Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 427
Filter
1.
Genetics ; 222(4)2022 11 30.
Article in English | MEDLINE | ID: mdl-36218393

ABSTRACT

Transcription factors play important roles in the development of the intestinal epithelium and its ability to respond to endocrine, nutritional, and microbial signals. Hepatocyte nuclear factor 4 family nuclear receptors are liganded transcription factors that are critical for the development and function of multiple digestive organs in vertebrates, including the intestinal epithelium. Zebrafish have 3 hepatocyte nuclear factor 4 homologs, of which, hnf4a was previously shown to mediate intestinal responses to microbiota in zebrafish larvae. To discern the functions of other hepatocyte nuclear factor 4 family members in zebrafish development and intestinal function, we created and characterized mutations in hnf4g and hnf4b. We addressed the possibility of genetic redundancy amongst these factors by creating double and triple mutants which showed different rates of survival, including apparent early lethality in hnf4a; hnf4b double mutants and triple mutants. RNA sequencing performed on digestive tracts from single and double mutant larvae revealed extensive changes in intestinal gene expression in hnf4a mutants that were amplified in hnf4a; hnf4g mutants, but limited in hnf4g mutants. Changes in hnf4a and hnf4a; hnf4g mutants were reminiscent of those seen in mice including decreased expression of genes involved in intestinal function and increased expression of cell proliferation genes, and were validated using transgenic reporters and EdU labeling in the intestinal epithelium. Gnotobiotics combined with RNA sequencing also showed hnf4g has subtler roles than hnf4a in host responses to microbiota. Overall, phenotypic changes in hnf4a single mutants were strongly enhanced in hnf4a; hnf4g double mutants, suggesting a conserved partial genetic redundancy between hnf4a and hnf4g in the vertebrate intestine.


Subject(s)
Hepatocyte Nuclear Factor 4 , Intestinal Mucosa , Zebrafish Proteins , Zebrafish , Animals , Hepatocyte Nuclear Factor 4/genetics , Hepatocyte Nuclear Factor 4/physiology , Intestinal Mucosa/embryology , Intestinal Mucosa/metabolism , Intestines/embryology , Intestines/metabolism , Zebrafish/embryology , Zebrafish/genetics , Zebrafish Proteins/genetics , Zebrafish Proteins/physiology
2.
Nat Commun ; 13(1): 715, 2022 02 07.
Article in English | MEDLINE | ID: mdl-35132078

ABSTRACT

Organs are anatomically compartmentalised to cater for specialised functions. In the small intestine (SI), regionalisation enables sequential processing of food and nutrient absorption. While several studies indicate the critical importance of non-epithelial cells during development and homeostasis, the extent to which these cells contribute to regionalisation during morphogenesis remains unexplored. Here, we identify a mesenchymal-epithelial crosstalk that shapes the developing SI during late morphogenesis. We find that subepithelial mesenchymal cells are characterised by gradients of factors supporting Wnt signalling and stimulate epithelial growth in vitro. Such a gradient impacts epithelial gene expression and regional villus formation along the anterior-posterior axis of the SI. Notably, we further provide evidence that Wnt signalling directly regulates epithelial expression of Sonic Hedgehog (SHH), which, in turn, acts on mesenchymal cells to drive villi formation. Taken together our results uncover a mechanistic link between Wnt and Hedgehog signalling across different cellular compartments that is central for anterior-posterior regionalisation and correct formation of the SI.


Subject(s)
Hedgehog Proteins/metabolism , Intestinal Mucosa/metabolism , Intestine, Small/embryology , Mesenchymal Stem Cells/metabolism , Wnt Signaling Pathway/physiology , Animals , Cell Lineage , Gene Expression Regulation, Developmental , Hedgehog Proteins/genetics , Intestinal Mucosa/cytology , Intestinal Mucosa/embryology , Intestine, Small/cytology , Intestine, Small/metabolism , Mesenchymal Stem Cells/cytology , Mice , Morphogenesis , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Wnt Signaling Pathway/genetics
3.
Sci Rep ; 12(1): 2645, 2022 02 16.
Article in English | MEDLINE | ID: mdl-35173228

ABSTRACT

Initial nutritional stimulation is a key driving force for small intestinal maturation. In chick embryos, administration of l-glutamine (Gln) into the amniotic fluid stimulates early development of the small intestinal epithelium by promoting enterocyte differentiation. In this study, we evaluated the effects of intra-amniotic administration of Gln on enterocyte morphology and function, and elucidated a potential enteroendocrine pathway through which Gln stimulates small intestinal maturation. Our results show that Gln stimulation at embryonic day 17 significantly increased enterocyte and microvilli dimensions by 10 and 20%, respectively, within 48 h. Post-hatch, enterocytes and microvilli were 20% longer in Gln-treated chicks. Correspondingly, Gln stimulation significantly upregulated mRNA expression of brush border nutrient transporters PepT-1 and SGLT-1 and tight junction proteins TJP-1 and TJP-2, before and after hatch (P < 0.05). Since GLP-2 signaling from intestinal L-cells is associated with enterocyte growth, functionality and integrity, we examined the effects of Gln stimulation on mRNA expression of key hormones and receptors within this enteroendocrine pathway and found significant increases in GLP-2R, IGF-1 and IGF-1R expression before and after hatch (P < 0.05). In conclusion, our findings link primary nutrient stimulation in the developing small intestine with enterocyte morphological and functional maturation and enteroendocrine signaling.


Subject(s)
Animal Nutritional Physiological Phenomena/physiology , Chick Embryo/embryology , Enteroendocrine Cells/drug effects , Glutamine/administration & dosage , Glutamine/pharmacology , Intestinal Mucosa/embryology , Intestinal Mucosa/growth & development , Intestine, Small/embryology , Intestine, Small/growth & development , Amniotic Fluid , Animals , Chick Embryo/cytology , Chick Embryo/metabolism , Enteroendocrine Cells/metabolism , Enteroendocrine Cells/physiology , Glucagon-Like Peptide-2 Receptor/metabolism , Injections , Insulin-Like Growth Factor I/metabolism , Receptor, IGF Type 1/metabolism , Stimulation, Chemical
4.
PLoS One ; 16(10): e0259195, 2021.
Article in English | MEDLINE | ID: mdl-34710177

ABSTRACT

The intestinal epithelium requires self-renewal and differentiation in order to function and adapt to pathological diseases such as inflammatory bowel disease, short gut syndrome, and ulcers. The rodent Slfn3 protein and the human Slfn12 analog are known to regulate intestinal epithelial differentiation. Previous work utilizing a pan-Slfn3 knockout (KO) mouse model revealed sex-dependent gene expression disturbances in intestinal differentiation markers, metabolic pathways, Slfn family member mRNA expression, adaptive immune cell proliferation/functioning genes, and phenotypically less weight gain and sex-dependent changes in villus length and crypt depth. We have now created a Vil-Cre specific Slfn3KO (VC-Slfn3KO) mouse to further evaluate its role in intestinal differentiation. There were increases in Slfn1, Slfn2, Slfn4, and Slfn8 and decreases in Slfn5 and Slfn9 mRNA expression that were intestinal region and sex-specific. Differentiation markers, sucrase isomaltase (SI), villin 1, and dipeptidyl peptidase 4 and glucose transporters, glucose transporter 1 (Glut1), Glut2, and sodium glucose transporter 1 (SGLT1), were increased in expression in VC-Slfn3KO mice based on intestinal region and were also highly female sex-biased, except for SI in the ileum was also increased for male VC-Slfn3KO mice and SGLT1 was decreased for both sexes. Overall, the variations that we observed in these VC-Slfn3KO mice indicate a complex regulation of intestinal gene expression that is sex-dependent.


Subject(s)
Cell Cycle Proteins/genetics , Intestinal Mucosa/metabolism , Animals , Cell Differentiation , Cell Self Renewal , Female , Glucose Transport Proteins, Facilitative/genetics , Glucose Transport Proteins, Facilitative/metabolism , Intestinal Mucosa/cytology , Intestinal Mucosa/embryology , Male , Mice , Mice, Inbred C57BL , Sex Factors
5.
Science ; 373(6558)2021 08 27.
Article in English | MEDLINE | ID: mdl-34446580

ABSTRACT

The immune system has evolved in the face of microbial exposure. How maternal infection experienced at distinct developmental stages shapes the offspring immune system remains poorly understood. Here, we show that during pregnancy, maternally restricted infection can have permanent and tissue-specific impacts on offspring immunity. Mechanistically, maternal interleukin-6 produced in response to infection can directly impose epigenetic changes on fetal intestinal epithelial stem cells, leading to long-lasting impacts on intestinal immune homeostasis. As a result, offspring of previously infected dams develop enhanced protective immunity to gut infection and increased inflammation in the context of colitis. Thus, maternal infection can be coopted by the fetus to promote long-term, tissue-specific fitness, a phenomenon that may come at the cost of predisposition to inflammatory disorders.


Subject(s)
Colitis/immunology , Immunity , Interleukin-6/immunology , Intestines/immunology , Pregnancy Complications, Infectious/immunology , Th17 Cells/immunology , Yersinia pseudotuberculosis Infections/immunology , Animals , Candidiasis/immunology , Chromatin/metabolism , Epigenesis, Genetic , Epigenome , Female , Fetal Development , Gastrointestinal Microbiome/immunology , Gastrointestinal Microbiome/physiology , Interleukin-6/blood , Interleukin-6/pharmacology , Intestinal Mucosa/cytology , Intestinal Mucosa/embryology , Intestinal Mucosa/immunology , Intestines/embryology , Intestines/microbiology , Mice , Pregnancy , Prenatal Exposure Delayed Effects , Salmonella Infections, Animal/immunology , Stem Cells/immunology , Stem Cells/physiology , T-Lymphocyte Subsets/immunology
6.
Cell Rep ; 36(5): 109484, 2021 08 03.
Article in English | MEDLINE | ID: mdl-34348153

ABSTRACT

We lack a holistic understanding of the genetic programs orchestrating embryonic colon morphogenesis and governing damage response in the adult. A window into these programs is the transcriptomes of the epithelial and mesenchymal cell populations in the colon. Performing unbiased single-cell transcriptomic analyses of the developing mouse colon at different embryonic stages (embryonic day 14.5 [E14.5], E15.5, and E18.5), we capture cellular and molecular profiles of the stages before, during, and after the appearance of crypt structures, as well as in a model of adult colitis. The data suggest most adult lineages are established by E18.5. We find embryonic-specific gene expression profiles and cell populations that reappear in response to tissue damage. Comparison of the datasets from mice and human colitis suggests the processes are conserved. In this study, we provide a comprehensive single-cell atlas of the developing mouse colon and evidence for the reactivation of embryonic genes in disease.


Subject(s)
Colon/embryology , Colon/pathology , Gene Expression Profiling , Animals , Cell Differentiation , Colitis/genetics , Disease Models, Animal , Embryo, Mammalian/metabolism , Gene Expression Regulation, Developmental , Humans , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/pathology , Intestinal Mucosa/embryology , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Mesoderm/embryology , Mice, Inbred C57BL , Single-Cell Analysis
7.
Nat Immunol ; 22(6): 699-710, 2021 06.
Article in English | MEDLINE | ID: mdl-34040226

ABSTRACT

It is increasingly recognized that immune development within mucosal tissues is under the control of environmental factors during early life. However, the cellular mechanisms that underlie such temporally and regionally restrictive governance of these processes are unclear. Here, we uncover an extrathymic pathway of immune development within the colon that is controlled by embryonic but not bone marrow-derived macrophages, which determines the ability of these organs to receive invariant natural killer T (iNKT) cells and allow them to establish local residency. Consequently, early-life perturbations of fetal-derived macrophages result in persistent decreases of mucosal iNKT cells and is associated with later-life susceptibility or resistance to iNKT cell-associated mucosal disorders. These studies uncover a host developmental program orchestrated by ontogenically distinct macrophages that is regulated by microbiota, and they reveal an important postnatal function of macrophages that emerge in fetal life.


Subject(s)
Colitis/immunology , Intestinal Mucosa/immunology , Listeriosis/immunology , Macrophages/immunology , Mucosal-Associated Invariant T Cells/immunology , Animals , Cell Proliferation/genetics , Colitis/microbiology , Colitis/pathology , Colon/cytology , Colon/embryology , Colon/immunology , Colon/pathology , Cytokines/metabolism , Diphtheria Toxin/administration & dosage , Diphtheria Toxin/immunology , Disease Models, Animal , Embryo, Mammalian , Female , Gastrointestinal Microbiome/immunology , Gene Expression Regulation, Developmental/immunology , Germ-Free Life , Humans , Intestinal Mucosa/cytology , Intestinal Mucosa/embryology , Intestinal Mucosa/pathology , Listeriosis/microbiology , Listeriosis/pathology , Macrophages/metabolism , Male , Membrane Proteins/genetics , Mice , Mice, Knockout , RNA-Seq , Signal Transduction/genetics , Signal Transduction/immunology
8.
Anat Sci Int ; 96(3): 450-460, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33630273

ABSTRACT

Integral analysis of the development of the epithelium, mesenchyme, and smooth muscle cell (SMC) layers, i.e., the inner circular (IC) and outer longitudinal layers, as well as their relation with the mesentery is necessary to understand macroscopic gut development. We here focused on the proximal duodenum with the characteristic "C"-shaped loop and analyzed the duodenum down to the duodenojejunal flexure in C57BL/6J mouse embryos at embryonic days (E) 13.5, 15.5, and 17.5 by histomorphometric analysis. We examined the angle of the axis of the epithelial lumen, which was oval at E13.5 against the mesentery, along with the epithelial cell nuclear shape, the adjacent mesenchymal cell density in relation to the epithelial lumen axis, and the development of SMC layers. The luminal axis of the oval epithelial lumen at E13.5 rotated clockwise against the mesentery in the proximal duodenum. The shape of epithelial nuclei was longer and thinner at the long axis but shorter and broader at the short axis, whereas mesenchymal density was significantly lower in the area on the luminal long axis than that on the short axis. The number of SMC layers in the IC at E13.5, E15.5, and E17.5 showed a regional difference in relation to the mesentery, but no regional difference along the long axis of the duodenum. These findings suggest that epithelial lumen winding against the mesentery and the corresponding changes in the epithelial cell shape and surrounding mesenchymal density may be involved in the formation of the "C" loop of the proximal duodenum.


Subject(s)
Duodenum/embryology , Mesoderm/embryology , Myocytes, Smooth Muscle/cytology , Organogenesis/physiology , Animals , Intestinal Mucosa/embryology , Mice
9.
BMC Mol Cell Biol ; 22(1): 12, 2021 Feb 12.
Article in English | MEDLINE | ID: mdl-33579204

ABSTRACT

BACKGROUND: Enterocytes exert an absorptive and protective function in the intestine, and they encounter many different challenging factors such as feed, bacteria, and parasites. An intestinal epithelial in vitro model can help to understand how enterocytes are affected by these factors and contribute to the development of strategies against pathogens. RESULTS: The present study describes a novel method to culture and maintain primary chicken enterocytes and their characterization by immunofluorescence and biomolecular approaches. Starting from 19-day-old chicken embryos it was possible to isolate viable intestinal cell aggregates that can expand and produce a self-maintaining intestinal epithelial cell population that survives until 12 days in culture. These cells resulted positive in immunofluorescence to Cytokeratin 18, Zonula occludens 1, Villin, and Occludin that are common intestinal epithelial markers, and negative to Vimentin that is expressed by endothelial cells. Cells were cultured also on Transwell® permeable supports and trans-epithelial electrical resistance, was measured. This value gradually increased reaching 64 Ω*cm2 7 days after seeding and it remained stable until day 12. CONCLUSIONS: Based on these results it was confirmed that it is possible to isolate and maintain chicken intestinal epithelial cells in culture and that they can be suitable as in vitro intestinal model for further studies.


Subject(s)
Cell Culture Techniques/methods , Cell Separation/methods , Enterocytes/cytology , Intestinal Mucosa/cytology , Intestinal Mucosa/embryology , Animals , Cell Proliferation , Cells, Cultured , Chick Embryo , Chickens , Collagenases/metabolism , Culture Media , Embryonic Development , Intestinal Mucosa/enzymology , Trypsin/pharmacology
10.
Am J Physiol Gastrointest Liver Physiol ; 320(4): G506-G520, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33470182

ABSTRACT

The stem/progenitor cells of the developing intestine are biologically distinct from their adult counterparts. Here, we examine the microenvironmental cues that regulate the embryonic stem/progenitor population, focusing on the role of Notch pathway factor delta-like protein-1 (DLK1). mRNA-seq analyses of intestinal mesenchymal cells (IMCs) collected from embryonic day 14.5 (E14.5) or adult IMCs and a novel coculture system with E14.5 intestinal epithelial organoids were used. Following addition of recombinant DLK1 (rDLK) or Dlk1 siRNA (siDlk1), epithelial characteristics were compared using imaging, replating efficiency assays, qPCR, and immunocytochemistry. The intestinal phenotypes of littermate Dlk1+/+ and Dlk1-/- mice were compared using immunohistochemistry. Using transcriptomic analyses, we identified morphogens derived from the embryonic mesenchyme that potentially regulate the developing epithelial cells, to focus on Notch family candidate DLK1. Immunohistochemistry indicated that DLK1 was expressed exclusively in the intestinal stroma at E14.5 at the top of emerging villi, decreased after birth, and shifted to the intestinal epithelium in adulthood. In coculture experiments, addition of rDLK1 to adult IMCs inhibited organoid differentiation, whereas Dlk1 knockdown in embryonic IMCs increased epithelial differentiation to secretory lineage cells. Dlk1-/- mice had restricted Ki67+ cells in the villi base and increased secretory lineage cells compared with Dlk1+/+ embryos. Mesenchyme-derived DLK1 plays an important role in the promotion of epithelial stem/precursor expansion and prevention of differentiation to secretory lineages in the developing intestine.NEW & NOTEWORTHY Using a novel coculture system, transcriptomics, and transgenic mice, we investigated differential molecular signaling between the intestinal epithelium and mesenchyme during development and in the adult. We show that the Notch pathway factor delta-like protein-1 (DLK1) is stromally produced during development and uncover a new role for DLK1 in the regulation of intestinal epithelial stem/precursor expansion and differentiation to secretory lineages.


Subject(s)
Calcium-Binding Proteins/metabolism , Cell Communication , Cell Differentiation , Cell Proliferation , Embryonic Stem Cells/enzymology , Epithelial Cells/enzymology , Intestinal Mucosa/enzymology , Stromal Cells/enzymology , Animals , Calcium-Binding Proteins/deficiency , Calcium-Binding Proteins/genetics , Cell Lineage , Cells, Cultured , Coculture Techniques , Gene Expression Regulation, Developmental , Intestinal Mucosa/embryology , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Organoids , Secretory Pathway , Signal Transduction , Stem Cell Niche , Transcriptome
11.
Mol Nutr Food Res ; 65(7): e2000425, 2021 04.
Article in English | MEDLINE | ID: mdl-33465830

ABSTRACT

SCOPE: Human milk oligosaccharides (hMOs) can attenuate inflammation by modulating intestinal epithelial cells, but the mechanisms of action are not well-understood. Here, the effects of hMOs on tumor necrosis factor-α (TNF-α) induced inflammatory events in gut epithelial cells are studied. METHODS AND RESULTS: The modulatory effects of 2'-fucosyllactose, 3-fucosyllactose (3-FL), 6'-sialyllactose, lacto-N-tetraose, lacto-N-neotetraose (LNnT), lactodifucotetraose (LDFT), and lacto-N-triaose (LNT2) on immature (FHs 74 Int) and adult (T84) intestinal epithelial cells with or without TNF-α are determined. Interleukin-8 (IL-8) secretion in FHs 74 Int and T84 are quantified to determine hMO induced attenuation of inflammatory events by ELISA. 3-FL, LNnT, and LDFT significantly attenuate TNF-α induced inflammation in FHs 74 Int, while LNT2 induces IL-8 secretion in T84. In addition, microscale thermophoresis assays and ELISA are used to study the possible mechanisms of interaction between effective hMOs and tumor necrosis factor receptor 1 (TNFR1). 3-FL, LNnT, and LDFT exert TNFR1 ectodomain shedding while LNnT also shows binding affinity to TNFR1 with a Kd of 900 ± 660 nM. CONCLUSION: The findings indicate that specific hMO types attenuate TNF-α induced inflammation in fetal gut epithelial cells through TNFR1 in a hMO structure-dependent fashion suggest possibilities to apply hMOs in management of TNF-α dependent diseases.


Subject(s)
Intestinal Mucosa/cytology , Milk, Human/chemistry , Oligosaccharides/pharmacology , Receptors, Tumor Necrosis Factor, Type I/metabolism , Cell Line , Cell Survival , Gastroenteritis/drug therapy , Humans , Hydrolysis , Interleukin-8/metabolism , Intestinal Mucosa/drug effects , Intestinal Mucosa/embryology , Oligosaccharides/chemistry , Protein Domains , Receptors, Tumor Necrosis Factor, Type I/chemistry , Structure-Activity Relationship , Tumor Necrosis Factor-alpha/adverse effects
12.
Dev Cell ; 55(6): 771-783.e5, 2020 12 21.
Article in English | MEDLINE | ID: mdl-33290721

ABSTRACT

Human gut development requires the orchestrated interaction of differentiating cell types. Here, we generate an in-depth single-cell map of the developing human intestine at 6-10 weeks post-conception. Our analysis reveals the transcriptional profile of cycling epithelial precursor cells; distinct from LGR5-expressing cells. We propose that these cells may contribute to differentiated cell subsets via the generation of LGR5-expressing stem cells and receive signals from surrounding mesenchymal cells. Furthermore, we draw parallels between the transcriptomes of ex vivo tissues and in vitro fetal organoids, revealing the maturation of organoid cultures in a dish. Lastly, we compare scRNA-seq profiles from pediatric Crohn's disease epithelium alongside matched healthy controls to reveal disease-associated changes in the epithelial composition. Contrasting these with the fetal profiles reveals the re-activation of fetal transcription factors in Crohn's disease. Our study provides a resource available at www.gutcellatlas.org, and underscores the importance of unraveling fetal development in understanding disease.


Subject(s)
Crohn Disease/genetics , Intestinal Mucosa/metabolism , Transcriptome , Adolescent , Cells, Cultured , Child , Crohn Disease/metabolism , Humans , Intestinal Mucosa/embryology , RNA-Seq , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Single-Cell Analysis , Transcription Factors/genetics , Transcription Factors/metabolism
13.
Int J Mol Sci ; 21(21)2020 11 03.
Article in English | MEDLINE | ID: mdl-33153048

ABSTRACT

The intestine is an organ essential to organismal nutrient absorption, metabolic control, barrier function and immunoprotection. The Caenorhabditis elegans intestine consists of 20 cells harboring a dense intermediate filament network positioned below the apical plasma membrane that forms a junction-anchored sheath around the intestinal lumen. This evolutionarily conserved arrangement provides mechanical and overall stress-protection, and it serves as an important model for deciphering the role of intestinal architecture in metazoan biology. We recently reported that the loss-of-function mutation of the intestinal intermediate filament organizer IFO-1 perturbs this architecture, leading to reduced body size and reproduction. Here, we demonstrate that the IFO-1 mutation dramatically affects cholesterol metabolism. Mutants showed an increased sensitivity to cholesterol depletion, reduced cholesterol uptake, and cholesterol transfer to the gonads, which is also observed in worms completely lacking an intermediate filament network. Accordingly, we found striking similarities to transcriptome and lipidome profiles of a nuclear hormone receptor (NHR)-8 mutant. NHR-8 is homologous to mammalian LXR (liver X receptor) that serves as a sterol sensor and transcriptional regulator of lipid metabolism. Remarkably, increasing exogenous cholesterol partially rescues the developmental retardation in IFO-1 mutants. Our results uncover a novel link of the intestinal intermediate filament cytoskeleton to cholesterol metabolism that contributes to compromised growth and reproduction.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans , Cholesterol/metabolism , Intermediate Filament Proteins/genetics , Lipid Metabolism/genetics , Animals , Animals, Genetically Modified , Caenorhabditis elegans/drug effects , Caenorhabditis elegans/embryology , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/genetics , Cholesterol/pharmacology , Embryo, Nonmammalian , Gene Expression Regulation, Developmental/drug effects , Intermediate Filament Proteins/metabolism , Intermediate Filaments/metabolism , Intestinal Mucosa/embryology , Intestinal Mucosa/metabolism , Intestinal Mucosa/ultrastructure , Intestines/embryology , Intestines/physiology , Intestines/ultrastructure , Lipid Metabolism/drug effects , Lipidomics , Receptors, Cytoplasmic and Nuclear/physiology , Transcriptome/drug effects
14.
Life Sci ; 260: 118428, 2020 Nov 01.
Article in English | MEDLINE | ID: mdl-32931798

ABSTRACT

AIMS: The benefits of utilizing laboratory mice include low cost, ease of maintenance, and accessibility of molecular tools. However, the ages of experimental mice in the literature vary drastically. We hypothesized that there exists age-related variation in the murine small intestine across developmental stages. MATERIALS AND METHODS: Segments of small intestine were harvested from C57BL/6J mice of varying ages (E17 to 24 weeks; n = 3-4/group). Slides were analyzed for morphometric parameters, cell types, and crypt proliferation index (CPI). Secondary analysis comparing age-matched males and females (n = 4/group) was performed. Means were compared with Student's t-test and variance of proportions with the Chi-squared test to a significance of p < 0.05. KEY FINDINGS: There were small but significant differences including regional variation in villus height, which abolished when examining the small intestine as a whole. Sexually immature mice had increased CPI compared to mature animals. The most dramatic differences were seen in mice at weaning, which demonstrated shallower crypts, increased CPI, fewer Paneth and goblet cells, and more enterochromaffin cells. Examination of embryonic intestine revealed an underdeveloped mucosa lacking differentiated cells. There were minimal differences when comparing age-matched males and females. SIGNIFICANCE: Small, but statistically significant differences in villus height, crypt depth, and crypt proliferation are present in mice across early developmental stages. Mice at weaning exhibit variation in crypt-villus cell composition compared to older animals, which may explain the propensity for certain intestinal conditions in the very young. Investigators studying the GI mucosa should employ consistent age-matching in order to allow direct comparison between studies.


Subject(s)
Intestinal Mucosa/cytology , Intestinal Mucosa/growth & development , Intestine, Small/growth & development , Age Factors , Animals , Female , Intestinal Mucosa/embryology , Intestine, Small/cytology , Intestine, Small/embryology , Male , Mice, Inbred C57BL , Microscopy, Electron, Scanning
15.
Cell Mol Gastroenterol Hepatol ; 10(3): 623-637, 2020.
Article in English | MEDLINE | ID: mdl-32474164

ABSTRACT

BACKGROUND AND AIMS: The Extra-Uterine Environment for Neonatal Development (EXTEND) aims to avoid the complications of prematurity, such as NEC. Our goal was to determine if bowel development occurs normally in EXTEND-supported lambs, with specific emphasis on markers of immaturity associated with NEC. METHODS: We compared terminal ileum from 17 pre-term lambs supported on EXTEND for 2- 4 weeks to bowel from age-matched fetal lambs that developed in utero. We evaluated morphology, markers of epithelial integrity and maturation, enteric nervous system structure, and bowel motility. RESULTS: EXTEND-supported lamb ileum had normal villus height, crypt depth, density of mucin-containing goblet cells, and enteric neuron density. Expression patterns for I-FABP, activated caspase-3 and EGFR were normal in bowel epithelium. Transmural resistance assessed in Ussing chambers was normal. Bowel motility was also normal as assessed by ex vivo organ bath and video imaging. However, Peyer's patch organization did not occur normally in EXTEND ileum, resulting in fewer circulating B cells in experimental animals. CONCLUSION: EXTEND supports normal ileal epithelial and enteric nervous system maturation in pre-term lambs. The classic morphologic changes and cellular expression profiles associated with NEC are not seen. However, immune development within the EXTEND supported lamb bowel does not progress normally.


Subject(s)
Enterocolitis, Necrotizing/prevention & control , Extracorporeal Membrane Oxygenation/methods , Fetal Organ Maturity/immunology , Ileum/embryology , Premature Birth/therapy , Animals , Animals, Newborn , Disease Models, Animal , Enterocolitis, Necrotizing/immunology , Female , Fetus/immunology , Humans , Ileum/immunology , Infant, Newborn , Intestinal Mucosa/embryology , Intestinal Mucosa/immunology , Premature Birth/immunology , Sheep , Umbilical Cord/blood supply
17.
J Vis Exp ; (153)2019 11 15.
Article in English | MEDLINE | ID: mdl-31789309

ABSTRACT

At the end of the suckling period, many mammalian species undergo major changes in the intestinal epithelium that are associated with the capability to digest solid food. This process is termed suckling-to-weaning transition and results in the replacement of neonatal epithelium with adult epithelium which goes hand in hand with metabolic and morphological adjustments. These complex developmental changes are the result of a genetic program that is intrinsic to the intestinal epithelial cells but can, to some extent, be modulated by extrinsic factors. Prolonged culture of mouse primary intestinal epithelial cells from late fetal period, recapitulates suckling-to-weaning transition in vitro. Here, we describe a detailed protocol for mouse fetal intestinal organoid culture best suited to model this process in vitro. We describe several useful assays designed to monitor the change of intestinal functions associated with suckling-to-weaning transition over time. Additionally, we include an example of an extrinsic factor that is capable to affect suckling-to-weaning transition in vivo, as a representation of modulating the timing of suckling-to-weaning transition in vitro. This in vitro approach can be used to study molecular mechanisms of the suckling-to-weaning transition as well as modulators of this process. Importantly, with respect to animal ethics in research, replacing in vivo models by this in vitro model contributes to refinement of animal experiments and possibly to a reduction in the use of animals to study gut maturation processes.


Subject(s)
Animals, Suckling/physiology , Fetal Development/physiology , Intestinal Mucosa/embryology , Intestinal Mucosa/physiology , Organoids/embryology , Organoids/physiology , Animals , Cells, Cultured , Epithelial Cells/physiology , Intestinal Mucosa/cytology , Mice , Organ Culture Techniques , Organoids/cytology , Weaning
18.
Curr Opin Immunol ; 58: 83-88, 2019 06.
Article in English | MEDLINE | ID: mdl-31146182

ABSTRACT

The intestinal epithelium is the outermost cellular layer that separates the body from the gut lumen. The integrity of this protective mucosal barrier is crucial and maintained by specialized cells-intraepithelial lymphocytes (IEL). Much research has been conducted on these cells and our overall understanding of them is increasing rapidly. In this review we focus on the TCRαß+ subset of CD8αα IEL. We discuss recent studies that shed light on the development, ontogeny, maintenance, and functional characteristics of CD8αα IEL, and highlight yet unanswered questions for future studies.


Subject(s)
CD8 Antigens/immunology , CD8-Positive T-Lymphocytes/immunology , Intraepithelial Lymphocytes/immunology , Receptors, Antigen, T-Cell, alpha-beta/immunology , T-Lymphocyte Subsets/immunology , Animals , CD8 Antigens/metabolism , CD8-Positive T-Lymphocytes/metabolism , Humans , Intestinal Mucosa/embryology , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Intraepithelial Lymphocytes/metabolism , Morphogenesis/immunology , Receptors, Antigen, T-Cell, alpha-beta/metabolism , T-Lymphocyte Subsets/metabolism , Thymus Gland/embryology , Thymus Gland/immunology , Thymus Gland/metabolism
19.
Nat Commun ; 10(1): 1071, 2019 03 06.
Article in English | MEDLINE | ID: mdl-30842416

ABSTRACT

Lgr5+ stem cells are crucial to gut epithelium homeostasis; however, how these cells are maintained is not fully understood. Zinc finger HIT-type containing 1 (Znhit1) is an evolutionarily conserved subunit of the SRCAP chromosome remodeling complex. Currently, the function of Znhit1 in vivo and its working mechanism in the SRCAP complex are unknown. Here we show that deletion of Znhit1 in intestinal epithelium depletes Lgr5+ stem cells thus disrupts intestinal homeostasis postnatal establishment and maintenance. Mechanistically, Znhit1 incorporates histone variant H2A.Z into TSS region of genes involved in Lgr5+ stem cell fate determination, including Lgr5, Tgfb1 and Tgfbr2, for subsequent transcriptional regulation. Importantly, Znhit1 promotes the interaction between H2A.Z and YL1 (H2A.Z chaperone) by controlling YL1 phosphorylation. These results demonstrate that Znhit1/H2A.Z is essential for Lgr5+ stem cell maintenance and intestinal homeostasis. Our findings identified a dominant role of Znhit1/H2A.Z in controlling mammalian organ development and tissue homeostasis in vivo.


Subject(s)
Carrier Proteins/metabolism , Histones/metabolism , Intestinal Mucosa/metabolism , Repressor Proteins/metabolism , Stem Cells/physiology , Animals , Carrier Proteins/genetics , Cell Differentiation/physiology , Embryo, Mammalian , Epithelial Cells/physiology , Female , Intestinal Mucosa/cytology , Intestinal Mucosa/embryology , Mice , Mice, Inbred C57BL , Mice, Knockout , Organogenesis/physiology , Organoids , Phosphorylation , Receptors, G-Protein-Coupled/metabolism , Tissue Culture Techniques
20.
Immunity ; 50(2): 462-476.e8, 2019 02 19.
Article in English | MEDLINE | ID: mdl-30770246

ABSTRACT

Although the fetal immune system is considered tolerogenic, preterm infants can suffer from severe intestinal inflammation, including necrotizing enterocolitis (NEC). Here, we demonstrate that human fetal intestines predominantly contain tumor necrosis factor-α (TNF-α)+CD4+CD69+ T effector memory (Tem) cells. Single-cell RNA sequencing of fetal intestinal CD4+ T cells showed a T helper 1 phenotype and expression of genes mediating epithelial growth and cell cycling. Organoid co-cultures revealed a dose-dependent, TNF-α-mediated effect of fetal intestinal CD4+ T cells on intestinal stem cell (ISC) development, in which low T cell numbers supported epithelial development, whereas high numbers abrogated ISC proliferation. CD4+ Tem cell frequencies were higher in inflamed intestines from preterm infants with NEC than in healthy infant intestines and showed enhanced TNF signaling. These findings reveal a distinct population of TNF-α-producing CD4+ T cells that promote mucosal development in fetal intestines but can also mediate inflammation upon preterm birth.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Fetus/immunology , Immunologic Memory/immunology , Intestines/immunology , Tumor Necrosis Factor-alpha/immunology , Animals , CD4-Positive T-Lymphocytes/metabolism , Epithelial Cells/cytology , Epithelial Cells/immunology , Epithelial Cells/metabolism , Female , Fetus/metabolism , Humans , Infant, Newborn , Intestinal Mucosa/embryology , Intestinal Mucosa/growth & development , Intestinal Mucosa/immunology , Intestines/embryology , Intestines/growth & development , Mice, Inbred C57BL , Pregnancy , Stem Cells/cytology , Stem Cells/immunology , Stem Cells/metabolism , Th1 Cells/immunology , Th1 Cells/metabolism , Tumor Necrosis Factor-alpha/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...