Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 74
Filter
1.
Food Funct ; 12(24): 12634-12646, 2021 Dec 13.
Article in English | MEDLINE | ID: mdl-34821899

ABSTRACT

Research has shown that maternal sucralose (MS) exposure alters the gut microbiota of offspring at weaning and predisposes the offspring to developing obesity, non-alcoholic fatty liver disease and metabolic syndrome later in life. However, the underlying mechanism remains unclear. Paneth cells are thought to critically influence the gut microbiota. This study aimed to investigate whether MS exposure induced Paneth cell defects and exacerbated gut dysbiosis of offspring. Female C57BL/6 mice were divided into the MS and control (water) groups during pregnancy and lactation. Progeny mice were fed a normal sucralose-free diet after weaning until adulthood. MS inhibited intestinal development and increased the expression of proinflammatory cytokines in the small intestines of 3-week-old progeny mice. MS increased the proportions of abnormal granule secretion by Paneth cells. The number of Paneth cells and mRNA expression of AMPs such as cryptdins and lysozyme were reduced in the MS group. MS disturbed the gut microbiota composition and diversity in the 3-week-old offspring mice. The relative abundances of pro-inflammatory bacteria, such as Desulfovibrionales, Helicobacter, Pasteurellales and Campylobacterales were significantly increased in the MS group, while anti-inflammatory bacteria, including Clostridium XI, were decreased. This dysbiosis continued into adulthood. These findings showed that MS exposure induced Paneth cell defects and exacerbated gut dysbiosis in offspring mice. Sucralose should be consumed with caution, especially during pregnancy and in early life.


Subject(s)
Gastrointestinal Microbiome/drug effects , Maternal Exposure/adverse effects , Paneth Cells/drug effects , Sucrose/analogs & derivatives , Sweetening Agents/administration & dosage , Sweetening Agents/adverse effects , Animals , Disease Models, Animal , Female , Humans , Male , Mice , Mice, Inbred C57BL , Pregnancy , Sucrose/administration & dosage , Sucrose/adverse effects
2.
Gut Microbes ; 13(1): 1957408, 2021.
Article in English | MEDLINE | ID: mdl-34375154

ABSTRACT

Vitamin D receptor (VDR) deficiency is associated with cancer, infection, and chronic inflammation. Prior research has demonstrated VDR regulation of bacteria; however, little is known regarding VDR and viruses. We hypothesize that VDR deficiency impacts on the intestinal virome and viral-bacterial interactions. We specifically deleted VDR from intestinal epithelial cells (VDRΔIEC), Paneth cells (VDRΔPC), and myeloid cells (VDRΔLyz) in mice. Feces were collected for shotgun metagenomic sequencing and metabolite profiling. To test the functional changes, we evaluated pattern recognition receptors (PRRs) and analyzed microbial metabolites. Vibrio phages, Lactobacillus phages, and Escherichia coli typing phages were significantly enriched in all three conditional VDR-knockout mice. In the VDRΔLyz mice, the levels of eight more virus species (2 enriched, 6 depleted) were significantly changed. Altered virus species were primarily observed in female VDRΔLyz (2 enriched, 3 depleted) versus male VDRΔLyz (1 enriched, 1 depleted). Altered alpha and beta diversity (family to species) were found in VDRΔLyz. In VDRΔIEC mice, bovine viral diarrhea virus 1 was significantly enriched. A significant correlation between viral and bacterial alterations was found in conditional VDR knockout mice. There was a positive correlation between Vibrio phage JSF5 and Cutibacterium acnes in VDRΔPC and VDRΔLyz mice. Also, there were more altered viral species in female conditional VDR knockout mice. Notably, there were significant changes in PRRs: upregulated TLR3, TLR7, and NOD2 in VDRΔLyz mice and increased CLEC4L expression in VDRΔIEC and VDRΔPC mice. Furthermore, we identified metabolites related to virus infection: decreased glucose in VDRΔIEC mice, increased ribulose/xylulose and xylose in VDRΔLyz mice, and increased long-chain fatty acids in VDRΔIEC and VDRΔLyz female mice. Tissue-specific deletion of VDR changes the virome and functionally changes viral receptors, which leads to dysbiosis, metabolic dysfunction, and infection risk. This study helps to elucidate VDR regulating the virome in a tissue-specific and sex-specific manner.


Subject(s)
Deficiency Diseases/physiopathology , Gastrointestinal Microbiome/drug effects , Intestines/virology , Microbial Interactions/drug effects , Receptors, Calcitriol/deficiency , Virome/drug effects , Animals , Feces/virology , Female , Male , Mice , Mice, Knockout , Myeloid Cells/drug effects , Myeloid Cells/virology , Paneth Cells/drug effects , Paneth Cells/virology
3.
Toxicol Appl Pharmacol ; 422: 115561, 2021 07 01.
Article in English | MEDLINE | ID: mdl-33957193

ABSTRACT

Arsenic is a global health concern that causes toxicity through ingestion of contaminated water and food. In vitro studies suggest that arsenic reduces stem and progenitor cell differentiation. Thus, this study determined if arsenic disrupted intestinal stem cell (ISC) differentiation, thereby altering the number, location, and/or function of intestinal epithelial cells. Adult male C57BL/6 mice were exposed to 0 or 100 ppb sodium arsenite (AsIII) through drinking water for 5 weeks. Duodenal sections were collected to assess changes in morphology, proliferation, and cell types. qPCR analysis revealed a 40% reduction in Lgr5 transcripts, an ISC marker, in the arsenic-exposed mice, although there were no changes in the protein expression of Olfm4. Secretory cell-specific transcript markers of Paneth (Defa1), Goblet (Tff3), and secretory transit amplifying (Math1) cells were reduced by 51%, 44%, and 30% respectively, in the arsenic-exposed mice, indicating significant impacts on the Wnt-dependent differentiation pathway. Further, protein levels of phosphorylated ß-catenin were reduced in the arsenic-exposed mice, which increased the expression of Wnt-dependent transcripts CD44 and c-myc. PCA analysis, followed by MANOVA and regression analyses, revealed significant changes and correlations between Lgr5 and the transit amplifying (TA) cell markers Math1 and Hes1, which are in the secretory cell pathway. Similar comparisons between Math1 and Defa1 show that terminal differentiation into Paneth cells is also reduced in the arsenic-exposed mice. The data suggests that ISCs are not lost following arsenic exposure, but rather, specific Wnt-dependent progenitor cell formation and terminal differentiation in the small intestine is reduced.


Subject(s)
Arsenites/toxicity , Cell Differentiation/drug effects , Duodenum/drug effects , Paneth Cells/drug effects , Receptors, G-Protein-Coupled/metabolism , Sodium Compounds/toxicity , Stem Cells/drug effects , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Down-Regulation , Duodenum/metabolism , Duodenum/pathology , Male , Mice, Inbred C57BL , Paneth Cells/metabolism , Paneth Cells/pathology , Receptors, G-Protein-Coupled/genetics , Stem Cells/metabolism , Stem Cells/pathology , Trefoil Factor-3/genetics , Trefoil Factor-3/metabolism , Wnt Signaling Pathway , alpha-Defensins/genetics , alpha-Defensins/metabolism
4.
Cell Host Microbe ; 29(6): 988-1001.e6, 2021 06 09.
Article in English | MEDLINE | ID: mdl-34010595

ABSTRACT

Intestinal Paneth cells modulate innate immunity and infection. In Crohn's disease, genetic mutations together with environmental triggers can disable Paneth cell function. Here, we find that a western diet (WD) similarly leads to Paneth cell dysfunction through mechanisms dependent on the microbiome and farnesoid X receptor (FXR) and type I interferon (IFN) signaling. Analysis of multiple human cohorts suggests that obesity is associated with Paneth cell dysfunction. In mouse models, consumption of a WD for as little as 4 weeks led to Paneth cell dysfunction. WD consumption in conjunction with Clostridium spp. increased the secondary bile acid deoxycholic acid levels in the ileum, which in turn inhibited Paneth cell function. The process required excess signaling of both FXR and IFN within intestinal epithelial cells. Our findings provide a mechanistic link between poor diet and inhibition of gut innate immunity and uncover an effect of FXR activation in gut inflammation.


Subject(s)
Diet, Western/adverse effects , Gastrointestinal Microbiome/drug effects , Interferon Type I/metabolism , Obesity/metabolism , Paneth Cells/drug effects , Paneth Cells/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Animals , Bile Acids and Salts/metabolism , Cells, Cultured , Diet, High-Fat/adverse effects , Disease Models, Animal , Gene Expression Profiling , Humans , Immunity, Innate/drug effects , Intestinal Mucosa/metabolism , Mice , Mice, Inbred C57BL , Signal Transduction
5.
Cell Rep ; 35(3): 109026, 2021 04 20.
Article in English | MEDLINE | ID: mdl-33882314

ABSTRACT

Organoids allow the recapitulation of intestinal homeostasis and cancerogenesis in vitro; however, RNA sequencing (RNA-seq)-based methods for drug screens are missing. We develop targeted organoid sequencing (TORNADO-seq), a high-throughput, high-content drug discovery platform that uses targeted RNA-seq to monitor the expression of large gene signatures for the detailed evaluation of cellular phenotypes in organoids. TORNADO-seq is a fast, highly reproducible time- and cost-effective ($5 per sample) method that can probe cell mixtures and their differentiation state in the intestinal system. We apply this method to isolate drugs that enrich for differentiated cell phenotypes and show that these drugs are highly efficacious against cancer compared to wild-type organoids. Furthermore, TORNADO-seq facilitates in-depth insight into the mode of action of these drugs. Our technology can easily be adapted to many other systems and will allow for more systematic, large-scale, and quantitative approaches to study the biology of complex cellular systems.


Subject(s)
Antineoplastic Agents/pharmacology , Early Detection of Cancer/methods , Gene Expression Regulation, Neoplastic/drug effects , Organoids/drug effects , Prescription Drugs/pharmacology , Small Molecule Libraries/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/classification , Cell Differentiation/drug effects , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Drug Discovery/methods , Drug Repositioning , Enterocytes/drug effects , Enterocytes/metabolism , Enterocytes/pathology , Gene Regulatory Networks , Goblet Cells/drug effects , Goblet Cells/metabolism , Goblet Cells/pathology , High-Throughput Screening Assays , Humans , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Organoids/metabolism , Organoids/pathology , Paneth Cells/drug effects , Paneth Cells/metabolism , Paneth Cells/pathology , Prescription Drugs/chemistry , Prescription Drugs/classification , RNA-Seq , Sequence Analysis, RNA , Small Molecule Libraries/chemistry , Small Molecule Libraries/classification
6.
Stem Cell Reports ; 16(5): 1290-1301, 2021 05 11.
Article in English | MEDLINE | ID: mdl-33891868

ABSTRACT

Non-muscle myosin IIA plays an important role in cell adhesion, cell migration, and tissue architecture. We previously showed that low activity of the heavy chain of non-muscle myosin II Myh9 is beneficial to LGR5+ intestinal stem cell maintenance. However, the function of Myh9 in adult mouse intestinal epithelium is largely unclear. In this study, we used the inducible Villin-creERT2 knockout approach to delete Myh9 in adult mouse intestinal epithelium and observed that homozygous deletion of Myh9 causes colitis-like morphologic changes in intestine, leads to a high sensitivity to dextran sulfate sodium and promotes colitis-related adenoma formation in the colon. Myh9 deletion disturbs cell junctions and impairs intestinal lumen barrier integrity, promoting the necroptosis of epithelial cells. Consistently, these changes can be partially rescued by Ripk3 knockout. Our results indicate that Myh9 is required for the maintenance of intestinal epithelium integrity and the prevention of cell necroptosis.


Subject(s)
Adenoma/pathology , Colitis/pathology , Colonic Neoplasms/pathology , Homeostasis , Intestinal Mucosa/pathology , Myosin Heavy Chains/metabolism , Necroptosis , Animals , Anti-Bacterial Agents/pharmacology , Gastrointestinal Microbiome/drug effects , Gene Deletion , Homeostasis/drug effects , Homozygote , Intestinal Mucosa/drug effects , Mice, Inbred C57BL , Mice, Knockout , Myosin Heavy Chains/deficiency , Necroptosis/drug effects , Paneth Cells/drug effects , Paneth Cells/metabolism , Receptors, G-Protein-Coupled/metabolism , Stem Cells/drug effects , Stem Cells/metabolism
7.
Biochem Biophys Res Commun ; 545: 14-19, 2021 03 19.
Article in English | MEDLINE | ID: mdl-33529805

ABSTRACT

Paneth cells and Lgr5+ intestinal stem cells (Lgr5+ ISCs) constitute the stem cell niche and maintain small intestinal epithelial integrity by recognizing various niche factors derived from subepithelial cells and external antigens. Although it has been known that interferon-γ (IFN-γ), a Th1 cytokine, is associated with intestinal epithelial disruption during inflammation as a niche factor, dynamics of Paneth cells and Lgr5+ ISCs in response to IFN-γ remain to be understood. Here we show that CAG-tdTomato;Lgr5-EGFP (CT-LE) mice generated in this study enable to identify Paneth cells and Lgr5+ ISCs separately by fluorescence signals. Lgr5+ ISCs underwent cell death a little earlier than Paneth cells in response to IFN-γ by simultaneous tracking using CT-LE mice. In addition, the timing of cell death in most Paneth cells overlapped with Lgr5+ ISCs, suggesting that Paneth cell depletion is induced directly by IFN-γ. Taken together, we established a novel simultaneous stem cell niche tracking method and clarified the involvement of both Paneth cells and Lgr5+ ISCs in stem cell niche damage induced by IFN-γ, further contribute to understanding the mechanism for maintaining intestinal homeostasis by stem cell niche.


Subject(s)
Interferon-gamma/pharmacology , Intestinal Mucosa/drug effects , Intestinal Mucosa/pathology , Paneth Cells/drug effects , Paneth Cells/pathology , Stem Cells/drug effects , Stem Cells/pathology , Animals , Cell Death/drug effects , Cell Death/physiology , Computer Systems , Homeostasis/drug effects , Homeostasis/physiology , Interferon-gamma/physiology , Intestinal Mucosa/physiology , Mice , Mice, Transgenic , Paneth Cells/physiology , Receptors, G-Protein-Coupled/metabolism , Receptors, Interferon/metabolism , Stem Cell Niche/drug effects , Stem Cell Niche/physiology , Stem Cells/physiology , Interferon gamma Receptor
8.
Cells ; 10(2)2021 01 23.
Article in English | MEDLINE | ID: mdl-33498747

ABSTRACT

We have previously reported that histone deacetylase epigenetic regulator Hdac1 and Hdac2 deletion in intestinal epithelial cells (IEC) disrupts mucosal tissue architecture and barrier, causing chronic inflammation. In this study, proteome and transcriptome analysis revealed the importance of signaling pathways induced upon genetic IEC-Hdac1 and Hdac2 deletion. Indeed, Gene Ontology biological process analysis of enriched deficient IEC RNA and proteins identified common pathways, including lipid metabolic and oxidation-reduction process, cell adhesion, and antigen processing and presentation, related to immune responses, correlating with dysregulation of major histocompatibility complex (MHC) class II genes. Top upstream regulators included regulators associated with environmental sensing pathways to xenobiotics, microbial and diet-derived ligands, and endogenous metabolites. Proteome analysis revealed mTOR signaling IEC-specific defects. In addition to mTOR, the STAT and Notch pathways were dysregulated specifically in jejunal IEC. To determine the impact of pathway dysregulation on mutant jejunum alterations, we treated mutant mice with Tofacitinib, a JAK inhibitor. Treatment with the inhibitor partially corrected proliferation and tight junction defects, as well as niche stabilization by increasing Paneth cell numbers. Thus, IEC-specific histone deacetylases 1 (HDAC1) and 2 (HDAC2) support intestinal homeostasis by regulating survival and translation processes, as well as differentiation and metabolic pathways. HDAC1 and HDAC2 may play an important role in the regulation of IEC-specific inflammatory responses by controlling, directly or indirectly, the JAK/STAT pathway. IEC-specific JAK/STAT pathway deregulation may be, at least in part, responsible for intestinal homeostasis disruption in mutant mice.


Subject(s)
Epithelial Cells/metabolism , Histone Deacetylase 1/deficiency , Histone Deacetylase 2/deficiency , Homeostasis , Intestines/cytology , Janus Kinases/metabolism , STAT Transcription Factors/metabolism , Animals , Cell Differentiation/drug effects , Epithelial Cells/drug effects , Gene Deletion , Histone Deacetylase 1/genetics , Histone Deacetylase 1/metabolism , Histone Deacetylase 2/genetics , Histone Deacetylase 2/metabolism , Homeostasis/drug effects , Lymphocyte Count , Mice, Inbred C57BL , Mice, Transgenic , Organoids/drug effects , Organoids/growth & development , Paneth Cells/drug effects , Paneth Cells/metabolism , Piperidines/pharmacology , Pyrimidines/pharmacology , T-Lymphocytes/drug effects
9.
Anat Sci Int ; 96(1): 142-156, 2021 Jan.
Article in English | MEDLINE | ID: mdl-32931001

ABSTRACT

Omeprazole is a potent inhibitor of gastric acid secretion. It was reported that omeprazole induced dramatic gastric mucosa morphologic changes from the resting state to the stimulated state. However, the effect of omeprazole administration on the ultrastructure and absorptive function of small intestines was largely unknown. Here, male Sprague-Dawley rats were daily treated with a single dose of omeprazole for 12 or 24 weeks. Ultrastructure intestinal mucosal change in duodenum, jejunum, and ileum was observed. We also determined small intestine inflammation, using intraepithelial lymphocytes activation. Finally, magnesium levels were measured in plasma, urine, feces, muscle, and bone to determine systemic magnesium balance. Omeprazole-treated rats had significantly decreased the width of tight junction, villous length, and absorptive area of duodenum, jejunum, and ileum compared to control rats. The small intestine of the omeprazole-treated group showed significantly higher intraepithelial lymphocytes activation levels compared with the control group. Lower secretory granules of Paneth cells at the base of the crypts were showed in omeprazole-treated rats. They also had significantly lower plasma, urinary, bone, and muscle Mg2+ contents indicating hypomagnesemia with systemic magnesium deficiency. In conclusion, prolonged omeprazole treatment-induced small intestinal inflammation and villous atrophy, which led to decrease small intestinal magnesium absorption in the condition of proton pump inhibitor-induced hypomagnesemia.


Subject(s)
Gastric Acid/metabolism , Intestinal Mucosa/drug effects , Intestinal Mucosa/ultrastructure , Omeprazole/administration & dosage , Omeprazole/adverse effects , Proton Pump Inhibitors/administration & dosage , Proton Pump Inhibitors/adverse effects , Animals , Atrophy , Hypercalciuria/chemically induced , Inflammation , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Lymphocyte Activation , Magnesium/metabolism , Male , Microscopy, Electron, Transmission , Nephrocalcinosis/chemically induced , Paneth Cells/drug effects , Paneth Cells/pathology , Rats, Sprague-Dawley , Renal Tubular Transport, Inborn Errors/chemically induced , Time Factors
10.
Biochem Biophys Res Commun ; 533(4): 1330-1337, 2020 12 17.
Article in English | MEDLINE | ID: mdl-33066957

ABSTRACT

Epithelial regeneration is essential for homeostasis and mucosal barrier repair. In this study, we aimed to define the effect of IL-10 on mucosal healing. Intestinal stem cells (ISCs) cultures and mice were treated with recombinant mice IL-10 (rmIL-10). The level of cell proliferation, differentiation, death and related signaling pathways for self-renewal of ISCs were measured in vitro and in vivo. It was uncovered that rmIL-10 increased the size and death, but reduced the total number of organoids. In addition, rmIL-10 depleted Lgr5+ ISCs and reduced epithelial proliferation, but enhanced the differentiation of epithelial cells and expanded numbers of transit-amplifying (TA) cells. These changes are related to the decrease of Wnt and Notch signals in vivo and in vitro. Meanwhile, increased expression of Paneth cells and decreased expression of enteroendocrine cells and goblet cells were induced by rmIL-10. Thus, our data indicate that IL-10 reduces the survival of Lgr5+ ISCs and proliferation of epithelial cells by inhibiting Notch and Wnt signaling, but promotes enhanced the differentiation of epithelial cells and expanded numbers of TA cells. Therefore, IL-10 acts as an anti-inflammatory factor, but may damage intestinal mucosa repair and maybe a potential target for the treatment of intestinal injury.


Subject(s)
Interleukin-10/pharmacology , Intestinal Mucosa/cytology , Receptors, G-Protein-Coupled/genetics , Stem Cells/cytology , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Self Renewal/drug effects , Enteroendocrine Cells/drug effects , Gene Expression Regulation/drug effects , Goblet Cells/drug effects , Male , Mice, Inbred C57BL , Mice, Transgenic , Organ Culture Techniques , Paneth Cells/drug effects , Receptors, G-Protein-Coupled/metabolism , Receptors, Notch/metabolism , Recombinant Proteins/pharmacology , Stem Cells/drug effects , Stem Cells/metabolism , Wnt Signaling Pathway/drug effects , Wnt Signaling Pathway/genetics
11.
Blood ; 136(12): 1442-1455, 2020 09 17.
Article in English | MEDLINE | ID: mdl-32542357

ABSTRACT

Acute graft-versus-host disease (GVHD) is a life-threatening complication after allogeneic hematopoietic cell transplantation (allo-HCT). Although currently used GVHD treatment regimens target the donor immune system, we explored here an approach that aims at protecting and regenerating Paneth cells (PCs) and intestinal stem cells (ISCs). Glucagon-like-peptide-2 (GLP-2) is an enteroendocrine tissue hormone produced by intestinal L cells. We observed that acute GVHD reduced intestinal GLP-2 levels in mice and patients developing GVHD. Treatment with the GLP-2 agonist, teduglutide, reduced de novo acute GVHD and steroid-refractory GVHD, without compromising graft-versus-leukemia (GVL) effects in multiple mouse models. Mechanistically GLP-2 substitution promoted regeneration of PCs and ISCs, which enhanced production of antimicrobial peptides and caused microbiome changes. GLP-2 expanded intestinal organoids and reduced expression of apoptosis-related genes. Low numbers of L cells in intestinal biopsies and high serum levels of GLP-2 were associated with a higher incidence of nonrelapse mortality in patients undergoing allo-HCT. Our findings indicate that L cells are a target of GVHD and that GLP-2-based treatment of acute GVHD restores intestinal homeostasis via an increase of ISCs and PCs without impairing GVL effects. Teduglutide could become a novel combination partner for immunosuppressive GVHD therapy to be tested in clinical trials.


Subject(s)
Glucagon-Like Peptide 2/therapeutic use , Graft vs Host Disease/drug therapy , Hematopoietic Stem Cell Transplantation/adverse effects , Intestines/drug effects , Paneth Cells/drug effects , Peptides/therapeutic use , Stem Cells/drug effects , Animals , Female , Gastrointestinal Agents/therapeutic use , Graft vs Host Disease/pathology , Humans , Intestines/cytology , Intestines/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Paneth Cells/pathology , Stem Cells/pathology , Transplantation, Homologous/adverse effects
12.
Nutrients ; 12(4)2020 Mar 26.
Article in English | MEDLINE | ID: mdl-32224880

ABSTRACT

Necrotizing enterocolitis (NEC) remains a significant cause of morbidity and mortality in preterm infants. Formula feeding is a risk factor for NEC and osmolality, which is increased by the fortification that is required for adequate growth of the infant, has been suggested as a potential cause. Our laboratory has shown that Paneth cell disruption followed by induction of dysbiosis can induce NEC-like pathology in the absence of feeds. We hypothesized adding formula feeds to the model would exacerbate intestinal injury and inflammation in an osmolality-dependent manner. NEC-like injury was induced in 14-16 day-old C57Bl/6J mice by Paneth cell disruption with dithizone or diphtheria toxin, followed by feeding rodent milk substitute with varying osmolality (250-1491 mOsm/kg H2O). Animal weight, serum cytokines and osmolality, small intestinal injury, and cecal microbial composition were quantified. Paneth cell-disrupted mice fed formula had significant NEC scores compared to controls and no longer required induction of bacterial dysbiosis. Significant increases in serum inflammatory markers, small intestinal damage, and overall mortality were osmolality-dependent and not related to microbial changes. Overall, formula feeding in combination with Paneth cell disruption induced NEC-like injury in an osmolality-dependent manner, emphasizing the importance of vigilance in designing preterm infant feeds.


Subject(s)
Dysbiosis/metabolism , Enterocolitis, Necrotizing , Infant Formula/adverse effects , Inflammation/metabolism , Paneth Cells , Animals , Animals, Newborn , Disease Models, Animal , Enterocolitis, Necrotizing/metabolism , Enterocolitis, Necrotizing/pathology , Gastrointestinal Microbiome/drug effects , Gastrointestinal Microbiome/physiology , Humans , Infant, Newborn , Infant, Newborn, Diseases , Inflammation/chemically induced , Intestine, Small/metabolism , Intestine, Small/pathology , Mice , Mice, Inbred C57BL , Osmolar Concentration , Paneth Cells/drug effects , Paneth Cells/metabolism , Paneth Cells/pathology
13.
PLoS One ; 15(4): e0230231, 2020.
Article in English | MEDLINE | ID: mdl-32240190

ABSTRACT

Enteroids are cultured primary intestinal epithelial cells that recapitulate epithelial lineage development allowing for a more complex and physiologically relevant model for scientific study. The large presence of intestinal stem cells (ISC) in these enteroids allows for the study of metabolite effects on cellular processes and resulting progeny cells. Short-chain fatty acids (SCFA) such as butyrate (BUT) are bacterial metabolites produced in the gastrointestinal tract that are considered to be beneficial to host cells. Therefore, the objective was to study the effects of SCFAs on biomarkers of ISC activity, differentiation, barrier function and epithelial defense in the intestine using mouse and human enteroid models. Enteroids were treated with two concentrations of acetate (ACET), propionate (PROP), or BUT for 24 h. Enteroids treated with BUT or PROP showed a decrease in proliferation via EdU uptake relative to the controls in both mouse and human models. Gene expression of Lgr5 was shown to decrease with BUT and PROP treatments, but increased with ACET. As a result of BUT and PROP treatments, there was an increase in differentiation markers for enterocyte, Paneth, goblet, and enteroendocrine cells. Gene expression of antimicrobial proteins Reg3ß, Reg3γ, and Defb1 were stimulated by BUT and PROP, but not by ACET which had a greater effect on expression of tight junction genes Cldn3 and Ocln in 3D enteroids. Similar results were obtained with human enteroids treated with 10 mM SCFAs and grown in either 3D or Transwell™ model cultures, although tight junctions were influenced by BUT and PROP, but not ACET in monolayer format. Furthermore, BUT and PROP treatments increased transepithelial electrical resistance after 24 h compared to ACET or control. Overall, individual SCFAs are potent stimulators of cellular gene expression, however, PROP and especially BUT show great efficacy for driving cell differentiation and gene expression.


Subject(s)
Acetic Acid/pharmacology , Butyric Acid/pharmacology , Gene Expression Regulation/drug effects , Propionates/pharmacology , Spheroids, Cellular/drug effects , Animals , Cell Culture Techniques , Cell Differentiation/drug effects , Claudin-3/genetics , Claudin-3/metabolism , Enterocytes/cytology , Enterocytes/drug effects , Enterocytes/metabolism , Enteroendocrine Cells/cytology , Enteroendocrine Cells/drug effects , Enteroendocrine Cells/metabolism , Goblet Cells/cytology , Goblet Cells/drug effects , Goblet Cells/metabolism , Humans , Mice , Occludin/genetics , Occludin/metabolism , Pancreatitis-Associated Proteins/genetics , Pancreatitis-Associated Proteins/metabolism , Paneth Cells/cytology , Paneth Cells/drug effects , Paneth Cells/metabolism , Primary Cell Culture , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Spheroids, Cellular/cytology , Spheroids, Cellular/metabolism , Tight Junctions/drug effects , beta-Defensins/genetics , beta-Defensins/metabolism
14.
Food Funct ; 11(4): 3371-3381, 2020 Apr 01.
Article in English | MEDLINE | ID: mdl-32232254

ABSTRACT

Increasing pressure of life may bring some disease risks and stress injuries, which may destroy the immune system and result in intestinal mucosal immune disorders. In this study, the effects of different doses of ATX (30 mg per kg b.w., 60 mg per kg b.w. and 120 mg per kg b.w.) on intestinal mucosal functions were explored in cyclophosphamide (Cy)-induced immunodeficient mice. The results showed that continuous intraperitoneal injection of 100 mg per kg b.w. Cy for three days led to a persistent decrease of body weight and a range of abnormalities in the intestine of C57BL/6 mice. However, administration of ATX at 60 and 120 mg per kg b.w. could effectively prevent intestinal mucosa from this damage, including reduced levels of oxidative stress (MDA, GSH and GSH-PX), increased intestinal morphological structural integrity, stimulative growth of goblet cells and mucous secretion, decreased development of Paneth cells and expression levels of antimicrobial peptides (AMPs) (Reg-3γ and lysozyme), increased IgA secretion, ameliorative main gut flora (especially total bacteria, Lactobacillus and Enterobacteriaceae spp. ) and its metabolites (acetic acid, propionic acid and butyric acid). These protective effects of ATX were better than those of control-ß-carotene in general. Our results may provide a new protective measure to keep intestinal mucosal barriers, which is of great significance for maintaining immune function in the body.


Subject(s)
Intestinal Mucosa/drug effects , Animals , Body Weight/drug effects , Butyric Acid/metabolism , Cytokines/metabolism , Fatty Acids, Volatile/analysis , Feces/chemistry , Gastrointestinal Microbiome/drug effects , Gene Expression Regulation , Goblet Cells/drug effects , Goblet Cells/metabolism , Intestinal Mucosa/metabolism , Lactobacillus/drug effects , Lactobacillus/metabolism , Male , Mice , Mice, Inbred C57BL , Organ Size/drug effects , Oxidative Stress/drug effects , Paneth Cells/drug effects , Paneth Cells/metabolism , RNA, Messenger/metabolism , Xanthophylls/pharmacology
15.
J Hepatol ; 73(3): 628-639, 2020 09.
Article in English | MEDLINE | ID: mdl-32205193

ABSTRACT

BACKGROUND & AIMS: Paneth cells (PCs) synthesize and secrete antimicrobial peptides that are key mediators of host-microbe interactions, establishing a balance between intestinal microflora and enteric pathogens. We observed that their number increases in experimental portal hypertension and aimed to investigate the mechanisms by which these cells can contribute to the regulation of portal pressure. METHODS: We first treated Math1Lox/LoxVilcreERT2 mice with tamoxifen to induce the complete depletion of intestinal PCs. Subsequently, we performed partial portal vein or bile duct ligation. We then studied the effects of these interventions on hemodynamic parameters, proliferation of blood vessels and the expression of genes regulating angiogenesis. Intestinal organoids were cultured and exposed to different microbial products to study the composition of their secreted products (by proteomics) and their effects on the proliferation and tube formation of endothelial cells (ECs). In vivo confocal laser endomicroscopy was used to confirm the findings on blood vessel proliferation. RESULTS: Portal hypertension was significantly attenuated in PC-depleted mice compared to control mice and was associated with a decrease in portosystemic shunts. Depletion of PCs also resulted in a significantly decreased density of blood vessels in the intestinal wall and mesentery. Furthermore, we observed reduced expression of intestinal genes regulating angiogenesis in Paneth cell depleted mice using arrays and next generation sequencing. Tube formation and wound healing responses were significantly decreased in ECs treated with conditioned media from PC-depleted intestinal organoids exposed to intestinal microbiota-derived products. Proteomic analysis of conditioned media in the presence of PCs revealed an increase in factors regulating angiogenesis and additional metabolic processes. In vivo endomicroscopy showed decreased vascular proliferation in the absence of PCs. CONCLUSIONS: These results suggest that in response to intestinal flora and microbiota-derived factors, PCs secrete not only antimicrobial peptides, but also pro-angiogenic signaling molecules, thereby promoting intestinal and mesenteric angiogenesis and regulating portal hypertension. LAY SUMMARY: Paneth cells are present in the lining of the small intestine. They prevent the passage of bacteria from the intestine into the blood circulation by secreting substances to fight bacteria. In this paper, we discovered that these substances not only act against bacteria, but also increase the quantity of blood vessels in the intestine and blood pressure in the portal vein. This is important, because high blood pressure in the portal vein may result in several complications which could be targeted with novel approaches.


Subject(s)
Escherichia coli Infections/metabolism , Escherichia coli/metabolism , Gastrointestinal Microbiome/genetics , Hypertension, Portal/metabolism , Hypertension, Portal/microbiology , Neovascularization, Pathologic/metabolism , Paneth Cells/metabolism , Animals , Culture Media, Conditioned , Disease Models, Animal , Escherichia coli Infections/microbiology , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Intestine, Small/metabolism , Intestine, Small/microbiology , Male , Mice , Mice, Transgenic , Organoids/metabolism , Organoids/microbiology , Paneth Cells/drug effects , Pore Forming Cytotoxic Proteins/metabolism , Proteome , Proteomics/methods , Tamoxifen/pharmacology
16.
Biosci Biotechnol Biochem ; 84(3): 594-597, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31760857

ABSTRACT

Reg3ß, a lectin, displays antibacterial activity. This study investigated Reg3ß-expressing cells using IL-22-stimulated enteroids. IL-22 stimulation elevated the mRNA and protein levels of Reg3ß. IL-22 also increased the mRNA levels of CD133 (a transit-amplifying cell marker) and lysozyme (a Paneth cell marker). Immunohistochemistry showed partial colocalization of Reg3ß- and lysozyme-positive cells, suggesting that Paneth cells are one of Reg3ß-producing cells.


Subject(s)
Lectins/biosynthesis , Paneth Cells/drug effects , Animals , Biomarkers/metabolism , Interleukins/pharmacology , Lectins/genetics , Lectins/metabolism , Paneth Cells/metabolism , Interleukin-22
17.
Mediators Inflamm ; 2019: 8474523, 2019.
Article in English | MEDLINE | ID: mdl-31827383

ABSTRACT

We previously reported that acute necrotizing pancreatitis (ANP) after normal or high-fat diet is associated with a decreased number of Paneth cells in ileal crypts. Here, we ablated Paneth cells in a rat model of ANP after normal and high-fat diet to investigate the effects on disease symptoms. Adult male Sprague-Dawley rats received standard rat chow or a high-fat diet for 2 weeks, after which they were treated with dithizone to deplete Paneth cells. Six hours later, ANP was established by retrograde injection of sodium taurocholate into the biliopancreatic duct. Rats were sacrificed at 6, 12, and 24 h for assessment. We found dithizone aggravated ANP-associated pathological injuries to the pancreas and ileum in rats on high-fat or standard diets. Lysozyme expression in ileal crypts was decreased, while serum inflammatory cytokines (TNFα, IL-1ß, and IL-17A) and intestinal permeability (serum DAO activity and D-lactate) were increased. Expression of tight junction proteins (claudin-1, zo-1, and occludin) was decreased. Using high-throughput 16S rRNA sequencing, we found dithizone reduced microbiota diversity and altered microbiota composition in rats on high-fat or standard diets. Dithizone decreased fecal short-chain fatty acids (SCFAs) in rats on high-fat or standard diets. Changes in intestinal microbiota correlated significantly with SCFAs, lysozyme, DAO activity, D-lactate, inflammatory cytokines, and pathological injury to the pancreas and ileum in rats on high-fat or standard diets. In conclusion, ablation of Paneth cells exacerbates pancreatic and intestinal injuries in ANP after normal and high-fat diet. These symptoms may be related to changes in the intestinal microbiota.


Subject(s)
Dithizone/pharmacology , Dithizone/therapeutic use , Pancreatitis, Acute Necrotizing/metabolism , Paneth Cells/drug effects , RNA, Ribosomal, 16S/metabolism , Animals , Blotting, Western , Diet, High-Fat , Enzyme-Linked Immunosorbent Assay , Immunohistochemistry , Intestines/drug effects , Intestines/injuries , Male , Muramidase/drug effects , Muramidase/metabolism , Pancreatitis, Acute Necrotizing/drug therapy , Rats , Rats, Sprague-Dawley , Taurocholic Acid/pharmacology
18.
Cell Death Dis ; 10(12): 896, 2019 11 26.
Article in English | MEDLINE | ID: mdl-31772152

ABSTRACT

Alternative pathway NF-κB signalling regulates susceptibility towards developing inflammatory bowel disease (IBD), colitis-associated cancer and sepsis-associated intestinal epithelial cell apoptosis and shedding. However, the cell populations responsible for the perturbed alternative pathway NF-κB signalling in intestinal mucosal pathology remain unclear. In order to investigate the contribution of the epithelial compartment, we have tested whether NF-κB2 regulated transcription in intestinal epithelial cells controls the intestinal epithelial response to cytokines that are known to disrupt intestinal barrier permeability. Enteroids were generated from the proximal, middle and distal regions of small intestine (SI) from C57BL/6J wild-type mice and displayed region-specific morphology that was maintained during sub-culture. Enteroids treated with 100 ng/mL TNF were compared with corresponding regions of SI from C57BL/6J mice treated systemically with 0.33 mg/kg TNF for 1.5 h. TNF-induced apoptosis in all regions of the intestine in vitro and in vivo but resulted in Paneth cell degranulation only in proximal tissue-derived SI and enteroids. TNF also resulted in increased enteroid sphericity (quantified as circularity from two-dimensional bright field images). This response was dose and time-dependent and correlated with active caspase-3 immunopositivity. Proximal tissue-derived enteroids generated from Nfκb2-/- mice showed a significantly blunted circularity response following the addition of TNF, IFNγ, lipopolysaccharide (LPS) activated C57BL/6J-derived bone marrow-derived dendritic cells (BMDC) and secreted factors from LPS-activated BMDCs. However, Nfκb1-/- mouse-derived enteroids showed no significant changes in response to these stimuli. In conclusion, the selection of SI region is important when designing enteroid studies as region-specific identity and response to stimuli such as TNF are maintained in culture. Intestinal epithelial cells are at least partially responsible for regulating their own fate by modulating NF-κB2 signalling in response to stimuli known to be involved in multiple intestinal and systemic diseases. Future studies are warranted to investigate the therapeutic potential of intestinal epithelial NF-κB2 inhibition.


Subject(s)
Bone Marrow Cells/cytology , Dendritic Cells/metabolism , Enterocytes/metabolism , NF-kappa B p52 Subunit/metabolism , Organoids/metabolism , Signal Transduction , Animals , Apoptosis/drug effects , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Cell Degranulation/drug effects , Cell Proliferation/drug effects , Culture Media, Conditioned/pharmacology , Dendritic Cells/drug effects , Enterocytes/cytology , Enterocytes/drug effects , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Interferon-gamma/pharmacology , Intestine, Small/metabolism , Lipopolysaccharides/pharmacology , Mice, Inbred C57BL , Paneth Cells/drug effects , Paneth Cells/metabolism , Reproducibility of Results , Tumor Necrosis Factor-alpha/pharmacology
19.
Mucosal Immunol ; 12(4): 930-944, 2019 07.
Article in English | MEDLINE | ID: mdl-31105269

ABSTRACT

Gut microbial translocation contributes to alcoholic hepatitis. Using a mouse model of alcoholic hepatitis, we investigated the effects of chronic alcohol plus binge and found increased abundance of Paneth cells and IL-17A in the proximal small intestine (PSI). Alcohol increased IL-17A production and pro-apoptotic signaling evidenced by Bax, Bim, caspase-3, and caspase-8 increases via endoplasmic reticulum (ER) stress indicated by C/EBP homologous protein (CHOP) upregulation; this was prevented by the ER stress inhibitor, 4-PBA, in isolated crypts in vitro and in vivo. Mechanistically, IL-17 augmented alcohol-induced ER stress in isolated crypts. In vivo IL-17A blocking antibody administration in alcohol-treated mice attenuated ER stress-mediated apoptosis and IL-18 induction and prevented alcohol-induced impairment of tight junctions in the PSI and LPS translocation to the liver. Acute-on-chronic alcohol resulted in inflammasome activation, caspase-1 cleavage, and IL-18 production in the PSI. In vivo treatment with antibiotics or 4-PBA prevented CHOP upregulation and inflammasome activation. Our data suggest that alcohol upregulates innate immune mechanisms by increasing Paneth cell numbers and IL-17A release contributing to apoptosis amplification, inflammasome activation, and gut leakiness in the PSI. Binge alcohol-induced Paneth cell expansion, ER stress, and inflammasome activation in the PSI are modulated by the gut microbiome.


Subject(s)
Apoptosis , Endoplasmic Reticulum Stress , Inflammasomes/metabolism , Interleukin-17/biosynthesis , Interleukin-18/metabolism , Intestine, Small/metabolism , Paneth Cells/metabolism , Alcohol Drinking , Animals , Apoptosis/drug effects , Biopsy , Caspases/metabolism , Cell Degranulation , Endoplasmic Reticulum Stress/drug effects , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Mice , Paneth Cells/drug effects , Signal Transduction , Transcription Factor CHOP/metabolism
20.
Biomed Res Int ; 2019: 7084734, 2019.
Article in English | MEDLINE | ID: mdl-30941370

ABSTRACT

Butyrate produced by the intestinal microbiota is essential for proper functioning of the intestinal immune system. Total dependence on parenteral nutrition (PN) is associated with numerous adverse effects, including severe microbial dysbiosis and loss of important butyrate producers. We hypothesised that a lack of butyrate produced by the gut microbiota may be compensated by its supplementation in PN mixtures. We tested whether i.v. butyrate administration would (a) positively modulate intestinal defence mechanisms and (b) counteract PN-induced dysbiosis. Male Wistar rats were randomised to chow, PN, and PN supplemented with 9 mM butyrate (PN+But) for 12 days. Antimicrobial peptides, mucins, tight junction proteins, and cytokine expression were assessed by RT-qPCR. T-cell subpopulations in mesenteric lymph nodes (MLN) were analysed by flow cytometry. Microbiota composition was assessed in caecum content. Butyrate supplementation resulted in increased expression of tight junction proteins (ZO-1, claudin-7, E-cadherin), antimicrobial peptides (Defa 8, Rd5, RegIIIγ), and lysozyme in the ileal mucosa. Butyrate partially alleviated PN-induced intestinal barrier impairment and normalised IL-4, IL-10, and IgA mRNA expression. PN administration was associated with an increase in Tregs in MLN, which was normalised by butyrate. Butyrate increased the total number of CD4+ and decreased a relative amount of CD8+ memory T cells in MLN. Lack of enteral nutrition and PN administration led to a shift in caecal microbiota composition. Butyrate did not reverse the altered expression of most taxa but did influence the abundance of some potentially beneficial/pathogenic genera, which might contribute to its overall beneficial effect.


Subject(s)
Butyrates/pharmacology , Dietary Supplements , Gastrointestinal Microbiome , Intestines/pathology , Parenteral Nutrition , Animals , Biodiversity , Colon/drug effects , Colon/pathology , Gastrointestinal Microbiome/drug effects , Gene Expression Regulation/drug effects , Ileum/drug effects , Ileum/pathology , Intestine, Small/drug effects , Lymph Nodes/drug effects , Lymph Nodes/metabolism , Lymphocytes/drug effects , Lymphocytes/metabolism , Male , Models, Animal , Mucins/biosynthesis , Paneth Cells/drug effects , Paneth Cells/metabolism , Peptides/genetics , Peptides/metabolism , Permeability , Phenotype , Phylogeny , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats, Wistar , Tight Junction Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...