Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 60
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Mol Cell ; 64(5): 900-912, 2016 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-27867006

RESUMEN

Circadian clock-gated cell division cycles are observed from cyanobacteria to mammals via intracellular molecular connections between these two oscillators. Here we demonstrate WNT-mediated intercellular coupling between the cell cycle and circadian clock in 3D murine intestinal organoids (enteroids). The circadian clock gates a population of cells with heterogeneous cell-cycle times that emerge as 12-hr synchronized cell division cycles. Remarkably, we observe reduced-amplitude oscillations of circadian rhythms in intestinal stem cells and progenitor cells, indicating an intercellular signal arising from differentiated cells governing circadian clock-dependent synchronized cell division cycles. Stochastic simulations and experimental validations reveal Paneth cell-secreted WNT as the key intercellular coupling component linking the circadian clock and cell cycle in enteroids.


Asunto(s)
Ciclo Celular/fisiología , Relojes Circadianos/fisiología , Mucosa Intestinal/fisiología , Vía de Señalización Wnt/fisiología , Células Madre Adultas/fisiología , Animales , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Ritmo Circadiano , Yeyuno/metabolismo , Ratones , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Organoides , Proteínas Circadianas Period/genética , Proteínas Circadianas Period/metabolismo , Proteínas Tirosina Quinasas/genética , Proteínas Tirosina Quinasas/metabolismo , Técnicas de Cultivo de Tejidos
2.
Nature ; 541(7636): 182-187, 2017 01 12.
Artículo en Inglés | MEDLINE | ID: mdl-28052057

RESUMEN

Despite the global prevalence of gastric disease, there are few adequate models in which to study the fundus epithelium of the human stomach. We differentiated human pluripotent stem cells (hPSCs) into gastric organoids containing fundic epithelium by first identifying and then recapitulating key events in embryonic fundus development. We found that disruption of Wnt/ß-catenin signalling in mouse embryos led to conversion of fundic to antral epithelium, and that ß-catenin activation in hPSC-derived foregut progenitors promoted the development of human fundic-type gastric organoids (hFGOs). We then used hFGOs to identify temporally distinct roles for multiple signalling pathways in epithelial morphogenesis and differentiation of fundic cell types, including chief cells and functional parietal cells. hFGOs are a powerful model for studying the development of the human fundus and the molecular bases of human gastric physiology and pathophysiology, and also represent a new platform for drug discovery.


Asunto(s)
Fundus Gástrico/metabolismo , Proteínas Wnt/metabolismo , Vía de Señalización Wnt , beta Catenina/metabolismo , Animales , Tipificación del Cuerpo , Diferenciación Celular , Linaje de la Célula , Descubrimiento de Drogas/métodos , Células Epiteliales/citología , Células Epiteliales/metabolismo , Epitelio/embriología , Epitelio/metabolismo , Femenino , Fundus Gástrico/citología , Fundus Gástrico/embriología , Proteínas de Homeodominio/metabolismo , Humanos , Masculino , Ratones , Organoides/citología , Organoides/embriología , Organoides/metabolismo , Células Parietales Gástricas/citología , Células Parietales Gástricas/metabolismo , Células Madre Pluripotentes/citología , Factores de Transcripción SOXB1/metabolismo , Esferoides Celulares/citología , Esferoides Celulares/metabolismo , Transactivadores/metabolismo , Vía de Señalización Wnt/genética , beta Catenina/agonistas
3.
Annu Rev Physiol ; 79: 357-380, 2017 02 10.
Artículo en Inglés | MEDLINE | ID: mdl-27992733

RESUMEN

Trefoil factor (TFF) peptides, with a 40-amino acid motif and including six conserved cysteine residues that form intramolecular disulfide bonds, are a family of mucin-associated secretory molecules mediating many physiological roles that maintain and restore gastrointestinal (GI) mucosal homeostasis. TFF peptides play important roles in response to GI mucosal injury and inflammation. In response to acute GI mucosal injury, TFF peptides accelerate cell migration to seal the damaged area from luminal contents, whereas chronic inflammation leads to increased TFF expression to prevent further progression of disease. Although much evidence supports the physiological significance of TFF peptides in mucosal defenses, the molecular and cellular mechanisms of TFF peptides in the GI epithelium remain largely unknown. In this review, we summarize the functional roles of TFF1, 2, and 3 and illustrate their action mechanisms, focusing on defense mechanisms in the GI tract.


Asunto(s)
Tracto Gastrointestinal/metabolismo , Tracto Gastrointestinal/fisiología , Factores Trefoil/metabolismo , Animales , Movimiento Celular/fisiología , Homeostasis/fisiología , Humanos , Inflamación/metabolismo , Inflamación/patología
4.
J Cell Sci ; 131(16)2018 08 20.
Artículo en Inglés | MEDLINE | ID: mdl-30072444

RESUMEN

The role of the actin cytoskeleton in the sequence of physiological epithelial repair in the intact epithelium has yet to be elucidated. Here, we explore the role of actin in gastric repair in vivo and in vitro gastric organoids (gastroids). In response to two-photon-induced cellular damage of either an in vivo gastric or in vitro gastroid epithelium, actin redistribution specifically occurred in the lateral membranes of cells neighboring the damaged cell. This was followed by their migration inward to close the gap at the basal pole of the dead cell, in parallel with exfoliation of the dead cell into the lumen. The repair and focal increase of actin was significantly blocked by treatment with EDTA or the inhibition of actin polymerization. Treatment with inhibitors of myosin light chain kinase, myosin II, trefoil factor 2 signaling or phospholipase C slowed both the initial actin redistribution and the repair. While Rac1 inhibition facilitated repair, inhibition of RhoA/Rho-associated protein kinase inhibited it. Inhibitors of focal adhesion kinase and Cdc42 had negligible effects. Hence, initial actin polymerization occurs in the lateral membrane, and is primarily important to initiate dead cell exfoliation and cell migration to close the gap.


Asunto(s)
Actinas/metabolismo , Mucosa Gástrica/lesiones , Organoides/lesiones , Multimerización de Proteína/fisiología , Repitelización/fisiología , Estómago/citología , Animales , Movimiento Celular , Células Cultivadas , Células Epiteliales/fisiología , Femenino , Mucosa Gástrica/metabolismo , Mucosa Gástrica/fisiología , Masculino , Ratones , Ratones Transgénicos , Organoides/citología , Organoides/fisiología , Polimerizacion , Regeneración/fisiología , Estómago/lesiones
5.
Dig Dis Sci ; 65(1): 119-131, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31515722

RESUMEN

BACKGROUND: Peptic ulcers recur, suggesting that ulcer healing may leave tissue predisposed to subsequent damage. In mice, we have identified that the regenerated epithelium found after ulcer healing will remain abnormal for months after healing. AIM: To determine whether healed gastric mucosa has altered epithelial function, as measured by electrophysiologic parameters. METHOD: Ulcers were induced in mouse gastric corpus by serosal local application of acetic acid. Thirty days or 8 months after ulcer induction, tissue was mounted in an Ussing chamber. Transepithelial electrophysiologic parameters (short-circuit current, Isc. resistance, R) were compared between the regenerated healed ulcer region and the non-ulcerated contralateral region, in response to luminal hyperosmolar NaCl challenge (0.5 M). RESULTS: In unperturbed stomach, luminal application of hyperosmolar NaCl transiently dropped Isc followed by gradual recovery over 2 h. Compared to the starting baseline Isc, percent Isc recovery was reduced in 30-day healing mucosa, but not at 8 months. Prior to NaCl challenge, a lower baseline Isc was observed in trefoil factor 2 (TFF2) knockout (KO) versus wild type (WT), with no Isc recovery in either non-ulcerated or healing mucosa of KO. Inhibiting Na/H exchanger (NHE) transport in WT mucosa inhibited Isc recovery in response to luminal challenge. NHE2-KO baseline Isc was reduced versus NHE2-WT. In murine gastric organoids, NHE inhibition slowed recovery of intracellular pH and delayed the repair of photic induced damage. CONCLUSION: Healing gastric mucosa has deficient electrophysiological recovery in response to hypertonic NaCl. TFF2 and NHE2 contribute to Isc regulation, and the recovery and healing of transepithelial function.


Asunto(s)
Células Epiteliales/metabolismo , Mucosa Gástrica/metabolismo , Cloruro de Sodio/metabolismo , Intercambiadores de Sodio-Hidrógeno/deficiencia , Úlcera Gástrica/metabolismo , Cicatrización de Heridas , Ácido Acético , Animales , Modelos Animales de Enfermedad , Impedancia Eléctrica , Células Epiteliales/patología , Femenino , Mucosa Gástrica/patología , Concentración de Iones de Hidrógeno , Soluciones Hipertónicas , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Repitelización , Intercambiadores de Sodio-Hidrógeno/genética , Úlcera Gástrica/inducido químicamente , Úlcera Gástrica/genética , Úlcera Gástrica/patología , Factores de Tiempo , Factor Trefoil-2/deficiencia , Factor Trefoil-2/genética
6.
J Physiol ; 597(10): 2673-2690, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30912855

RESUMEN

KEY POINTS: Determining the signalling cascade of epithelial repair, using murine gastric organoids, allows definition of regulatory processes intrinsic to epithelial cells, at the same time as validating and dissecting the signalling cascade with more precision than is possible in vivo Following single cell damage, intracellular calcium selectively increases within cells adjacent to the damage site and is essential for promoting repair. Trefoil factor 2 (TFF2) acts via chemokine C-X-C receptor 4 and epidermal growth factor receptor signalling, including extracellular signal-regulated kinase activation, to drive calcium mobilization and promote gastric repair. Sodium hydrogen exchanger 2, although essential for repair, acts downstream of TFF2 and calcium mobilization. ABSTRACT: The gastric mucosa of the stomach is continually exposed to environmental and physiological stress factors that can cause local epithelial damage. Although much is known about the complex nature of gastric wound repair, the stepwise process that characterizes epithelial restitution remains poorly defined. The present study aimed to determine the effectors that drive gastric epithelial repair using a reductionist culture model. To determine the role of trefoil factor 2 (TFF2) and intracellular calcium (Ca2+ ) mobilization in gastric restitution, gastric organoids were derived from TFF2 knockout (KO) mice and yellow Cameleon-Nano15 (fluorescent calcium reporter) transgenic mice, respectively. Inhibitors and recombinant protein were used to determine the upstream and downstream effectors of gastric restitution following photodamage (PD) to single cells within the gastric organoids. Single cell PD resulted in parallel events of dead cell exfoliation and migration of intact neighbouring cells to restore a continuous epithelium in the damage site. Under normal conditions following PD, Ca2+ levels increased within neighbour migrating cells, peaking at ∼1 min, suggesting localized Ca2+ mobilization at the site of cell protrusion/migration. TFF2 KO organoids exhibit delayed repair; however, this delay can be rescued by the addition of exogenous TFF2. Inhibition of epidermal growth factor receptor (EGFR), extracellular signal-regulated kinase (ERK)1/2 or a TFF2 receptor, chemokine C-X-C receptor 4 (CXCR4), resulted in significant delay and dampened Ca2+ mobilization. Inhibition of sodium hydrogen exchanger 2 (NHE2) caused significant delay but did not affect Ca2+ mobilization. A similar delay was observed in NHE2 KO organoids. In TFF2 KO gastric organoids, the addition of exogenous TFF2 in the presence of EGFR or CXCR4 inhibition was unable to rescue repair. The present study demonstrates that intracellular Ca2+ mobilization occurs within gastric epithelial cells adjacent to the damage site to promote repair by mechanisms that involve TFF2 signalling via CXCR4, as well as activation of EGFR and ERK1/2. Furthermore NHE2 is shown to be important for efficient repair and to operate via a mechanism either downstream or independent of calcium mobilization.


Asunto(s)
Calcio/metabolismo , Organoides/metabolismo , Receptores CXCR4/metabolismo , Factor Trefoil-2/metabolismo , Animales , Calcio/farmacología , Epitelio , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Receptores CXCR4/genética , Intercambiadores de Sodio-Hidrógeno/antagonistas & inhibidores , Intercambiadores de Sodio-Hidrógeno/genética , Intercambiadores de Sodio-Hidrógeno/metabolismo , Estómago , Factor Trefoil-2/administración & dosificación , Factor Trefoil-2/genética , Factor Trefoil-2/farmacología
7.
Infect Immun ; 87(9)2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31262979

RESUMEN

Helicobacter pylori is a pathogen that chronically colonizes the stomachs of approximately half of the world's population and contributes to the development of gastric inflammation. We demonstrated previously in vivo that H. pylori uses motility to preferentially colonize injury sites in the mouse stomach. However, the chemoreceptor responsible for sensing gastric injury has not yet been identified. In this study, we utilized murine gastric organoids (gastroids) and mutant H. pylori strains to investigate the components necessary for H. pylori chemotaxis. High-intensity 730-nm light (two-photon photodamage) was used to cause single-cell damage in gastroids, and repair of the damage was monitored over time; complete repair occurred within ∼10 min in uninfected gastroids. Wild-type H. pylori accumulated at the damage site after gastric damage induction. In contrast, mutants lacking motility (ΔmotB) or chemotaxis (ΔcheY) did not accumulate at the injury site. Using mutants lacking individual chemoreceptors, we found that only TlpB was required for H. pylori accumulation, while TlpA, TlpC, and TlpD were dispensable. All strains that were able to accumulate at the damage site limited repair. When urea (an identified chemoattractant sensed by TlpB) was microinjected into the gastroid lumen, it prevented the accumulation of H. pylori at damage sites. Overall, our findings demonstrate that H. pylori colonizes and limits repair at damage sites via chemotactic motility that requires the TlpB chemoreceptor to sense signals generated by gastric epithelial cells.


Asunto(s)
Proteínas Bacterianas/fisiología , Factores Quimiotácticos/farmacología , Quimiotaxis/fisiología , Infecciones por Helicobacter/microbiología , Helicobacter pylori/efectos de los fármacos , Gastropatías/microbiología , Animales , Modelos Animales de Enfermedad , Mucosa Gástrica/microbiología , Ratones
9.
PLoS Pathog ; 10(7): e1004275, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25033386

RESUMEN

Helicobacter pylori (H. pylori) is a pathogen contributing to peptic inflammation, ulceration, and cancer. A crucial step in the pathogenic sequence is when the bacterium first interacts with gastric tissue, an event that is poorly understood in vivo. We have shown that the luminal space adjacent to gastric epithelial damage is a microenvironment, and we hypothesized that this microenvironment might enhance H. pylori colonization. Inoculation with 106 H. pylori (wild-type Sydney Strain 1, SS1) significantly delayed healing of acetic-acid induced ulcers at Day 1, 7 and 30 post-inoculation, and wild-type SS1 preferentially colonized the ulcerated area compared to uninjured gastric tissue in the same animal at all time points. Gastric resident Lactobacillus spp. did not preferentially colonize ulcerated tissue. To determine whether bacterial motility and chemotaxis are important to ulcer healing and colonization, we analyzed isogenic H. pylori mutants defective in motility (ΔmotB) or chemotaxis (ΔcheY). ΔmotB (10(6)) failed to colonize ulcerated or healthy stomach tissue. ΔcheY (10(6)) colonized both tissues, but without preferential colonization of ulcerated tissue. However, ΔcheY did modestly delay ulcer healing, suggesting that chemotaxis is not required for this process. We used two-photon microscopy to induce microscopic epithelial lesions in vivo, and evaluated accumulation of fluorescently labeled H. pylori at gastric damage sites in the time frame of minutes instead of days. By 5 min after inducing damage, H. pylori SS1 preferentially accumulated at the site of damage and inhibited gastric epithelial restitution. H. pylori ΔcheY modestly accumulated at the gastric surface and inhibited restitution, but did not preferentially accumulate at the injury site. H. pylori ΔmotB neither accumulated at the surface nor inhibited restitution. We conclude that bacterial chemosensing and motility rapidly promote H. pylori colonization of injury sites, and thereby biases the injured tissue towards sustained gastric damage.


Asunto(s)
Mucosa Gástrica , Infecciones por Helicobacter/inmunología , Helicobacter pylori/inmunología , Úlcera Gástrica/inmunología , Úlcera Gástrica/microbiología , Ácido Acético/efectos adversos , Ácido Acético/farmacología , Animales , Mucosa Gástrica/inmunología , Mucosa Gástrica/lesiones , Mucosa Gástrica/microbiología , Mucosa Gástrica/patología , Infecciones por Helicobacter/patología , Indicadores y Reactivos/efectos adversos , Indicadores y Reactivos/farmacología , Ratones , Úlcera Gástrica/inducido químicamente , Úlcera Gástrica/patología
10.
Gut ; 64(5): 720-30, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25123931

RESUMEN

OBJECTIVE: Helicobacter pylori strains that express the oncoprotein CagA augment risk for gastric cancer. However, the precise mechanisms through which cag(+) strains heighten cancer risk have not been fully delineated and model systems that recapitulate the gastric niche are critical for understanding pathogenesis. Gastroids are three-dimensional organ-like structures that provide unique opportunities to study host-H. pylori interactions in a preclinical model. We used gastroids to inform and direct in vitro studies to define mechanisms through which H. pylori modulates expression of the cancer-associated tight junction protein claudin-7. DESIGN: Gastroids were infected by luminal microinjection, and MKN28 gastric epithelial cells were cocultured with H. pylori wild-type cag(+) strains or isogenic mutants. ß-catenin, claudin-7 and snail localisation was determined by immunocytochemistry. Proliferation was assessed using 5-ethynyl-2'-deoxyuridine, and levels of claudin-7 and snail were determined by western blot and flow cytometry. RESULTS: Gastroids developed into a self-organising differentiation axis and H. pylori induced mislocalisation of claudin-7 and increased proliferation in a CagA- and ß-catenin-dependent manner. In MKN28 cells, H pylori-induced suppression of claudin-7 was regulated by ß-catenin and snail. Similarly, snail expression was increased and claudin-7 levels were decreased among H. pylori-infected individuals. CONCLUSIONS: H. pylori increase proliferation in a strain-specific manner in a novel gastroid system. H. pylori also alter expression and localisation of claudin-7 in gastroids and human epithelial cells, which is mediated by ß-catenin and snail activation. These data provide new insights into molecular interactions with carcinogenic potential that occur between H. pylori and epithelial cells within the gastric niche.


Asunto(s)
Claudinas/metabolismo , Mucosa Gástrica/metabolismo , Mucosa Gástrica/microbiología , Infecciones por Helicobacter/metabolismo , Helicobacter pylori/patogenicidad , Animales , Proliferación Celular , Células Cultivadas , Técnicas de Cocultivo , Células Epiteliales/metabolismo , Células Epiteliales/microbiología , Humanos , Ratones Endogámicos C57BL , Factores de Transcripción de la Familia Snail , Factores de Transcripción/metabolismo , beta Catenina/metabolismo
11.
J Physiol ; 593(8): 1809-27, 2015 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-25605613

RESUMEN

KEY POINTS: An in vitro approach to study gastric development is primary mouse-derived epithelium cultured as three-dimensional spheroids known as organoids. We have devised two unique gastric fundic-derived organoid cultures: model 1 for the expansion of gastric fundic stem cells, and model 2 for the maintenance of mature cell lineages. Organoids maintained in co-culture with immortalized stomach mesenchymal cells express robust numbers of surface pit, mucous neck, chief, endocrine and parietal cells. Histamine induced a significant decrease in intraluminal pH that was reversed by omeprazole in fundic organoids and indicated functional activity and regulation of parietal cells. Localized photodamage resulted in rapid cell exfoliation coincident with migration of neighbouring cells to the damaged area, sustaining epithelial continuity. We report the use of these models for studies of epithelial cell biology and cell damage and repair. ABSTRACT: Studies of gastric function and disease have been limited by the lack of extended primary cultures of the epithelium. An in vitro approach to study gastric development is primary mouse-derived antral epithelium cultured as three-dimensional spheroids known as organoids. There have been no reports on the use of organoids for gastric function. We have devised two unique gastric fundic-derived organoid cultures: model 1 for the expansion of gastric fundic stem cells, and model 2 for the maintenance of mature cell lineages. Both models were generated from single glands dissociated from whole fundic tissue and grown in basement membrane matrix (Matrigel) and organoid growth medium. Model 1 enriches for a stem cell-like niche via simple passage of the organoids. Maintained in Matrigel and growth medium, proliferating organoids expressed high levels of stem cell markers CD44 and Lgr5. Model 2 is a system of gastric organoids co-cultured with immortalized stomach mesenchymal cells (ISMCs). Organoids maintained in co-culture with ISMCs express robust numbers of surface pit, mucous neck, chief, endocrine and parietal cells. Histamine induced a significant decrease in intraluminal pH that was reversed by omeprazole in fundic organoids and indicated functional activity and regulation of parietal cells. Localized photodamage resulted in rapid cell exfoliation coincident with migration of neighbouring cells to the damaged area, sustaining epithelial continuity. Thus, we report the use of these models for studies of epithelial cell biology and cell damage and repair.


Asunto(s)
Técnicas de Cocultivo/métodos , Células Epiteliales/citología , Mucosa Gástrica/citología , Organoides/citología , Células Madre/citología , Animales , Diferenciación Celular , Ratones
12.
Gastroenterology ; 147(3): 655-666.e9, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24859162

RESUMEN

BACKGROUND & AIMS: Loss of expression of Sonic Hedgehog (Shh) from parietal cells results in hypergastrinemia in mice, accompanied by increased expression of Indian Hedgehog (Ihh) and hyperproliferation of surface mucous cells. We investigated whether hypergastrinemia induces gastric epithelial proliferation by activating Ihh signaling in mice. METHODS: We studied mice with parietal cell-specific deletion of Shh (PC-Shh(KO)) and hypergastrinemia, crossed with gastrin-deficient (GKO) mice (PC-Shh(KO)/GKO). When mice were 3-4 months old, gastric tissues were collected and analyzed by histology, for incorporation of bromodeoxyuridine, and for expression of the surface mucous cell marker Ulex europaeus. PC-Shh(KO)/GKO mice were given gastrin infusions for 7 days; gastric surface epithelium was collected and expression of Ihh was quantified by laser capture microdissection followed by quantitative reverse transcriptase polymerase chain reaction. Mouse stomach-derived organoids were incubated with or without inhibitors of WNT (DKK1) or Smoothened (vismodegib) and then cocultured with immortalized stomach mesenchymal cells, to assess proliferative responses to gastrin. RESULTS: Gastric tissues from PC-Shh(KO)/GKO mice with hypergastrinemia had an expanded surface pit epithelium, indicated by a significant increase in numbers of bromodeoxyuridine- and Ulex europaeus-positive cells, but there was no evidence for hyperproliferation. Gastrin infusion of PC PC-Shh(KO)/GKO mice increased expression of Ihh and proliferation within the surface epithelium compared with mice given infusions of saline. In gastric organoids cocultured with immortalized stomach mesenchymal cells, antagonists of WNT and Smoothened inhibited gastrin-induced proliferation and WNT activity. Activity of WNT in media collected from immortalized stomach mesenchymal cells correlated with increased expression of glioma-associated oncogene homolog 1, and was inhibited by DKK1 or vismodegib. CONCLUSIONS: Ihh signaling mediates gastrin-induced proliferation of epithelial cells in stomachs of adult mice.


Asunto(s)
Proliferación Celular , Células Epiteliales/metabolismo , Mucosa Gástrica/metabolismo , Gastrinas/metabolismo , Proteínas Hedgehog/metabolismo , Gastropatías/metabolismo , Animales , Línea Celular , Técnicas de Cocultivo , Modelos Animales de Enfermedad , Células Epiteliales/patología , Mucosa Gástrica/patología , Gastrinas/administración & dosificación , Gastrinas/deficiencia , Gastrinas/genética , Proteínas Hedgehog/deficiencia , Proteínas Hedgehog/genética , Infusiones Parenterales , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Factores de Transcripción de Tipo Kruppel/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Organoides , Receptores Acoplados a Proteínas G/metabolismo , Receptor Smoothened , Gastropatías/genética , Gastropatías/patología , Factores de Tiempo , Vía de Señalización Wnt , Proteína con Dedos de Zinc GLI1
13.
Helicobacter ; 20(1): 19-28, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25495001

RESUMEN

BACKGROUND: Helicobacter pylori (H. pylori) infection leads to acute induction of Sonic Hedgehog (Shh) in the stomach that is associated with the initiation of gastritis. The mechanism by which H. pylori induces Shh is unknown. Shh is a target gene of transcription factor Nuclear Factor-κB (NFκB). We hypothesize that NFκB mediates H. pylori-induced Shh. MATERIALS AND METHODS: To visualize Shh ligand expression in response to H. pylori infection in vivo, we used a mouse model that expresses Shh fused to green fluorescent protein (Shh::GFP mice) in place of wild-type Shh. In vitro, changes in Shh expression were measured in response to H. pylori infection using 3-dimensional epithelial cell cultures grown from whole dissociated gastric glands (organoids). Organoids were generated from stomachs collected from the fundic region of control and mice expressing a parietal cell-specific deletion of Shh (PC-Shh(KO) mice). RESULTS: Within 2 days of infection, H. pylori induced Shh expression within parietal cells of Shh::GFP mice. Organoids expressed all major gastric cell markers, including parietal cell marker H(+) ,K(+) -ATPase and Shh. H. pylori infection of gastric organoids induced Shh expression; a response that was blocked by inhibiting NFκB signaling and correlated with IκB degradation. H. pylori infection of PC-Shh(KO) mouse-derived organoids did not result in the induction of Shh expression. CONCLUSION: Gastric organoids allow for the study of the interaction between H. pylori and the differentiated gastric epithelium independent of the host immune response. H. pylori induces Shh expression from the parietal cells, a response mediated via activation of NFκB signaling.


Asunto(s)
Técnicas Citológicas/métodos , Células Epiteliales/microbiología , Células Epiteliales/patología , Proteínas Hedgehog/biosíntesis , Helicobacter pylori/fisiología , Interacciones Huésped-Patógeno , FN-kappa B/metabolismo , Animales , Fusión Artificial Génica , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Proteínas Fluorescentes Verdes/análisis , Infecciones por Helicobacter/microbiología , Infecciones por Helicobacter/patología , Ratones , Ratones Noqueados , Organoides , Proteínas Recombinantes de Fusión/análisis
14.
J Biol Chem ; 288(47): 33585-33597, 2013 Nov 22.
Artículo en Inglés | MEDLINE | ID: mdl-24121509

RESUMEN

We report that a localized intracellular and extracellular Ca(2+) mobilization occurs at the site of microscopic epithelial damage in vivo and is required to mediate tissue repair. Intravital confocal/two-photon microscopy continuously imaged the surgically exposed stomach mucosa of anesthetized mice while photodamage of gastric epithelial surface cells created a microscopic lesion that healed within 15 min. Transgenic mice with an intracellular Ca(2+)-sensitive protein (yellow cameleon 3.0) report that intracellular Ca(2+) selectively increases in restituting gastric epithelial cells adjacent to the damaged cells. Pretreatment with U-73122, indomethacin, 2-aminoethoxydiphenylborane, or verapamil inhibits repair of the damage and also inhibits the intracellular Ca(2+) increase. Confocal imaging of Fura-Red dye in luminal superfusate shows a localized extracellular Ca(2+) increase at the gastric surface adjacent to the damage that temporally follows intracellular Ca(2+) mobilization. Indomethacin and verapamil also inhibit the luminal Ca(2+) increase. Intracellular Ca(2+) chelation (1,2-bis(o-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid/acetoxymethyl ester, BAPTA/AM) fully inhibits intracellular and luminal Ca(2+) increases, whereas luminal calcium chelation (N-(2-hydroxyetheyl)-ethylendiamin-N,N,N'-triacetic acid trisodium, HEDTA) blocks the increase of luminal Ca(2+) and unevenly inhibits late-phase intracellular Ca(2+) mobilization. Both modes of Ca(2+) chelation slow gastric repair. In plasma membrane Ca-ATPase 1(+/-) mice, but not plasma membrane Ca-ATPase 4(-/-) mice, there is slowed epithelial repair and a diminished gastric surface Ca(2+) increase. We conclude that endogenous Ca(2+), mobilized by signaling pathways and transmembrane Ca(2+) transport, causes increased Ca(2+) levels at the epithelial damage site that are essential to gastric epithelial cell restitution in vivo.


Asunto(s)
Señalización del Calcio , Calcio/metabolismo , Mucosa Gástrica/lesiones , Cicatrización de Heridas , Animales , Antiinflamatorios no Esteroideos/farmacología , Compuestos de Boro/farmacología , Bloqueadores de los Canales de Calcio/farmacología , ATPasas Transportadoras de Calcio/antagonistas & inhibidores , ATPasas Transportadoras de Calcio/genética , ATPasas Transportadoras de Calcio/metabolismo , Quelantes/farmacología , Ácido Edético/análogos & derivados , Ácido Edético/farmacología , Ácido Egtácico/análogos & derivados , Ácido Egtácico/farmacología , Estrenos/farmacología , Mucosa Gástrica/metabolismo , Mucosa Gástrica/patología , Indometacina/farmacología , Ratones , Ratones Noqueados , Inhibidores de Fosfodiesterasa/farmacología , Pirrolidinonas/farmacología , Verapamilo/farmacología
15.
Am J Physiol Gastrointest Liver Physiol ; 306(11): G1002-10, 2014 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-24742986

RESUMEN

Pharmacotherapy based on 5-aminosalicylic acid (5-ASA) is a preferred treatment for ulcerative colitis, but variable patient response to this therapy is observed. Inflammation can affect therapeutic outcomes by regulating the expression and activity of drug-metabolizing enzymes; its effect on 5-ASA metabolism by the colonic arylamine N-acetyltransferase (NAT) enzyme isoforms is not firmly established. We examined if inflammation affects the capacity for colonic 5-ASA metabolism and NAT enzyme expression. 5-ASA metabolism by colonic mucosal homogenates was directly measured with a novel fluorimetric rate assay. 5-ASA metabolism reported by the assay was dependent on Ac-CoA, inhibited by alternative NAT substrates (isoniazid, p-aminobenzoylglutamate), and saturable with Km (5-ASA) = 5.8 µM. A mouse model of acute dextran sulfate sodium (DSS) colitis caused pronounced inflammation in central and distal colon, and modest inflammation of proximal colon, defined by myeloperoxidase activity and histology. DSS colitis reduced capacity for 5-ASA metabolism in central and distal colon segments by 52 and 51%, respectively. Use of selective substrates of NAT isoforms to inhibit 5-ASA metabolism suggested that mNAT2 mediated 5-ASA metabolism in normal and colitis conditions. Western blot and real-time RT-PCR identified that proximal and distal mucosa had a decreased mNAT2 protein-to-mRNA ratio after DSS. In conclusion, an acute colonic inflammation impairs the expression and function of mNAT2 enzyme, thereby diminishing the capacity for 5-ASA metabolism by colonic mucosa.


Asunto(s)
Arilamina N-Acetiltransferasa/metabolismo , Colitis/patología , Colon/metabolismo , Regulación Enzimológica de la Expresión Génica/fisiología , Mesalamina/metabolismo , Animales , Antiinflamatorios no Esteroideos/metabolismo , Arilamina N-Acetiltransferasa/genética , Colitis/inducido químicamente , Sulfato de Dextran/toxicidad , Humanos , Mucosa Intestinal/enzimología , Mucosa Intestinal/patología , Masculino , Ratones , Ratones Endogámicos C57BL
16.
Am J Physiol Gastrointest Liver Physiol ; 305(10): G697-711, 2013 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-24072680

RESUMEN

Changes in the intestinal microbiota have been linked to diabetes, obesity, inflammatory bowel disease, and Clostridium difficile (C. difficile)-associated disease. Despite this, it remains unclear how the intestinal environment, set by ion transport, affects luminal and mucosa-associated bacterial composition. Na(+)/H(+)-exchanger isoform 3 (NHE3), a target of C. difficile toxin B, plays an integral role in intestinal Na(+) absorption. Thus the NHE3-deficient mouse model was chosen to examine the effect of pH and ion composition on bacterial growth. We hypothesized that ion transport-induced change in the intestinal environment would lead to alteration of the microbiota. Region-specific changes in ion composition and pH correlated with region-specific alteration of luminal and mucosal-associated bacteria with general decreases in Firmicutes and increases in Bacteroidetes members. Bacteroides thetaiotaomicron (B. thetaiotaomicron) increased in NHE3(-/-) terminal ileum and was examined in vitro to determine whether altered Na(+) was sufficient to affect growth. Increased in vitro growth of B. thetaiotaomicron occurred in 43 mM Na(+) correlating with the NHE3(-/-) mouse terminal ileum [Na(+)]. NHE3(-/-) terminal ileum displayed increased fut2 mRNA and fucosylation correlating with B. thetaiotaomicron growth. Inoculation of B. thetaiotaomicron in wild-type and NHE3(-/-) terminal ileum organoids displayed increased fut2 and fucosylation, indicating that B. thetaiotaomicron alone is sufficient for the increased fucosylation seen in vivo. These data demonstrate that loss of NHE3 alters the intestinal environment, leading to region-specific changes in bacteria, and shed light on the growth requirements of some gut microbiota members, which is vital for creating better treatments of complex diseases with an altered gut microbiota.


Asunto(s)
Bacteroides/clasificación , Bacteroides/crecimiento & desarrollo , Intercambiadores de Sodio-Hidrógeno/metabolismo , Animales , Medios de Cultivo , Concentración de Iones de Hidrógeno , Intestinos/microbiología , Intestinos/fisiología , Ratones , Sodio , Intercambiador 3 de Sodio-Hidrógeno , Intercambiadores de Sodio-Hidrógeno/genética
17.
Gut ; 61(6): 804-11, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21997560

RESUMEN

BACKGROUND AND AIMS: The authors' goal was to measure pH at the gastric surface (pH0) to understand how acid secretion affects the repair of microscopic injury to the gastric epithelium. METHODS: Microscopic gastric damage was induced by laser light, during confocal/two-photon imaging of pH-sensitive dyes (Cl-NERF, BCECF) that were superfused over the mucosal surface of the exposed gastric corpus of anaesthetised mice. The progression of repair was measured in parallel with pH0. Experimental conditions included varying pH of luminal superfusates, and using omeprazole (60 mg/kg ip) or famotidine (30 mg/kg ip) to inhibit acid secretion. RESULTS: Similar rates of epithelial repair and resting pH0 values (∼pH 4) were reported in the presence of luminal pH 3 or pH 5. Epithelial repair was unreliable at luminal pH 2 and pH0 was lower (2.5±0.2, P <0.05 vs pH 3). Epithelial repair was slower at luminal pH 7 and pH0 was higher (6.4±0.1, P<0.001). In all conditions, pH0 increased adjacent to damage. At luminal pH 3 or pH 7, omeprazole reduced maximal damage size and accelerated epithelial repair, although only at pH 3 did omeprazole further increase surface pH above the level caused by imposed damage. At luminal pH 7, famotidine also reduced maximal damage size and accelerated epithelial repair. Neither famotidine nor omeprazole raised plasma gastrin levels during the time course of the experiments. CONCLUSIONS: Epithelial repair in vivo is affected by luminal pH variation, but the beneficial effects of acutely blocking acid secretion extend beyond simply raising luminal and/or surface pH.


Asunto(s)
Famotidina/uso terapéutico , Ácido Gástrico/metabolismo , Omeprazol/uso terapéutico , Estómago/lesiones , Cicatrización de Heridas/efectos de los fármacos , Animales , Epitelio/lesiones , Femenino , Determinación de la Acidez Gástrica , Gastrinas/sangre , Concentración de Iones de Hidrógeno , Masculino , Ratones , Ratones Endogámicos C57BL
18.
J Biol Chem ; 286(44): 38375-38382, 2011 Nov 04.
Artículo en Inglés | MEDLINE | ID: mdl-21900251

RESUMEN

Trefoil factor (TFF) peptides are pivotal for gastric restitution after surface epithelial damage, but TFF cellular targets that promote cell migration are poorly understood. Conversely, Na/H exchangers (NHE) are often implicated in cellular migration but have a controversial role in gastric restitution. Using intravital microscopy to create microscopic lesions in the mouse gastric surface epithelium and directly measure epithelial restitution, we evaluated whether TFFs and NHE isoforms share a common pathway to promote epithelial repair. Blocking Na/H exchange (luminal 10 µm 5-(N-ethyl-N-isopropyl) amiloride or 25 µm HOE694) slows restitution 72-83% in wild-type or NHE1(-/-) mice. In contrast, HOE694 has no effect on the intrinsically defective gastric restitution in NHE2(-/-) mice or TFF2(-/-) mice. In TFF2(-/-) mice, NHE2 protein is reduced 23%, NHE2 remains localized to apical membranes of surface epithelium, and NHE1 protein amount or localization is unchanged. The action of topical rat TFF3 to accelerate restitution in TFF2(-/-) mice was inhibited by AMD3100 (CXCR4 receptor antagonist). Furthermore, rat TFF3 did not rescue restitution when NHE2 was inhibited [TFF2(-/-) mice +HOE694, or NHE2(-/-) mice]. HOE694 had no effect on pH at the juxtamucosal surface before or after damage. We conclude that functional NHE2, but not NHE1, is essential for mouse gastric epithelial restitution and that TFFs activate epithelial repair via NHE2.


