Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
PLoS Genet ; 19(11): e1011030, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37948459

ABSTRACT

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.


Subject(s)
Enhancer Elements, Genetic , Gastrointestinal Tract , Proto-Oncogene Proteins c-ret , Haplotypes , Humans , Proto-Oncogene Proteins c-ret/genetics , Neuroblastoma , Cell Line, Tumor , Hirschsprung Disease/genetics , Fetus , Gastrointestinal Tract/embryology , Neural Crest/cytology , Enteric Nervous System/embryology , Single-Cell Gene Expression Analysis , Gene Expression Regulation, Developmental
2.
Proc Natl Acad Sci U S A ; 120(34): e2211986120, 2023 08 22.
Article in English | MEDLINE | ID: mdl-37585461

ABSTRACT

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.


Subject(s)
Enteric Nervous System , Hirschsprung Disease , Proto-Oncogene Proteins c-ret , Animals , Mice , Cell Differentiation , Cell Proliferation , Hirschsprung Disease/genetics , Neural Crest , Proto-Oncogene Proteins c-ret/genetics
3.
PLoS One ; 18(1): e0277804, 2023.
Article in English | MEDLINE | ID: mdl-36701283

ABSTRACT

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.


Subject(s)
Plant Breeding , Zea mays , Zea mays/genetics , Edible Grain
4.
Genome Res ; 31(12): 2199-2208, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34782358

ABSTRACT

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.
Hum Genomics ; 15(1): 44, 2021 07 13.
Article in English | MEDLINE | ID: mdl-34256850

ABSTRACT

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.


Subject(s)
Autistic Disorder/genetics , Genetic Predisposition to Disease , Muscle Hypotonia/genetics , Neurodevelopmental Disorders/genetics , Transcription Factors/genetics , Autistic Disorder/epidemiology , Autistic Disorder/pathology , Enhancer Elements, Genetic/genetics , Exome/genetics , Female , Gene Regulatory Networks/genetics , Humans , Male , Muscle Hypotonia/epidemiology , Muscle Hypotonia/pathology , Mutation/genetics , Neurodevelopmental Disorders/epidemiology , Neurodevelopmental Disorders/pathology , Neurons/metabolism , Neurons/pathology
6.
Proc Natl Acad Sci U S A ; 116(52): 26697-26708, 2019 Dec 26.
Article in English | MEDLINE | ID: mdl-31818953

ABSTRACT

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.

7.
Hum Mol Genet ; 28(18): 3137-3147, 2019 09 15.
Article in English | MEDLINE | ID: mdl-31313802

ABSTRACT

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.


Subject(s)
Epistasis, Genetic , Gene Expression Regulation , Gene Regulatory Networks , Hirschsprung Disease/genetics , Proto-Oncogene Proteins c-ret/genetics , Receptor, Endothelin B/genetics , Animals , Disease Models, Animal , Enhancer Elements, Genetic , Epigenesis, Genetic , Genetic Association Studies , Genetic Predisposition to Disease , Humans , Mice , Models, Biological , Regulatory Sequences, Nucleic Acid , Transcription Factors/metabolism
8.
N Engl J Med ; 380(15): 1421-1432, 2019 Apr 11.
Article in English | MEDLINE | ID: mdl-30970187

ABSTRACT

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.).


Subject(s)
Genetic Variation , Genotype , Hirschsprung Disease/genetics , Exome , Female , Genetic Predisposition to Disease , Humans , Male , Mutation , Odds Ratio , Penetrance , Sequence Analysis, DNA , Exome Sequencing
9.
Eur J Hum Genet ; 26(4): 561-569, 2018 04.
Article in English | MEDLINE | ID: mdl-29379196

ABSTRACT

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.


Subject(s)
Hirschsprung Disease/genetics , Polymorphism, Single Nucleotide , Proto-Oncogene Proteins c-ret/genetics , Haplotypes , Humans
10.
Biomed Res Int ; 2017: 8932583, 2017.
Article in English | MEDLINE | ID: mdl-28630873

ABSTRACT

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.


Subject(s)
Embryo, Mammalian/embryology , Gene Expression Regulation, Developmental/physiology , Organogenesis/physiology , SOX Transcription Factors/metabolism , Systems Biology , Animals , Mice , SOX Transcription Factors/genetics , Zebrafish/embryology
11.
Annu Rev Genomics Hum Genet ; 18: 45-63, 2017 08 31.
Article in English | MEDLINE | ID: mdl-28399667

ABSTRACT

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.


Subject(s)
Enhancer Elements, Genetic , Gene Regulatory Networks , Promoter Regions, Genetic , CRISPR-Cas Systems , High-Throughput Nucleotide Sequencing/methods , Humans , Sequence Analysis, DNA/methods
12.
Hum Mol Genet ; 26(10): 1811-1820, 2017 05 15.
Article in English | MEDLINE | ID: mdl-28334784

ABSTRACT

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.


Subject(s)
Proto-Oncogene Proteins c-ret/genetics , Semaphorins/genetics , Acetylcholinesterase , Animals , Enteric Nervous System/metabolism , Epistasis, Genetic , Genotype , Heterozygote , Hirschsprung Disease/genetics , Homozygote , Mice , Mice, Knockout , Mutation , Penetrance , Phenotype , Proto-Oncogene Proteins c-ret/metabolism , Semaphorins/metabolism
13.
Cell ; 167(2): 355-368.e10, 2016 Oct 06.
Article in English | MEDLINE | ID: mdl-27693352

ABSTRACT

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.


Subject(s)
Enhancer Elements, Genetic , Gene Expression Regulation , Gene Regulatory Networks , Hirschsprung Disease/genetics , Proto-Oncogene Proteins c-ret/genetics , Alleles , Animals , Binding Sites , Disease Models, Animal , GATA2 Transcription Factor/genetics , GATA2 Transcription Factor/metabolism , Gastrointestinal Tract/embryology , Humans , Mice , Mice, Transgenic , RNA, Untranslated/genetics , Receptors, Retinoic Acid/genetics , Receptors, Retinoic Acid/metabolism , SOXE Transcription Factors/genetics , SOXE Transcription Factors/metabolism
14.
Genom Data ; 10: 51-3, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27672560

ABSTRACT

This work pertains to GEO submission GSE36672, in vivo and in vitro genome wide binding (ChIP-Seq) of Bapx1/Nkx3.2 and Sox9 proteins. We have previously shown that data from a genome wide binding assay combined with transcriptional profiling is an insightful means to divulge the mechanisms directing cell type specification and the generation of tissues and subsequent organs [1]. Our earlier work identified the role of the DNA-binding homeodomain containing protein Bapx1/Nkx3.2 in midgestation murine embryos. Microarray analysis of EGFP-tagged cells (both wildtype and null) was integrated using ChIP-Seq analysis of Bapx1/Nkx3.2 and Sox9 DNA-binding proteins in living tissue.

15.
J Invest Surg ; 29(2): 88-92, 2016.
Article in English | MEDLINE | ID: mdl-26375922

ABSTRACT

BACKGROUND: Gastroschisis is a developmental disorder involving the extrusion of fetal intestines through a defect in the abdominal wall. The mechanism is presumed to be a dual vascular/thrombotic pathogenesis, where normal right umbilical vein involution forms a possible site for thrombosis adjacent to the umbilical ring. PURPOSE: The aim of this study was to demonstrate that the 3 common prothrombotic polymorphisms, MTHFR c.677C>T, F2 c.20210G>A, and F5 Leiden, were elevated in frequency in Indonesian gastroschisis patients. MATERIAL AND METHODS: Three genetic markers were investigated in 46 patients with gastroschisis and 89 ethnicity-matched controls for association studies using polymerase chain reaction-restriction fragment length polymorphisms (PCR-RFLP) or TaqMan Genotyping Assays on genomic DNA. RESULTS: MTHFR c.677C>T showed a significant association with gastroschisis (OR = 2.1, 95% CI = 1.13-3.86; p = .018) but no affected infants had risk alleles for either F2 c.20210G>A or F5 Leiden. Further, the frequency of MTHFR risk allele (T) in patients with maternal age <25 years is marginally significant higher than those in cases with maternal age ≥25 years (p = .069) with an OR of 2.7 (95% CI = 0.90-8.07). CONCLUSIONS: MTHFR is a common susceptibility factor for gastroschisis in Indonesia. The increased gastroschisis risk in offspring of younger maternal age suggests the thrombotic pathogenesis model. A founder effect is the most likely explanation for the rarity of the F2 and F5 Leiden polymorphisms in Indonesian population.


Subject(s)
Factor V/genetics , Gastroschisis/genetics , Methylenetetrahydrofolate Reductase (NADPH2)/genetics , Prothrombin/genetics , Thromboembolism/genetics , Thrombosis/genetics , Adult , Alleles , Amplified Fragment Length Polymorphism Analysis , Female , Founder Effect , Genetic Association Studies , Genetic Predisposition to Disease , Genotype , Humans , Indonesia , Infant , Male , Maternal Age , Polymorphism, Single Nucleotide
16.
Pediatr Dev Pathol ; 18(6): 466-76, 2015.
Article in English | MEDLINE | ID: mdl-26699691

ABSTRACT

Intestinal neuronal dysplasia type B (IND) denotes an increased proportion of hyperplastic submucosal ganglia, as resolved histochemically in 15-µm-thick frozen sections. IND has been reported proximal to the aganglionic segment in patients with Hirschsprung disease (HSCR) and is putatively associated with a higher rate of postsurgical dysmotility. We developed and validated histological criteria to diagnose IND-like submucosal ganglion cell hyperplasia (IND-SH) in paraffin sections and used the approach to study the incidence and clinical and/or genetic associations of IND-SH at the proximal margins of HSCR pull-through resection specimens. Full-circumference paraffin sections from the proximal margins of 64 HSCR colonic pull-through specimens and 24 autopsy controls were immunostained for neuron-specific Hu antigen, and nucleated ganglion cells in each submucosal ganglion were counted. In controls, an age-related decline in the relative abundance of "giant" ganglia (≥7 nucleated Hu-positive [Hu+] ganglion cells) was observed. A conservative diagnostic threshold for IND-SH (control mean ± 3× standard deviation) was derived from 15 controls less than 25 weeks of age. No control exceeded this threshold, whereas in the same age range, IND-SH was observed at the proximal margins in 15% (7 of 46) of HSCR resections, up to 15 cm proximal to the aganglionic segment. No significant correlation was observed between IND-SH and length of or distance from the aganglionic segment, sex, trisomy 21, RET or SEMA3C/D polymorphisms, or clinical outcome, but analysis of more patients, with better long-term follow-up will be required to clarify the significance of this histological phenotype.


Subject(s)
Colectomy , Colon/innervation , Enteric Nervous System/pathology , Hirschsprung Disease/pathology , Intestinal Diseases/pathology , Nervous System Diseases/pathology , Neurons/pathology , Biomarkers/analysis , Case-Control Studies , Cell Count , Colon/pathology , Colon/surgery , ELAV Proteins/analysis , Female , Hirschsprung Disease/genetics , Hirschsprung Disease/surgery , Humans , Hyperplasia , Immunohistochemistry , Infant, Newborn , Intestinal Diseases/genetics , Male , Nervous System Diseases/genetics , Paraffin Embedding , Predictive Value of Tests , Reproducibility of Results
17.
Genom Data ; 5: 103-105, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26101748

ABSTRACT

The data described in this article refers to Chatterjee et al. (2015) "In vivo genome-wide analysis of multiple tissues identifies gene regulatory networks, novel functions and downstream regulatory genes for Bapx1 and its co-regulation with Sox9 in the mammalian vertebral column" (GEO GSE35649) [1]. Transcriptional profiling combined with genome wide binding data is a powerful tool to elucidate the molecular mechanism behind vertebrate organogenesis. It also helps to uncover multiple roles of a single gene in different organs. In the above mentioned report we reveal the function of the homeobox gene Bapx1 during the embryogenesis of five distinct organs (vertebral column, spleen, gut, forelimb and hindlimb) at a relevant developmental stage (E12.5), microarray analysis of isolated wildtype and mutant cells in is compared in conjunction with ChIP-Seq analysis. We also analyzed the development of the vertebral column by comparing microarray and ChIP-Seq data for Bapx1 with similarly generated data sets for Sox9 to generate a gene regulatory network controlling various facets of the organogenesis.

18.
Hum Mol Genet ; 24(10): 2997-3003, 2015 May 15.
Article in English | MEDLINE | ID: mdl-25666438

ABSTRACT

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.


Subject(s)
Hirschsprung Disease/genetics , Neuregulin-1/genetics , Polymorphism, Genetic , Proto-Oncogene Proteins c-ret/genetics , Semaphorin-3A/genetics , Female , Genetic Association Studies , Genetic Predisposition to Disease , Humans , Male , Risk Factors , Sex Factors , White People/genetics
19.
J Pediatr Surg ; 49(11): 1614-8, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25475805

ABSTRACT

BACKGROUND: Hirschsprung disease (HSCR) is a neurocristopathy characterized by absence of intramural ganglion cells along variable lengths of the gastrointestinal tract in neonates. Three polymorphisms, rs2435357, within a conserved transcriptional enhancer of RET, and, rs7835688 and rs16879552, within intron 1 of NRG1, have been shown to be associated with isolated forms of HSCR. We wished to replicate these findings, and study the interactions between these variants, in Indonesian HSCR patients. METHODS: Sixty isolated HSCR patients and 124 controls were ascertained for this study. The three genetic markers were examined using TaqMan Genotyping Assays in genomic DNA for association studies. RESULTS: RET rs2435357 showed the strongest association with HSCR both by case-control analysis (p=2.5 × 10(-8)) and transmission disequilibrium test (p=4.2 × 10(-6)). NRG1 rs7835688 was modestly associated with HSCR only by case-control analysis (p=4.3 × 10(-3)), whereas rs16879552 demonstrated no association (p>0.097). Two locus analyses of variants showed significant interactions with increased and decreased disease risks of HSCR at NRG1 but conditional on rs2435357 genotype. CONCLUSIONS: RET and NRG1 variants are common susceptibility factors for HSCR in Indonesia. These common variants demonstrate that development of HSCR requires joint effects of RET and NRG1 early in gut development.


Subject(s)
DNA/genetics , Genetic Predisposition to Disease , Hirschsprung Disease/genetics , Neuregulin-1/genetics , Polymorphism, Genetic , Proto-Oncogene Proteins c-ret/genetics , Female , Genetic Markers , Genotype , Growth Substances , Hirschsprung Disease/epidemiology , Hirschsprung Disease/metabolism , Humans , Incidence , Indonesia/epidemiology , Introns , Male , Mitogens , Neuregulin-1/metabolism , Proto-Oncogene Proteins c-ret/metabolism
20.
BMC Genomics ; 15: 1072, 2014 Dec 05.
Article in English | MEDLINE | ID: mdl-25480362

ABSTRACT

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.


Subject(s)
Gene Regulatory Networks , Genome , Homeodomain Proteins/genetics , SOX9 Transcription Factor/genetics , Spine/metabolism , Alleles , Animals , Cell Survival , Chondrocytes/cytology , Chromatin Immunoprecipitation , Embryo, Mammalian/metabolism , Embryonic Development , Enzyme Inhibitors/metabolism , Gene Expression Profiling , Homeodomain Proteins/metabolism , Mice , Mice, Transgenic , Protein Binding , SOX9 Transcription Factor/metabolism , Sequence Analysis, DNA
SELECTION OF CITATIONS
SEARCH DETAIL
...