Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
1.
Genome Res ; 30(11): 1618-1632, 2020 11.
Article in English | MEDLINE | ID: mdl-32948616

ABSTRACT

It is widely recognized that noncoding genetic variants play important roles in many human diseases, but there are multiple challenges that hinder the identification of functional disease-associated noncoding variants. The number of noncoding variants can be many times that of coding variants; many of them are not functional but in linkage disequilibrium with the functional ones; different variants can have epistatic effects; different variants can affect the same genes or pathways in different individuals; and some variants are related to each other not by affecting the same gene but by affecting the binding of the same upstream regulator. To overcome these difficulties, we propose a novel analysis framework that considers convergent impacts of different genetic variants on protein binding, which provides multiscale information about disease-associated perturbations of regulatory elements, genes, and pathways. Applying it to our whole-genome sequencing data of 918 short-segment Hirschsprung disease patients and matched controls, we identify various novel genes not detected by standard single-variant and region-based tests, functionally centering on neural crest migration and development. Our framework also identifies upstream regulators whose binding is influenced by the noncoding variants. Using human neural crest cells, we confirm cell stage-specific regulatory roles of three top novel regulatory elements on our list, respectively in the RET, RASGEF1A, and PIK3C2B loci. In the PIK3C2B regulatory element, we further show that a noncoding variant found only in the patients affects the binding of the gliogenesis regulator NFIA, with a corresponding up-regulation of multiple genes in the same topologically associating domain.


Subject(s)
Enhancer Elements, Genetic , Hirschsprung Disease/genetics , Promoter Regions, Genetic , Class II Phosphatidylinositol 3-Kinases/genetics , Class II Phosphatidylinositol 3-Kinases/metabolism , Genetic Variation , Humans , Introns , NFI Transcription Factors/metabolism , Proto-Oncogene Proteins c-ret/genetics , Whole Genome Sequencing , ras Guanine Nucleotide Exchange Factors/genetics
2.
Gastroenterology ; 157(6): 1556-1571.e5, 2019 12.
Article in English | MEDLINE | ID: mdl-31442438

ABSTRACT

BACKGROUND & AIMS: It has been a challenge to develop fully functioning cells from human pluripotent stem cells (hPSCs). We investigated how activation of hedgehog signaling regulates derivation of enteric neural crest (NC) cells from hPSCs. METHODS: We analyzed transcriptomes of mouse and hPSC-derived enteric NCs using single-cell RNA sequencing (scRNA-seq) to identify the changes in expression associated with lineage differentiation. Intestine tissues were collected from Tg(GBS-GFP), Sufuf/f; Wnt1-cre, Ptch1+/-, and Gli3Δ699/Δ699 mice and analyzed by flow cytometry and immunofluorescence for levels of messenger RNAs encoding factors in the hedgehog signaling pathway during differentiation of enteric NCs. Human NC cells (HNK-1+p75NTR+) were derived from IMR90 and UE02302 hPSC lines. hPSCs were incubated with a hedgehog agonist (smoothened agonist [SAG]) and antagonists (cyclopamine) and analyzed for differentiation. hPSC-based innervated colonic organoids were derived from these hPSC lines and analyzed by immunofluorescence and neuromuscular coupling assay for expression of neuronal subtype markers and assessment of the functional maturity of the hPSC-derived neurons, respectively. RESULTS: Single-cell RNA sequencing analysis showed that neural fate acquisition by human and mouse enteric NC cells requires reduced expression of NC- and cell cycle-specific genes and up-regulation of neuronal or glial lineage-specific genes. Activation of the hedgehog pathway was associated with progression of mouse enteric NCs to the more mature state along the neuronal and glial lineage differentiation trajectories. Activation of the hedgehog pathway promoted development of cultured hPSCs into NCs of greater neurogenic potential by activating expression of genes in the neurogenic lineage. The hedgehog agonist increased differentiation of hPSCs into cells of the neuronal lineage by up-regulating expression of GLI2 target genes, including INSM1, NHLH1, and various bHLH family members. The hedgehog agonist increased expression of late neuronal markers and neuronal activities in hPSC-derived neurons. CONCLUSIONS: In enteric NCs from humans and mice, activation of hedgehog signaling promotes differentiation into neurons by promoting cell-state transition, expression of genes in the neurogenic lineage, and functional maturity of enteric neurons.


Subject(s)
Cell Differentiation , Hedgehog Proteins/metabolism , Induced Pluripotent Stem Cells/physiology , Neurons/physiology , Signal Transduction/physiology , Animals , Cell Line , Enteric Nervous System/cytology , Gene Expression Profiling/methods , Hedgehog Proteins/genetics , Humans , Intestinal Mucosa/cytology , Intestinal Mucosa/innervation , Male , Mice , Mice, Transgenic , Neural Crest/cytology , Sequence Analysis, RNA/methods , Single-Cell Analysis/methods
3.
Gastroenterology ; 155(6): 1908-1922.e5, 2018 12.
Article in English | MEDLINE | ID: mdl-30217742

ABSTRACT

BACKGROUND & AIMS: Hirschsprung disease, or congenital aganglionosis, is believed to be oligogenic-that is, caused by multiple genetic factors. We performed whole-genome sequence analyses of patients with Hirschsprung disease to identify genetic factors that contribute to disease development and analyzed the functional effects of these variants. METHODS: We performed whole-genome sequence analyses of 443 patients with short-segment disease, recruited from hospitals in China and Vietnam, and 493 ethnically matched individuals without Hirschsprung disease (controls). We performed genome-wide association analyses and gene-based rare-variant burden tests to identify rare and common disease-associated variants and study their interactions. We obtained induced pluripotent stem cell (iPSC) lines from 4 patients with Hirschsprung disease and 2 control individuals, and we used these to generate enteric neural crest cells for transcriptomic analyses. We assessed the neuronal lineage differentiation capability of iPSC-derived enteric neural crest cells using an in vitro differentiation assay. RESULTS: We identified 4 susceptibility loci, including 1 in the phospholipase D1 gene (PLD1) (P = 7.4 × 10-7). The patients had a significant excess of rare protein-altering variants in genes previously associated with Hirschsprung disease and in the ß-secretase 2 gene (BACE2) (P = 2.9 × 10-6). The epistatic effects of common and rare variants across these loci provided a sensitized background that increased risk for the disease. In studies of the iPSCs, we observed common and distinct pathways associated with variants in RET that affect risk. In functional assays, we found variants in BACE2 to protect enteric neurons from apoptosis. We propose that alterations in BACE1 signaling via amyloid ß precursor protein and BACE2 contribute to pathogenesis of Hirschsprung disease. CONCLUSIONS: In whole-genome sequence analyses of patients with Hirschsprung disease, we identified rare and common variants associated with disease risk. Using iPSC cells, we discovered some functional effects of these variants.


Subject(s)
Enteric Nervous System/growth & development , Hirschsprung Disease/genetics , Amyloid Precursor Protein Secretases/metabolism , Aspartic Acid Endopeptidases/metabolism , Case-Control Studies , Cell Differentiation , China , Genetic Predisposition to Disease , Genetic Variation , Humans , Induced Pluripotent Stem Cells , Neural Crest/physiology , Phospholipase D/metabolism , Proto-Oncogene Proteins c-ret/metabolism , Signal Transduction/genetics , Vietnam , Whole Genome Sequencing
4.
Gastroenterology ; 153(1): 139-153.e8, 2017 07.
Article in English | MEDLINE | ID: mdl-28342760

ABSTRACT

BACKGROUND & AIMS: Hirschsprung disease is caused by failure of enteric neural crest cells (ENCCs) to fully colonize the bowel, leading to bowel obstruction and megacolon. Heterozygous mutations in the coding region of the RET gene cause a severe form of Hirschsprung disease (total colonic aganglionosis). However, 80% of HSCR patients have short-segment Hirschsprung disease (S-HSCR), which has not been associated with genetic factors. We sought to identify mutations associated with S-HSCR, and used the clustered regularly interspaced short palindromic repeats (CRISPR)/Cas9 gene editing system to determine how mutations affect ENCC function. METHODS: We created induced pluripotent stem cell (iPSC) lines from 1 patient with total colonic aganglionosis (with the G731del mutation in RET) and from 2 patients with S-HSCR (without a RET mutation), as well as RET+/- and RET-/- iPSCs. IMR90-iPSC cells were used as the control cell line. Migration and differentiation capacities of iPSC-derived ENCCs were analyzed in differentiation and migration assays. We searched for mutation(s) associated with S-HSCR by combining genetic and transcriptome data from patient blood- and iPSC-derived ENCCs, respectively. Mutations in the iPSCs were corrected using the CRISPR/Cas9 system. RESULTS: ENCCs derived from all iPSC lines, but not control iPSCs, had defects in migration and neuronal lineage differentiation. RET mutations were associated with differentiation and migration defects of ENCCs in vitro. Genetic and transcriptome analyses associated a mutation in the vinculin gene (VCL M209L) with S-HSCR. CRISPR/Cas9 correction of the RET G731del and VCL M209L mutations in iPSCs restored the differentiation and migration capacities of ENCCs. CONCLUSIONS: We identified mutations in VCL associated with S-HSCR. Correction of this mutation in iPSC using CRISPR/Cas9 editing, as well as the RET G731del mutation that causes Hirschsprung disease with total colonic aganglionosis, restored ENCC function. Our study demonstrates how human iPSCs can be used to identify disease-associated mutations and determine how they affect cell functions and contribute to pathogenesis.


