Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 771
Filter
1.
Sci Rep ; 13(1): 13701, 2023 08 22.
Article in English | MEDLINE | ID: mdl-37607995

ABSTRACT

To close the gap between ultra-hygienic research mouse models and the much more environmentally exposed conditions of humans, we have established a system where laboratory mice are raised under a full set of environmental factors present in a naturalistic, farmyard-type habitat-a process we have called feralization. In previous studies we have shown that feralized (Fer) mice were protected against colorectal cancer when compared to conventionally reared laboratory mice (Lab). However, the protective mechanisms remain to be elucidated. Disruption of the protective intestinal barrier is an acknowledged player in colorectal carcinogenesis, and in the current study we assessed colonic mucosal barrier properties in healthy, feralized C57BL/6JRj male mice. While we found no effect of feralization on mucus layer properties, higher expression of genes encoding the mucus components Fcgbp and Clca1 still suggested mucus enforcement due to feralization. Genes encoding other proteins known to be involved in bacterial defense (Itln1, Ang1, Retnlb) and inflammatory mechanisms (Zbp1, Gsdmc2) were also higher expressed in feralized mice, further suggesting that the Fer mice have an altered intestinal mucosal barrier. These findings demonstrate that microbial experience conferred by housing in a farmyard-type environment alters the intestinal barrier properties in mice possibly leading to a more robust protection against disease. Future studies to unravel regulatory roles of feralization on intestinal barrier should aim to conduct proteomic analyses and in vivo performance of the feralized mice intestinal barrier.


Subject(s)
Animals, Laboratory , Colon , Farms , Housing, Animal , Intestinal Mucosa , Laboratories , Animals , Female , Male , Mice , Animals, Laboratory/microbiology , Animals, Laboratory/physiology , Colon/microbiology , Colon/physiology , Gastrointestinal Microbiome , Gene Expression Regulation , Ileum/microbiology , Ileum/physiology , Intestinal Mucosa/anatomy & histology , Intestinal Mucosa/growth & development , Intestinal Mucosa/microbiology , Intestinal Mucosa/physiology , Mice, Inbred C57BL
2.
J Appl Microbiol ; 133(2): 503-514, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35429125

ABSTRACT

AIM: The intestinal microbiota contributes to infant's intestine homeostasis. This study aimed to analyse how probiotics derived from breast milk promote infant intestinal development in rat pups. METHODS AND RESULTS: The effect of potential probiotics derived from breast milk on development of intrauterine growth retardation (IUGR) newborn rats' intestine was investigated. Limosilactobacillus oris ML-329 and Lacticaseibacillus paracasei ML-446 exhibited good hydrophobicity percentages (p < 0.05). ML-446 showed a significant effect on intestinal length and weight (p < 0.05). Meanwhile, the villus height of the IUGR newborn rats fed with ML-329 was significantly higher compared with those fed with Lacticaseibacillus rhamnosus GG (p < 0.05). Moreover, ML-329 and ML-446 both significantly stimulated the proliferation and differentiation of intestinal epithelial cells by increasing the number of ki67-positive cells, goblet cells, and lysozyme-positive Paneth cells (p < 0.05) through Wnt and Notch pathway. CONCLUSIONS: The proliferation and differentiation stimulating effects of ML-329 and ML-446 on IECs in the jejunum, ileum, and colon were mediated by activating the Wnt pathway with increased expression of wnt, lrp5, and ß-catenin genes and accumulation of ß-catenin, and by downregulating the Notch signalling pathway with decreased expression of the activated notch protein. SIGNIFICANCE AND IMPACT OF THE STUDY: Lactobacillus could facilitate IUGR rat pups' intestinal development and enhance the proliferation of Paneth cells and goblet cells. These findings provide further insights into promotion of the intestinal development by breast milk-derived beneficial microbes in early life of the IUGR newborn rats.


Subject(s)
Fetal Growth Retardation , Intestinal Mucosa , Lactobacillus , Milk, Human , Animals , Female , Humans , Intestinal Mucosa/growth & development , Lactobacillus/metabolism , Milk, Human/microbiology , Rats , beta Catenin/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.
Science ; 375(6576): eaaw9021, 2022 Jan 07.
Article in English | MEDLINE | ID: mdl-34990240

ABSTRACT

Epithelial organoids are stem cell­derived tissues that approximate aspects of real organs, and thus they have potential as powerful tools in basic and translational research. By definition, they self-organize, but the structures formed are often heterogeneous and irreproducible, which limits their use in the lab and clinic. We describe methodologies for spatially and temporally controlling organoid formation, thereby rendering a stochastic process more deterministic. Bioengineered stem cell microenvironments are used to specify the initial geometry of intestinal organoids, which in turn controls their patterning and crypt formation. We leveraged the reproducibility and predictability of the culture to identify the underlying mechanisms of epithelial patterning, which may contribute to reinforcing intestinal regionalization in vivo. By controlling organoid culture, we demonstrate how these structures can be used to answer questions not readily addressable with the standard, more variable, organoid models.


Subject(s)
Intestinal Mucosa/growth & development , Organogenesis , Organoids/growth & development , Tissue Engineering , Animals , Cell Differentiation , Cell Shape , Epithelial Cells/cytology , Hydrogels , Intestinal Mucosa/anatomy & histology , Intestinal Mucosa/cytology , Intestinal Mucosa/metabolism , Mice , Organoids/anatomy & histology , Organoids/cytology , Organoids/metabolism , Paneth Cells/cytology , Receptors, Notch/metabolism , Signal Transduction , Stem Cells/cytology , Stem Cells/physiology , Tissue Culture Techniques , YAP-Signaling Proteins/metabolism
5.
Oxid Med Cell Longev ; 2021: 9424582, 2021.
Article in English | MEDLINE | ID: mdl-34938384

ABSTRACT

Parkinson's disease (PD) is a common degenerative disease of the central nervous system. Although some drugs can alleviate the progress of PD, their long-term use will lead to complications, so it is still necessary to find new drugs to delay or cure PD effectively. In view of the difficulty in developing new drugs, it is imperative to discover new functions of existing compounds that could be used to treat PD. In this study, 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) was used to induce PD symptoms in a mouse model. Subsequently, these mice were treated with the antibiotic ceftriaxone. Ceftriaxone alleviated the behavioural and neuropathological changes induced by MPTP, downregulated the expression of glial fibrillary acidic protein (GFAP) and ionised calcium-binding adapter molecule 1 (Iba1) as markers of astroglia and microglia, respectively, and reduced the expression of neuroinflammation-related Toll-like receptor 4 (TLR4), myeloid differentiation primary response 88 (MyD88), and phosphorylated nuclear factor kappa-B (p-NF-κB)/NF-κB in the brain of PD mice. In addition, ceftriaxone reduced the abundance of pathogenic bacteria of the genus Proteus and increased the abundance of probiotic Akkermansia. Finally, ceftriaxone treatment increased the expression of the tight junction proteins zona occludens-1(ZO-1) and occludin in the colon, decreased the expression of the inflammation-related proteins TLR4, MyD88, and NF-κB in the colon, and decreased the serum concentration of the proinflammatory cytokines interleukin-1ß (IL-1ß), IL-6, and tumour necrosis factor-α (TNF-α). These results indicate that ceftriaxone had a neuroprotective effect on MPTP-induced PD mice, and its neuroprotective effect could be through regulating inflammation and intestinal microbiota. While we showed that ceftriaxone exerts a neuroprotective effect in an MPTP-induced PD mouse model, our findings are limited to the short-term effects of ceftriaxone. Additional work using transgenic mice is required to determine the long-term effects of ceftriaxone. In addition, the dose and frequency of ceftriaxone use should be evaluated.


Subject(s)
1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/adverse effects , Ceftriaxone/administration & dosage , Gastrointestinal Microbiome , Intestinal Mucosa/drug effects , Neuroinflammatory Diseases/drug therapy , Neuroprotective Agents/administration & dosage , Parkinson Disease/drug therapy , Animals , Anti-Bacterial Agents/administration & dosage , Disease Models, Animal , Intestinal Mucosa/growth & development , Intestinal Mucosa/microbiology , Male , Mice , Mice, Inbred C57BL , Neuroinflammatory Diseases/etiology , Neuroinflammatory Diseases/metabolism , Neuroinflammatory Diseases/pathology , Neurotoxins/adverse effects , Parkinson Disease/etiology , Parkinson Disease/metabolism , Parkinson Disease/pathology
6.
Development ; 148(21)2021 11 01.
Article in English | MEDLINE | ID: mdl-34758081

ABSTRACT

The developmental programs that build and sustain animal forms also encode the capacity to sense and adapt to the microbial world within which they evolved. This is abundantly apparent in the development of the digestive tract, which typically harbors the densest microbial communities of the body. Here, we review studies in human, mouse, zebrafish and Drosophila that are revealing how the microbiota impacts the development of the gut and its communication with the nervous system, highlighting important implications for human and animal health.


Subject(s)
Brain-Gut Axis/physiology , Gastrointestinal Microbiome/physiology , Gastrointestinal Tract/growth & development , Animals , Cell Lineage , Enteric Nervous System/cytology , Enteric Nervous System/growth & development , Enteric Nervous System/physiology , Gastrointestinal Motility , Gastrointestinal Tract/innervation , Gastrointestinal Tract/microbiology , Humans , Intestinal Mucosa/cytology , Intestinal Mucosa/growth & development , Neurons/cytology , Neurons/physiology
7.
PLoS One ; 16(8): e0256774, 2021.
Article in English | MEDLINE | ID: mdl-34437645

ABSTRACT

Cross talk between different signaling pathways is thought to be important for regulation of homeostasis of, as well as oncogenesis of, the intestinal epithelium. Expression of an active form of K-Ras specifically in intestinal epithelial cells (IECs) of mice (IEC-RasDA mice) resulted in the development of hyperplasia in the small intestine and colon of mice. IEC-RasDA mice also manifested the increased proliferation of IECs. In addition, the number of goblet cells markedly increased, while that of Paneth cells decreased in IEC-RasDA mice. Development of intestinal organoids was markedly enhanced for IEC-RasDA mice compared with control mice. Whereas, the expression of Wnt target genes was significantly reduced in the in intestinal crypts from IEC-RasDA mice compared with that apparent for the control. Our results thus suggest that K-Ras promotes the proliferation of IECs as well as generation of goblet cells. By contrast, Ras counter-regulates the Wnt signaling and thereby contribute to the proper regulation of intestinal epithelial cell homeostasis.


Subject(s)
Cell Proliferation/genetics , Homeostasis/genetics , Intestinal Mucosa/growth & development , Organoids/metabolism , Proto-Oncogene Proteins p21(ras)/genetics , Animals , Carcinogenesis/genetics , Colon/growth & development , Colon/pathology , Gene Expression Regulation, Neoplastic/genetics , Goblet Cells/metabolism , Humans , Intestinal Mucosa/pathology , Intestine, Small/metabolism , Mice , Wnt Signaling Pathway/genetics
8.
Nutrients ; 13(8)2021 Jul 26.
Article in English | MEDLINE | ID: mdl-34444719

ABSTRACT

Low birthweight (LBW) is associated with metabolic complications, such as glucose and lipid metabolism disturbances in early life. The objective of this study was to assess: (1) the effect of dietary tryptophan (Trp) on glucose and fat metabolism in an LBW piglet model, and (2) the role peripheral 5-hydroxytryptamine type 3 (5HT3) receptors in regulating the feeding behavior in LBW piglets fed with Trp-supplemented diets. Seven-day-old piglets were assigned to 4 treatments: normal birthweight-0%Trp (NBW-T0), LBW-0%Trp (LBW-T0), LBW-0.4%Trp (LBW-T0.4), and LBW-0.8%Trp (LBW-T0.8) for 3 weeks. Compared to LBW-T0, the blood glucose was decreased in LBW-T0.8 at 60 min following the meal test, and the triglycerides were lower in LBW-T0.4 and LBW-T0.8. Relative to LBW-T0, LBW-T0.8 had a lower transcript and protein abundance of hepatic glucose transporter-2, a higher mRNA abundance of glucokinase, and a lower transcript of phosphoenolpyruvate carboxykinase. LBW-T0.4 tended to have a lower protein abundance of sodium-glucose co-transporter 1 in the jejunum. In comparison with LBW-T0, LBW-T0.4 and LBW-T0.8 had a lower transcript of hepatic acetyl-CoA carboxylase, and LBW-T0.4 had a higher transcript of 3-hydroxyacyl-CoA dehydrogenase. Blocking 5-HT3 receptors with ondansetron reduced the feed intake in all groups, with a transient effect on LBW-T0, but more persistent effect on LBW-T0.8 and NBW-T0. In conclusion, Trp supplementation reduced the hepatic lipogenesis and gluconeogenesis, but increased the glycolysis in LBW piglets. Peripheral serotonin is likely involved in the regulation of feeding behavior, particularly in LBW piglets fed diets supplemented with a higher dose of Trp.


Subject(s)
Dietary Supplements , Glucose/metabolism , Lipid Metabolism , Liver/metabolism , Tryptophan/administration & dosage , Adipose Tissue, White/metabolism , Animals , Animals, Newborn , Birth Weight , Blood Glucose/analysis , Body Weight , Cholesterol/blood , Diet , Hypothalamus/metabolism , Insulin/blood , Intestinal Mucosa/anatomy & histology , Intestinal Mucosa/growth & development , Intestine, Small/anatomy & histology , Intestine, Small/growth & development , Models, Animal , Ondansetron/pharmacology , Serotonin 5-HT3 Receptor Antagonists/pharmacology , Swine/growth & development , Triglycerides/blood
9.
Int J Mol Sci ; 22(11)2021 Jun 02.
Article in English | MEDLINE | ID: mdl-34199463

ABSTRACT

Little is known about the ability for epithelial regeneration and wound healing in patients with inflammatory bowel diseases. We evaluated the epithelial proliferation and wound healing ability of patients with Crohn's disease (CD) using patient-derived intestinal organoids. Human intestinal organoids were constructed in a three-dimensional intestinal crypt culture of enteroscopic biopsy samples from controls and CD patients. The organoid-forming efficiency of ileal crypts derived from CD patients was reduced compared with those from control subjects (p < 0.001). Long-term cultured organoids (≥6 passages) derived from controls and CD patients showed an indistinguishable microscopic appearance and culturing behavior. Under TNFα-enriched conditions (30 ng/mL), the organoid reconstitution rate and cell viability of CD patient-derived organoids were significantly lower than those of the control organoids (p < 0.05 for each). The number of EdU+ cells was significantly lower in TNFα-treated organoids derived from CD patients than in TNFα-treated control organoids (p < 0.05). In a wound healing assay, the unhealed area in TNFα-treated CD patient-derived organoids was significantly larger than that of TNFα-treated control organoids (p < 0.001). The wound healing ability of CD patient-derived organoids is reduced in TNFα-enriched conditions, due to reduced cell proliferation. Epithelial regeneration ability may be impaired in patients with CD.


Subject(s)
Cell Proliferation/genetics , Crohn Disease/therapy , Epithelial Cells/metabolism , Organoids/growth & development , Adult , Crohn Disease/metabolism , Crohn Disease/pathology , Epithelial Cells/pathology , Female , Humans , Ileum/growth & development , Ileum/injuries , Ileum/pathology , Intestinal Mucosa/growth & development , Intestinal Mucosa/pathology , Intestines/diagnostic imaging , Intestines/injuries , Male , Middle Aged , Organoids/metabolism , Regeneration/genetics , Signal Transduction/genetics , Stem Cells/cytology , Stem Cells/metabolism , Tumor Necrosis Factor-alpha/genetics , Wound Healing/genetics
10.
Int J Mol Sci ; 22(14)2021 Jul 08.
Article in English | MEDLINE | ID: mdl-34298973

ABSTRACT

Intestinal cylindrical growth peaks in mice a few weeks after birth, simultaneously with crypt fission activity. It nearly stops after weaning and cannot be reactivated later. Transgenic mice expressing Cd97/Adgre5 in the intestinal epithelium develop a mega-intestine with normal microscopic morphology in adult mice. Here, we demonstrate premature intestinal differentiation in Cd97/Adgre5 transgenic mice at both the cellular and molecular levels until postnatal day 14. Subsequently, the growth of the intestinal epithelium becomes activated and its maturation suppressed. These changes are paralleled by postnatal regulation of growth factors and by an increased expression of secretory cell markers, suggesting growth activation of non-epithelial tissue layers as the origin of enforced tissue growth. To understand postnatal intestinal growth mechanistically, we study epithelial fate decisions during this period with the use of a 3D individual cell-based computer model. In the model, the expansion of the intestinal stem cell (SC) population, a prerequisite for crypt fission, is largely independent of the tissue growth rate and is therefore not spontaneously adaptive. Accordingly, the model suggests that, besides the growth activation of non-epithelial tissue layers, the formation of a mega-intestine requires a released growth control in the epithelium, enabling accelerated SC expansion. The similar intestinal morphology in Cd97/Adgre5 transgenic and wild type mice indicates a synchronization of tissue growth and SC expansion, likely by a crypt density-controlled contact inhibition of growth of intestinal SC proliferation. The formation of a mega-intestine with normal microscopic morphology turns out to originate in changes of autonomous and conditional specification of the intestinal cell fate induced by the activation of Cd97/Adgre5.


Subject(s)
Computer Simulation , Intestinal Mucosa/growth & development , Intestine, Small/growth & development , Models, Biological , Receptors, G-Protein-Coupled/metabolism , Stem Cells/metabolism , Animals , Kruppel-Like Factor 4 , Mice , Mice, Transgenic , Organ Culture Techniques , Receptors, G-Protein-Coupled/genetics
12.
Nat Commun ; 12(1): 2105, 2021 04 08.
Article in English | MEDLINE | ID: mdl-33833232

ABSTRACT

Intestinal microbiota-derived metabolites have biological importance for the host. Polyamines, such as putrescine and spermidine, are produced by the intestinal microbiota and regulate multiple biological processes. Increased colonic luminal polyamines promote longevity in mice. However, no direct evidence has shown that microbial polyamines are incorporated into host cells to regulate cellular responses. Here, we show that microbial polyamines reinforce colonic epithelial proliferation and regulate macrophage differentiation. Colonisation by wild-type, but not polyamine biosynthesis-deficient, Escherichia coli in germ-free mice raises intracellular polyamine levels in colonocytes, accelerating epithelial renewal. Commensal bacterium-derived putrescine increases the abundance of anti-inflammatory macrophages in the colon. The bacterial polyamines ameliorate symptoms of dextran sulfate sodium-induced colitis in mice. These effects mainly result from enhanced hypusination of eukaryotic initiation translation factor. We conclude that bacterial putrescine functions as a substrate for symbiotic metabolism and is further absorbed and metabolised by the host, thus helping maintain mucosal homoeostasis in the intestine.


Subject(s)
Colon/metabolism , Escherichia coli/metabolism , Intestinal Mucosa/metabolism , Peptide Initiation Factors/metabolism , Putrescine/metabolism , RNA-Binding Proteins/metabolism , Animals , Cell Differentiation/physiology , Cell Proliferation/physiology , Colitis/chemically induced , Colitis/pathology , Dextran Sulfate/toxicity , Epithelial Cells/metabolism , Female , Gastrointestinal Microbiome/physiology , Homeostasis , Intestinal Mucosa/cytology , Intestinal Mucosa/growth & development , Macrophages/cytology , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Eukaryotic Translation Initiation Factor 5A
13.
Endocrinology ; 162(5)2021 05 01.
Article in English | MEDLINE | ID: mdl-33675223

ABSTRACT

Hypothyroidism is a common pathological condition characterized by insufficient activity of the thyroid hormones (THs), thyroxine (T4), and 3,5,3'-triiodothyronine (T3), in the whole body or in specific tissues. Hypothyroidism is associated with inadequate development of the intestine as well as gastrointestinal diseases. We used a zebrafish model of hypothyroidism to identify and characterize TH-modulated genes and cellular pathways controlling intestine development. In the intestine of hypothyroid juveniles and adults, the number of mucus-secreting goblet cells was reduced, and this phenotype could be rescued by T3 treatment. Transcriptome profiling revealed dozens of differentially expressed genes in the intestine of hypothyroid adults compared to controls. Notably, the expression of genes encoding to Fgf19 and its receptor Fgfr4 was markedly increased in the intestine of hypothyroid adults, and treatment with T3 normalized it. Blocking fibroblast growth factor (FGF) signaling, using an inducible dominant-negative Fgfr transgenic line, rescued the number of goblet cells in hypothyroid adults. These results show that THs inhibit the Fgf19-Fgfr4 signaling pathway, which is associated with inhibition of goblet cell differentiation in hypothyroidism. Both the TH and Fgf19-Fgfr4 signaling pathways can be pharmaceutical targets for the treatment of TH-related gastrointestinal diseases.


Subject(s)
Fibroblast Growth Factors/metabolism , Goblet Cells/metabolism , Hypothyroidism/metabolism , Receptor, Fibroblast Growth Factor, Type 4/metabolism , Thyroxine/metabolism , Triiodothyronine/metabolism , Zebrafish Proteins/metabolism , Zebrafish/metabolism , Animals , Cell Proliferation , Disease Models, Animal , Fibroblast Growth Factors/genetics , Goblet Cells/cytology , Humans , Hypothyroidism/genetics , Hypothyroidism/physiopathology , Intestinal Mucosa/growth & development , Intestinal Mucosa/metabolism , Receptor, Fibroblast Growth Factor, Type 4/genetics , Signal Transduction , Zebrafish/genetics , Zebrafish/growth & development , Zebrafish Proteins/genetics
14.
Biomed Res Int ; 2021: 9823969, 2021.
Article in English | MEDLINE | ID: mdl-33532501

ABSTRACT

The present study was conducted to investigate the effects of early transplantation of the faecal microbiota from Tibetan pigs on the gut development of dextran sulphate sodium- (DSS-) challenged piglets. In total, 24 3-day-old DLY piglets were divided into four groups (n = 6 per group); a 2 × 2 factorial arrangement was used, which included faecal microbiota transplantation (FMT) (from Tibetan pigs) and DSS challenge. The whole trial lasted for 55 days. DSS infusion increased the intestinal density, serum diamine oxidase (DAO) activity, and colonic Escherichia coli count (P < 0.05), and decreased the Lactobacillus spp. count and mRNA abundances of epidermal growth factor (EGF), glucagon-like peptide-2 (GLP-2), insulin-like growth factor 1 (IGF-1), occludin, mucin 2 (MUC2), regeneration protein IIIγ (RegIIIγ), and interleukin-10 (IL-10) in the colon (P < 0.05). FMT increased the Lactobacillus spp. count and mRNA abundances of GLP-2, RegIIIγ, and IL-10 in the colon (P < 0.05), and decreased the intestinal density, serum DAO activity, and colonic E. coli number (P < 0.05). In addition, in DSS-challenged piglets, FMT decreased the disease activity index (P < 0.05) and attenuated the effect of DSS challenge on the intestinal density, serum DAO activity, and colonic E. coli number (P < 0.05). These data indicated that the faecal microbiota from Tibetan pigs could attenuate the negative effect of DSS challenge on the gut development of piglets.


Subject(s)
Fecal Microbiota Transplantation , Gastrointestinal Microbiome , Intestinal Mucosa , Animals , Animals, Suckling/growth & development , Animals, Suckling/physiology , Dextran Sulfate , Escherichia coli/genetics , Feces/microbiology , Gastrointestinal Microbiome/genetics , Gastrointestinal Microbiome/physiology , Intestinal Mucosa/growth & development , Intestinal Mucosa/physiology , Lactobacillus/genetics , Swine
15.
Cell ; 184(3): 810-826.e23, 2021 02 04.
Article in English | MEDLINE | ID: mdl-33406409

ABSTRACT

Development of the human intestine is not well understood. Here, we link single-cell RNA sequencing and spatial transcriptomics to characterize intestinal morphogenesis through time. We identify 101 cell states including epithelial and mesenchymal progenitor populations and programs linked to key morphogenetic milestones. We describe principles of crypt-villus axis formation; neural, vascular, mesenchymal morphogenesis, and immune population of the developing gut. We identify the differentiation hierarchies of developing fibroblast and myofibroblast subtypes and describe diverse functions for these including as vascular niche cells. We pinpoint the origins of Peyer's patches and gut-associated lymphoid tissue (GALT) and describe location-specific immune programs. We use our resource to present an unbiased analysis of morphogen gradients that direct sequential waves of cellular differentiation and define cells and locations linked to rare developmental intestinal disorders. We compile a publicly available online resource, spatio-temporal analysis resource of fetal intestinal development (STAR-FINDer), to facilitate further work.


Subject(s)
Intestines/cytology , Intestines/growth & development , Single-Cell Analysis , Endothelial Cells/cytology , Enteric Nervous System/cytology , Fetus/embryology , Fibroblasts/cytology , Humans , Immunity , Intestinal Diseases/congenital , Intestinal Diseases/pathology , Intestinal Mucosa/growth & development , Intestines/blood supply , Ligands , Mesoderm/cytology , Neovascularization, Physiologic , Pericytes/cytology , Stem Cells/cytology , Time Factors , Transcription Factors/metabolism
16.
Cell Mol Gastroenterol Hepatol ; 11(2): 503-524, 2021.
Article in English | MEDLINE | ID: mdl-32896624

ABSTRACT

BACKGROUND & AIMS: Notch signaling coordinates cell differentiation processes in the intestinal epithelium. The transcription factor Nrf2 orchestrates defense mechanisms by regulating cellular redox homeostasis, which, as shown previously in murine liver, can be amplified through signaling crosstalk with the Notch pathway. However, interplay between these 2 signaling pathways in the gut is unknown. METHODS: Mice modified genetically to amplify Nrf2 in the intestinal epithelium (Keap1f/f::VilCre) were generated as well as pharmacological activation of Nrf2 and subjected to phenotypic and cell lineage analyses. Cell lines were used for reporter gene assays together with Nrf2 overexpression to study transcriptional regulation of the Notch downstream effector. RESULTS: Constitutive activation of Nrf2 signaling caused increased intestinal length along with expanded cell number and thickness of enterocytes without any alterations of secretory lineage, outcomes abrogated by concomitant disruption of Nrf2. The Nrf2 and Notch pathways in epithelium showed inverse spatial profiles, where Nrf2 activity in crypts was lower than villi. In progenitor cells of Keap1f/f::VilCre mice, Notch downstream effector Math1, which regulates a differentiation balance of cell lineage through lateral inhibition, showed suppressed expression. In vitro results demonstrated Nrf2 negatively regulated Math1, where 6 antioxidant response elements located in the regulatory regions contributed to this repression. CONCLUSIONS: Activation of Nrf2 perturbed the dialog of the Notch cascade though negative regulation of Math1 in progenitor cells, leading to enhanced enterogenesis. The crosstalk between the Nrf2 and Notch pathways could be critical for fine-tuning intestinal homeostasis and point to new approaches for the pharmacological management of absorptive deficiencies.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Intestinal Mucosa/growth & development , Intestine, Small/growth & development , NF-E2-Related Factor 2/metabolism , Regeneration/genetics , Animals , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Line , Enterocytes/drug effects , Enterocytes/physiology , Female , Intestinal Mucosa/drug effects , Intestine, Small/drug effects , Male , Mice , Models, Animal , NF-E2-Related Factor 2/agonists , NF-E2-Related Factor 2/genetics , Regeneration/drug effects , Stem Cells/drug effects , Stem Cells/physiology
17.
J Cell Physiol ; 236(4): 2631-2648, 2021 04.
Article in English | MEDLINE | ID: mdl-32853405

ABSTRACT

Individuals with postnatal growth retardation (PGR) are prone to developing chronic diseases. Abnormal development in small intestine is casually implicated in impaired growth. However, the exact mechanism is still implausible. In this present study, PGR piglets (aged 42 days) were employed as a good model to analyze developmental changes in intestinal mucosal barrier function. Our data demonstrated that PGR piglets exhibited impaired jejunal and ileal epithelial villous morphology and permeability, accompanied by decreased cell proliferation ability and increased apoptosis rate. In addition, the expression of tight junction proteins (ZO-1, claudin 1, and occludin) and E-cadherin was markedly inhibited by PGR. The expression of P-glycoprotein was significantly reduced in PGR piglets, as well as decreased activity of lysozyme. Moreover, the mRNA abundance and content of inflammatory cytokines were significantly increased in the intestinal mucosa and plasma of PGR piglets, respectively. PGR also contributed to lower level of sIgA, and higher level of CD68-positive rate, ß-defensins, and protein expression involved p38 MAPK/NF-κB pathway. Furthermore, PGR altered the intestinal microbial community such as decreased genus Alloprevotella and Oscillospira abundances, and led to lower microbial-derived butyrate production, which may be potential targets for treatment. Collectively, our findings indicated that the intestinal mucosal barrier function of PGR piglets could develop the nutritional intervention strategies in prevention and treatment of the intestinal mucosal barrier dysfunction in piglets and humans.


Subject(s)
Growth Disorders/metabolism , Intestinal Mucosa/metabolism , Intestine, Small/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Animals , Animals, Newborn , Apoptosis , Bacteria/metabolism , Butyrates/metabolism , Cell Proliferation , Cytokines/metabolism , Disease Models, Animal , Gastrointestinal Microbiome , Growth Disorders/microbiology , Growth Disorders/pathology , Growth Disorders/physiopathology , Inflammation Mediators/metabolism , Intestinal Mucosa/growth & development , Intestinal Mucosa/microbiology , Intestinal Mucosa/ultrastructure , Intestine, Small/growth & development , Intestine, Small/microbiology , Intestine, Small/ultrastructure , Muramidase/metabolism , NF-kappa B/metabolism , Permeability , Sus scrofa , Tight Junction Proteins/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
18.
J Appl Toxicol ; 41(2): 291-302, 2021 02.
Article in English | MEDLINE | ID: mdl-33107989

ABSTRACT

The use of CuO nanoparticles (NPs) has increased greatly and their potential effects on human health need to be investigated. Differentiated Caco-2 cells were treated from the apical (Ap) and the basolateral (Bl) compartment with different concentrations (0, 10, 50 and 100 µg/mL) of commercial or sonochemically synthesized (sono) CuO NPs. Sono NPs were prepared in ethanol (CuOe) or in water (CuOw), obtaining CuO NPs differing in size and shape. The effects on the Caco-2 cell barrier were assessed via transepithelial electrical resistance (TEER) evaluation just before and after 1, 2 and 24 hours of exposure and through the analysis of cytokine release and biomarkers of oxidative damage to proteins after 24 hours. Sono CuOe and CuOw NPs induced a TEER decrease with a dose-dependent pattern after Bl exposure. Conversely, TEER values were not affected by the Ap exposure to commercial CuO NPs and, concerning the Bl exposure, only the lowest concentration tested (10 µg/mL) caused a TEER decrease after 24 hours of exposure. An increased release of interleukin-8 was induced by sono CuO NPs after the Ap exposure to 100 µg/mL and by sono and commercial CuO after the Bl exposure to all the concentrations. No effects of commercial and sono CuO NPs on interleukin-6 (with the only exception of 100 µg/mL Bl commercial CuO) and tumor necrosis factor-α release were observed. Ap treatment with commercial and CuOw NPs was able to induce significant alterations on specific biomarkers of protein oxidative damage (protein sulfhydryl group oxidation and protein carbonylation).


Subject(s)
Caco-2 Cells/drug effects , Cell Differentiation/drug effects , Cell Survival/drug effects , Copper/toxicity , Intestinal Mucosa/drug effects , Intestinal Mucosa/growth & development , Metal Nanoparticles/toxicity , Humans
19.
FASEB J ; 35(1): e21178, 2021 01.
Article in English | MEDLINE | ID: mdl-33190300

ABSTRACT

Food withdrawal is usually used for accurate feed metabolizable energy (ME) assessment in poultry, but its effects on intestinal structure and the absorption of nutrients are unclear. In this study, broilers were fed ad libitum (CT) or withdrew food for 12 (FH12), 24 (FH24), 36 (FH36), or 48 hours (FH48). We showed that food withdrawal increased the energy assimilation when compared with the CT. Food withdrawal improved the digestibility of ether extract and the level of lipid substances and fatty acid-derived ß-hydroxybutyrate in serum. Compared to the CT, food withdrawal did not influence the digestibility of starch. Due to 12 hours or longer food withdrawal duration increased glutamate oxidation and uric acid excretion, the analyzed digestibility of crude protein was underestimated, although the upregulated amino acid transporter genes. In addition, histological analysis showed that short-term food withdrawal (12 hours) increased intestinal villus height, crypt depth, and proliferative cell, whereas prolonged food withdrawal (more than 24 hours) impaired villus structure due to the decreased cell proliferation. Moreover, proteomics analysis revealed upregulated pathways in birds withdrawn food for 36 hours involved in nutrient absorption and amino acid oxidation. In conclusion, food withdrawal changes nutrient absorption and utilization, especially for amino acid and ether extract, and results in increased ME. Both glutamate oxidation and fatty acid incomplete oxidation are involved in energy supply after refeeding. In contrast to short-term food withdrawal, prolonged food withdrawal impairs the intestinal structure and villus renewal. Our findings deserve attention from nutritionists who are analyzing food digestibility.


Subject(s)
Animal Feed , Animal Nutritional Physiological Phenomena , Chickens/metabolism , Energy Metabolism , Intestinal Mucosa , Animals , Intestinal Mucosa/anatomy & histology , Intestinal Mucosa/growth & development , Intestinal Mucosa/metabolism , Male
20.
Drug Metab Dispos ; 49(1): 12-19, 2021 01.
Article in English | MEDLINE | ID: mdl-33154041

ABSTRACT

UDP-glucuronosyltransferase (UGT) 1A1 is the only transferase capable of conjugating serum bilirubin. However, temporal delay in the development of the UGT1A1 gene leads to an accumulation of serum bilirubin in newborn children. Neonatal humanized UGT1 (hUGT1) mice, which accumulate severe levels of total serum bilirubin (TSB), were treated by oral gavage with obeticholic acid (OCA), a potent FXR agonist. OCA treatment led to dramatic reduction in TSB levels. Analysis of UGT1A1 expression confirmed that OCA induced intestinal and not hepatic UGT1A1. Interestingly, Cyp2b10, a target gene of the nuclear receptor CAR, was also induced by OCA in intestinal tissue. In neonatal hUGT1/Car -/- mice, OCA was unable to induce CYP2B10 and UGT1A1, confirming that CAR and not FXR is involved in the induction of intestinal UGT1A1. However, OCA did induce FXR target genes, such as Shp, in both intestines and liver with induction of Fgf15 in intestinal tissue. Circulating FGF15 activates hepatic FXR and, together with hepatic Shp, blocks Cyp7a1 and Cyp7b1 gene expression, key enzymes in bile acid metabolism. Importantly, the administration of OCA in neonatal hUGT1 mice accelerates intestinal epithelial cell maturation, which directly impacts on induction of the UGT1A1 gene and the reduction in TSB levels. Accelerated intestinal maturation is directly controlled by CAR, since induction of enterocyte marker genes sucrase-isomaltase, alkaline phosphatase 3, and keratin 20 by OCA does not occur in hUGT1/Car -/- mice. Thus, new findings link an important role for CAR in intestinal UGT1A1 induction and its role in the intestinal maturation pathway. SIGNIFICANCE STATEMENT: Obeticholic acid (OCA) activates FXR target genes in both liver and intestinal tissues while inducing intestinal UGT1A1, which leads to the elimination of serum bilirubin in humanized UGT1 mice. However, the induction of intestinal UGT1A1 and the elimination of bilirubin by OCA is driven entirely by activation of intestinal CAR and not FXR. The elimination of serum bilirubin is based on a CAR-dependent mechanism that facilitates the acceleration of intestinal epithelium cell differentiation, an event that underlies the induction of intestinal UGT1A1.


Subject(s)
Bilirubin/metabolism , Chenodeoxycholic Acid/analogs & derivatives , Constitutive Androstane Receptor/metabolism , Glucuronosyltransferase/metabolism , Intestines , Liver/metabolism , Receptors, Cytoplasmic and Nuclear , Animals , Animals, Newborn , Cell Differentiation/physiology , Chenodeoxycholic Acid/pharmacokinetics , Gastrointestinal Agents/pharmacokinetics , Humans , Intestinal Mucosa/growth & development , Intestinal Mucosa/physiology , Intestines/growth & development , Intestines/metabolism , Mice , Receptors, Cytoplasmic and Nuclear/agonists , Receptors, Cytoplasmic and Nuclear/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...