Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
Int J Mol Sci ; 24(7)2023 Apr 04.
Article in English | MEDLINE | ID: mdl-37047673

ABSTRACT

Insights into the role of microRNAs (miRNAs) in disease pathogenesis have made them attractive therapeutic targets, and numerous miRNAs have been functionally linked to Hirschsprung disease (HSCR), a life-threatening genetic disorder due to defective migration, proliferation, and colonization of enteric neural crest cells (ENCCs) in the gut. Recent studies have demonstrated that miR-424 strongly inhibits migration in a variety of cell types and its potential target RICTOR is essential for neural crest cell development. We therefore sought to interrogate how miR-424 and RICTOR contribute to the pathogenesis of HSCR. We utilized HSCR cases and human neural cells to evaluate the miR-424-mediated regulation of RICTOR and the downstream AKT phosphorylation. We further developed an ex vivo model to assess the effects of miR-424 on ENCC migration and proliferation. Then, single-cell atlases of gene expression in both human and mouse fetal intestines were used to determine the characteristics of RICTOR and AKT expression in the developing gut. Our findings demonstrate that miR-424 levels are markedly increased in the colonic tissues of patients with HSCR and that it regulates human neural cell migration by directly targeting RICTOR. Up-regulation of miR-424 leads to decreased AKT phosphorylation levels in a RICTOR-dependent manner, and this, in turn, impairs ENCC proliferation and migration in the developing gut. Interestingly, we further identified prominent RICTOR and AKT expressions in the enteric neurons and other types of enteric neural cells in human and mouse fetal intestines. Our present study reveals the role of the miR-424/RICTOR axis in HSCR pathogenesis and indicates that miR-424 is a promising candidate for the development of targeted therapies against HSCR.


Subject(s)
Enteric Nervous System , Hirschsprung Disease , MicroRNAs , Mice , Animals , Humans , Hirschsprung Disease/metabolism , Neural Crest/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Up-Regulation , Phosphorylation , Cell Movement/genetics , MicroRNAs/genetics , MicroRNAs/metabolism , Transcription Factors/metabolism , Enteric Nervous System/metabolism
2.
Pediatr Dev Pathol ; 24(1): 19-26, 2021.
Article in English | MEDLINE | ID: mdl-32975470

ABSTRACT

BACKGROUND: It can be challenging to recognize undifferentiated/immature ganglion cells, especially single forms. Ganglion cells and glia are derived from enteric neural crest cells (ENCCs), a group of autonomic nervous system (ANS)-lineage neural crest progenitors that PHOX2B regulates. Phox2b is an excellent marker for neoplastic and non-neoplastic ANS cells (eg, peripheral neuroblastic tumors [pNTs]). We hypothesized that Phox2b immunohistochemical staining (IHC) would also be useful for detecting ENCCs. METHODS: Hematoxylin and eosin, calretinin IHC, and Phox2b IHC were reviewed on 21 pull-through specimens and on a cohort of 12 rectal biopsies. RESULTS: Phox2b IHC demonstrated nuclear positivity in all of the ganglion cells across the different phases of differentiation without background staining. The Phox2b result correlated with the morphological findings, calretinin IHC results, and diagnoses based on the routine diagnostic method. The intensity was uniformly strong in the undifferentiated/immature forms and became variable in the mature forms; this pattern was similar to that seen in pNTs. CONCLUSION: Phox2b IHC was highly sensitive and specific for detecting ganglion cells. It worked especially well for immature ganglion cells, seen in premature neonates, and scattered single forms in transition zones. In basic research settings, Phox2b can be a useful marker for early differentiation of ENCCs.


Subject(s)
Enteric Nervous System/chemistry , Hirschsprung Disease/metabolism , Homeodomain Proteins/analysis , Immunohistochemistry , Neural Crest/chemistry , Rectum/innervation , Transcription Factors/analysis , Biopsy , Child , Child, Preschool , Enteric Nervous System/pathology , Female , Hirschsprung Disease/pathology , Humans , Infant , Infant, Newborn , Male , Neural Crest/pathology
3.
Int J Mol Sci ; 22(18)2021 Sep 07.
Article in English | MEDLINE | ID: mdl-34575824

ABSTRACT

The development of the enteric nervous system (ENS) is highly modulated by the synchronized interaction between the enteric neural crest cells (ENCCs) and the neural stem cell niche comprising the gut microenvironment. Genetic defects dysregulating the cellular behaviour(s) of the ENCCs result in incomplete innervation and hence ENS dysfunction. Hirschsprung disease (HSCR) is a rare complex neurocristopathy in which the enteric neural crest-derived cells fail to colonize the distal colon. In addition to ENS defects, increasing evidence suggests that HSCR patients may have intrinsic defects in the niche impairing the extracellular matrix (ECM)-cell interaction and/or dysregulating the cellular niche factors necessary for controlling stem cell behaviour. The niche defects in patients may compromise the regenerative capacity of the stem cell-based therapy and advocate for drug- and niche-based therapies as complementary therapeutic strategies to alleviate/enhance niche-cell interaction. Here, we provide a summary of the current understandings of the role of the enteric neural stem cell niche in modulating the development of the ENS and in the pathogenesis of HSCR. Deciphering the contribution of the niche to HSCR may provide important implications to the development of regenerative medicine for HSCR.


Subject(s)
Enteric Nervous System/cytology , Enteric Nervous System/metabolism , Hirschsprung Disease/genetics , Neural Stem Cells/cytology , Neural Stem Cells/physiology , Stem Cell Niche , Animals , Biomarkers , Cell Differentiation , Disease Management , Disease Susceptibility , Endothelin-3/metabolism , Genetic Predisposition to Disease , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Hirschsprung Disease/diagnosis , Hirschsprung Disease/metabolism , Hirschsprung Disease/therapy , Humans , Neural Crest/cytology , Neural Crest/metabolism , Receptor, Endothelin B/metabolism , Regenerative Medicine , Signal Transduction
4.
Cell Mol Neurobiol ; 40(4): 617-628, 2020 May.
Article in English | MEDLINE | ID: mdl-31760535

ABSTRACT

Although approximately 50% of cases have a known genetic defect, the precise pathogenesis of Hirschsprung disease (HSCR) is still unclear. We recently reported that expression of fibronectin (FN), which is involved in the migration, colonization, and differentiation of enteric neural crest cells (ENCCs), is increased in aganglionic colonic segments obtained from patients. We hypothesized that abnormally high levels of FN might play a role in the etiology of HSCR. Here, to test this hypothesis, we investigated aganglionic, transitional, and ganglionic colon segments from 63 children with HSCR and distal colon from thirty healthy Wistar rats at embryonic day 20, in addition to in vitro studies with PC12 Adh neural crest cells. We measured the protein and mRNA expression levels of FN, together with a panel of excitatory (VGLUT1, GluA1, GluN1, PSD-95, and NL-1) and inhibitory (GAD67, GABA AR-α1, NL-2, and SLC32) synaptic markers. Expression of all these synaptic markers was significantly decreased in aganglionic colon, compared to ganglionic colon, whereas expression of FN was significantly increased. In a neural crest cell line, PC12 Adh, knockdown of FN with small-interfering RNA increased the expression of synaptic markers. Co-culture of colons from embryonic day 20 rats with RGD recombinant protein, which contains the RGD motif of FN, reduced the expression of excitatory and inhibitory synaptic markers. These results are consistent with the idea that the etiology of HSCR involves aberrant overexpression of FN, which may impair synaptic function and enteric nervous system development, leading to motor dysfunction of intestinal muscles.


Subject(s)
Fibronectins/metabolism , Hirschsprung Disease/metabolism , Neural Inhibition , Synapses/metabolism , Animals , Biomarkers/metabolism , Child , Child, Preschool , Colon/pathology , Female , Hirschsprung Disease/pathology , Humans , Infant , Infant, Newborn , Male , Myenteric Plexus/metabolism , PC12 Cells , Rats , Rats, Wistar
5.
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
6.
Development ; 143(2): 264-75, 2016 Jan 15.
Article in English | MEDLINE | ID: mdl-26674309

ABSTRACT

The enteric nervous system (ENS) develops from neural crest cells that migrate along the intestine, differentiate into neurons and glia, and pattern into two plexuses within the gut wall. Inductive interactions between epithelium and mesenchyme regulate gut development, but the influence of these interactions on ENS development is unknown. Epithelial-mesenchymal recombinations were constructed using avian hindgut mesenchyme and non-intestinal epithelium from the bursa of Fabricius. These recombinations led to abnormally large and ectopically positioned ganglia. We hypothesized that sonic hedgehog (Shh), a secreted intestinal epithelial protein not expressed in the bursa, mediates this effect. Inhibition of Shh signaling, by addition of cyclopamine or a function-blocking antibody, resulted in large, ectopic ganglia adjacent to the epithelium. Shh overexpression, achieved in ovo using Shh-encoding retrovirus and in organ culture using recombinant protein, led to intestinal aganglionosis. Shh strongly induced the expression of versican and collagen type IX, whereas cyclopamine reduced expression of these chondroitin sulfate proteoglycans that are known to be inhibitory to neural crest cell migration. Shh also inhibited enteric neural crest-derived cell (ENCC) proliferation, promoted neuronal differentiation, and reduced expression of Gdnf, a key regulator of ENS formation. Ptc1 and Ptc2 were not expressed by ENCCs, and migration of isolated ENCCs was not inhibited by Shh protein. These results suggest that epithelial-derived Shh acts indirectly on the developing ENS by regulating the composition of the intestinal microenvironment.


Subject(s)
Enteric Nervous System/metabolism , Extracellular Matrix/metabolism , Hedgehog Proteins/metabolism , Animals , Apoptosis/physiology , Cell Movement , Chickens , Hedgehog Proteins/genetics , Immunohistochemistry , In Situ Hybridization , Mice , Patched Receptors , Patched-1 Receptor , Quail , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Signal Transduction/physiology
7.
Development ; 142(2): 331-42, 2015 Jan 15.
Article in English | MEDLINE | ID: mdl-25519241

ABSTRACT

In vertebrates, the digestive tract develops from a uniform structure where reciprocal epithelial-mesenchymal interactions pattern this complex organ into regions with specific morphologies and functions. Concomitant with these early patterning events, the primitive GI tract is colonized by the vagal enteric neural crest cells (vENCCs), a population of cells that will give rise to the enteric nervous system (ENS), the intrinsic innervation of the GI tract. The influence of vENCCs on early patterning and differentiation of the GI tract has never been evaluated. In this study, we report that a crucial number of vENCCs is required for proper chick stomach development, patterning and differentiation. We show that reducing the number of vENCCs by performing vENCC ablations induces sustained activation of the BMP and Notch pathways in the stomach mesenchyme and impairs smooth muscle development. A reduction in vENCCs also leads to the transdifferentiation of the stomach into a stomach-intestinal mixed phenotype. In addition, sustained Notch signaling activity in the stomach mesenchyme phenocopies the defects observed in vENCC-ablated stomachs, indicating that inhibition of the Notch signaling pathway is essential for stomach patterning and differentiation. Finally, we report that a crucial number of vENCCs is also required for maintenance of stomach identity and differentiation through inhibition of the Notch signaling pathway. Altogether, our data reveal that, through the regulation of mesenchyme identity, vENCCs act as a new mediator in the mesenchymal-epithelial interactions that control stomach development.


Subject(s)
Cell Differentiation/physiology , Enteric Nervous System/embryology , Morphogenesis/physiology , Neural Crest/embryology , Signal Transduction/physiology , Stomach/embryology , Animals , Bone Morphogenetic Proteins/metabolism , Chick Embryo , Fluorescent Antibody Technique , In Situ Hybridization , Muscle, Smooth/embryology , Organ Culture Techniques , Receptors, Notch/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Stomach/innervation
8.
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
9.
Exp Mol Pathol ; 95(1): 111-6, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23769877

ABSTRACT

C-X-C chemokine receptor type 4 (CXCR4) plays a crucial role in a wide range of physiological and pathological processes, including the migration of stem cells, such as neural crest-derived cells. Hirschsprung's disease (HSCR), a developmental disorder characterized by the absence of ganglion cells, is regarded as the consequence of the premature arrest of the craniocaudal migration of neural crest-derived cells (NCDCs) in the gastrointestinal tract during the development of the enteric nervous system (ENS). In this study, colon tissue samples from 61 HSCR patients were surgically collected and divided into aganglionic, oligoganglionic and normal ganglionic segments. Quantitative real-time polymerase chain reactions (PCR), Western blotting, and immunohistochemical and immunofluorescence staining were performed to analyze the expression levels and patterns of CXCR4 in different colon tissue segments. The expression levels of CXCR4 mRNA and protein in the aganglionic segments were decreased compared to the normal ganglionic and oligoganglionic colon segments (p<0.01). Immunohistochemical staining showed that intensive CXCR4 staining was detected in the ganglion cells and the supporting glial cells in the ganglion in control colon specimens and normal ganglionic and oligoganglionic colon segments from the HSCR patients; however, CXCR4 staining was significantly decreased in the aganglionic colon segments. Immunofluorescence staining showed that CXCR4 staining was mainly detected in the ganglia where RET-positive ganglion cells were observed. Elucidating CXCR4 expression patterns in colon segments could be the basis for further investigations of the potential role of CXCR4 in ENS development.


Subject(s)
Colon/metabolism , Hirschsprung Disease/metabolism , Receptors, CXCR4/metabolism , Colon/pathology , Female , Ganglia/metabolism , Ganglia/pathology , Gene Expression , Hirschsprung Disease/pathology , Humans , Male , Proto-Oncogene Proteins c-ret/metabolism , Receptors, CXCR4/genetics
10.
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.

11.
Mol Med Rep ; 17(4): 5844-5850, 2018 04.
Article in English | MEDLINE | ID: mdl-29484400

ABSTRACT

Hirschsprung disease (HSCR) is a newborn colorectal disease characterized by an absence of ganglia in the distal gut. Hedgehog (Hh) and endothelin signaling serve important roles in gastrointestinal tract formation. Alterations in the signaling pathways disrupt the development of enteric neural crest cells (ENCCs). It is not known whether there is any coordination between these pathways in the pathogenesis of HSCR. In the present study, tissue samples from 35 patients with HSCR, including stenotic aganglionosis gut and normal ganglionic gut, were obtained. The expression of Gli family zinc finger 1 (Gli1) and endothelin receptor type B (EDNRB) was determined using reverse transcription­quantitative polymerase chain reaction, immunohistochemistry and western blotting. In addition, the SK­N­SH cell line was used to investigate the association between Hh signaling and the expression of EDNRB. The results revealed aberrant expression of Gli1 in the aganglionic segments, as well as decreased expression of Gli1 in tissues from 7 patients with HSCR exhibited, whereas tissues from 9 patients with HSCR exhibited increased Gli1 expression compared with the expression in the normal tissues. There was a negative association between EDNRB expression and Gli1 expression in the same sample. Knockdown of Gli1 by small interfering RNA and inhibition of Hh signaling by Vismodegib in SK­N­SH cells increased EDNRB expression. By contrast, upregulation of Gli1 expression by plasmids and activation of Hh signaling by Purmorphamine decreased EDNRB expression. Furthermore, premature enteric ganglia were observed in 4 patients with HSCR with decreased Gli1 expression. Thus, the results of the present study suggest that altered Gli1 expression negatively regulates EDNRB expression in patients with HSCR. The increased expression of EDNRB induced by decreased Gli1 expression may represent a novel mechanism in HSCR.


Subject(s)
Hirschsprung Disease/genetics , Hirschsprung Disease/metabolism , Receptor, Endothelin B/genetics , Receptor, Endothelin B/metabolism , Zinc Finger Protein GLI1/genetics , Zinc Finger Protein GLI1/metabolism , Adolescent , Cell Line , Child , Child, Preschool , Female , Gene Expression , Gene Expression Regulation , Hirschsprung Disease/pathology , Humans , Immunohistochemistry , Infant , Male , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL