Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
Cell Stem Cell ; 29(1): 86-100.e6, 2022 01 06.
Article in English | MEDLINE | ID: mdl-34727519

ABSTRACT

The high turnover and regenerative capacity of the adult intestine relies on resident stem cells located at the bottom of the crypt. The enteric nervous system consists of an abundant network of enteric glial cells (EGCs) and neurons. Despite the close proximity of EGCs to stem cells, their in vivo role as a stem cell niche is still unclear. By analyzing the mouse and human intestinal mucosa transcriptomes at the single-cell level, we defined the regulation of EGC heterogeneity in homeostasis and chronic inflammatory bowel disease. Ablation of EGC subpopulations revealed that the repair potential of intestinal stem cells (ISCs) is regulated by a specific subset of glial fibrillary acidic protein (GFAP)+ EGCs. Mechanistically, injury induces expansion of GFAP+ EGCs, which express several WNT ligands to promote LGR5+ ISC self-renewal. Our work reveals the dynamically regulated heterogeneity of EGCs as a key part of the intestinal stem cell niche in regeneration and disease.


Subject(s)
Enteric Nervous System , Stem Cell Niche , Animals , Intestinal Mucosa , Intestines , Mice , Neuroglia
2.
Dev Cell ; 52(5): 647-658.e6, 2020 03 09.
Article in English | MEDLINE | ID: mdl-32155439

ABSTRACT

During development, intestinal epithelia undergo dramatic morphogenesis mediated by mesenchymal signaling to form villi, which are required for efficient nutrient absorption and host defense. Although both smooth-muscle-induced physical forces and mesenchymal cell clustering beneath emerging villi are implicated in epithelial folding, the underlying cellular mechanisms are unclear. Hedgehog (Hh) signaling can mediate both processes. We therefore analyzed its direct targetome and revealed GLI2 transcriptional activation of atypical cadherin and planar cell polarity (PCP) genes. By examining Fat4 and Dchs1 knockout mice, we demonstrate their critical roles in villus formation. Analyses of PCP-mutant mice and genetic interaction studies show that the Fat4-Dchs1 axis acts in parallel to the core-Vangl2 PCP axis to control mesenchymal cell clustering. Moreover, live light-sheet fluorescence microscopy and cultured PDGFRα+ cells reveal a requirement for PCP in their oriented cell migration guided by WNT5A. Therefore, mesenchymal PCP induced by Hh signaling drives cell clustering and subsequent epithelial remodeling.


Subject(s)
Cadherins/metabolism , Cell Polarity , Hedgehog Proteins/metabolism , Intestinal Mucosa/growth & development , Mesenchymal Stem Cells/metabolism , Microvilli/metabolism , Animals , Cadherins/genetics , Cell Differentiation , Cell Movement , Cells, Cultured , Female , Hedgehog Proteins/genetics , Intestinal Mucosa/cytology , Intestinal Mucosa/metabolism , Male , Mesenchymal Stem Cells/cytology , Mice , Mice, Inbred C57BL , Morphogenesis , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Signal Transduction , Wnt-5a Protein/genetics , Wnt-5a Protein/metabolism , Zinc Finger Protein Gli2/genetics , Zinc Finger Protein Gli2/metabolism
3.
Nat Commun ; 11(1): 334, 2020 01 17.
Article in English | MEDLINE | ID: mdl-31953387

ABSTRACT

Stomach and intestinal stem cells are located in discrete niches called the isthmus and crypt, respectively. Recent studies have demonstrated a surprisingly conserved role for Wnt signaling in gastrointestinal development. Although intestinal stromal cells secrete Wnt ligands to promote stem cell renewal, the source of stomach Wnt ligands is still unclear. Here, by performing single cell analysis, we identify gastrointestinal stromal cell populations with transcriptome signatures that are conserved between the stomach and intestine. In close proximity to epithelial cells, these perictye-like cells highly express telocyte and pericyte markers as well as Wnt ligands, and they are enriched for Hh signaling. By analyzing mice activated for Hh signaling, we show a conserved mechanism of GLI2 activation of Wnt ligands. Moreover, genetic inhibition of Wnt secretion in perictye-like stromal cells or stromal cells more broadly demonstrates their essential roles in gastrointestinal regeneration and development, respectively, highlighting a redundancy in gastrointestinal stem cell niches.


Subject(s)
Gastrointestinal Tract/metabolism , Genetic Testing , Stem Cell Niche/genetics , Stromal Cells/metabolism , Animals , Cell Self Renewal/genetics , Epithelial Cells/metabolism , Gastrointestinal Tract/cytology , Homeostasis , Ligands , Male , Mice , Mice, Knockout , Regeneration , Stromal Cells/cytology , Telocytes/metabolism , Transcriptome , Wnt Proteins/metabolism , Wnt Signaling Pathway , Zinc Finger Protein Gli2/metabolism
4.
Nat Commun ; 10(1): 4647, 2019 10 11.
Article in English | MEDLINE | ID: mdl-31604927

ABSTRACT

Human embryonic stem cell-derived beta cells offer a promising cell-based therapy for diabetes. However, efficient stem cell to beta cell differentiation has proven difficult, possibly due to the lack of cross-talk with the appropriate mesenchymal niche. To define organ-specific niche signals, we isolated pancreatic and gastrointestinal stromal cells, and analyzed their gene expression during development. Our genetic studies reveal the importance of tightly regulated Hedgehog signaling in the pancreatic mesenchyme: inactivation of mesenchymal signaling leads to annular pancreas, whereas stroma-specific activation of signaling via loss of Hedgehog regulators, Sufu and Spop, impairs pancreatic growth and beta cell genesis. Genetic rescue and transcriptome analyses show that these Sufu and Spop knockout defects occur through Gli2-mediated activation of gastrointestinal stromal signals such as Wnt ligands. Importantly, inhibition of Wnt signaling in organoid and human stem cell cultures significantly promotes insulin-producing cell generation, altogether revealing the requirement for organ-specific regulation of stromal niche signals.


Subject(s)
Embryonic Stem Cells/cytology , Hedgehog Proteins/metabolism , Insulin-Secreting Cells/cytology , Nuclear Proteins/physiology , Repressor Proteins/physiology , Cell Culture Techniques , Cell Differentiation , Cell- and Tissue-Based Therapy/methods , Diabetes Mellitus/therapy , Down-Regulation , Humans , Insulin-Secreting Cells/transplantation , Nuclear Proteins/metabolism , Organoids/cytology , Repressor Proteins/metabolism , Wnt Proteins/metabolism
5.
Cell Rep ; 27(10): 3006-3018.e4, 2019 06 04.
Article in English | MEDLINE | ID: mdl-31167144

ABSTRACT

Gut mesenchyme provides key stem cell niche signals such as Wnt ligands, but how these signals are regulated is unclear. Because Hedgehog (Hh) signaling is critical for gut mesenchymal development and tumorigenesis, we investigated Hh-mediated mechanisms by analyzing mice deleted for key negative regulators of Hh signaling, Sufu and/or Spop, in the gut mesenchyme, and demonstrated their dosage-dependent roles. Although these mutants exhibit abnormal mesenchymal cell growth and functionally defective muscle layers, villification is completed with proper mesenchymal clustering, implying a permissive role for Hh signaling. These mesenchymal defects are partially rescued by Gli2 reduction. Consistent with increased epithelial proliferation caused by abnormal Hh activation in development, Sufu reduction promotes intestinal tumorigenesis, whereas Gli2 heterozygosity suppresses it. Our analyses of chromatin and GLI2 binding genomic regions reveal its transcriptional regulation of stem cell niche signals through enhancers, providing mechanistic insight into the intestinal stem cell niche in development and tumorigenesis.


Subject(s)
Cell Transformation, Neoplastic , Intestine, Small/metabolism , Repressor Proteins/metabolism , Ubiquitin-Protein Ligase Complexes/metabolism , Zinc Finger Protein Gli2/metabolism , Actins/metabolism , Animals , Cell Proliferation , Embryo, Mammalian/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Hedgehog Proteins/metabolism , Intestine, Small/growth & development , Intestine, Small/pathology , Mice , Mice, Knockout , Microfilament Proteins/metabolism , Muscle Contraction , Muscle Proteins/metabolism , Muscles/metabolism , Muscles/physiology , Repressor Proteins/deficiency , Repressor Proteins/genetics , Signal Transduction , Stem Cell Niche , Transforming Growth Factor beta/metabolism , Ubiquitin-Protein Ligase Complexes/deficiency , Ubiquitin-Protein Ligase Complexes/genetics , Wnt Proteins/metabolism , Zinc Finger Protein Gli2/genetics
6.
Am J Physiol Gastrointest Liver Physiol ; 315(1): G1-G11, 2018 07 01.
Article in English | MEDLINE | ID: mdl-29517926

ABSTRACT

Gone are the days when enteric glial cells (EGC) were considered merely satellites of enteric neurons. Like their brain counterpart astrocytes, EGC express an impressive number of receptors for neurotransmitters and intercellular messengers, thereby contributing to neuroprotection and to the regulation of neuronal activity. EGC also produce different soluble factors that regulate neighboring cells, among which are intestinal epithelial cells. A better understanding of EGC response to an inflammatory environment, often referred to as enteric glial reactivity, could help define the physiological role of EGC and the importance of this reactivity in maintaining gut functions. In chronic inflammatory disorders of the gut such as Crohn's disease (CD) and ulcerative colitis, EGC exhibit abnormal phenotypes, and their neighboring cells are dysfunctional; however, it remains unclear whether EGC are only passive bystanders or active players in the pathophysiology of both disorders. The aim of the present study is to review the physiological roles and properties of EGC, their response to inflammation, and their role in the regulation of the intestinal epithelial barrier and to discuss the emerging concept of CD as an enteric gliopathy.


Subject(s)
Crohn Disease , Enteric Nervous System/immunology , Intestines , Neuroglia/immunology , Crohn Disease/immunology , Crohn Disease/physiopathology , Enteric Nervous System/physiopathology , Humans , Inflammation , Intestines/immunology , Intestines/innervation
7.
Sci Rep ; 6: 25203, 2016 05 03.
Article in English | MEDLINE | ID: mdl-27140063

ABSTRACT

In healthy gut enteric glial cells (EGC) are essential to intestinal epithelial barrier (IEB) functions. In Crohn's Disease (CD), both EGC phenotype and IEB functions are altered, but putative involvement of EGC in CD pathogenesis remains unknown and study of human EGC are lacking. EGC isolated from CD and control patients showed similar expression of glial markers and EGC-derived soluble factors (IL6, TGF-ß, proEGF, GSH) but CD EGC failed to increase IEB resistance and healing. Lipid profiling showed that CD EGC produced decreased amounts of 15-HETE, 18-HEPE, 15dPGJ2 and 11ßPGF2α as compared to healthy EGC. They also had reduced expression of the L-PGDS and AKR1C3 enzymes. Produced by healthy EGC, the 11ßPGF2 activated PPARγ receptor of intestinal epithelial cells to induce cell spreading and IEB wound repair. In addition to this novel healing mechanism our data show that CD EGC presented impaired ability to promote IEB functions through defect in L-PGDS-AKR1C3-11ßPGF2α dependent pathway.


Subject(s)
Crohn Disease/metabolism , Dinoprost/metabolism , Hydroxyeicosatetraenoic Acids/metabolism , Intestines/cytology , Adolescent , Adult , Aged , Aged, 80 and over , Aldo-Keto Reductase Family 1 Member C3/metabolism , Caco-2 Cells , Cells, Cultured , Coculture Techniques , Eicosapentaenoic Acid/analogs & derivatives , Eicosapentaenoic Acid/metabolism , Epithelial Cells/cytology , Epithelial Cells/metabolism , Female , Humans , Intestinal Mucosa/metabolism , Male , Middle Aged , Prostaglandin D2/analogs & derivatives , Prostaglandin D2/metabolism , Wound Healing , Young Adult
8.
Gastroenterology ; 150(1): 168-80, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26433161

ABSTRACT

BACKGROUND & AIMS: Enteric glial cells (EGCs) produce soluble mediators that regulate homeostasis and permeability of the intestinal epithelial barrier (IEB). We investigated the profile of polyunsaturated fatty acid (PUFA) metabolites produced by EGCs from rats and from patients with Crohn's disease (CD), compared with controls, along with the ability of one of these metabolites, 15-hydroxyeicosatetraenoic acid (15-HETE), to regulate the permeability of the IEB. METHODS: We isolated EGCs from male Sprague-Dawley rats, intestinal resections of 6 patients with CD, and uninflamed healthy areas of intestinal tissue from 6 patients who underwent surgery for colorectal cancer (controls). EGC-conditioned media was analyzed by high-sensitivity liquid-chromatography tandem mass spectrometry to determine PUFA signatures. We used immunostaining to identify 15-HETE-producing enzymes in EGCs and tissues. The effects of human EGCs and 15-HETE on permeability and transepithelial electrical resistance of the IEB were measured using Caco-2 cells; effects on signal transduction proteins were measured with immunoblots. Levels of proteins were reduced in Caco-2 cells using short-hairpin RNAs or proteins were inhibited pharmacologically. Rats were given intraperitoneal injections of 15-HETE or an inhibitor of 15-lipoxygenase (the enzyme that produces 15-HETE); colons were collected and permeability was measured. RESULTS: EGCs expressed 15-lipoxygenase-2 and produced high levels of 15-HETE, which increased IEB resistance and reduced IEB permeability. 15-HETE production was reduced in EGCs from patients with CD compared with controls. EGCs from patients with CD were unable to reduce the permeability of the IEB; the addition of 15-HETE restored permeability to levels of control tissues. Inhibiting 15-HETE production in rats increased the permeability of the IEB in colon tissues. We found that 15-HETE regulates IEB permeability by inhibiting an adenosine monophosphate-activated protein kinase and increasing expression of zonula occludens-1. CONCLUSIONS: Enteric glial cells from patients with CD have reduced production of 15-HETE, which controls IEB permeability by inhibiting adenosine monophosphate-activated protein kinase and increasing expression of zonula occludens-1.


Subject(s)
Cell Membrane Permeability/physiology , Crohn Disease/metabolism , Hydroxyeicosatetraenoic Acids/metabolism , Neuroglia/metabolism , Analysis of Variance , Animals , Blotting, Western , Caco-2 Cells/metabolism , Cells, Cultured , Disease Models, Animal , Humans , Immunohistochemistry , Intestinal Mucosa/cytology , Intestinal Mucosa/metabolism , Male , Random Allocation , Rats , Rats, Sprague-Dawley , Reference Values
9.
J Neuroinflammation ; 11: 202, 2014 Dec 12.
Article in English | MEDLINE | ID: mdl-25497784

ABSTRACT

BACKGROUND: Evidence continues to mount concerning the importance of the enteric nervous system (ENS) in controlling numerous intestinal functions in addition to motility and epithelial functions. Nevertheless, little is known concerning the direct participation of the ENS in the inflammatory response of the gut during infectious or inflammatory insults. In the present study we analyzed the ENS response to bacterial lipopolysaccharide, in particular the production of a major proinflammatory cytokine, tumor necrosis factor-alpha (TNF-α). METHODS: TNF-α expression (measured by qPCR, quantitative Polymerase Chain Reaction) and production (measured by ELISA) were measured in human longitudinal muscle-myenteric plexus (LMMP) and rat ENS primary cultures (rENSpc). They were either treated or not treated with lipopolysaccharide (LPS) in the presence or not of electrical field stimulation (EFS). Activation of extracellular signal-regulated kinase (ERK) and 5'-adenosine monophosphate-activated protein kinase (AMPK) pathways was analyzed by immunocytochemistry and Western blot analysis. Their implications were studied using specific inhibitors (U0126, mitogen-activated protein kinase kinase, MEK, inhibitor and C compound, AMPK inhibitor). We also analyzed toll-like receptor 2 (TLR2) expression and interleukin-6 (IL-6) production after LPS treatment simultaneously with EFS or TNF-α-neutralizing antibody. RESULTS: Treatment of human LMMP or rENSpc with LPS induced an increase in TNF-α production. Activation of the ENS by EFS significantly inhibited TNF-α production. This regulation occurred at the transcriptional level. Signaling analyses showed that LPS induced activation of ERK but not AMPK, which was constitutively activated in rENSpc neurons. Both U0126 and C compound almost completely prevented LPS-induced TNF-α production. In the presence of LPS, EFS inhibited the ERK and AMPK pathways. In addition, we demonstrated using TNF-α-neutralizing antibody that LPS-induced TNF-α production increased TLR2 expression and reduced IL-6 production. CONCLUSIONS: Our results show that LPS induced TNF-α production by enteric neurons through activation of the canonical ERK pathway and also in an AMPK-dependent manner. ENS activation through the inhibition of these pathways decreased TNF-α production, thereby modulating the inflammatory response induced by endotoxin.


Subject(s)
Enteric Nervous System/metabolism , Inflammation Mediators/metabolism , Lipopolysaccharides/toxicity , Neurons/metabolism , Animals , Cells, Cultured , Enteric Nervous System/drug effects , Humans , Neurons/drug effects , Organ Culture Techniques , Rats , Rats, Sprague-Dawley , Tumor Necrosis Factor-alpha/biosynthesis
10.
Dig Liver Dis ; 45(6): 487-92, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23466186

ABSTRACT

BACKGROUND: Probe-based confocal laser endomicroscopy enables microscopic examination of the digestive mucosa. AIMS: (1) To identify and validate quantitative endomicroscopic criteria for evaluation of the colonic mucosa and (2) to compare these criteria between healthy and Crohn's disease patients in clinical remission. METHODS: Six healthy controls and ten Crohn's disease patients in clinical remission were included in this prospective study. Methylene blue-stained biopsies of the right colon and corresponding endomicroscopic images were analyzed. Major axis, minor axis, and major axis/minor axis ratio of crypt lumens were quantified. RESULTS: Quantitative assessment was performed on 21 ± 4 crypt lumens per patient. Major axis/minor axis ratio values measured with endomicroscopy or methylene blue-stained biopsies were linearly correlated (r=0.63, p=0.01). All macroscopically inflamed mucosa had values of major axis/minor axis ratio higher than the median of controls. Interestingly, 50% (3/6) of Crohn's disease patients with macroscopically normal mucosa had also a higher ratio than pooled controls. Histological analysis showed that 6/7 patients with major axis/minor axis ratio superior to 1.7 had microscopic inflammation. CONCLUSION: Probe-based confocal laser endomicroscopy allows quantitative analysis of colonic pit structure. Endomicroscopic analysis of major axis/minor axis ratio allows the detection of microscopic residual inflammation with greater accuracy than standard endoscopy in Crohn's disease patients in clinical remission.


Subject(s)
Colon/pathology , Crohn Disease/pathology , Intestinal Mucosa/pathology , Microscopy, Confocal/methods , Adolescent , Adult , Aged , Biopsy , Case-Control Studies , Evaluation Studies as Topic , Female , Humans , Inflammation/pathology , Male , Middle Aged , Prospective Studies , Young Adult
11.
Gastrointest Endosc ; 73(5): 1029-34, 2011 May.
Article in English | MEDLINE | ID: mdl-21521570

ABSTRACT

BACKGROUND: Better understanding of the pathophysiological mechanisms involved in severe dysmotility disorders is crucial to improve patient management and identify novel therapeutic targets. Recent studies suggested that endoscopic full-thickness biopsies (eFTBs) could be developed as an alternative to surgical biopsies. However, currently it remains unknown whether eFTBs would allow myenteric plexus analysis on whole mounts and the evaluation of neuromuscular transmission. OBJECTIVE: To determine with eFTB specimens the ability to analyze on whole mounts the key parameters of the myenteric plexus, ie, ganglia and neurons, and to perform functional evaluation of neuromuscular transmission. DESIGN: An experimental pilot study in 6 pigs was conducted in accordance with French institutional guidelines. INTERVENTION: Under general anesthesia, pigs underwent a rectosigmoidoscopy. In each pig, an eFTB was performed at 25, 30, and 35 cm from the anal margin with an EMR-based technique. Tissue specimens were immediately processed for immunohistochemical and/or functional ex vivo analysis of neuromuscular transmission. In 2 pigs, over-the-scope clips were used to seal the perforation. MAIN OUTCOME MEASUREMENTS: Feasibility of obtaining specimens containing myenteric plexus and muscularis propria, quantitative and standardized immunohistochemical evaluation of ganglia and myenteric neurons, ex vivo assessment of neuromuscular transmission and its pharmacology, and closure rate (ancillary study). RESULTS: Adequate tissue specimens were obtained in 100% of the procedures, on average, in 6±2 minutes. Immunohistochemical analysis of a whole mount of the myenteric plexus showed that each eFTB contained 14±5 ganglia and 1562±1066 myenteric neurons. In circular muscle strips, electrical field stimulation or exposure to a pharmacological agent induced a specific tissue response. A successful closure was achieved in 50% of cases. LIMITATIONS: Nonsurvival study; safety of the procedure needs to be specifically assessed and compared with recently published data. CONCLUSIONS: We demonstrate, for the first time, that full-thickness biopsy specimens obtained by using an endoscopic approach allow the performance of a precise study of the ENS phenotype on whole mounts of the myenteric plexus and the performance of functional studies such as evaluation of neuromuscular transmission. However, further studies are warranted to identify the optimal and safest endoscopic procedure before application of eFTB in humans.


Subject(s)
Biopsy/methods , Colon/innervation , Colonoscopy/methods , Myenteric Plexus/physiology , Nerve Tissue Proteins/metabolism , Neuromuscular Junction/physiology , Synaptic Transmission/physiology , Animals , Colon/cytology , Colon/physiology , Female , Immunohistochemistry , Intestinal Mucosa/cytology , Intestinal Mucosa/innervation , Intestinal Mucosa/metabolism , Male , Muscle Contraction , Muscle, Smooth/innervation , Muscle, Smooth/physiology , Myenteric Plexus/ultrastructure , Neurofilament Proteins/metabolism , Neuromuscular Junction/ultrastructure , Phosphoproteins/metabolism , Photomicrography , Swine
SELECTION OF CITATIONS
SEARCH DETAIL
...