Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 346
Filter
1.
Dev Cell ; 59(13): 1689-1706.e8, 2024 Jul 08.
Article in English | MEDLINE | ID: mdl-38636517

ABSTRACT

During enteric nervous system (ENS) development, pioneering wavefront enteric neural crest cells (ENCCs) initiate gut colonization. However, the molecular mechanisms guiding their specification and niche interaction are not fully understood. We used single-cell RNA sequencing and spatial transcriptomics to map the spatiotemporal dynamics and molecular landscape of wavefront ENCCs in mouse embryos. Our analysis shows a progressive decline in wavefront ENCC potency during migration and identifies transcription factors governing their specification and differentiation. We further delineate key signaling pathways (ephrin-Eph, Wnt-Frizzled, and Sema3a-Nrp1) utilized by wavefront ENCCs to interact with their surrounding cells. Disruptions in these pathways are observed in human Hirschsprung's disease gut tissue, linking them to ENS malformations. Additionally, we observed region-specific and cell-type-specific transcriptional changes in surrounding gut tissues upon wavefront ENCC arrival, suggesting their role in shaping the gut microenvironment. This work offers a roadmap of ENS development, with implications for understanding ENS disorders.


Subject(s)
Cell Movement , Enteric Nervous System , Neural Crest , Signal Transduction , Animals , Neural Crest/metabolism , Neural Crest/cytology , Mice , Enteric Nervous System/metabolism , Enteric Nervous System/embryology , Enteric Nervous System/cytology , Embryo, Mammalian/metabolism , Embryo, Mammalian/cytology , Cell Differentiation , Gene Expression Regulation, Developmental , Hirschsprung Disease/genetics , Hirschsprung Disease/metabolism , Hirschsprung Disease/pathology , Humans
2.
Gastroenterology ; 166(6): 1085-1099, 2024 06.
Article in English | MEDLINE | ID: mdl-38452824

ABSTRACT

BACKGROUND & AIMS: The enteric nervous system (ENS), the gut's intrinsic nervous system critical for gastrointestinal function and gut-brain communication, is believed to mainly originate from vagal neural crest cells (vNCCs) and partially from sacral NCCs (sNCCs). Resolving the exact origins of the ENS is critical for understanding congenital ENS diseases but has been confounded by the inability to distinguish between both NCC populations in situ. Here, we aimed to resolve the exact origins of the mammalian ENS. METHODS: We genetically engineered mouse embryos facilitating comparative lineage-tracing of either all (pan-) NCCs including vNCCs or caudal trunk and sNCCs (s/tNCCs) excluding vNCCs. This was combined with dual-lineage tracing and 3-dimensional reconstruction of pelvic plexus and hindgut to precisely pinpoint sNCC and vNCC contributions. We further used coculture assays to determine the specificity of cell migration from different neural tissues into the hindgut. RESULTS: Both pan-NCCs and s/tNCCs contributed to established NCC derivatives but only pan-NCCs contributed to the ENS. Dual-lineage tracing combined with 3-dimensional reconstruction revealed that s/tNCCs settle in complex patterns in pelvic plexus and hindgut-surrounding tissues, explaining previous confusion regarding their contributions. Coculture experiments revealed unspecific cell migration from autonomic, sensory, and neural tube explants into the hindgut. Lineage tracing of ENS precursors lastly provided complimentary evidence for an exclusive vNCC origin of the murine ENS. CONCLUSIONS: sNCCs do not contribute to the murine ENS, suggesting that the mammalian ENS exclusively originates from vNCCs. These results have immediate implications for comprehending (and devising treatments for) congenital ENS disorders, including Hirschsprung's disease.


Subject(s)
Cell Lineage , Cell Movement , Enteric Nervous System , Neural Crest , Animals , Neural Crest/cytology , Neural Crest/embryology , Enteric Nervous System/embryology , Mice , Coculture Techniques , Mice, Transgenic , Vagus Nerve/embryology , Sacrum/innervation
3.
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
4.
Elife ; 112022 03 24.
Article in English | MEDLINE | ID: mdl-35324425

