Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 167(2): 355-368.e10, 2016 Oct 06.
Artículo en Inglés | MEDLINE | ID: mdl-27693352

RESUMEN

Common sequence variants in cis-regulatory elements (CREs) are suspected etiological causes of complex disorders. We previously identified an intronic enhancer variant in the RET gene disrupting SOX10 binding and increasing Hirschsprung disease (HSCR) risk 4-fold. We now show that two other functionally independent CRE variants, one binding Gata2 and the other binding Rarb, also reduce Ret expression and increase risk 2- and 1.7-fold. By studying human and mouse fetal gut tissues and cell lines, we demonstrate that reduced RET expression propagates throughout its gene regulatory network, exerting effects on both its positive and negative feedback components. We also provide evidence that the presence of a combination of CRE variants synergistically reduces RET expression and its effects throughout the GRN. These studies show how the effects of functionally independent non-coding variants in a coordinated gene regulatory network amplify their individually small effects, providing a model for complex disorders.


Asunto(s)
Elementos de Facilitación Genéticos , Regulación de la Expresión Génica , Redes Reguladoras de Genes , Enfermedad de Hirschsprung/genética , Proteínas Proto-Oncogénicas c-ret/genética , Alelos , Animales , Sitios de Unión , Modelos Animales de Enfermedad , Factor de Transcripción GATA2/genética , Factor de Transcripción GATA2/metabolismo , Tracto Gastrointestinal/embriología , Humanos , Ratones , Ratones Transgénicos , ARN no Traducido/genética , Receptores de Ácido Retinoico/genética , Receptores de Ácido Retinoico/metabolismo , Factores de Transcripción SOXE/genética , Factores de Transcripción SOXE/metabolismo
2.
PLoS Genet ; 19(11): e1011030, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37948459

RESUMEN

Hirschsprung disease (HSCR) is associated with deficiency of the receptor tyrosine kinase RET, resulting in loss of cells of the enteric nervous system (ENS) during fetal gut development. The major contribution to HSCR risk is from common sequence variants in RET enhancers with additional risk from rare coding variants in many genes. Here, we demonstrate that these RET enhancer variants specifically alter the human fetal gut development program through significant decreases in gene expression of RET, members of the RET-EDNRB gene regulatory network (GRN), other HSCR genes, with an altered transcriptome of 2,382 differentially expressed genes across diverse neuronal and mesenchymal functions. A parsimonious hypothesis for these results is that beyond RET's direct effect on its GRN, it also has a major role in enteric neural crest-derived cell (ENCDC) precursor proliferation, its deficiency reducing ENCDCs with relative expansion of non-ENCDC cells. Thus, genes reducing RET proliferative activity can potentially cause HSCR. One such class is the 23 RET-dependent transcription factors enriched in early gut development. We show that their knockdown in human neuroblastoma SK-N-SH cells reduces RET and/or EDNRB gene expression, expanding the RET-EDNRB GRN. The human embryos we studied had major remodeling of the gut transcriptome but were unlikely to have had HSCR: thus, genetic or epigenetic changes in addition to those in RET are required for aganglionosis.


Asunto(s)
Elementos de Facilitación Genéticos , Tracto Gastrointestinal , Proteínas Proto-Oncogénicas c-ret , Haplotipos , Humanos , Proteínas Proto-Oncogénicas c-ret/genética , Neuroblastoma , Línea Celular Tumoral , Enfermedad de Hirschsprung/genética , Feto , Tracto Gastrointestinal/embriología , Cresta Neural/citología , Sistema Nervioso Entérico/embriología , Análisis de Expresión Génica de una Sola Célula , Regulación del Desarrollo de la Expresión Génica
3.
Proc Natl Acad Sci U S A ; 120(34): e2211986120, 2023 08 22.
Artículo en Inglés | MEDLINE | ID: mdl-37585461

RESUMEN

The receptor tyrosine kinase RET plays a critical role in the fate specification of enteric neural crest-derived cells (ENCDCs) during enteric nervous system (ENS) development. RET loss of function (LoF) is associated with Hirschsprung disease (HSCR), which is marked by aganglionosis of the gastrointestinal (GI) tract. Although the major phenotypic consequences and the underlying transcriptional changes from Ret LoF in the developing ENS have been described, cell type- and state-specific effects are unknown. We performed single-cell RNA sequencing on an enriched population of ENCDCs from the developing GI tract of Ret null heterozygous and homozygous mice at embryonic day (E)12.5 and E14.5. We demonstrate four significant findings: 1) Ret-expressing ENCDCs are a heterogeneous population comprising ENS progenitors as well as glial- and neuronal-committed cells; 2) neurons committed to a predominantly inhibitory motor neuron developmental trajectory are not produced under Ret LoF, leaving behind a mostly excitatory motor neuron developmental program; 3) expression patterns of HSCR-associated and Ret gene regulatory network genes are impacted by Ret LoF; and 4) Ret deficiency leads to precocious differentiation and reduction in the number of proliferating ENS precursors. Our results support a model in which Ret contributes to multiple distinct cellular phenotypes during development of the ENS, including the specification of inhibitory neuron subtypes, cell cycle dynamics of ENS progenitors, and the developmental timing of neuronal and glial commitment.


Asunto(s)
Sistema Nervioso Entérico , Enfermedad de Hirschsprung , Proteínas Proto-Oncogénicas c-ret , Animales , Ratones , Diferenciación Celular , Proliferación Celular , Enfermedad de Hirschsprung/genética , Cresta Neural , Proteínas Proto-Oncogénicas c-ret/genética
4.
Genome Res ; 31(12): 2199-2208, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34782358

RESUMEN

The major genetic risk factors for Hirschsprung disease (HSCR) are three common polymorphisms within cis-regulatory elements (CREs) of the receptor tyrosine kinase gene RET, which reduce its expression during enteric nervous system (ENS) development. These risk variants attenuate binding of the transcription factors RARB, GATA2, and SOX10 to their cognate CREs, reduce RET gene expression, and dysregulate other ENS and HSCR genes in the RET-EDNRB gene regulatory network (GRN). Here, we use siRNA, ChIP, and CRISPR-Cas9 deletion analyses in the SK-N-SH cell line to ask how many additional HSCR-associated risk variants reside in RET CREs that affect its gene expression. We identify 22 HSCR-associated variants in candidate RET CREs, of which seven have differential allele-specific in vitro enhancer activity, and four of these seven affect RET gene expression; of these, two enhancers are bound by the transcription factor PAX3. We also show that deleting multiple variant-containing enhancers leads to synergistic effects on RET gene expression. These, coupled with our prior results, show that common sequence variants in at least 10 RET enhancers affect HSCR risk, seven with experimental evidence of affecting RET gene expression, extending the known RET-EDNRB GRN to reveal an extensive regulatory code modulating disease risk at a single gene.

5.
N Engl J Med ; 380(15): 1421-1432, 2019 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-30970187

RESUMEN

BACKGROUND: Hirschsprung's disease, or congenital aganglionosis, is a developmental disorder of the enteric nervous system and is the most common cause of intestinal obstruction in neonates and infants. The disease has more than 80% heritability, including significant associations with rare and common sequence variants in genes related to the enteric nervous system, as well as with monogenic and chromosomal syndromes. METHODS: We genotyped and exome-sequenced samples from 190 patients with Hirschsprung's disease to quantify the genetic burden in patients with this condition. DNA sequence variants, large copy-number variants, and karyotype variants in probands were considered to be pathogenic when they were significantly associated with Hirschsprung's disease or another neurodevelopmental disorder. Novel genes were confirmed by functional studies in the mouse and human embryonic gut and in zebrafish embryos. RESULTS: The presence of five or more variants in four noncoding elements defined a widespread risk of Hirschsprung's disease (48.4% of patients and 17.1% of controls; odds ratio, 4.54; 95% confidence interval [CI], 3.19 to 6.46). Rare coding variants in 24 genes that play roles in enteric neural-crest cell fate, 7 of which were novel, were also common (34.7% of patients and 5.0% of controls) and conferred a much greater risk than noncoding variants (odds ratio, 10.02; 95% CI, 6.45 to 15.58). Large copy-number variants, which were present in fewer patients (11.4%, as compared with 0.2% of controls), conferred the highest risk (odds ratio, 63.07; 95% CI, 36.75 to 108.25). At least one identifiable genetic risk factor was found in 72.1% of the patients, and at least 48.4% of patients had a structural or regulatory deficiency in the gene encoding receptor tyrosine kinase (RET). For individual patients, the estimated risk of Hirschsprung's disease ranged from 5.33 cases per 100,000 live births (approximately 1 per 18,800) to 8.38 per 1000 live births (approximately 1 per 120). CONCLUSIONS: Among the patients in our study, Hirschsprung's disease arose from common noncoding variants, rare coding variants, and copy-number variants affecting genes involved in enteric neural-crest cell fate that exacerbate the widespread genetic susceptibility associated with RET. For individual patients, the genotype-specific odds ratios varied by a factor of approximately 67, which provides a basis for risk stratification and genetic counseling. (Funded by the National Institutes of Health.).


Asunto(s)
Variación Genética , Genotipo , Enfermedad de Hirschsprung/genética , Exoma , Femenino , Predisposición Genética a la Enfermedad , Humanos , Masculino , Mutación , Oportunidad Relativa , Penetrancia , Análisis de Secuencia de ADN , Secuenciación del Exoma
6.
Hum Genomics ; 15(1): 44, 2021 07 13.
Artículo en Inglés | MEDLINE | ID: mdl-34256850

RESUMEN

BACKGROUND: Previous research in autism and other neurodevelopmental disorders (NDDs) has indicated an important contribution of protein-coding (coding) de novo variants (DNVs) within specific genes. The role of de novo noncoding variation has been observable as a general increase in genetic burden but has yet to be resolved to individual functional elements. In this study, we assessed whole-genome sequencing data in 2671 families with autism (discovery cohort of 516 families, replication cohort of 2155 families). We focused on DNVs in enhancers with characterized in vivo activity in the brain and identified an excess of DNVs in an enhancer named hs737. RESULTS: We adapted the fitDNM statistical model to work in noncoding regions and tested enhancers for excess of DNVs in families with autism. We found only one enhancer (hs737) with nominal significance in the discovery (p = 0.0172), replication (p = 2.5 × 10-3), and combined dataset (p = 1.1 × 10-4). Each individual with a DNV in hs737 had shared phenotypes including being male, intact cognitive function, and hypotonia or motor delay. Our in vitro assessment of the DNVs showed they all reduce enhancer activity in a neuronal cell line. By epigenomic analyses, we found that hs737 is brain-specific and targets the transcription factor gene EBF3 in human fetal brain. EBF3 is genome-wide significant for coding DNVs in NDDs (missense p = 8.12 × 10-35, loss-of-function p = 2.26 × 10-13) and is widely expressed in the body. Through characterization of promoters bound by EBF3 in neuronal cells, we saw enrichment for binding to NDD genes (p = 7.43 × 10-6, OR = 1.87) involved in gene regulation. Individuals with coding DNVs have greater phenotypic severity (hypotonia, ataxia, and delayed development syndrome [HADDS]) in comparison to individuals with noncoding DNVs that have autism and hypotonia. CONCLUSIONS: In this study, we identify DNVs in the hs737 enhancer in individuals with autism. Through multiple approaches, we find hs737 targets the gene EBF3 that is genome-wide significant in NDDs. By assessment of noncoding variation and the genes they affect, we are beginning to understand their impact on gene regulatory networks in NDDs.


Asunto(s)
Trastorno Autístico/genética , Predisposición Genética a la Enfermedad , Hipotonía Muscular/genética , Trastornos del Neurodesarrollo/genética , Factores de Transcripción/genética , Trastorno Autístico/epidemiología , Trastorno Autístico/patología , Elementos de Facilitación Genéticos/genética , Exoma/genética , Femenino , Redes Reguladoras de Genes/genética , Humanos , Masculino , Hipotonía Muscular/epidemiología , Hipotonía Muscular/patología , Mutación/genética , Trastornos del Neurodesarrollo/epidemiología , Trastornos del Neurodesarrollo/patología , Neuronas/metabolismo , Neuronas/patología
7.
Proc Natl Acad Sci U S A ; 116(52): 26697-26708, 2019 Dec 26.
Artículo en Inglés | MEDLINE | ID: mdl-31818953

RESUMEN

The development of the gut from endodermal tissue to an organ with multiple distinct structures and functions occurs over a prolonged time during embryonic days E10.5-E14.5 in the mouse. During this process, one major event is innervation of the gut by enteric neural crest cells (ENCCs) to establish the enteric nervous system (ENS). To understand the molecular processes underpinning gut and ENS development, we generated RNA-sequencing profiles from wild-type mouse guts at E10.5, E12.5, and E14.5 from both sexes. We also generated these profiles from homozygous Ret null embryos, a model for Hirschsprung disease (HSCR), in which the ENS is absent. These data reveal 4 major features: 1) between E10.5 and E14.5 the developmental genetic programs change from expression of major transcription factors and its modifiers to genes controlling tissue (epithelium, muscle, endothelium) specialization; 2) the major effect of Ret is not only on ENCC differentiation to enteric neurons but also on the enteric mesenchyme and epithelium; 3) a muscle genetic program exerts significant effects on ENS development; and 4) sex differences in gut development profiles are minor. The genetic programs identified, and their changes across development, suggest that both cell autonomous and nonautonomous factors, and interactions between the different developing gut tissues, are important for normal ENS development and its disorders.

8.
Hum Mol Genet ; 28(18): 3137-3147, 2019 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-31313802

RESUMEN

Disruptions in gene regulatory networks (GRNs), driven by multiple deleterious variants, potentially underlie complex traits and diseases. Hirschsprung disease (HSCR), a multifactorial disorder of enteric nervous system (ENS) development, is associated with at least 24 genes and seven chromosomal loci, with RET and EDNRB as its major genes. We previously demonstrated that RET transcription in the ENS is controlled by an extensive GRN involving the transcription factors (TFs) RARB, GATA2 and SOX10 and other HSCR genes. We now demonstrate, using human and mouse cellular and animal models, that EDNRB is transcriptionally regulated in the ENS by GATA2, SOX10 and NKX2.5 TFs. Significantly, RET and EDNRB expression is regulated by their shared use of GATA2 and SOX10, and in turn, these TFs are controlled by EDNRB and RET in a dose-dependent manner. This study expands the ENS development GRN to include both RET and EDNRB, uncovers the mechanistic basis for RET-EDNRB epistasis and emphasizes how functionally different genes associated with a complex disorder can be united through a common GRN.


Asunto(s)
Epistasis Genética , Regulación de la Expresión Génica , Redes Reguladoras de Genes , Enfermedad de Hirschsprung/genética , Proteínas Proto-Oncogénicas c-ret/genética , Receptor de Endotelina B/genética , Animales , Modelos Animales de Enfermedad , Elementos de Facilitación Genéticos , Epigénesis Genética , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Humanos , Ratones , Modelos Biológicos , Secuencias Reguladoras de Ácidos Nucleicos , Factores de Transcripción/metabolismo
9.
Annu Rev Genomics Hum Genet ; 18: 45-63, 2017 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-28399667

RESUMEN

Gene expression changes, the driving forces for cellular diversity in multicellular organisms, are regulated by a diverse set of gene regulatory elements that direct transcription in specific cells. Mutations in these elements, ranging from chromosomal aberrations to single-nucleotide polymorphisms, are a major cause of human disease. However, we currently have a very limited understanding of how regulatory element genotypes lead to specific phenotypes. In this review, we discuss the various methods of regulatory element identification, the different types of mutations they harbor, and their impact on human disease. We highlight how these variations can affect transcription of multiple genes in gene regulatory networks. In addition, we describe how novel technologies, such as massively parallel reporter assays and CRISPR/Cas9 genome editing, are beginning to provide a better understanding of the functional roles that these elements have and how their alteration can lead to specific phenotypes.


Asunto(s)
Elementos de Facilitación Genéticos , Redes Reguladoras de Genes , Regiones Promotoras Genéticas , Sistemas CRISPR-Cas , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Humanos , Análisis de Secuencia de ADN/métodos
10.
Hum Mol Genet ; 26(10): 1811-1820, 2017 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-28334784

RESUMEN

For most multigenic disorders, clinical manifestation (penetrance) and presentation (expressivity) are likely to be an outcome of genetic interaction between multiple susceptibility genes. Here, using gene knockouts in mice, we evaluated genetic interaction between loss of Ret and loss of Sema3d, two Hirschsprung disease susceptibility genes. We intercrossed Ret and Sema3d double null heterozygotes to generate mice with the nine possible genotypes and assessed survival by counting various genotypes, myenteric plexus presence by acetylcholinesterase staining and embryonic day 12.5 (E12.5) intestine transcriptome by RNA-sequencing. Survival rates of Ret wild-type, null heterozygote and null homozygote mice at E12.5, birth and weaning were not influenced by the genotypes at Sema3d locus and vice versa. Loss of myenteric plexus was observed only in all Ret null homozygotes, irrespective of the genotypes at Sema3d locus, and Sema3d null heterozygote and homozygote mice had normal intestinal innervation. As compared with wild-type mice intestinal gene expression, loss of Ret in null homozygotes led to differential expression of ∼300 genes, whereas loss of Sema3d in null homozygotes had no major consequence and there was no evidence supporting major interaction between the two genes influencing intestine transcriptome. Overall, given the null alleles and phenotypic assays used, we did not find evidence for genetic interaction between Ret and Sema3d affecting survival, presence of myenteric plexus or intestine transcriptome.


Asunto(s)
Proteínas Proto-Oncogénicas c-ret/genética , Semaforinas/genética , Acetilcolinesterasa , Animales , Sistema Nervioso Entérico/metabolismo , Epistasis Genética , Genotipo , Heterocigoto , Enfermedad de Hirschsprung/genética , Homocigoto , Ratones , Ratones Noqueados , Mutación , Penetrancia , Fenotipo , Proteínas Proto-Oncogénicas c-ret/metabolismo , Semaforinas/metabolismo
11.
Hum Mol Genet ; 24(10): 2997-3003, 2015 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-25666438

RESUMEN

The risk of Hirschsprung disease (HSCR) is ∼15/100 000 live births per newborn but has been reported to show significant inter-individual variation from the effects of seven common susceptibility alleles at the RET, SEMA3 and NRG1 loci. We show, by analyses of these variants in 997 samples from 376 HSCR families of European ancestry, that significant genetic risk can only be detected at RET (rs2435357 and rs2506030) and at SEMA3 (rs11766001), but not at NRG1. RET rs2435357 also showed significant frequency differences by gender, segment length of aganglionosis and familiality. Further, in combination, disease risk varied >30-fold between individuals with none and up to 6 susceptibility alleles. Thus, these polymorphisms can be used to stratify the newborn population into distinct phenotypic classes with defined risks to understand HSCR etiology.


Asunto(s)
Enfermedad de Hirschsprung/genética , Neurregulina-1/genética , Polimorfismo Genético , Proteínas Proto-Oncogénicas c-ret/genética , Semaforina-3A/genética , Femenino , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Humanos , Masculino , Factores de Riesgo , Factores Sexuales , Población Blanca/genética
12.
Am J Hum Genet ; 94(6): 854-69, 2014 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-24857694

RESUMEN

QT interval variation is assumed to arise from variation in repolarization as evidenced from rare Na- and K-channel mutations in Mendelian QT prolongation syndromes. However, in the general population, common noncoding variants at a chromosome 1q locus are the most common genetic regulators of QT interval variation. In this study, we use multiple human genetic, molecular genetic, and cellular assays to identify a functional variant underlying trait association: a noncoding polymorphism (rs7539120) that maps within an enhancer of NOS1AP and affects cardiac function by increasing NOS1AP transcript expression. We further localized NOS1AP to cardiomyocyte intercalated discs (IDs) and demonstrate that overexpression of NOS1AP in cardiomyocytes leads to altered cellular electrophysiology. We advance the hypothesis that NOS1AP affects cardiac electrical conductance and coupling and thereby regulates the QT interval through propagation defects. As further evidence of an important role for propagation variation affecting QT interval in humans, we show that common polymorphisms mapping near a specific set of 170 genes encoding ID proteins are significantly enriched for association with the QT interval, as compared to genome-wide markers. These results suggest that focused studies of proteins within the cardiomyocyte ID are likely to provide insights into QT prolongation and its associated disorders.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Síndrome de QT Prolongado/genética , Miocitos Cardíacos/metabolismo , Sitios de Carácter Cuantitativo , Animales , Estudios de Cohortes , Electrocardiografía , Regulación de la Expresión Génica , Estudio de Asociación del Genoma Completo , Genotipo , Células HEK293 , Humanos , Lentivirus/genética , Ratones , Fenotipo , Polimorfismo de Nucleótido Simple , Pez Cebra/embriología , Pez Cebra/genética
13.
BMC Genomics ; 15: 1072, 2014 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-25480362

RESUMEN

BACKGROUND: Vertebrate organogenesis is a highly complex process involving sequential cascades of transcription factor activation or repression. Interestingly a single developmental control gene can occasionally be essential for the morphogenesis and differentiation of tissues and organs arising from vastly disparate embryological lineages. RESULTS: Here we elucidated the role of the mammalian homeobox gene Bapx1 during the embryogenesis of five distinct organs at E12.5 - vertebral column, spleen, gut, forelimb and hindlimb - using expression profiling of sorted wildtype and mutant cells combined with genome wide binding site analysis. Furthermore we analyzed the development of the vertebral column at the molecular level by combining transcriptional profiling and genome wide binding data for Bapx1 with similarly generated data sets for Sox9 to assemble a detailed gene regulatory network revealing genes previously not reported to be controlled by either of these two transcription factors. CONCLUSIONS: The gene regulatory network appears to control cell fate decisions and morphogenesis in the vertebral column along with the prevention of premature chondrocyte differentiation thus providing a detailed molecular view of vertebral column development.


Asunto(s)
Redes Reguladoras de Genes , Genoma , Proteínas de Homeodominio/genética , Factor de Transcripción SOX9/genética , Columna Vertebral/metabolismo , Alelos , Animales , Supervivencia Celular , Condrocitos/citología , Inmunoprecipitación de Cromatina , Embrión de Mamíferos/metabolismo , Desarrollo Embrionario , Inhibidores Enzimáticos/metabolismo , Perfilación de la Expresión Génica , Proteínas de Homeodominio/metabolismo , Ratones , Ratones Transgénicos , Unión Proteica , Factor de Transcripción SOX9/metabolismo , Análisis de Secuencia de ADN
14.
Biotechnol Lett ; 35(12): 1991-6, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23907671

RESUMEN

Traditionally, conditional knockout studies in mouse have utilized the Cre or Flpe technology to activate the expression of reporter genes such as lacZ or PLAP. Employing these reporter genes, however, requires tissue fixation. To make way for downstream in vivo or in vitro applications, we have inserted enhanced green fluorescent protein (EGFP) into the endogenous Sox9 locus and generated a novel conditional Sox9 null allele, by flanking the entire Sox9 coding region with loxP sites and inserting an EGFP reporter gene into the 3'-UTR allowing for EGFP to be expressed upon Sox9 loss of function yet under the control of the endogenous Sox9 promoter. Mating this new allele to any Cre-expressing line, the fate of Sox9 null cells can be traced in the cell type of interest in vivo or in vitro after fluorescence-activated cell sorting.


Asunto(s)
Ingeniería Genética/métodos , Proteínas Fluorescentes Verdes/genética , Factor de Transcripción SOX9/genética , Animales , Línea Celular , Clonación Molecular , Colágeno Tipo II/genética , Colágeno Tipo II/metabolismo , Embrión de Mamíferos , Femenino , Técnicas de Inactivación de Genes , Proteínas Fluorescentes Verdes/química , Proteínas Fluorescentes Verdes/metabolismo , Masculino , Ratones , Ratones Noqueados , Factor de Transcripción SOX9/química , Factor de Transcripción SOX9/metabolismo
15.
PLoS One ; 18(1): e0277804, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36701283

RESUMEN

Unoccupied aerial systems (UAS) based high throughput phenotyping studies require further investigation to combine different environments and planting times into one model. Here 100 elite breeding hybrids of maize (Zea mays L.) were evaluated in two environment trials-one with optimal planting and irrigation (IHOT), and one dryland with delayed planting (DHOT). RGB (Red-Green-Blue) based canopy height measurement (CHM) and vegetation indices (VIs) were estimated from a UAS platform. Time series and cumulative VIs, by both summation (ΣVI-SUMs) and area under the curve (ΣVI-AUCs), were fit via machine learning regression modeling (random forest, linear, ridge, lasso, elastic net regressions) to estimate grain yield. VIs were more valuable predictors of yield to combine different environments than CHM. Time series VIs and CHM produced high accuracies (~68-72%), but inconsistent models. A little sacrifice in accuracy (~60-65%) produced consistent models using ΣVI-SUMs and CHM during pre-reproductive vegetative growth. Absence of VIs produced poorer accuracies (by about ~5-10%). Normalized difference type VIs produced maximum accuracies, and flowering times were the best times for UAS data acquisition. This study suggests that the best yielding varieties can be accurately predicted in new environments at or before flowering when combining multiple temporal flights and predictors.


Asunto(s)
Fitomejoramiento , Zea mays , Zea mays/genética , Grano Comestible
16.
BMC Dev Biol ; 11: 63, 2011 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-22011226

RESUMEN

BACKGROUND: Identifying DNA sequences (enhancers) that direct the precise spatial and temporal expression of developmental control genes remains a significant challenge in the annotation of vertebrate genomes. Locating these sequences, which in many cases lie at a great distance from the transcription start site, has been a major obstacle in deciphering gene regulation. Coupling of comparative genomics with functional validation to locate such regulatory elements has been a successful method in locating many such regulatory elements. But most of these studies looked either at a single gene only or the whole genome without focusing on any particular process. The pressing need is to integrate the tools of comparative genomics with knowledge of developmental biology to validate enhancers for developmental transcription factors in greater detail RESULTS: Our results show that near four different genes (nkx3.2, pax9, otx1b and foxa2) in zebrafish, only 20-30% of highly conserved DNA sequences can act as developmental enhancers irrespective of the tissue the gene expresses in. We find that some genes also have multiple conserved enhancers expressing in the same tissue at the same or different time points in development. We also located non-conserved enhancers for two of the genes (pax9 and otx1b). Our modified Bacterial artificial chromosome (BACs) studies for these 4 genes revealed that many of these enhancers work in a synergistic fashion, which cannot be captured by individual DNA constructs and are not conserved at the sequence level. Our detailed biochemical and transgenic analysis revealed Foxa1 binds to the otx1b non-conserved enhancer to direct its activity in forebrain and otic vesicle of zebrafish at 24 hpf. CONCLUSION: Our results clearly indicate that high level of functional conservation of genes is not necessarily associated with sequence conservation of its regulatory elements. Moreover certain non conserved DNA elements might have role in gene regulation. The need is to bring together multiple approaches to bear upon individual genes to decipher all its regulatory elements.


Asunto(s)
Elementos de Facilitación Genéticos/genética , Regulación del Desarrollo de la Expresión Génica , Estratos Germinativos/metabolismo , Secuencias Reguladoras de Ácidos Nucleicos/genética , Transcripción Genética/genética , Pez Cebra/embriología , Pez Cebra/genética , Animales , Cromosomas Artificiales Bacterianos/genética , Secuencia Conservada/genética , Genes del Desarrollo , Genómica/métodos , Factor Nuclear 3-alfa del Hepatocito/genética , Factor Nuclear 3-alfa del Hepatocito/metabolismo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Morfolinos/genética , Factores de Transcripción Otx/genética , Factores de Transcripción Otx/metabolismo , Factor de Transcripción PAX9/genética , Factor de Transcripción PAX9/metabolismo , Alineación de Secuencia , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Pez Cebra/metabolismo , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
17.
BMC Genet ; 11: 68, 2010 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-20637105

RESUMEN

The inner ear is one of the most complex and detailed organs in the vertebrate body and provides us with the priceless ability to hear and perceive linear and angular acceleration (hence maintain balance). The development and morphogenesis of the inner ear from an ectodermal thickening into distinct auditory and vestibular components depends upon precise temporally and spatially coordinated gene expression patterns and well orchestrated signaling cascades within the otic vesicle and upon cellular movements and interactions with surrounding tissues. Gene loss of function analysis in mice has identified homeobox genes along with other transcription and secreted factors as crucial regulators of inner ear morphogenesis and development. While otic induction seems dependent upon fibroblast growth factors, morphogenesis of the otic vesicle into the distinct vestibular and auditory components appears to be clearly dependent upon the activities of a number of homeobox transcription factors. The Pax2 paired-homeobox gene is crucial for the specification of the ventral otic vesicle derived auditory structures and the Dlx5 and Dlx6 homeobox genes play a major role in specification of the dorsally derived vestibular structures. Some Micro RNAs have also been recently identified which play a crucial role in the inner ear formation.


Asunto(s)
Oído Interno/embriología , Genes del Desarrollo , Animales , Regulación del Desarrollo de la Expresión Génica , Humanos , Ratones , Morfogénesis/genética , Factor de Transcripción PAX2 , Proteómica , Vestíbulo del Laberinto/embriología
18.
Transgenic Res ; 19(2): 299-304, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19662507

RESUMEN

A tissue-specific transgenic model was employed to test the effects of intron and vector sequences on transgene expression in zebrafish after microinjection. In this model, the 2.3 kb promoter taken from the 5' upstream region of the transcription initiation site of keratin 4 (krt4) was used to drive the enhanced green fluorescence protein (EGFP) reporter gene in a transgenic vector. For assaying the strength of EGFP expression, the effects of including an intron before the EGFP coding region or using different forms of DNA, including circular plasmid, linear full-length plasmid, and the linear transgene coding region without any prokaryotic vector sequence, were tested. After microinjection, the transgene expression was analyzed using transient assays. Consequently, further comparative analysis supported by Fisher's exact test was performed based on the data generated by analyzing the strength of the transgene expression. It was shown that inclusion of an intron in the construct increases the transgene expression in a transient transgenic zebrafish assay. Furthermore, the circular plasmid containing the transgene produced the strongest EGFP expression.


Asunto(s)
Secuencia de Bases , Vectores Genéticos/genética , Plásmidos/genética , Procesamiento Postranscripcional del ARN , Transgenes/fisiología , Pez Cebra/metabolismo , Animales , Animales Modificados Genéticamente , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Intrones/genética , Queratina-4/genética , Queratina-4/metabolismo , Microinyecciones , Conejos , Transgenes/genética , Pez Cebra/embriología , Pez Cebra/genética , Globinas beta/genética
19.
Eur J Hum Genet ; 26(4): 561-569, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29379196

RESUMEN

Hirschsprung disease (HSCR) is a congenital disorder with a population incidence of ~1/5000 live births, defined by an absence of enteric ganglia along variable lengths of the colon. HSCR genome-wide association studies (GWAS) have found common associated variants at RET, SEMA3, and NRG1, but they still fail to explain all of its heritability. To enhance gene discovery, we performed a GWAS of 170 cases identified from the Danish nationwide pathology registry with 4717 controls, based on 6.2 million variants imputed from the haplotype reference consortium panel. We found a novel low-frequency variant (rs144432435), which, when conditioning on the lead RET single-nucleotide polymorphism (SNP), was of genome-wide significance in the discovery analysis. This conditional association signal was replicated in a Swedish HSCR cohort with discovery plus replication meta-analysis conditional odds ratio of 6.6 (P = 7.7 × 10-10; 322 cases and 4893 controls). The conditional signal was, however, not replicated in two HSCR cohorts from USA and Finland, leading to the hypothesis that rs144432435 tags a rare haplotype present in Denmark and Sweden. Using the genome-wide complex trait analysis method, we estimated the SNP heritability of HSCR to be 88%, close to estimates based on classical family studies. Moreover, by using Lasso (least absolute shrinkage and selection operator) regression we were able to construct a genetic HSCR predictor with a area under the receiver operator characteristics curve of 76% in an independent validation set. In conclusion, we combined the largest collection of sporadic Hirschsprung cases to date (586 cases) to further elucidate HSCR's genetic architecture.


Asunto(s)
Enfermedad de Hirschsprung/genética , Polimorfismo de Nucleótido Simple , Proteínas Proto-Oncogénicas c-ret/genética , Haplotipos , Humanos
20.
Biomed Res Int ; 2017: 8932583, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28630873

RESUMEN

Embryogenesis is an intricate process involving multiple genes and pathways. Some of the key transcription factors controlling specific cell types are the Sox trio, namely, Sox5, Sox6, and Sox9, which play crucial roles in organogenesis working in a concerted manner. Much however still needs to be learned about their combinatorial roles during this process. A developmental genomics and systems biology approach offers to complement the reductionist methodology of current developmental biology and provide a more comprehensive and integrated view of the interrelationships of complex regulatory networks that occur during organogenesis. By combining cell type-specific transcriptome analysis and in vivo ChIP-Seq of the Sox trio using mouse embryos, we provide evidence for the direct control of Sox5 and Sox6 by the transcriptional trio in the murine model and by Morpholino knockdown in zebrafish and demonstrate the novel role of Tgfb2, Fbxl18, and Tle3 in formation of Sox5, Sox6, and Sox9 dependent tissues. Concurrently, a complete embryonic gene regulatory network has been generated, identifying a wide repertoire of genes involved and controlled by the Sox trio in the intricate process of normal embryogenesis.


Asunto(s)
Embrión de Mamíferos/embriología , Regulación del Desarrollo de la Expresión Génica/fisiología , Organogénesis/fisiología , Factores de Transcripción SOX/metabolismo , Biología de Sistemas , Animales , Ratones , Factores de Transcripción SOX/genética , Pez Cebra/embriología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA