Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
1.
Nucleic Acids Res ; 50(6): e34, 2022 04 08.
Article in English | MEDLINE | ID: mdl-34931221

ABSTRACT

Identifying rare variants that contribute to complex diseases is challenging because of the low statistical power in current tests comparing cases with controls. Here, we propose a novel and powerful rare variants association test based on the deviation of the observed mutation burden of a gene in cases from a baseline predicted by a weighted recursive truncated negative-binomial regression (RUNNER) on genomic features available from public data. Simulation studies show that RUNNER is substantially more powerful than state-of-the-art rare variant association tests and has reasonable type 1 error rates even for stratified populations or in small samples. Applied to real case-control data, RUNNER recapitulates known genes of Hirschsprung disease and Alzheimer's disease missed by current methods and detects promising new candidate genes for both disorders. In a case-only study, RUNNER successfully detected a known causal gene of amyotrophic lateral sclerosis. The present study provides a powerful and robust method to identify susceptibility genes with rare risk variants for complex diseases.


Subject(s)
Genetic Predisposition to Disease , Genetic Variation , Models, Genetic , Software , Case-Control Studies , Computer Simulation , Humans , Mutation
2.
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
3.
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
4.
World J Surg ; 39(10): 2484-91, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26071011

ABSTRACT

BACKGROUND: Bilateral pheochromocytoma (PHEO) is more frequently found in patients with multiple endocrine neoplasia 2A carrying a RET germline mutation located in codon 634 (C634). However, it is unclear whether different amino acid substitutions within C634 cause differences in bilateral PHEOs expression. We aimed to answer this by pooling data from two Asian institutions. METHODS: Sixty-seven patients had confirmed C634 germline mutation. Age-dependent penetrance of bilateral PHEO was calculated from date of birth to the date when bilateral PHEO was first diagnosed or when the contralateral gland became a PHEO (if the patient already had one adrenal gland removed). Age-dependent penetrance was estimated by the Kaplan-Meier method and compared by log-rank test. RESULTS: The 4 different amino acid substitutions included C634R (arginine) (n = 19, 28.4 %), C634Y (tyrosine) (n = 36, 38.8 %), C634G (glycine) (n = 4, 6.0 %), and C634W (tryptophan) (n = 8, 11.9 %). The age-related penetrance of PHEO was similar between C634R, C634Y, C634G, and C634W (by age 40, 69.8, 55.2, 25.0, and 56.2 %, respectively) (p = 0.529). However, the age-related penetrance of bilateral PHEO in C634R was significantly higher than C634Y (by age of 40, 59.3 % vs. 25.2 %, p = 0.046) or C634Y, C634G, and C634W combined (59.3 % vs. 21.5 %, p = 0.024). Nevertheless, the accumulative risk of bilateral PHEOs across all four C634 mutations almost approached 100 % over time. CONCLUSION: The accumulative risk of bilateral PHEOs almost reached 100 % but its onset was significantly earlier in C634R mutation. These findings implied that those with C634R mutation might benefit from earlier screening of contralateral PHEO than other C634 mutations after an unilateral adrenalectomy.


Subject(s)
Adrenal Gland Neoplasms/genetics , Multiple Endocrine Neoplasia Type 2a/genetics , Penetrance , Pheochromocytoma/genetics , Proto-Oncogene Proteins c-ret/genetics , Adolescent , Adrenal Gland Neoplasms/surgery , Adrenalectomy , Adult , Age Factors , Aged , Aged, 80 and over , Amino Acid Substitution/genetics , Arginine , Child , Child, Preschool , Codon , Female , Germ-Line Mutation , Glycine , Humans , Male , Middle Aged , Pheochromocytoma/surgery , Tryptophan , Tyrosine , Young Adult
5.
J Pediatr Surg ; 56(2): 352-359, 2021 Feb.
Article in English | MEDLINE | ID: mdl-32709532

ABSTRACT

PURPOSE: Biliary atresia (BA) is a devastating obstructive bile duct disease of newborns. BA has the highest incidence in Asians (1/5000), and its pathogenesis is unclear. We identified BA-private rare copy number variants (CNVs; 22 duplications and 6 deletions). ILF2 gene locates in the chromosome region (Chr1:153410347-153,634,058) which was deleted in a nonsyndromic BA patient. However, it is still not known whether ILF2 plays a role in hepatobiliary development and its deletion impacts on the bile duct development. METHODS: To investigate if ILF2 is required for biliary development, we knock-out the zebrafish homologs of ILF2 by CRISPR/Cas9 approach, and discover that deletion of ILF2 causes a defective biliary development and a lack of bile flow from the liver to the gall bladder in zebrafish, which is a resemblance of phenotypes of BA. RESULTS: Our data indicate that ILF2 gene is required for biliary development; deletion of ILF2 impairs bile duct development and could contribute to BA pathogenesis. This will be the first study to functionally evaluate the genes interfered by BA-private CNVs in hepatobiliary development and in BA pathogenesis. CONCLUSIONS: Such functional study may reveal the potential value of these BA-private CNVs in the disease pathogenesis for BA. LEVEL OF EVIDENCE: N/A (animal and laboratory study).


Subject(s)
Biliary Atresia , Zebrafish , Animals , Bile , Bile Ducts , Biliary Atresia/genetics , Humans , Infant, Newborn , Nuclear Factor 45 Protein , Zebrafish/genetics
6.
Gastroenterology ; 134(4): 1104-15, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18395091

ABSTRACT

BACKGROUND & AIMS: The enteric nervous system (ENS) controls intestinal peristalsis, and defective development of this system results in hypo/aganglionosis, as seen in Hirschsprung's disease. In the embryo, vagal neural crest cells (NCC) migrate and colonize the intestine rostrocaudally then differentiate into the ganglia of the ENS. Vagal NCC express the homeobox gene Hoxb5, a transcriptional activator, in human and mouse, so we used transgenic mice to investigate the function of Hoxb5 and the receptor tyrosine kinase gene Ret, which is affected in many patients with Hirschsprung's disease, in ENS development. METHODS: We perturbed the Hoxb5 pathway by expressing a chimeric protein enb5, in which the transcription activation domain of Hoxb5 was replaced with the repressor domain of the Drosophila engrailed protein (en), in vagal NCC. This enb5 transcriptional repressor competes with wild-type Hoxb5 for binding to target genes, exerting a dominant negative effect. RESULTS: We observed that 30.6% +/- 2.3% of NCC expressed enb5 and that these enb5-expressing NCC failed to migrate to the distal intestine. A 34%-37% reduction of ganglia (hypoganglionosis) and slow peristalsis and, occasionally, absence of ganglia and intestinal obstruction were observed in enb5-expressing mice. Ret expression was markedly reduced or absent in NCC and ganglia, and enb5 blocked Hoxb5 induction of Ret in neuroblastoma cells. CONCLUSIONS: Our data indicate that Ret is a downstream target of Hoxb5 whose perturbation causes Ret haploinsufficiency, impaired NCC migration, and hypo/aganglionosis, suggesting that Hoxb5 may contribute to the etiology of Hirschsprung's disease.


Subject(s)
DNA/genetics , Down-Regulation , Homeodomain Proteins/genetics , Intestines/innervation , Neural Crest/metabolism , Proto-Oncogene Proteins c-ret/genetics , Vagus Nerve/metabolism , Animals , Disease Models, Animal , Enteric Nervous System/abnormalities , Enteric Nervous System/metabolism , Female , Gene Expression Regulation, Developmental , Hirschsprung Disease/embryology , Hirschsprung Disease/genetics , Hirschsprung Disease/metabolism , Homeodomain Proteins/biosynthesis , Intestines/physiopathology , Male , Mice , Mice, Transgenic , Neural Crest/abnormalities , Neural Crest/embryology , Peristalsis/physiology , Proto-Oncogene Proteins c-ret/metabolism , Signal Transduction/physiology , Vagus Nerve/abnormalities , Vagus Nerve/embryology
7.
PLoS One ; 6(6): e20815, 2011.
Article in English | MEDLINE | ID: mdl-21677782

ABSTRACT

The enteric nervous system (ENS) regulates peristaltic movement of the gut, and abnormal ENS causes Hirschsprung's disease (HSCR) in newborns. HSCR is a congenital complex genetic disorder characterised by a lack of enteric ganglia along a variable length of the intestine. The receptor tyrosine kinase gene (RET) is the major HSCR gene and its expression is crucial for ENS development. We have previously reported that (i) HOXB5 transcription factor mediates RET expression, and (ii) mouse with defective HOXB5 activity develop HSCR phenotype. In this study, we (i) elucidate the underlying mechanisms that HOXB5 mediate RET expression, and (ii) examine the interactions between HOXB5 and other transcription factors implicated in RET expression. We show that human HOXB5 binds to the promoter region 5' upstream of the binding site of NKX2-1 and regulates RET expression. HOXB5 and NKX2-1 form a protein complex and mediate RET expression in a synergistic manner. HSCR associated SNPs at the NKX2-1 binding site (-5G>A rs10900296; -1A>C rs10900297), which reduce NKX2-1 binding, abolish the synergistic trans-activation of RET by HOXB5 and NKX2-1. In contrast to the synergistic activation of RET with NKX2-1, HOXB5 cooperates in an additive manner with SOX10, PAX3 and PHOX2B in trans-activation of RET promoter. Taken together, our data suggests that HOXB5 in coordination with other transcription factors mediates RET expression. Therefore, defects in cis- or trans-regulation of RET by HOXB5 could lead to reduction of RET expression and contribute to the manifestation of the HSCR phenotype.


Subject(s)
Homeodomain Proteins/metabolism , Nuclear Proteins/metabolism , Proto-Oncogene Proteins c-ret/metabolism , Transcription Factors/metabolism , Transcription, Genetic , Animals , Base Sequence , Binding Sites , Cell Line, Tumor , Enteric Nervous System/physiology , Gene Expression Regulation, Developmental , Hirschsprung Disease/genetics , Hirschsprung Disease/metabolism , Homeodomain Proteins/genetics , Humans , Molecular Sequence Data , Nuclear Proteins/genetics , Phenotype , Promoter Regions, Genetic , Proto-Oncogene Proteins c-ret/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Thyroid Nuclear Factor 1 , Transcription Factors/genetics
8.
J Pediatr Surg ; 44(10): 1904-12, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19853745

ABSTRACT

BACKGROUND: The rearranged during transfection (RET) gene encodes a single-pass receptor whose proper expression and function are essential for the development of enteric nervous system. Mutations in RET regulatory regions are also associated with Hirschsprung disease (HSCR) (aganglionosis of the colon). We previously showed that 2 polymorphisms in RET promoter are associated with the increased risk of HSCR. These single nucleotide polymorphisms overlap with the NK2 homeobox 1 (Nkx2-1) binding motif interrupting the physical interaction of NKX2-1 with the RET promoter and result in reduced RET transcription. In this study, we further delineated Nkx2-1-mediated RET Transcription. METHODS AND RESULTS: First, we demonstrated that PHOX2B, like SOX10 and NKX2-1, is expressed in the mature enteric ganglions of human gut by immunohistochemistry. Second, subsequent dual-luciferase-reporter studies indicated that Nkx2-1 indeed works coordinately with Phox2b and Sox10, but not Pax3, to mediate RET transcription. In addition, identification of Phox2b responsive region in RET promoter further provides solid evidence of the potential functional interaction between Phox2b and RET. CONCLUSION: In sum, Phox2b and Sox10 act together with Nkx2.1 to modify RET signaling and this interaction may also contribute to HSCR susceptibility.


Subject(s)
Hirschsprung Disease/genetics , Homeodomain Proteins/genetics , Proto-Oncogene Proteins c-ret/genetics , Transcription Factors/genetics , Animals , Cell Line, Tumor/metabolism , Enteric Nervous System/metabolism , Epistasis, Genetic/genetics , Gene Expression Regulation/genetics , Genetic Predisposition to Disease/genetics , Genotype , Homeodomain Proteins/metabolism , Humans , Mice , Mutation/genetics , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , PAX3 Transcription Factor , Paired Box Transcription Factors/genetics , Paired Box Transcription Factors/metabolism , Polymorphism, Single Nucleotide , Promoter Regions, Genetic , Proto-Oncogene Proteins c-ret/metabolism , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/metabolism , SOXE Transcription Factors/genetics , SOXE Transcription Factors/metabolism , Signal Transduction/genetics , Thyroid Nuclear Factor 1 , Transcription Factors/metabolism , Transcriptional Activation/genetics , Transfection/statistics & numerical data
SELECTION OF CITATIONS
SEARCH DETAIL