Try a new search

Format these results:

Searched for:

person:chatts03

in-biosketch:yes

Total Results:

32


RET enhancer haplotype-dependent remodeling of the human fetal gut development program

Chatterjee, Sumantra; Fries, Lauren E; Yaacov, Or; Hu, Nan; Berk-Rauch, Hanna E; Chakravarti, Aravinda
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.
PMCID:10664930
PMID: 37948459
ISSN: 1553-7404
CID: 5607952

Ret deficiency decreases neural crest progenitor proliferation and restricts fate potential during enteric nervous system development

Vincent, Elizabeth; Chatterjee, Sumantra; Cannon, Gabrielle H; Auer, Dallas; Ross, Holly; Chakravarti, Aravinda; Goff, Loyal A
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.
PMCID:10451519
PMID: 37585461
ISSN: 1091-6490
CID: 5595682

RET enhancer haplotype-dependent remodeling of the human fetal gut development program [PrePrint]

Chatterjee, Sumantra; Fries, Lauren E; Yaacov, Or; Hu, Nan; Berk-Rauch, Hanna E; Chakravarti, Aravinda
ORIGINAL:0015750
ISSN: 2692-8205
CID: 5294112

Ret loss-of-function decreases neural crest progenitor proliferation and restricts developmental fate potential during enteric nervous system development [PrePrint]

Vincent, Elizabeth; Chatterjee, Sumantra; Cannon, Gabrielle H; Auer, Dallas; Ross, Holly; Chakravarti, Aravinda; Goff, Loyal A
ORIGINAL:0015751
ISSN: 2692-8205
CID: 5294122

A multi-enhancer RET regulatory code is disrupted in Hirschsprung disease

Chatterjee, Sumantra; Karasaki, Kameko M; Fries, Lauren E; Kapoor, Ashish; Chakravarti, Aravinda
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.
PMID: 34782358
ISSN: 1549-5469
CID: 5049002

Coding and noncoding variants in EBF3 are involved in HADDS and simplex autism

Padhi, Evin M; Hayeck, Tristan J; Cheng, Zhang; Chatterjee, Sumantra; Mannion, Brandon J; Byrska-Bishop, Marta; Willems, Marjolaine; Pinson, Lucile; Redon, Sylvia; Benech, Caroline; Uguen, Kevin; Audebert-Bellanger, Séverine; Le Marechal, Cédric; Férec, Claude; Efthymiou, Stephanie; Rahman, Fatima; Maqbool, Shazia; Maroofian, Reza; Houlden, Henry; Musunuri, Rajeeva; Narzisi, Giuseppe; Abhyankar, Avinash; Hunter, Riana D; Akiyama, Jennifer; Fries, Lauren E; Ng, Jeffrey K; Mehinovic, Elvisa; Stong, Nick; Allen, Andrew S; Dickel, Diane E; Bernier, Raphael A; Gorkin, David U; Pennacchio, Len A; Zody, Michael C; Turner, Tychele N
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:, 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.
PMCID:8278787
PMID: 34256850
ISSN: 1479-7364
CID: 4965282

Gene- and tissue-level interactions in normal gastrointestinal development and Hirschsprung disease

Chatterjee, Sumantra; Nandakumar, Priyanka; Auer, Dallas R; Gabriel, Stacey B; Chakravarti, Aravinda
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.
PMID: 31818953
ISSN: 1091-6490
CID: 4238762

A gene regulatory network explains RET-EDNRB epistasis in Hirschsprung disease

Chatterjee, Sumantra; Chakravarti, Aravinda
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 7 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 (TF) 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.
PMID: 31313802
ISSN: 1460-2083
CID: 3977902

The gastrointestinal development ‘parts list’: transcript profiling of embryonic gut development in wildtype and Ret-deficient mice [PrePrint]

Chatterjee, Sumantra; Nandakumar, Priyanka; Auer, Dallas R; Gabriel, Stacey B; Chakravarti, Aravinda
ORIGINAL:0014922
ISSN: 2692-8205
CID: 4772442

Molecular Genetic Anatomy and Risk Profile of Hirschsprung's Disease

Tilghman, Joseph M; Ling, Albee Y; Turner, Tychele N; Sosa, Maria X; Krumm, Niklas; Chatterjee, Sumantra; Kapoor, Ashish; Coe, Bradley P; Nguyen, Khanh-Dung H; Gupta, Namrata; Gabriel, Stacey; Eichler, Evan E; Berrios, Courtney; Chakravarti, Aravinda
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:). 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:. 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.).
PMID: 30970187
ISSN: 1533-4406
CID: 3826322