionada a Receptor de Lipoproteína de Baja Densidad/genética , Espectrometría de Masas , Fosforilación , Proteínas Tirosina Quinasas/genética , Transducción de Señal , Proteínas Wnt/genética , beta Catenina/genética , Familia-src Quinasas/genética
17.
Development ; 139(5): 917-28, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22318626

RESUMEN

During embryogenesis, tissue specification is triggered by the expression of a unique combination of developmental genes and their expression in time and space is crucial for successful development. Synexpression groups are batteries of spatiotemporally co-expressed genes that act in shared biological processes through their coordinated expression. Although several synexpression groups have been described in numerous vertebrate species, the regulatory mechanisms that orchestrate their common complex expression pattern remain to be elucidated. Here we performed a pilot screen on 560 genes of the vertebrate model system medaka (Oryzias latipes) to systematically identify synexpression groups and investigate their regulatory properties by searching for common regulatory cues. We find that synexpression groups share DNA motifs that are arranged in various combinations into cis-regulatory modules that drive co-expression. In contrast to previous assumptions that these genes are located randomly in the genome, we discovered that genes belonging to the same synexpression group frequently occur in synexpression clusters in the genome. This work presents a first repertoire of synexpression group common signatures, a resource that will contribute to deciphering developmental gene regulatory networks.


Asunto(s)
Desarrollo Embrionario/genética , Regulación del Desarrollo de la Expresión Génica , Redes Reguladoras de Genes , Oryzias/embriología , Oryzias/genética , Animales , Secuencia de Bases , Biología Computacional/métodos , Bases de Datos Factuales , Embrión no Mamífero/anatomía & histología , Embrión no Mamífero/fisiología , Genes Reporteros , Genoma , Datos de Secuencia Molecular , Familia de Multigenes , Motivos de Nucleótidos , Oryzias/anatomía & histología , Sintenía
18.
Comput Biol Med ; 171: 108068, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38354497

RESUMEN

The availability of large-scale epigenomic data from various cell types and conditions has yielded valuable insights for evaluating and learning features predicting the co-binding of transcription factors (TF). However, prior attempts to develop models predicting motif co-occurrence lacked scalability for globally analyzing any motif combination or making cross-species predictions. Moreover, mapping co-regulatory modules (CRM) to gene regulatory networks (GRN) is crucial for understanding underlying function. Currently, no comprehensive pipeline exists for large-scale, rapid, and accurate CRM and GRN identification. In this study, we analyzed and evaluated different TF binding characteristics facilitating biologically significant co-binding to identify all potential clusters of co-binding TFs. We curated the UniBind database, containing ChIP-Seq data from over 1983 samples and 232 TFs, and implemented two machine learning models to predict CRMs and the potential regulatory networks they operate on. Two machine learning models, Convolution Neural Networks (CNN) and Random Forest Classifier(RFC), used to predict co-binding between TFs, were compared using precision-recall Receiver Operating Characteristic (ROC) curves. CNN outperformed RFC (AUC 0.94 vs. 0.88) and achieved higher F1 scores (0.938 vs. 0.872). The CRMs generated by the clustering algorithm were validated against ChipAtlas and MCOT, revealing additional motifs forming CRMs. We predicted 200k CRMs for 50k+ human genes, validated against recent CRM prediction methods with 100% overlap. Further, we narrowed our focus to study heart-related regulatory motifs, filtering the generated CRMs to report 1784 Cardiac CRMs containing at least four cardiac TFs. Identified cardiac CRMs revealed potential novel regulators like ARID3A and RXRB for SCAD, including known TFs like PPARG for F11R. Our findings highlight the importance of the NKX family of transcription factors in cardiac development and provide potential targets for further investigation in cardiac disease.


Asunto(s)
Epigenómica , Redes Reguladoras de Genes , Humanos , Redes Reguladoras de Genes/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Algoritmos , Corazón , Proteínas de Unión al ADN/genética
19.
Aging Cell ; 23(1): e13862, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37183563

RESUMEN

Sarcopenia, the age-related decline in muscle function, places a considerable burden on health-care systems. While the stereotypic hallmarks of sarcopenia are well characterized, their contribution to muscle wasting remains elusive, which is partly due to the limited availability of animal models. Here, we have performed cellular and molecular characterization of skeletal muscle from the African killifish-an extremely short-lived vertebrate-revealing that while many characteristics deteriorate with increasing age, supporting the use of killifish as a model for sarcopenia research, some features surprisingly reverse to an "early-life" state in the extremely old stages. This suggests that in extremely old animals, there may be mechanisms that prevent further deterioration of skeletal muscle, contributing to an extension of life span. In line with this, we report a reduction in mortality rates in extremely old killifish. To identify mechanisms for this phenomenon, we used a systems metabolomics approach, which revealed that during aging there is a striking depletion of triglycerides, mimicking a state of calorie restriction. This results in the activation of mitohormesis, increasing Sirt1 levels, which improves lipid metabolism and maintains nutrient homeostasis in extremely old animals. Pharmacological induction of Sirt1 in aged animals was sufficient to induce a late life-like metabolic profile, supporting its role in life span extension in vertebrate populations that are naturally long-lived. Collectively, our results demonstrate that killifish are not only a novel model to study the biological processes that govern sarcopenia, but they also provide a unique vertebrate system to dissect the regulation of longevity.


Asunto(s)
Longevidad , Sarcopenia , Animales , Sarcopenia/metabolismo , Sirtuina 1/metabolismo , Envejecimiento , Músculo Esquelético/metabolismo , Fundulus heteroclitus , Vertebrados , Biología
20.
Commun Biol ; 7(1): 371, 2024 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-38575811

RESUMEN

Cardiac function requires appropriate proteins in each chamber. Atria requires slow myosin to act as reservoirs, while ventricles demand fast myosin for swift pumping. Myosins are thus under chamber-biased cis-regulation, with myosin gene expression imbalances leading to congenital heart dysfunction. To identify regulatory inputs leading to cardiac chamber-biased expression, we computationally and molecularly dissected the quail Slow Myosin Heavy Chain III (SMyHC III) promoter that drives preferential expression to the atria. We show that SMyHC III gene states are orchestrated by a complex Nuclear Receptor Element (cNRE) of 32 base pairs. Using transgenesis in zebrafish and mice, we demonstrate that preferential atrial expression is achieved by a combinatorial regulatory input composed of atrial activation motifs and ventricular repression motifs. Using comparative genomics, we show that the cNRE might have emerged from an endogenous viral element through infection of an ancestral host germline, revealing an evolutionary pathway to cardiac chamber-specific expression.


Asunto(s)
Atrios Cardíacos , Pez Cebra , Ratones , Animales , Pez Cebra/genética , Atrios Cardíacos/metabolismo , Ventrículos Cardíacos , Miosinas/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA