Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 7 de 7
1.
EMBO J ; 41(2): e106973, 2022 12 17.
Article En | MEDLINE | ID: mdl-34704277

Circadian rhythms regulate diverse aspects of gastrointestinal physiology ranging from the composition of microbiota to motility. However, development of the intestinal circadian clock and detailed mechanisms regulating circadian physiology of the intestine remain largely unknown. In this report, we show that both pluripotent stem cell-derived human intestinal organoids engrafted into mice and patient-derived human intestinal enteroids possess circadian rhythms and demonstrate circadian phase-dependent necrotic cell death responses to Clostridium difficile toxin B (TcdB). Intriguingly, mouse and human enteroids demonstrate anti-phasic necrotic cell death responses to TcdB. RNA-Seq analysis shows that ~3-10% of the detectable transcripts are rhythmically expressed in mouse and human enteroids. Remarkably, we observe anti-phasic gene expression of Rac1, a small GTPase directly inactivated by TcdB, between mouse and human enteroids, and disruption of Rac1 abolishes clock-dependent necrotic cell death responses. Our findings uncover robust functions of circadian rhythms regulating clock-controlled genes in both mouse and human enteroids governing organism-specific, circadian phase-dependent necrotic cell death responses, and lay a foundation for human organ- and disease-specific investigation of clock functions using human organoids for translational applications.


Circadian Clocks , Jejunum/cytology , Organoids/metabolism , Animals , Bacterial Proteins/toxicity , Bacterial Toxins/toxicity , Cell Death , Cells, Cultured , Humans , Mice , Mice, Inbred C57BL , Organoids/drug effects , Organoids/physiology , rac1 GTP-Binding Protein/genetics , rac1 GTP-Binding Protein/metabolism
2.
Cancer Lett ; 518: 59-71, 2021 10 10.
Article En | MEDLINE | ID: mdl-34126195

Tumors evade immune surveillance by expressing Programmed Death-Ligand 1 (PD-L1), subsequently inhibiting CD8+ cytotoxic T lymphocyte function. Response of gastric cancer to immunotherapy is relatively low. Our laboratory has reported that Helicobacter pylori-induced PD-L1 expression within the gastric epithelium is mediated by the Hedgehog (Hh) signaling pathway. The PI3K/AKT/mTOR pathway is activated in gastric cancer and may have immunomodulatory potential. We hypothesize that Hh signaling mediates mTOR-induced PD-L1 expression. Patient-derived organoids (PDOs) were generated from gastric biopsies and resected tumor tissues. Autologous organoid/immune cell co-cultures were used to study the immunosuppressive function of MDSCs. NanoString Digital Spatial Profiling (DSP) of immune-related protein markers using FFPE slide-mounted tissues from gastric cancer patients was performed. DSP analysis showed infiltration of immunosuppressive MDSCs expressing Arg1, CD66b, VISTA and IDO1 within cancer tissues. Orthotopic transplantation of patient derived organoids (PDOs) resulted in the engraftment of organoids and the development of histology similar to that observed in the patient's tumor tissue. PDO/immune cell co-cultures revealed that PD-L1-expressing organoids were unresponsive to nivolumab in vitro in the presence of PMN-MDSCs. Depletion of PMN-MDSCs within these co-cultures sensitized the organoids to anti-PD-1/PD-L1-induced cancer cell death. Rapamycin decreased phosphorylated S6K, Gli2 and PD-L1 expression in PDO/immune cell co-cultures. Transcriptional regulation of PD-L1 by GLI1 and GLI2 was blocked by rapamycin. In conclusion, the PDO/immune cell co-cultures may be used to study immunosuppressive MDSC function within the gastric tumor microenvironment. The mTOR signaling pathway mediates GLI-induced PD-L1 expression in gastric cancer.


B7-H1 Antigen/genetics , Hedgehog Proteins/genetics , Organoids/metabolism , Stomach Neoplasms/genetics , TOR Serine-Threonine Kinases/genetics , Transcription, Genetic/genetics , Zinc Finger Protein GLI1/genetics , CD8-Positive T-Lymphocytes/metabolism , Cells, Cultured , Helicobacter pylori/pathogenicity , Humans , Immunotherapy/methods , Signal Transduction/genetics , Stomach Neoplasms/microbiology , T-Lymphocytes, Cytotoxic/metabolism , Tumor Microenvironment/genetics
3.
PLoS Pathog ; 15(1): e1007468, 2019 01.
Article En | MEDLINE | ID: mdl-30703170

Helicobacter pylori (H. pylori) is the major risk factor for the development of gastric cancer. Our laboratory has reported that the Sonic Hedgehog (Shh) signaling pathway is an early response to infection that is fundamental to the initiation of H. pylori-induced gastritis. H. pylori also induces programmed death ligand 1 (PD-L1) expression on gastric epithelial cells, yet the mechanism is unknown. We hypothesize that H. pylori-induced PD-L1 expression within the gastric epithelium is mediated by the Shh signaling pathway during infection. To identify the role of Shh signaling as a mediator of H. pylori-induced PD-L1 expression, human gastric organoids generated from either induced pluripotent stem cells (HGOs) or tissue (huFGOs) were microinjected with bacteria and treated with Hedgehog/Gli inhibitor GANT61. Gastric epithelial monolayers generated from the huFGOs were also infected with H. pylori and treated with GANT61 to study the role of Hedgehog signaling as a mediator of induced PD-1 expression. A patient-derived organoid/autologous immune cell co-culture system infected with H. pylori and treated with PD-1 inhibitor (PD-1Inh) was developed to study the protective mechanism of PD-L1 in response to bacterial infection. H. pylori significantly increased PD-L1 expression in organoid cultures 48 hours post-infection when compared to uninfected controls. The mechanism was cytotoxic associated gene A (CagA) dependent. This response was blocked by pretreatment with GANT61. Anti-PD-L1 treatment of H. pylori infected huFGOs, co-cultured with autologous patient cytotoxic T lymphocytes and dendritic cells, induced organoid death. H. pylori-induced PD-L1 expression is mediated by the Shh signaling pathway within the gastric epithelium. Cells infected with H. pylori that express PD-L1 may be protected from the immune response, creating premalignant lesions progressing to gastric cancer.


B7-H1 Antigen/metabolism , Helicobacter Infections/immunology , Adolescent , Antigens, Bacterial/genetics , B7-H1 Antigen/genetics , Epithelial Cells/metabolism , Gastric Mucosa/microbiology , Gastritis/microbiology , Gene Expression Regulation/genetics , Hedgehog Proteins/metabolism , Helicobacter Infections/genetics , Helicobacter pylori/metabolism , Helicobacter pylori/pathogenicity , Humans , Organoids/microbiology , Pyridines/pharmacology , Pyrimidines/pharmacology , Signal Transduction , Stomach , Young Adult
4.
Am J Physiol Gastrointest Liver Physiol ; 310(6): G427-38, 2016 Mar 15.
Article En | MEDLINE | ID: mdl-26702137

Intestinal resection resulting in short bowel syndrome (SBS) carries a heavy burden of long-term morbidity, mortality, and cost of care, which can be attenuated with strategies that improve intestinal adaptation. SBS infants fed human milk, compared with formula, have more rapid intestinal adaptation. We tested the hypothesis that the major noncaloric human milk oligosaccharide 2'-fucosyllactose (2'-FL) contributes to the adaptive response after intestinal resection. Using a previously described murine model of intestinal adaptation, we demonstrated increased weight gain from 21 to 56 days (P < 0.001) and crypt depth at 56 days (P < 0.0095) with 2'-FL supplementation after ileocecal resection. Furthermore, 2'-FL increased small bowel luminal content microbial alpha diversity following resection (P < 0.005) and stimulated a bloom in organisms of the genus Parabacteroides (log2-fold = 4.1, P = 0.035). Finally, transcriptional analysis of the intestine revealed enriched ontologies and pathways related to antimicrobial peptides, metabolism, and energy processing. We conclude that 2'-FL supplementation following ileocecal resection increases weight gain, energy availability through microbial community modulation, and histological changes consistent with improved adaptation.


Adaptation, Physiological/drug effects , Intestines/drug effects , Intestines/surgery , Milk, Human/chemistry , Short Bowel Syndrome/drug therapy , Trisaccharides/pharmacology , Animals , Cecum/surgery , Diet , Digestive System Surgical Procedures , Energy Metabolism/drug effects , Humans , Ileum/surgery , Male , Mice , Mice, Inbred C57BL , Microbiota , RNA, Ribosomal, 16S/biosynthesis , Trisaccharides/chemistry , Weight Gain/drug effects
5.
J Pediatr Gastroenterol Nutr ; 43 Suppl 1: S74-81, 2006 Jul.
Article En | MEDLINE | ID: mdl-16819406

OBJECTIVES AND METHODS: Guanylin and uroguanylin are peptides synthesized in the intestine and kidney that are postulated to have both paracrine and endocrine functions, forming a potential enteric-renal link to coordinate salt ingestion with natriuresis. To explore the in vivo role of guanylin and uroguanylin in the regulation of sodium excretion, we used gene-targeted mice in which the uroguanylin, guanylin or the peptide receptor guanylate cyclase C gene expression had been ablated. RESULTS: Metabolic balance studies demonstrated that there was impaired excretion of a sodium load in uroguanylin (but not in guanylin or guanylate cyclase C) knockout mice. Uroguanylin-dependent natriuresis occurred without an increase in circulating prouroguanylin. A distinct morphological phenotype was present in the proximal convoluted tubules of uroguanylin knockout animals after an enteral salt loading. Marked vacuolization of the proximal convoluted tubule epithelial cells was observed by using light and electron microscopy. There was also a change in the distribution of the sodium hydrogen exchanger 3 (NHE3) after an enteral salt loading. In wild-type animals, there was a partial redistribution of NHE3 from the villus fraction to the less accessible submicrovillus membrane compartment, but this effect was less apparent in uroguanylin knockout animals, presumably resulting in greater Na/H exchange. CONCLUSIONS: Together, these findings further establish a role for uroguanylin in fluid homeostasis and support a role for uroguanylin as an integral component of a signaling mechanism that mediates changes in Na excretion in response to an enteral salt loading. Proximal tubular NHE3 activity is a possible target for uroguanylin-mediated changes in Na excretion.


Gastrointestinal Hormones/metabolism , Kidney Tubules, Proximal/metabolism , Natriuresis , Natriuretic Peptides/metabolism , Analysis of Variance , Animals , Biomarkers/blood , Biomarkers/urine , Blotting, Western , Cyclic GMP/urine , Enteral Nutrition , Fluorescent Antibody Technique, Indirect , Gastrointestinal Hormones/blood , Guanylate Cyclase/drug effects , Guanylate Cyclase/metabolism , Kidney Tubules, Proximal/drug effects , Kidney Tubules, Proximal/physiopathology , Mice , Mice, Knockout , Microscopy, Electron , Models, Animal , Natriuresis/drug effects , Natriuretic Peptides/blood , Potassium Channels/drug effects , Potassium Channels/metabolism , Receptors, Enterotoxin , Receptors, Guanylate Cyclase-Coupled , Receptors, Peptide/drug effects , Receptors, Peptide/metabolism , Sodium Chloride, Dietary/administration & dosage , Sodium-Hydrogen Exchanger 3 , Sodium-Hydrogen Exchangers/drug effects , Sodium-Hydrogen Exchangers/metabolism , Time Factors , Water-Electrolyte Balance/drug effects
6.
J Clin Invest ; 112(8): 1244-54, 2003 Oct.
Article En | MEDLINE | ID: mdl-14561709

Guanylin and uroguanylin, peptides synthesized in the intestine and kidney, have been postulated to have both paracrine and endocrine functions, forming a potential enteric-renal link to coordinate salt ingestion with natriuresis. To explore the in vivo role of uroguanylin in the regulation of sodium excretion, we created gene-targeted mice in which uroguanylin gene expression had been ablated. Northern and Western analysis confirmed the absence of uroguanylin message and protein in knockout mice, and cGMP levels were decreased in the mucosa of the small intestine. Ussing chamber analysis of jejunum revealed that Na+/H+ exchanger-mediated Na+ absorption and tissue conductance was not altered in the knockout animals, but short-circuit current, an index of electrogenic anion secretion, was reduced. Renal clearance measurements showed that uroguanylin deficiency results in impaired ability to excrete an enteral load of NaCl, primarily due to an inappropriate increase in renal Na+ reabsorption. Finally, telemetric recordings of blood pressure demonstrated increased mean arterial pressure in uroguanylin knockout animals that was independent of the level of dietary salt intake. Together, these findings establish a role for uroguanylin in an enteric-renal communication axis as well as a fundamental principle of this axis in the maintenance of salt homeostasis in vivo.


Blood Pressure , Natriuresis , Peptides/physiology , Sodium Chloride/metabolism , Animals , Cyclic GMP/analysis , Genotype , Jejunum/metabolism , Kidney/physiology , Mice , Mice, Inbred BALB C , Mice, Knockout , Natriuretic Peptides
7.
Am J Physiol Gastrointest Liver Physiol ; 283(3): G695-702, 2002 Sep.
Article En | MEDLINE | ID: mdl-12181185

The mechanisms of proguanylin synthesis and secretion in the intestine are incompletely understood. We designed an in vitro model to study proguanylin secretion in a model of intestinal villous epithelial cells. The C2/bbe1 cell line, a differentiated subclone of Caco-2 cells, was used to examine the direction of proguanylin secretion and the potential for feedback regulation via activators of the guanylyl cyclase C signal transduction pathway. When cells were grown on Transwell inserts, proguanylin was secreted into the apical and basolateral media, consistent with other models of intestinal guanylin secretion. Proguanylin synthesis and secretion were not decreased on activation of guanylyl cyclase C-mediated chloride secretion, implying a regulatory system other than negative-feedback inhibition. These data describe the use of C2/bbe1 cells as a model for proguanylin secretion in villous epithelial cells and demonstrate their potential use for the study of the regulatory mechanisms governing proguanylin synthesis and secretion.


Caco-2 Cells/physiology , Gastrointestinal Hormones , Protein Precursors/metabolism , Cell Membrane/metabolism , Chlorides/metabolism , Cytological Techniques/instrumentation , Enterocytes/physiology , Feedback , Guanylate Cyclase/physiology , Humans , Intracellular Membranes/metabolism , Isoenzymes/physiology , Protein Precursors/genetics , RNA, Messenger/metabolism
...