ABSTRACT

Overarching themes in the terminal differentiation of the enteric nervous system, an autonomously acting unit of animal nervous systems, have so far eluded discovery. We describe here the overall regulatory logic of enteric nervous system differentiation of the nematode Caenorhabditis elegans that resides within the foregut (pharynx) of the worm. A C. elegans homolog of the Drosophila Sine oculis homeobox gene, ceh-34, is expressed in all 14 classes of interconnected pharyngeal neurons from their birth throughout their life time, but in no other neuron type of the entire animal. Constitutive and temporally controlled ceh-34 removal shows that ceh-34 is required to initiate and maintain the neuron type-specific terminal differentiation program of all pharyngeal neuron classes, including their circuit assembly. Through additional genetic loss of function analysis, we show that within each pharyngeal neuron class, ceh-34 cooperates with different homeodomain transcription factors to individuate distinct pharyngeal neuron classes. Our analysis underscores the critical role of homeobox genes in neuronal identity specification and links them to the control of neuronal circuit assembly of the enteric nervous system. Together with the pharyngeal nervous system simplicity as well as its specification by a Sine oculis homolog, our findings invite speculations about the early evolution of nervous systems.


Subject(s)
Caenorhabditis elegans Proteins , Enteric Nervous System , Gene Expression Regulation, Developmental , Homeodomain Proteins , Transcription Factors , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Enteric Nervous System/embryology , Enteric Nervous System/growth & development , Gene Expression Regulation, Developmental/genetics , Genes, Homeobox , Homeodomain Proteins/metabolism , Pharynx , Transcription Factors/genetics , Transcription Factors/metabolism
5.
Nature ; 597(7875): 250-255, 2021 09.
Article in English | MEDLINE | ID: mdl-34497389

ABSTRACT

The cellular landscape of the human intestinal tract is dynamic throughout life, developing in utero and changing in response to functional requirements and environmental exposures. Here, to comprehensively map cell lineages, we use single-cell RNA sequencing and antigen receptor analysis of almost half a million cells from up to 5 anatomical regions in the developing and up to 11 distinct anatomical regions in the healthy paediatric and adult human gut. This reveals the existence of transcriptionally distinct BEST4 epithelial cells throughout the human intestinal tract. Furthermore, we implicate IgG sensing as a function of intestinal tuft cells. We describe neural cell populations in the developing enteric nervous system, and predict cell-type-specific expression of genes associated with Hirschsprung's disease. Finally, using a systems approach, we identify key cell players that drive the formation of secondary lymphoid tissue in early human development. We show that these programs are adopted in inflammatory bowel disease to recruit and retain immune cells at the site of inflammation. This catalogue of intestinal cells will provide new insights into cellular programs in development, homeostasis and disease.


Subject(s)
Aging , Enteric Nervous System/cytology , Fetus/cytology , Health , Intestines/cytology , Intestines/growth & development , Lymph Nodes/cytology , Lymph Nodes/growth & development , Adult , Animals , Child , Crohn Disease/pathology , Datasets as Topic , Enteric Nervous System/anatomy & histology , Enteric Nervous System/embryology , Enteric Nervous System/growth & development , Epithelial Cells/cytology , Female , Fetus/anatomy & histology , Fetus/embryology , Humans , Intestines/embryology , Intestines/innervation , Lymph Nodes/embryology , Lymph Nodes/pathology , Mice , Mice, Inbred C57BL , Organogenesis , Receptors, IgG/metabolism , Signal Transduction , Spatio-Temporal Analysis , Time Factors
6.
JCI Insight ; 6(10)2021 05 24.
Article in English | MEDLINE | ID: mdl-33848271

ABSTRACT

Retinoic acid (RA) signaling is essential for enteric nervous system (ENS) development, since vitamin A deficiency or mutations in RA signaling profoundly reduce bowel colonization by ENS precursors. These RA effects could occur because of RA activity within the ENS lineage or via RA activity in other cell types. To define cell-autonomous roles for retinoid signaling within the ENS lineage at distinct developmental time points, we activated a potent floxed dominant-negative RA receptor α (RarαDN) in the ENS using diverse CRE recombinase-expressing mouse lines. This strategy enabled us to block RA signaling at premigratory, migratory, and postmigratory stages for ENS precursors. We found that cell-autonomous loss of RA receptor (RAR) signaling dramatically affected ENS development. CRE activation of RarαDN expression at premigratory or migratory stages caused severe intestinal aganglionosis, but at later stages, RarαDN induced a broad range of phenotypes including hypoganglionosis, submucosal plexus loss, and abnormal neural differentiation. RNA sequencing highlighted distinct RA-regulated gene sets at different developmental stages. These studies show complicated context-dependent RA-mediated regulation of ENS development.


Subject(s)
Enteric Nervous System , Receptors, Retinoic Acid , Signal Transduction , Animals , Embryo, Mammalian/innervation , Embryo, Mammalian/metabolism , Embryonic Development/genetics , Embryonic Development/physiology , Enteric Nervous System/embryology , Enteric Nervous System/metabolism , Female , Male , Mice , Neurogenesis/genetics , Neurogenesis/physiology , Receptors, Retinoic Acid/genetics , Receptors, Retinoic Acid/metabolism , Signal Transduction/genetics , Signal Transduction/physiology
7.
Nat Commun ; 12(1): 1894, 2021 03 25.
Article in English | MEDLINE | ID: mdl-33767165

ABSTRACT

Neural crest stem cells arising from caudal hindbrain (often called cardiac and posterior vagal neural crest) migrate long distances to form cell types as diverse as heart muscle and enteric ganglia, abnormalities of which lead to common congenital birth defects. Here, we explore whether individual caudal hindbrain neural crest precursors are multipotent or predetermined toward these particular fates and destinations. To this end, we perform lineage tracing of chick neural crest cells at single-cell resolution using two complementary approaches: retrovirally mediated multiplex clonal analysis and single-cell photoconversion. Both methods show that the majority of these neural crest precursors are multipotent with many clones producing mesenchymal as well as neuronal derivatives. Time-lapse imaging demonstrates that sister cells can migrate in distinct directions, suggesting stochasticity in choice of migration path. Perturbation experiments further identify guidance cues acting on cells in the pharyngeal junction that can influence this choice; loss of CXCR4 signaling results in failure to migrate to the heart but no influence on migration toward the foregut, whereas loss of RET signaling does the opposite. Taken together, the results suggest that environmental influences rather than intrinsic information govern cell fate choice of multipotent caudal hindbrain neural crest cells.


Subject(s)
Enteric Nervous System/embryology , Heart/embryology , Multipotent Stem Cells/cytology , Neural Crest/cytology , Neural Stem Cells/cytology , Animals , Cell Differentiation/physiology , Cell Movement/physiology , Chickens , Proto-Oncogene Proteins c-ret/genetics , Receptors, CXCR4/genetics , Rhombencephalon/cytology , Signal Transduction/genetics
8.
Development ; 148(3)2021 02 08.
Article in English | MEDLINE | ID: mdl-33558316

ABSTRACT

During embryonic development, the gut is innervated by intrinsic (enteric) and extrinsic nerves. Focusing on mammalian ENS development, in this Review we highlight how important the different compartments of this innervation are to assure proper gut function. We specifically address the three-dimensional architecture of the innervation, paying special attention to the differences in development along the longitudinal and circumferential axes of the gut. We review recent information about the formation of both intrinsic innervation, which is fairly well-known, as well as the establishment of the extrinsic innervation, which, despite its importance in gut-brain signaling, has received much less attention. We further discuss how external microbial and nutritional cues or neuroimmune interactions may influence development of gut innervation. Finally, we provide summary tables, describing the location and function of several well-known molecules, along with some newer factors that have more recently been implicated in the development of gut innervation.


Subject(s)
Embryonic Development/physiology , Enteric Nervous System/embryology , Enteric Nervous System/growth & development , Gastrointestinal Tract/innervation , Animals , Brain/physiology , Humans , Neurons/physiology , Organogenesis/physiology , Signal Transduction
9.
Nat Rev Gastroenterol Hepatol ; 18(6): 393-410, 2021 06.
Article in English | MEDLINE | ID: mdl-33514916

ABSTRACT

The enteric nervous system (ENS) is the largest division of the peripheral nervous system and closely resembles components and functions of the central nervous system. Although the central role of the ENS in congenital enteric neuropathic disorders, including Hirschsprung disease and inflammatory and functional bowel diseases, is well acknowledged, its role in systemic diseases is less understood. Evidence of a disordered ENS has accumulated in neurodegenerative diseases ranging from amyotrophic lateral sclerosis, Alzheimer disease and multiple sclerosis to Parkinson disease as well as neurodevelopmental disorders such as autism. The ENS is a key modulator of gut barrier function and a regulator of enteric homeostasis. A 'leaky gut' represents the gateway for bacterial and toxin translocation that might initiate downstream processes. Data indicate that changes in the gut microbiome acting in concert with the individual genetic background can modify the ENS, central nervous system and the immune system, impair barrier function, and contribute to various disorders such as irritable bowel syndrome, inflammatory bowel disease or neurodegeneration. Here, we summarize the current knowledge on the role of the ENS in gastrointestinal and systemic diseases, highlighting its interaction with various key players involved in shaping the phenotypes. Finally, current flaws and pitfalls related to ENS research in addition to future perspectives are also addressed.


Subject(s)
Enteric Nervous System/physiopathology , Colorectal Neoplasms/physiopathology , Diabetes Mellitus/physiopathology , Diet , Enteric Nervous System/embryology , Esophageal Achalasia/genetics , Esophageal Achalasia/physiopathology , Gastric Mucosa/physiology , Gastrointestinal Microbiome/physiology , Hirschsprung Disease/genetics , Hirschsprung Disease/physiopathology , Humans , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/physiopathology , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/physiopathology
10.
Int J Mol Sci ; 21(23)2020 Nov 28.
Article in English | MEDLINE | ID: mdl-33260622

ABSTRACT

Hirschsprung disease (HSCR) is a neurocristopathy characterized by intestinal aganglionosis which is attributed to a failure in neural crest cell (NCC) development during the embryonic stage. The colonization of the intestine by NCCs is a process finely controlled by a wide and complex gene regulatory system. Several genes have been associated with HSCR, but many aspects still remain poorly understood. The present study is focused on deciphering the PAX6 interaction network during enteric nervous system (ENS) formation. A combined experimental and computational approach was performed to identify PAX6 direct targets, as well as gene networks shared among such targets as potential susceptibility factors for HSCR. As a result, genes related to PAX6 either directly (RABGGTB and BRD3) or indirectly (TGFB1, HRAS, and GRB2) were identified as putative genes associated with HSCR. Interestingly, GRB2 is involved in the RET/GDNF/GFRA1 signaling pathway, one of the main pathways implicated in the disease. Our findings represent a new contribution to advance in the knowledge of the genetic basis of HSCR. The investigation of the role of these genes could help to elucidate their implication in HSCR onset.


Subject(s)
Chromatin Immunoprecipitation Sequencing , Enteric Nervous System/embryology , Hirschsprung Disease/embryology , Hirschsprung Disease/genetics , Animals , Base Sequence , Gene Expression Regulation , Genetic Predisposition to Disease , Genome , Mice , Nucleotide Motifs/genetics , PAX6 Transcription Factor/metabolism , Spheroids, Cellular/pathology
11.
Development ; 147(13)2020 07 13.
Article in English | MEDLINE | ID: mdl-32661018

ABSTRACT

The enteric nervous system (ENS) derives from the neural crest and innervates the gastrointestinal system, in which it is essential for gut function throughout life. A new paper in Development uses zebrafish to investigate the poorly understood process of post-embryonic ENS neurogenesis, in both development and injury contexts. To find out more, we met the paper's two authors, Wael Noor El-Nachef, Assistant Clinical Professor of Medicine at UCLA, and Marianne Bronner, Albert Billings Ruddock Professor of Biology and Biological Engineering at Caltech.


Subject(s)
Enteric Nervous System/embryology , Animals , History, 20th Century , History, 21st Century , Humans , Neural Crest/embryology , Neurogenesis/physiology , Zebrafish
12.
J Anat ; 237(4): 655-671, 2020 10.
Article in English | MEDLINE | ID: mdl-32598482

ABSTRACT

Compared to the intrinsic enteric nervous system (ENS), development of the extrinsic ENS is poorly documented, even though its presence is easily detectable with histological techniques. We visualised its development in human embryos and foetuses of 4-9.5 weeks post-fertilisation using Amira 3D-reconstruction and Cinema 4D-remodelling software. The extrinsic ENS originated from small, basophilic neural crest cells (NCCs) that migrated to the para-aortic region and then continued ventrally to the pre-aortic region, where they formed autonomic pre-aortic plexuses. From here, nerve fibres extended along the ventral abdominal arteries and finally connected to the intrinsic system. Schwann cell precursors (SCPs), a subgroup of NCCs that migrate on nerve fibres, showed region-specific differences in differentiation. SCPs developed into scattered chromaffin cells of the adrenal medulla dorsolateral to the coeliac artery (CA) and into more tightly packed chromaffin cells of the para-aortic bodies ventrolateral to the inferior mesenteric artery (IMA), with reciprocal topographic gradients between both fates. The extrinsic ENS first extended along the CA and then along the superior mesenteric artery (SMA) and IMA 5 days later. Apart from the branch to the caecum, extrinsic nerves did not extend along SMA branches in the herniated parts of the midgut until the gut loops had returned in the abdominal cavity, suggesting a permissive role of the intraperitoneal environment. Accordingly, extrinsic innervation had not yet reached the distal (colonic) loop of the midgut at 9.5 weeks development. Based on intrinsic ENS-dependent architectural remodelling of the gut layers, extrinsic innervation followed intrinsic innervation 3-4 Carnegie stages later.


Subject(s)
Embryonic Development/physiology , Enteric Nervous System/embryology , Intestines/innervation , Organogenesis/physiology , Cell Differentiation/physiology , Cell Movement/physiology , Humans , Intestines/embryology , Neural Crest/cytology
13.
Pediatr Dev Pathol ; 23(4): 285-295, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32212960

ABSTRACT

INTRODUCTION: The objective of this study is to investigate the role of thyroid hormone (TH) in the pathogenesis of intestinal dysganglionosis (ID). METHODS: A zebrafish model of congenital hypothyroidism (CH) was created by exposing the larvae to the 6-propyl-2-thiouracil (PTU). The enteric neurons were labeled with anti-HuC/D antibodies. The number of enteric neurons was counted. The larval intestine was dissociated and stained with anti-p75 and anti-α4 integrin antibodies. Mitosis and apoptosis of the p75+ α4 integrin+ enteric neural crest cells (ENCCs) were studied using flow cytometry. Intestinal motility was studied by analyzing the transit of fluorescent tracers. RESULTS: PTU (25 mg/L) significantly reduced TH production at 6- and 9-days post fertilization without changing the body length, body weight, and intestinal length of the larvae. Furthermore, PTU inhibited mitosis of ENCCs and reduced the number of enteric neurons throughout the larval zebrafish intestine. Importantly, PTU inhibited intestinal transit of fluorescent tracers. Finally, thyroxine supplementation restored ENCC mitosis, increased the number of enteric neurons, and recovered intestinal motility in the PTU-treated larvae. CONCLUSIONS: PTU inhibited TH production, reduced the number of enteric neurons, impaired intestinal motility, and impeded ENCC mitosis in zebrafish, suggesting a possible role of CH in the pathogenesis of ID.


Subject(s)
Congenital Hypothyroidism/complications , Enteric Nervous System/embryology , Hirschsprung Disease/embryology , Thyroid Hormones/metabolism , Animals , Biomarkers/metabolism , Cell Differentiation , Cell Movement , Cell Proliferation , Congenital Hypothyroidism/embryology , Congenital Hypothyroidism/metabolism , Congenital Hypothyroidism/pathology , Enteric Nervous System/metabolism , Enteric Nervous System/pathology , Flow Cytometry , Gastrointestinal Motility , Hirschsprung Disease/metabolism , Hirschsprung Disease/pathology , Neural Crest/embryology , Neural Crest/metabolism , Neural Crest/pathology , Zebrafish
14.
Dev Biol ; 455(2): 473-484, 2019 11 15.
Article in English | MEDLINE | ID: mdl-31394080

ABSTRACT

Intestinal tract development is a coordinated process involving signaling among the progenitors and developing cells from all three germ layers. Development of endoderm-derived intestinal epithelium has been shown to depend on epigenetic modifications, but whether that is also the case for intestinal tract cell types from other germ layers remains unclear. We found that functional loss of a DNA methylation machinery component, ubiquitin-like protein containing PHD and RING finger domains 1 (uhrf1), leads to reduced numbers of ectoderm-derived enteric neurons and severe disruption of mesoderm-derived intestinal smooth muscle. Genetic chimeras revealed that Uhrf1 functions both cell-autonomously in enteric neuron precursors and cell-non-autonomously in surrounding intestinal cells, consistent with what is known about signaling interactions between these cell types that promote one another's development. Uhrf1 recruits the DNA methyltransferase Dnmt1 to unmethylated DNA during replication. Dnmt1 is also expressed in enteric neurons and smooth muscle progenitors. dnmt1 mutants have fewer enteric neurons and disrupted intestinal smooth muscle compared to wildtypes. Because dnmt1;uhrf1 double mutants have a similar phenotype to dnmt1 and uhrf1 single mutants, Dnmt1 and Uhrf1 must function together during enteric neuron and intestinal muscle development. This work shows that genes controlling epigenetic modifications are important to coordinate intestinal tract development, provides the first demonstration that these genes influence development of the ENS, and advances uhrf1 and dnmt1 as potential new Hirschsprung disease candidates.


Subject(s)
DNA (Cytosine-5-)-Methyltransferase 1/physiology , Enteric Nervous System/embryology , Epigenesis, Genetic , Intestines/embryology , Trans-Activators/physiology , Zebrafish Proteins/physiology , Animals , Chimera , DNA (Cytosine-5-)-Methyltransferase 1/genetics , Embryonic Stem Cells/metabolism , Female , Gene Expression Regulation, Developmental , Intestines/cytology , Intestines/innervation , Male , Muscle, Smooth/embryology , Mutation , Neurons , Trans-Activators/genetics , Zebrafish , Zebrafish Proteins/genetics
15.
Dev Biol ; 455(2): 362-368, 2019 11 15.
Article in English | MEDLINE | ID: mdl-31306639

ABSTRACT

BACKGROUND: Cells derived from the neural crest colonize the developing gut and give rise to the enteric nervous system. The rate at which the ENCC population advances along the bowel will be affected by both the speed and directionality of individual ENCCs. The aim of the study was to use time-lapse imaging and pharmacological activators and inhibitors to examine the role of several intracellular signalling pathways in both the speed and the directionality of individual enteric neural crest-derived cells in intact explants of E12.5 mouse gut. Drugs that activate or inhibit intracellular components proposed to be involved in GDNF-RET and EDN3-ETB signalling in ENCCs were used. FINDINGS: Pharmacological inhibition of JNK significantly reduced ENCC speed but did not affect ENCC directionality. MEK inhibition did not affect ENCC speed or directionality. Pharmacological activation of adenylyl cyclase or PKA (a downstream cAMP-dependent kinase) resulted in a significant decrease in ENCC speed and an increase in caudal directionality of ENCCs. In addition, adenylyl cyclase activation also resulted in reduced cell-cell contact between ENCCs, however this was not observed following PKA activation, suggesting that the effects of cAMP on adhesion are not mediated by PKA. CONCLUSIONS: JNK is required for normal ENCC migration speed, but not directionality, while cAMP signalling appears to regulate ENCC migration speed, directionality and adhesion. Collectively, our data demonstrate that intracellular signalling pathways can differentially affect the speed and directionality of migrating ENCCs.


Subject(s)
Adenylyl Cyclases/metabolism , Cell Movement , JNK Mitogen-Activated Protein Kinases/metabolism , MAP Kinase Kinase Kinases/metabolism , MAP Kinase Signaling System , Neural Crest/cytology , Animals , Embryonic Induction , Enteric Nervous System/embryology , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , MAP Kinase Kinase Kinases/antagonists & inhibitors , MAP Kinase Signaling System/drug effects , Mice , Neural Crest/enzymology , Neural Crest/metabolism , Time Factors
16.
Mol Med Rep ; 20(2): 1297-1305, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31173231

ABSTRACT

Certain patients with anorectal malforma-tions (ARMs) continue to suffer from postoperative dysphoria. The enteric nervous system (ENS) is closely associated with defecation. The purinergic receptor P2Y2 (P2Y2) and Hu antigen D (HuD) proteins contain multiple motifs that enable their activation and direct coupling to integrin and growth factor receptor signaling pathways; thus, they may serve as key points in ENS development. The aim of the present study was to investigate the expression pattern of P2Y2 and HuD proteins during anorectal development in ARM embryos. The embryogenesis of ARM in rats was induced by ethylenethiourea (ETU) on the 10th gestational day. The expression patterns of P2Y2 and HuD proteins were evaluated by immunohistochemistry and western blot analysis in normal, ETU and ARM rat embryos on embryonic days E17, E19 and E21; their mRNA levels were assessed via reverse transcription­quantitative polymerase chain reaction (RT­qPCR) of the distal rectum of fetal rats. Immunohistochemistry of the distal rectum demonstrated that on E17, the expression levels of the two proteins were not different between the three groups. On E19, the expression of HuD was significantly decreased in the ARM group. On E21, the two proteins were significantly decreased in the ARM group. Additionally, the expression levels of the two proteins on E17 were significantly lower than on E21 in the ARM group. Western blotting and RT­qPCR also revealed that the P2Y2 and HuD proteins and mRNA expression levels were significantly decreased in the ARM groups when compared with the normal group on E17 and E21 (P<0.01). Thus, the present study demonstrated that downregulation of P2Y2 and HuD may partly be related to the development of the ENS in ARM embryos.


Subject(s)
Anorectal Malformations/embryology , Anorectal Malformations/genetics , Down-Regulation/genetics , ELAV-Like Protein 4/genetics , Enteric Nervous System/embryology , Fetus/metabolism , Gene Expression Regulation, Developmental , Receptors, Purinergic P2Y2/genetics , Animals , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats, Sprague-Dawley , Receptors, Purinergic P2Y2/metabolism
17.
Dev Dyn ; 248(6): 437-448, 2019 06.
Article in English | MEDLINE | ID: mdl-30958591

ABSTRACT

BACKGROUND: The enteric nervous system (ENS) is derived from enteric neural crest cells (ENCCs) that migrate into the gut. The zebrafish larva is a good model to study ENCC development due to its simplicity and transparency. However, little is known how individual ENCCs divide and become neurons. RESULTS: Here, by applying our new method of local heat-shock mediated Cre-recombination around the dorsal vagal area of zebrafish embryos we produced multicolored clones of ENCCs, and performed in vivo time-lapse imaging from ca. 3.5 to 4 days post-fertilization after arrival of ENCCs in the gut. Individual ENCCs migrated in various directions and were highly intermingled. The cell divisions were not restricted to a specific position in the gut. Antibody staining after imaging with anti-HuC/D and anti-Sox10 showed that an ENCC produced two neurons, or formed a neuron and an additional ENCC that further divided. At division, the daughter cells immediately separated. Afterward, some made soma-soma contact with other ENCCs. CONCLUSIONS: We introduced a new method of visualizing individual ENCCs in the zebrafish gut, describing their behaviors associated with cell division, providing a foundation to study the mechanism of proliferation and neurogenesis in the ENS in vertebrates.


Subject(s)
Enteric Nervous System/growth & development , Neural Crest/cytology , Neurogenesis , Time-Lapse Imaging/methods , Zebrafish/anatomy & histology , Animals , Cell Division , Cell Movement , Embryo, Nonmammalian , Enteric Nervous System/cytology , Enteric Nervous System/embryology , Gastrointestinal Tract , Heat-Shock Response , Neurons/cytology , Zebrafish/embryology
18.
Gastroenterology ; 156(5): 1483-1495.e6, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30610864

ABSTRACT

BACKGROUND & AIMS: The enteric nervous system (ENS) is the largest branch of the peripheral nervous system, comprising complex networks of neurons and glia, which are present throughout the gastrointestinal tract. Although development of a fully functional ENS is required for gastrointestinal motility, little is known about the ontogeny of ENS function in humans. We studied the development of neuronal subtypes and the emergence of evoked electrical activity in the developing human ENS. METHODS: Human fetal gut samples (obtained via the MRC-Wellcome Trust Human Developmental Biology Resource-UK) were characterized by immunohistochemistry, calcium imaging, RNA sequencing, and quantitative real-time polymerase chain reaction analyses. RESULTS: Human fetal colon samples have dense neuronal networks at the level of the myenteric plexus by embryonic week (EW) 12, with expression of excitatory neurotransmitter and synaptic markers. By contrast, markers of inhibitory neurotransmitters were not observed until EW14. Electrical train stimulation of internodal strands did not evoke activity in the ENS of EW12 or EW14 tissues. However, compound calcium activation was observed at EW16, which was blocked by the addition of 1 µmol/L tetrodotoxin. Expression analyses showed that this activity was coincident with increases in expression of genes encoding proteins involved in neurotransmission and action potential generation. CONCLUSIONS: In analyses of human fetal intestinal samples, we followed development of neuronal diversity, electrical excitability, and network formation in the ENS. These processes are required to establish the functional enteric circuitry. Further studies could increase our understanding of the pathogenesis of a range of congenital enteric neuropathies.


Subject(s)
Colon/innervation , Enteric Nervous System/physiology , Evoked Potentials , Nerve Net/physiology , Neurogenesis , Neurons/physiology , Calcium Signaling , Colon/embryology , Electric Stimulation , Enteric Nervous System/drug effects , Enteric Nervous System/embryology , Evoked Potentials/drug effects , Female , Gene Expression Regulation, Developmental , Gestational Age , Humans , Nerve Net/drug effects , Nerve Net/embryology , Neurogenesis/drug effects , Neurogenesis/genetics , Neurons/drug effects , Neurotransmitter Agents/pharmacology , Phenotype , Pregnancy , Pregnancy Trimester, Second , Synaptic Transmission
19.
Dev Biol ; 445(2): 256-270, 2019 01 15.
Article in English | MEDLINE | ID: mdl-30472119

ABSTRACT

The enteric nervous system is thought to originate solely from the neural crest. Transgenic lineage tracing revealed a novel population of clonal pancreatic duodenal homeobox-1 (Pdx1)-Cre lineage progenitor cells in the tunica muscularis of the gut that produced pancreatic descendants as well as neurons upon differentiation in vitro. Additionally, an in vivo subpopulation of endoderm lineage enteric neurons, but not glial cells, was seen especially in the proximal gut. Analysis of early transgenic embryos revealed Pdx1-Cre progeny (as well as Sox-17-Cre and Foxa2-Cre progeny) migrating from the developing pancreas and duodenum at E11.5 and contributing to the enteric nervous system. These results show that the mammalian enteric nervous system arises from both the neural crest and the endoderm. Moreover, in adult mice there are separate Wnt1-Cre neural crest stem cells and Pdx1-Cre pancreatic progenitors within the muscle layer of the gut.


Subject(s)
Enteric Nervous System/embryology , Animals , Cell Lineage/genetics , Duodenum/embryology , Duodenum/innervation , Duodenum/metabolism , Endoderm/cytology , Endoderm/embryology , Endoderm/metabolism , Enteric Nervous System/cytology , Enteric Nervous System/metabolism , Gene Expression Regulation, Developmental , HMGB Proteins/genetics , HMGB Proteins/metabolism , Hepatocyte Nuclear Factor 3-beta/genetics , Hepatocyte Nuclear Factor 3-beta/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Mice , Mice, Transgenic , Neural Crest/cytology , Neural Crest/embryology , Neural Crest/metabolism , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Pancreas/embryology , Pancreas/innervation , Pancreas/metabolism , SOXF Transcription Factors/genetics , SOXF Transcription Factors/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism , Wnt1 Protein/genetics , Wnt1 Protein/metabolism
20.
Cell Mol Life Sci ; 76(5): 921-940, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30470852

ABSTRACT

The transcription factor Ptf1a is a crucial helix-loop-helix (bHLH) protein selectively expressed in the pancreas, retina, spinal cord, brain, and enteric nervous system. Ptf1a is preferably assembled into a transcription trimeric complex PTF1 with an E protein and Rbpj (or Rbpjl). In pancreatic development, Ptf1a is indispensable in controlling the expansion of multipotent progenitor cells as well as the specification and maintenance of the acinar cells. In neural tissues, Ptf1a is transiently expressed in the post-mitotic cells and specifies the inhibitory neuronal cell fates, mostly mediated by downstream genes such as Tfap2a/b and Prdm13. Mutations in the coding and non-coding regulatory sequences resulting in Ptf1a gain- or loss-of-function are associated with genetic diseases such as pancreatic and cerebellar agenesis in the rodent and human. Surprisingly, Ptf1a alone is sufficient to reprogram mouse or human fibroblasts into tripotential neural stem cells. Its pleiotropic functions in many biological processes remain to be deciphered in the future.


Subject(s)
Cellular Reprogramming , Transcription Factors/physiology , Animals , Brain/embryology , Cell Transdifferentiation , Enteric Nervous System/embryology , Humans , Immunoglobulin J Recombination Signal Sequence-Binding Protein/physiology , Pancreas/embryology , Pancreas/physiology , Retina/embryology , Spinal Cord/embryology , Transcription Factors/chemistry , Transcription Factors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL