scholarly journals Enteric nervous system development: migration, differentiation, and disease

2013 ◽  
Vol 305 (1) ◽  
pp. G1-G24 ◽  
Author(s):  
Jonathan I. Lake ◽  
Robert O. Heuckeroth

The enteric nervous system (ENS) provides the intrinsic innervation of the bowel and is the most neurochemically diverse branch of the peripheral nervous system, consisting of two layers of ganglia and fibers encircling the gastrointestinal tract. The ENS is vital for life and is capable of autonomous regulation of motility and secretion. Developmental studies in model organisms and genetic studies of the most common congenital disease of the ENS, Hirschsprung disease, have provided a detailed understanding of ENS development. The ENS originates in the neural crest, mostly from the vagal levels of the neuraxis, which invades, proliferates, and migrates within the intestinal wall until the entire bowel is colonized with enteric neural crest-derived cells (ENCDCs). After initial migration, the ENS develops further by responding to guidance factors and morphogens that pattern the bowel concentrically, differentiating into glia and neuronal subtypes and wiring together to form a functional nervous system. Molecules controlling this process, including glial cell line-derived neurotrophic factor and its receptor RET, endothelin (ET)-3 and its receptor endothelin receptor type B, and transcription factors such as SOX10 and PHOX2B, are required for ENS development in humans. Important areas of active investigation include mechanisms that guide ENCDC migration, the role and signals downstream of endothelin receptor type B, and control of differentiation, neurochemical coding, and axonal targeting. Recent work also focuses on disease treatment by exploring the natural role of ENS stem cells and investigating potential therapeutic uses. Disease prevention may also be possible by modifying the fetal microenvironment to reduce the penetrance of Hirschsprung disease-causing mutations.

2008 ◽  
Vol 43 (7) ◽  
pp. 1308-1311 ◽  
Author(s):  
Macarena Ruiz-Ferrer ◽  
Raquel M. Fernandez ◽  
Guillermo Antiñolo ◽  
Manuel Lopez-Alonso ◽  
Salud Borrego

2019 ◽  
Vol 14 (1) ◽  
Author(s):  
Francesca Lantieri ◽  
Stefania Gimelli ◽  
Chiara Viaggi ◽  
Elissavet Stathaki ◽  
Michela Malacarne ◽  
...  

Abstract Background Hirschsprung Disease (HSCR) is a congenital defect of the intestinal innervations characterized by complex inheritance. Many susceptibility genes including RET, the major HSCR gene, and several linked regions and associated loci have been shown to contribute to disease pathogenesis. Nonetheless, a proportion of patients still remains unexplained. Copy Number Variations (CNVs) have already been involved in HSCR, and for this reason we performed Comparative Genomic Hybridization (CGH), using a custom array with high density probes. Results A total of 20 HSCR candidate regions/genes was tested in 55 sporadic patients and four patients with already known chromosomal aberrations. Among 83 calls, 12 variants were experimentally validated, three of which involving the HSCR crucial genes SEMA3A/3D, NRG1, and PHOX2B. Conversely RET involvement in HSCR does not seem to rely on the presence of CNVs while, interestingly, several gains and losses did co-occur with another RET defect, thus confirming that more than one predisposing event is necessary for HSCR to develop. New loci were also shown to be involved, such as ALDH1A2, already found to play a major role in the enteric nervous system. Finally, all the inherited CNVs were of maternal origin. Conclusions Our results confirm a wide genetic heterogeneity in HSCR occurrence and support a role of candidate genes in expression regulation and cell signaling, thus contributing to depict further the molecular complexity of the genomic regions involved in the Enteric Nervous System development. The observed maternal transmission bias for HSCR associated CNVs supports the hypothesis that in females these variants might be more tolerated, requiring additional alterations to develop HSCR disease.


2015 ◽  
Vol 149 (7) ◽  
pp. 1837-1848.e5 ◽  
Author(s):  
Jessica Ai-Jia Liu ◽  
Frank Pui-Ling Lai ◽  
Hong-Sheng Gui ◽  
Mai-Har Sham ◽  
Paul Kwong-Hang Tam ◽  
...  

2017 ◽  
Vol 7 (1) ◽  
Author(s):  
Leticia Villalba-Benito ◽  
Ana Torroglosa ◽  
Raquel María Fernández ◽  
Macarena Ruíz-Ferrer ◽  
María José Moya-Jiménez ◽  
...  

PLoS Genetics ◽  
2021 ◽  
Vol 17 (8) ◽  
pp. e1009698
Author(s):  
Laura E. Kuil ◽  
Katherine C. MacKenzie ◽  
Clara S. Tang ◽  
Jonathan D. Windster ◽  
Thuy Linh Le ◽  
...  

Hirschsprung disease (HSCR) is a complex genetic disease characterized by absence of ganglia in the intestine. HSCR etiology can be explained by a unique combination of genetic alterations: rare coding variants, predisposing haplotypes and Copy Number Variation (CNV). Approximately 18% of patients have additional anatomical malformations or neurological symptoms (HSCR-AAM). Pinpointing the responsible culprits within a CNV is challenging as often many genes are affected. Therefore, we selected candidate genes based on gene enrichment strategies using mouse enteric nervous system transcriptomes and constraint metrics. Next, we used a zebrafish model to investigate whether loss of these genes affects enteric neuron development in vivo. This study included three groups of patients, two groups without coding variants in disease associated genes: HSCR-AAM and HSCR patients without associated anomalies (HSCR-isolated). The third group consisted of all HSCR patients in which a confirmed pathogenic rare coding variant was identified. We compared these patient groups to unaffected controls. Predisposing haplotypes were determined, confirming that every HSCR subgroup had increased contributions of predisposing haplotypes, but their contribution was highest in isolated HSCR patients without RET coding variants. CNV profiling proved that specifically HSCR-AAM patients had larger Copy Number (CN) losses. Gene enrichment strategies using mouse enteric nervous system transcriptomes and constraint metrics were used to determine plausible candidate genes located within CN losses. Validation in zebrafish using CRISPR/Cas9 targeting confirmed the contribution of UFD1L, TBX2, SLC8A1, and MAPK8 to ENS development. In addition, we revealed epistasis between reduced Ret and Gnl1 expression and between reduced Ret and Tubb5 expression in vivo. Rare large CN losses—often de novo—contribute to HSCR in HSCR-AAM patients. We proved the involvement of six genes in enteric nervous system development and Hirschsprung disease.


2005 ◽  
Vol 233 (2) ◽  
pp. 473-483 ◽  
Author(s):  
Kwok Keung Chan ◽  
Yuk Shan Chen ◽  
Tai On Yau ◽  
Ming Fu ◽  
Vincent Chi Hang Lui ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document