Subject(s)
Clustered Regularly Interspaced Short Palindromic Repeats , Gene Editing/methods , Hirschsprung Disease/genetics , Neural Crest/physiopathology , Proto-Oncogene Proteins c-ret/genetics , Vinculin/genetics , Cell Differentiation/genetics , Cell Line , Cell Movement/genetics , DNA Mutational Analysis/methods , Humans , Induced Pluripotent Stem Cells/physiology , Phenotype
5.
Biochim Biophys Acta ; 1852(8): 1676-86, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25981745

ABSTRACT

A germline mutation (A339V) in thyroid transcription factor-1 (TITF1/NKX2.1) was shown to be associated with multinodular goiter (MNG) and papillary thyroid carcinoma (PTC) pathogenesis. The overexpression of A339V TTF1 significantly promoted hormone-independent growth of the normal thyroid cells, representing a cause of MNG and/or PTC. Nevertheless, the underlying mechanism still remains unclear. In this study, we used liquid chromatography (LC)-tandem mass spectrometry (MS/MS)-based shotgun proteomics comparing the global protein expression profiles of normal thyroid cells (PCCL3) that overexpressed the wild-type or A339V TTF1 to identify key proteins implicated in this process. Proteomic pathway analysis revealed that the aberrant activation of epidermal growth factor (EGF) signaling is significantly associated with the overexpression of A339V TTF1 in PCCL3, and clathrin heavy chain (Chc) is the most significantly up-regulated protein of the pathway. Intriguingly, dysregulated Chc expression facilitated a nuclear accumulation of pStat3, leading to an enhanced cell proliferation of the A339V clones. Down-regulation and abrogation of Chc-mediated cellular trafficking, respectively, by knocking-down Chc and ectopic expression of a dominant-negative (DN) form of Chc could significantly reduce the nuclear pStat3 and rescue the aberrant cell proliferation of the A339V clones. Subsequent expression analysis further revealed that CHC and pSTAT3 are co-overexpressed in 66.7% (10/15) MNG. Taken together, our results suggest that the A339V TTF1 mutant protein up-regulates the cellular expression of Chc, resulting in a constitutive activation of Stat3 pathway, and prompting the aberrant growth of thyroid cells. This extensive growth signal may promote the development of MNG.


Subject(s)
Cell Proliferation , Clathrin Heavy Chains/genetics , Clathrin Heavy Chains/metabolism , Goiter, Nodular/pathology , Thyroid Gland/cytology , Thyroid Gland/pathology , Adolescent , Adult , Aged , Aged, 80 and over , Animals , COS Cells , Carcinoma/genetics , Carcinoma/metabolism , Carcinoma/pathology , Carcinoma, Papillary , Cell Proliferation/genetics , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Cells, Cultured , Child , Chlorocebus aethiops , Female , Gene Expression Regulation, Neoplastic , Goiter, Nodular/genetics , Goiter, Nodular/metabolism , Humans , Male , Middle Aged , Thyroid Cancer, Papillary , Thyroid Neoplasms/genetics , Thyroid Neoplasms/metabolism , Thyroid Neoplasms/pathology , Young Adult
6.
Gastroenterology ; 149(7): 1837-1848.e5, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26261006

ABSTRACT

BACKGROUND & AIMS: Hirschsprung disease is characterized by a deficit in enteric neurons, which are derived from neural crest cells (NCCs). Aberrant hedgehog signaling disrupts NCC differentiation and might cause Hirschsprung disease. We performed genetic analyses to determine whether hedgehog signaling is involved in pathogenesis. METHODS: We performed deep-target sequencing of DNA from 20 patients with Hirschsprung disease (16 men, 4 women), and 20 individuals without (controls), and searched for mutation(s) in GLI1, GLI2, GLI3, SUFU, and SOX10. Biological effects of GLI mutations were tested in luciferase reporter assays using HeLa or neuroblastoma cell lines. Development of the enteric nervous system was studied in Sufu(f/f), Gli3(Δ699), Wnt1-Cre, and Sox10(NGFP) mice using immunohistochemical and whole-mount staining procedures to quantify enteric neurons and glia and analyze axon fasciculation, respectively. NCC migration was studied using time-lapse imaging. RESULTS: We identified 3 mutations in GLI in 5 patients with Hirschsprung disease but no controls; all lead to increased transcription of SOX10 in cell lines. SUFU, GLI, and SOX10 form a regulatory loop that controls the neuronal vs glial lineages and migration of NCCs. Sufu mutants mice had high Gli activity, due to loss of Sufu, disrupting the regulatory loop and migration of enteric NCCs, leading to defective axonal fasciculation, delayed gut colonization, or intestinal hypoganglionosis. The ratio of enteric neurons to glia correlated inversely with Gli activity. CONCLUSIONS: We identified mutations that increase GLI activity in patients with Hirschsprung disease. Disruption of the SUFU-GLI-SOX10 regulatory loop disrupts migration of NCCs and development of the enteric nervous system in mice.


Subject(s)
Enteric Nervous System/abnormalities , Hirschsprung Disease/genetics , Hirschsprung Disease/pathology , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Mutation , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neural Crest/pathology , Nuclear Proteins/genetics , Transcription Factors/genetics , Animals , Case-Control Studies , Cell Lineage , Cell Movement , DNA Mutational Analysis/methods , Disease Models, Animal , Enteric Nervous System/metabolism , Female , Gene Expression Regulation, Developmental , Genetic Predisposition to Disease , HeLa Cells , High-Throughput Nucleotide Sequencing , Hirschsprung Disease/diagnosis , Hirschsprung Disease/metabolism , Humans , Male , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Inbred ICR , Mice, Transgenic , Neural Crest/metabolism , Neurogenesis , Nuclear Proteins/metabolism , Phenotype , Repressor Proteins/genetics , Repressor Proteins/metabolism , SOXE Transcription Factors/genetics , SOXE Transcription Factors/metabolism , Transcription Factors/metabolism , Transfection , Wnt1 Protein/genetics , Wnt1 Protein/metabolism , Zinc Finger Protein GLI1 , Zinc Finger Protein Gli2 , Zinc Finger Protein Gli3
7.
Hum Mol Genet ; 22(3): 621-31, 2013 Feb 01.
Article in English | MEDLINE | ID: mdl-23108157

ABSTRACT

Anorectal malformations (ARMs, congenital obstruction of the anal opening) are among the most common birth defects requiring surgical treatment (2-5/10 000 live-births) and carry significant chronic morbidity. ARMs present either as isolated or as part of the phenotypic spectrum of some chromosomal abnormalities or monogenic syndromes. The etiology is unknown. To assess the genetic contribution to ARMs, we investigated single-nucleotide polymorphisms and copy number variations (CNVs) at genome-wide scale. A total of 363 Han Chinese sporadic ARM patients and 4006 Han Chinese controls were included. Overall, we detected a 1.3-fold significant excess of rare CNVs in patients. Stratification of patients by presence/absence of other congenital anomalies showed that while syndromic ARM patients carried significantly longer rare duplications than controls (P = 0.049), non-syndromic patients were enriched with both rare deletions and duplications when compared with controls (P = 0.00031). Twelve chromosomal aberrations and 114 rare CNVs were observed in patients but not in 868 controls nor 11 943 healthy individuals from the Database of Genomic Variants. Importantly, these aberrations were observed in isolated ARM patients. Gene-based analysis revealed 79 genes interfered by CNVs in patients only. In particular, we identified a de novo DKK4 duplication. DKK4 is a member of the WNT signaling pathway which is involved in the development of the anorectal region. In mice, Wnt disruption results in ARMs. Our data suggest a role for rare CNVs not only in syndromic but also in isolated ARM patients and provide a list of plausible candidate genes for the disorder.


Subject(s)
Anus, Imperforate/genetics , Anus, Imperforate/physiopathology , DNA Copy Number Variations , Gene Duplication , Intercellular Signaling Peptides and Proteins/genetics , Animals , Anorectal Malformations , Asian People , Chromosome Aberrations , Female , Gene Dosage , Gene Expression Regulation , Genome-Wide Association Study , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Male , Mice , Mice, Inbred ICR , Phenotype , Polymorphism, Single Nucleotide , Prospective Studies , Wnt Signaling Pathway
8.
PLoS Genet ; 8(5): e1002687, 2012.
Article in English | MEDLINE | ID: mdl-22589734

ABSTRACT

Hirschsprung disease (HSCR) is a congenital disorder characterized by aganglionosis of the distal intestine. To assess the contribution of copy number variants (CNVs) to HSCR, we analysed the data generated from our previous genome-wide association study on HSCR patients, whereby we identified NRG1 as a new HSCR susceptibility locus. Analysis of 129 Chinese patients and 331 ethnically matched controls showed that HSCR patients have a greater burden of rare CNVs (p = 1.50 × 10(-5)), particularly for those encompassing genes (p = 5.00 × 10(-6)). Our study identified 246 rare-genic CNVs exclusive to patients. Among those, we detected a NRG3 deletion (p = 1.64 × 10(-3)). Subsequent follow-up (96 additional patients and 220 controls) on NRG3 revealed 9 deletions (combined p = 3.36 × 10(-5)) and 2 de novo duplications among patients and two deletions among controls. Importantly, NRG3 is a paralog of NRG1. Stratification of patients by presence/absence of HSCR-associated syndromes showed that while syndromic-HSCR patients carried significantly longer CNVs than the non-syndromic or controls (p = 1.50 × 10(-5)), non-syndromic patients were enriched in CNV number when compared to controls (p = 4.00 × 10(-6)) or the syndromic counterpart. Our results suggest a role for NRG3 in HSCR etiology and provide insights into the relative contribution of structural variants in both syndromic and non-syndromic HSCR. This would be the first genome-wide catalog of copy number variants identified in HSCR.


Subject(s)
DNA Copy Number Variations/genetics , Gene Deletion , Hirschsprung Disease/genetics , Neuregulins/genetics , Female , Gene Frequency , Genome-Wide Association Study , Genotype , Haplotypes , Humans , Male , Polymorphism, Single Nucleotide
9.
Neurosignals ; 22(1): 1-13, 2014.
Article in English | MEDLINE | ID: mdl-24356576

ABSTRACT

The enteric nervous system (ENS) in mammals is derived from a small pool of progenitor cells, namely enteric neural crest cells (NCCs). These precursor cells proliferate extensively to expand, migrate over a long distance to fully colonize the developing gut and differentiate into millions of neurons and glia to form a functional ENS for regulating the complex behaviors of the gut. This developmental process relies on a precise regulation of the neuronal and glial differentiation and requires an appropriate balance between the migration, proliferation and differentiation of enteric NCCs and their progeny. Hedgehog (Hh) and Notch signalings are essential for almost every aspect of ENS development, and they confer both the long- and short-range signals to coordinate these seemingly diverse cellular processes. In this review, we summarize the roles of Hh and Notch signaling, particularly in the context of gut organogenesis and ENS development and emphasize how combinatory Hh and Notch signaling renders functional diversity as well as specificity.


Subject(s)
Enteric Nervous System/embryology , Enteric Nervous System/metabolism , Hedgehog Proteins/metabolism , Neural Crest/metabolism , Receptors, Notch/metabolism , Signal Transduction , Animals , Gastrointestinal Tract/embryology , Gastrointestinal Tract/metabolism , Humans
10.
Exp Cell Res ; 319(2): 177-84, 2013 Jan 15.
Article in English | MEDLINE | ID: mdl-23159386

ABSTRACT

The availability of human pluriopotent stem cells, embryonic (ESC) and induced pluriopotent (iPSC) stem cells, not only can be a renewable source for investigating the early human development, etiology and progression of different diseases but also recapitulating the disease with the same genomic materials of the patient. In particular, specific neuronal subtypes generated from the patient ESC/iPSCs has become a source for studying disease mechanisms underlying different neurological disorders and allowed drug discovery. In this review, we summarize the recent advances in establishing patient ESC/iPSC to model various neurological diseases. We will also discuss the challenges and limitations of the current disease models and their potential future applications for untangling the unknowns in neurological disorders.


Subject(s)
Models, Biological , Nervous System Diseases/pathology , Pluripotent Stem Cells/pathology , Animals , Cell Differentiation , Humans
11.
J Hepatol ; 59(6): 1285-91, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23872602

ABSTRACT

BACKGROUND & AIMS: Biliary atresia (BA) is a rare and most severe cholestatic disease in neonates, but the pathogenic mechanisms are unknown. Through a previous genome wide association study (GWAS) on Han Chinese, we discovered association of the 10q24.2 region encompassing ADD3 and XPNPEP1 genes, which was replicated in Chinese and Thai populations. This study aims to fully characterize the genetic architecture at 10q24.2 and to reveal the link between the genetic variants and BA. METHODS: We genotyped 107 single nucleotide polymorphisms (SNPs) in 10q24.2 in 339 Han Chinese patients and 401 matched controls using Sequenom. Exhaustive follow-up studies of the association signals were performed. RESULTS: The combined BA-association p-value of the GWAS SNP (rs17095355) achieved 6.06×10(-10). Further, we revealed the common risk haplotype encompassing 5 tagging-SNPs, capturing the risk-predisposing alleles in 10q24.2 [p=5.32×10(-11); odds ratio, OR: 2.38; confidence interval, CI: (2.14-2.62)]. Through Sanger sequencing, no deleterious rare variants (RVs) residing in the risk haplotype were found, dismissing the theory of "synthetic" association. Moreover, in bioinformatics and in vivo genotype-expression investigations, the BA-associated potentially regulatory SNPs correlated with ADD3 gene expression (n=36; p=0.0030). Remarkably, the risk haplotype frequency coincides with BA incidences in the population, and, positive selection (favoring the derived alleles that arose from mutations) was evident at the ADD3 locus, suggesting a possible role for the BA-associated common variants in shaping the general population diversity. CONCLUSIONS: Common genetic variants in 10q24.2 can alter BA risk by regulating ADD3 expression levels in the liver, and may exert an effect on disease epidemiology and on the general population.


Subject(s)
Biliary Atresia/genetics , Calmodulin-Binding Proteins/genetics , Genetic Predisposition to Disease , Polymorphism, Single Nucleotide , Biliary Atresia/etiology , Female , Genotype , Haplotypes , Humans , Infant , Male , Risk
12.
Hum Genet ; 132(5): 591-600, 2013 May.
Article in English | MEDLINE | ID: mdl-23400839

ABSTRACT

Hirschsprung disease (HSCR, aganglionic megacolon) is a complex genetic disorder of the enteric nervous system (ENS) characterized by the absence of enteric neurons along a variable length of the intestine. While rare variants (RVs) in the coding sequence (CDS) of several genes involved in ENS development lead to disease, the association of common variants (CVs) with HSCR has only been reported for RET (the major HSCR gene) and NRG1. Importantly, RVs in the CDS of these two genes are also associated with the disorder. To assess independent and joint effects between the different types of RET and NRG1 variants identified in HSCR patients, we used 254 Chinese sporadic HSCR patients and 143 ethnically matched controls for whom the RET and/or NRG1 variants genotypes (rare and common) were available. Four genetic risk factors were defined and interaction effects were modeled using conditional logistic regression analyses and pair-wise Kendall correlations. Our analysis revealed a joint effect of RET CVs with RET RVs, NRG1 CVs or NRG1 RVs. To assess whether the genetic interaction translated into functional interaction, mouse neural crest cells (NCCs; enteric neuron precursors) isolated from embryonic guts were treated with NRG1 (ErbB2 ligand) or/and GDNF (Ret ligand) and monitored during the subsequent neural differentiation process. Nrg1 inhibited the Gdnf-induced neuronal differentiation and Gdnf negatively regulated Nrg1-signaling by down-regulating the expression of its receptor, ErbB2. This preliminary data suggest that the balance neurogenesis/gliogenesis is critical for ENS development.


Subject(s)
Genetic Variation/genetics , Hirschsprung Disease/genetics , Neuregulin-1/genetics , Proto-Oncogene Proteins c-ret/genetics , Animals , Case-Control Studies , Cells, Cultured , China , Female , Genomics , Genotype , Glial Cell Line-Derived Neurotrophic Factor/genetics , Glial Cell Line-Derived Neurotrophic Factor/metabolism , HapMap Project , Hirschsprung Disease/metabolism , Humans , Intestines/cytology , Intestines/innervation , Linkage Disequilibrium , Male , Mice , Mutation , Neural Crest/cytology , Neuregulin-1/metabolism , Phenotype , Proto-Oncogene Proteins c-ret/metabolism , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism , Risk Factors , Transgenes
13.
J Pathol ; 226(4): 645-53, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22072235

ABSTRACT

Caveolin-1 (Cav1) has been implicated in diverse human cancers, yet its role in hepatocellular carcinoma (HCC) tumourigenesis and metastasis remains elusive. In the current study, we aim to provide a comprehensive understanding regarding the functional role of Cav1 in HCC tumourigenesis and metastasis. Cav1 expression was examined in a panel of human HCC cell lines using western blotting analysis and quantitative RT-PCR and human tissues by immunohistochemistry. Cav1 was not detected in normal liver cell line and all non-tumourous liver tissues but exclusively expressed in HCC cell lines and tissues. Dramatic expression of Cav1 was found in metastatic HCC cell lines and tumours, indicating a progressive increase of Cav1 expression along disease progression. Cav1 overexpression was significantly correlated with venous invasion (p = 0.036). To investigate the functions of Cav1 in HCC, Cav1 overexpressing and knockdown stable clones were established in HCC cells and their tumourigenicity and metastatic potential were examined. Overexpression of Cav1 promoted HCC cell growth, motility, and invasiveness, as well as tumourigenicity in vivo. Conversely, knockdown of Cav1 in metastatic HCC cells inhibited the motility and invasiveness and markedly suppressed the tumour growth and metastatic potential in vivo. Collectively, our findings have shown the exclusive expression of Cav1 in HCC cell lines and clinical samples and revealed an up-regulation of Cav1 along HCC progression. The definitive role of Cav1 in promoting HCC tumourigenesis was demonstrated, and we have shown for the first time in a mouse model that Cav1 promotes HCC metastasis.


Subject(s)
Carcinoma, Hepatocellular/secondary , Caveolin 1/metabolism , Liver Neoplasms/pathology , Adolescent , Adult , Aged , Animals , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/mortality , Caveolin 1/genetics , Cell Line, Tumor , Cell Movement , Cell Proliferation , Clone Cells , Disease Models, Animal , Disease Progression , Female , Gene Knockdown Techniques , Gene Silencing , Hong Kong/epidemiology , Humans , Liver/metabolism , Liver/pathology , Liver Neoplasms/genetics , Liver Neoplasms/mortality , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Neoplasm Transplantation , Survival Rate , Up-Regulation , Young Adult
14.
Stem Cell Reports ; 18(7): 1500-1515, 2023 07 11.
Article in English | MEDLINE | ID: mdl-37352849

ABSTRACT

Retrotrapezoid nucleus (RTN) neurons in the brainstem regulate the ventilatory response to hypercarbia. It is unclear how PHOX2B-polyalanine repeat mutations (PHOX2B-PARMs) alter the function of PHOX2B and perturb the formation of RTN neurons. Here, we generated human brainstem organoids (HBSOs) with RTN-like neurons from human pluripotent stem cells. Single-cell transcriptomics revealed that expression of PHOX2B+7Ala PARM alters the differentiation trajectories of the hindbrain neurons and hampers the formation of the RTN-like neurons in HBSOs. With the unguided cerebral organoids (HCOs), PHOX2B+7Ala PARM interrupted the patterning of PHOX2B+ neurons with dysregulation of Hedgehog pathway and HOX genes. With complementary use of HBSOs and HCOs with a patient and two mutant induced pluripotent stem cell lines carrying different polyalanine repetition in PHOX2B, we further defined the association between the length of polyalanine repetition and malformation of RTN-respiratory center and demonstrated the potential toxic gain of function of PHOX2B-PARMs, highlighting the uniqueness of these organoid models for disease modeling.


Subject(s)
Hedgehog Proteins , Homeodomain Proteins , Humans , Homeodomain Proteins/metabolism , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Transcription Factors/metabolism , Rhombencephalon/metabolism , Neurons/metabolism , Mutation
15.
Nat Commun ; 14(1): 2157, 2023 04 15.
Article in English | MEDLINE | ID: mdl-37061531

ABSTRACT

Hirschsprung disease is characterized by the absence of enteric neurons caused by the defects of enteric neural crest cells, leading to intestinal obstruction. Here, using induced pluripotent stem cell-based models of Hirschsprung and single-cell transcriptomic analysis, we identify a gene set of 118 genes commonly dysregulated in all patient enteric neural crest cells, and suggest HDAC1 may be a key regulator of these genes. Furthermore, upregulation of RNA splicing mediators and enhanced alternative splicing events are associated with severe form of Hirschsprung. In particular, the higher inclusion rate of exon 9 in PTBP1 and the perturbed expression of a PTBP1-target, PKM, are significantly enriched in these patient cells, and associated with the defective oxidative phosphorylation and impaired neurogenesis. Hedgehog-induced oxidative phosphorylation significantly enhances the survival and differentiation capacity of patient cells. In sum, we define various factors associated with Hirschsprung pathogenesis and demonstrate the implications of oxidative phosphorylation in enteric neural crest development and HSCR pathogenesis.


Subject(s)
Enteric Nervous System , Hirschsprung Disease , Humans , Hirschsprung Disease/genetics , Hirschsprung Disease/metabolism , Neural Crest/metabolism , Transcriptome , Oxidative Phosphorylation , Heterogeneous-Nuclear Ribonucleoproteins/metabolism , Polypyrimidine Tract-Binding Protein/genetics
16.
Hum Mol Genet ; 19(8): 1461-7, 2010 Apr 15.
Article in English | MEDLINE | ID: mdl-20089534

ABSTRACT

Receptor tyrosine kinase (RET) single nucleotide polymorphisms (SNPs) are associated with the Hirschsprung's disease (HSCR). We investigated whether the amount of RET expressed in the ganglionic gut of human was dependent on the genotype of three regulatory SNPs (-5G>A rs10900296 and -1A>C rs10900297 in the promoter, and C>T rs2435357 in intron 1). We examined the effects of three regulatory SNPs on the RET gene expression in 67 human ganglionic gut tissues using quantitative real-time PCR. Also, 315 Chinese HSCR patients and 325 ethnically matched controls were genotyped for the three SNPs by polymerase chain reaction (PCR) and direct sequencing. The expression of RET mRNA in human gut tissue did indeed correlate with the genotypes of the individuals. The lowest RET expression was found for those individuals homozygous for the three risk alleles (A-C-T/A-C-T), and the highest for those homozygous for the 'wild-type' counterpart (G-A-C/G-A-C), with expression values ranging from 218.32 +/- 125.69 (mean +/- SE) in tissues from individuals carrying G-A-C/G-A-C to 31.42 +/- 8.42 for individuals carrying A-C-T/A-C-T (P = 0.018). As expected, alleles -5A, -1C and intron 1 T were associated with HSCR (P = 5.94 x 10(-31), 3.12 x 10(-24) and 5.94 x 10(-37), respectively) as was the haplotype encompassing the three associated alleles (A-C-T) when compared with the wild-type counterpart G-A-C (chi2 = 155.29, P << 0.0001). To our knowledge, this is the first RET expression genotype-phenotype correlation study conducted on human subjects to indicate common genetic variants in the regulatory region of RET may play a role in mediating susceptibility to HSCR, by conferring a significant reduction of the RET expression.


Subject(s)
Down-Regulation , Genetic Predisposition to Disease , Hirschsprung Disease/genetics , Intestine, Large/enzymology , Receptor Protein-Tyrosine Kinases/genetics , Alleles , Asian People/genetics , Case-Control Studies , Female , Genotype , Hirschsprung Disease/enzymology , Humans , Male , Polymorphism, Single Nucleotide , Receptor Protein-Tyrosine Kinases/metabolism
17.
Hum Mol Genet ; 19(14): 2917-25, 2010 Jul 15.
Article in English | MEDLINE | ID: mdl-20460270

ABSTRACT

Biliary atresia (BA) is characterized by the progressive fibrosclerosing obliteration of the extrahepatic biliary system during the first few weeks of life. Despite early diagnosis and prompt surgical intervention, the disease progresses to cirrhosis in many patients. The current theory for the pathogenesis of BA proposes that during the perinatal period, a still unknown exogenous factor meets the innate immune system of a genetically predisposed individual and induces an uncontrollable and potentially self-limiting immune response, which becomes manifest in liver fibrosis and atresia of the extrahepatic bile ducts. Genetic factors that could account for the disease, let alone for its high incidence in Chinese, are to be investigated. To identify BA susceptibility loci, we carried out a genome-wide association study (GWAS) using the Affymetrix 5.0 and 500 K marker sets. We genotyped nearly 500 000 single-nucleotide polymorphisms (SNPs) in 200 Chinese BA patients and 481 ethnically matched control subjects. The 10 most BA-associated SNPs from the GWAS were genotyped in an independent set of 124 BA and 90 control subjects. The strongest overall association was found for rs17095355 on 10q24, downstream XPNPEP1, a gene involved in the metabolism of inflammatory mediators. Allelic chi-square test P-value for the meta-analysis of the GWAS and replication results was 6.94 x 10(-9). The identification of putative BA susceptibility loci not only opens new fields of investigation into the mechanisms underlying BA but may also provide new clues for the development of preventive and curative strategies.


Subject(s)
Biliary Atresia/genetics , Chromosomes, Human, Pair 10 , Genetic Loci , Genetic Predisposition to Disease , Asian People/genetics , Case-Control Studies , Chromosome Mapping , Chromosomes, Human, Pair 10/genetics , Female , Gene Frequency , Genome-Wide Association Study , Humans , Linkage Disequilibrium , Male , Polymorphism, Single Nucleotide
18.
Hum Genet ; 131(1): 67-76, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21706185

ABSTRACT

Hirschsprung disease (HSCR, congenital colon aganglionosis) is a relatively common complex genetic condition caused by abnormal development of the enteric nervous system (ENS). Through a recent genome-wide association study conducted on Chinese HSCR patients, we identified a new HSCR contributing locus, neuregulin 1 (NRG1; 8p12), a gene known to be involved in the development of the ENS. As genes in which disease-associated common variants are found are to be considered as candidates for the search of deleterious rare variants (RVs) in the coding sequences, we sequenced the NRG1 exons of 358 sporadic HSCR patients and 333 controls. We identified a total of 13 different heterozygous RVs including 8 non-synonymous (A28G, E134K, V266L, H347Y, P356L, V486M, A511T, P608A) and 3 synonymous amino acid substitutions (P24P, T169T, L483L), a frameshift (E239fsX10), and a c.503-4insT insertion. Functional analysis of the most conserved non-synonymous substitutions, H347Y and P356L, showed uneven intracellular distribution and aberrant expression of the mutant proteins. Except for T169T and V486M, all variants were exclusive to HSCR patients. Overall, there was a statistically significant over-representation of NRG1 RVs in HSCR patients (p = 0.008). We show here that not only common, but also rare variants of the NRG1 gene contribute to HSCR. This strengthens the role of NRG1.


Subject(s)
Hirschsprung Disease/genetics , Mutation/genetics , Neuregulin-1/genetics , Animals , COS Cells , Case-Control Studies , Chlorocebus aethiops , DNA Mutational Analysis , Female , Genome-Wide Association Study , Genotype , Humans , Immunoblotting , Immunoenzyme Techniques , Male
19.
Proc Natl Acad Sci U S A ; 106(8): 2694-9, 2009 Feb 24.
Article in English | MEDLINE | ID: mdl-19196962

ABSTRACT

Hirschsprung's disease (HSCR), or aganglionic megacolon, is a congenital disorder characterized by the absence of enteric ganglia in variable portions of the distal intestine. RET is a well-established susceptibility locus, although existing evidence strongly suggests additional loci contributing to sporadic HSCR. To identify these additional genetic loci, we carried out a genome-wide association study using the Affymetrix 500K marker set. We successfully genotyped 293,836 SNPs in 181 Chinese subjects with sporadic HSCR and 346 ethnically matched control subjects. The SNPs most associated with HSCR were genotyped in an independent set of 190 HSCR and 510 control subjects. Aside from SNPs in RET, the strongest overall associations in plausible candidate genes were found for 2 SNPs located in intron 1 of the neuregulin1 gene (NRG1) on 8p12, with rs16879552 and rs7835688 yielding odds ratios of 1.68 [CI(95%):(1.40, 2.00), P = 1.80 x 10(-8)] and 1.98 [CI(95%):(1.59, 2.47), P = 1.12 x 10(-9)], respectively, for the heterozygous risk genotypes under an additive model. There was also a significant interaction between RET and NRG1 (P = 0.0095), increasing the odds ratio 2.3-fold to 19.53 for the RET rs2435357 risk genotype (TT) in the presence of the NRG1 rs7835688 heterozygote, indicating that NRG1 is a modifier of HSRC penetrance. Our highly significant association findings are backed-up by the important role of NRG1 as regulator of the development of the enteric ganglia precursors. The identification of NRG1 as an additional HSCR susceptibility locus not only opens unique fields of investigation into the mechanisms underlying the HSCR pathology, but also the mechanisms by which a discrete number of loci interact with each other to cause disease.


Subject(s)
Genetic Predisposition to Disease , Genome, Human , Hirschsprung Disease/genetics , Nerve Tissue Proteins/genetics , Female , Genetic Markers , Genome-Wide Association Study , Humans , Male , Neuregulin-1 , Polymorphism, Single Nucleotide , Proto-Oncogene Proteins c-ret/genetics
20.
Comput Struct Biotechnol J ; 20: 2464-2472, 2022.
Article in English | MEDLINE | ID: mdl-35664232

ABSTRACT

With the rapid development of single-cell sequencing technologies, it has become a powerful strategy for the discovery of rare cells and delineating the molecular basis underlying various biological processes. Use of single-cell multimodal sequencing to explore the chromatin accessibility, gene expression and spatial transcriptome has propelled us to success in untangling the unknowns in the enteric nervous system (ENS) and provided unprecedented resources for building new diagnostic framework for enteric neuropathies. Here, we summarize the recent findings of single-cell multimodal sequencing, especially focusing on the most commonly used single-cell RNA sequencing (scRNA-seq) on ENS cells, ranged from the progenitors, neural crest (NC) cells, to the mature ENS circuit, in both human and mouse. These studies have highlighted the heterogeneity of ENS cells at various developmental stages and discovered numerous novel cell types. We will also discuss various computational methods that were used to reconstruct the differentiation trajectories of the developing ENS and to elucidate the cell fate decisions. Profiling disease mechanisms and cellular drug responses with single-cell multimodal omics techniques likely leads to a paradigm shift in the field of biomedical research. Further improvements in the high-resolution sequencing platforms and integrative computational tools will greatly hasten their applications in both the basic and translational medicine.

SELECTION OF CITATIONS
SEARCH DETAIL