Asunto(s)
Células Epiteliales/citología , Mucosa Gástrica/metabolismo , Mucinas/fisiología , Proteínas Musculares/fisiología , Péptidos/fisiología , Intercambiadores de Sodio-Hidrógeno/metabolismo , Animales , Concentración de Iones de Hidrógeno , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Confocal/métodos , Mucinas/genética , Proteínas Musculares/genética , Péptidos/genética , Fosfoproteínas/metabolismo , Ratas , Receptores CXCR4/metabolismo , Intercambiadores de Sodio-Hidrógeno/genética , Factor Trefoil-2 , Cicatrización de Heridas
19.
Gastroenterology ; 140(4): 1208-1218.e1-2, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21237166

RESUMEN

BACKGROUND & AIMS: Tumor necrosis factor (TNF) increases intestinal epithelial cell shedding and apoptosis, potentially challenging the barrier between the gastrointestinal lumen and internal tissues. We investigated the mechanism of tight junction remodeling and barrier maintenance as well as the roles of cytoskeletal regulatory molecules during TNF-induced shedding. METHODS: We studied wild-type and transgenic mice that express the fluorescent-tagged proteins enhanced green fluorescent protein-occludin or monomeric red fluorescent protein 1-ZO-1. After injection of high doses of TNF (7.5 µg intraperitoneally), laparotomies were performed and segments of small intestine were opened to visualize the mucosa by video confocal microscopy. Pharmacologic inhibitors and knockout mice were used to determine the roles of caspase activation, actomyosin, and microtubule remodeling and membrane trafficking in epithelial shedding. RESULTS: Changes detected included redistribution of the tight junction proteins ZO-1 and occludin to lateral membranes of shedding cells. These proteins ultimately formed a funnel around the shedding cell that defined the site of barrier preservation. Claudins, E-cadherin, F-actin, myosin II, Rho-associated kinase (ROCK), and myosin light chain kinase (MLCK) were also recruited to lateral membranes. Caspase activity, myosin motor activity, and microtubules were required to initiate shedding, whereas completion of the process required microfilament remodeling and ROCK, MLCK, and dynamin II activities. CONCLUSIONS: Maintenance of the epithelial barrier during TNF-induced cell shedding is a complex process that involves integration of microtubules, microfilaments, and membrane traffic to remove apoptotic cells. This process is accompanied by redistribution of apical junctional complex proteins to form intercellular barriers between lateral membranes and maintain mucosal function.


Asunto(s)
Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Uniones Estrechas/metabolismo , Uniones Estrechas/patología , Citoesqueleto de Actina/metabolismo , Animales , Apoptosis/fisiología , Caspasas/metabolismo , Dinamina II/metabolismo , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Células Epiteliales/patología , Proteínas Fluorescentes Verdes/genética , Mucosa Intestinal/efectos de los fármacos , Proteínas Luminiscentes/genética , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microtúbulos/metabolismo , Cadenas Ligeras de Miosina/metabolismo , Quinasa de Cadena Ligera de Miosina/metabolismo , Ocludina , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Transporte de Proteínas/fisiología , Uniones Estrechas/efectos de los fármacos , Factor de Necrosis Tumoral alfa/farmacología , Proteína de la Zonula Occludens-1 , Quinasas Asociadas a rho/genética , Quinasas Asociadas a rho/metabolismo , Proteína Fluorescente Roja
20.
Cell Rep ; 39(11): 110937, 2022 06 14.
Artículo en Inglés | MEDLINE | ID: mdl-35705057

RESUMEN

Intestinal epithelial tight junction disruption is a primary contributing factor in alcohol-associated endotoxemia, systemic inflammation, and multiple organ damage. Ethanol and acetaldehyde disrupt tight junctions by elevating intracellular Ca2+. Here we identify TRPV6, a Ca2+-permeable channel, as responsible for alcohol-induced elevation of intracellular Ca2+, intestinal barrier dysfunction, and systemic inflammation. Ethanol and acetaldehyde elicit TRPV6 ionic currents in Caco-2 cells. Studies in Caco-2 cell monolayers and mouse intestinal organoids show that TRPV6 deficiency or inhibition attenuates ethanol- and acetaldehyde-induced Ca2+ influx, tight junction disruption, and barrier dysfunction. Moreover, Trpv6-/- mice are resistant to alcohol-induced intestinal barrier dysfunction. Photoaffinity labeling of 3-azibutanol identifies a histidine as a potential alcohol-binding site in TRPV6. The substitution of this histidine, and a nearby arginine, reduces ethanol-activated currents. Our findings reveal that TRPV6 is required for alcohol-induced gut barrier dysfunction and inflammation. Molecules that decrease TRPV6 function have the potential to attenuate alcohol-associated tissue injury.


Asunto(s)
Endotoxemia , Etanol , Histidina , Mucosa Intestinal , Canales Catiónicos TRPV , Acetaldehído/toxicidad , Animales , Células CACO-2 , Canales de Calcio/efectos de los fármacos , Canales de Calcio/metabolismo , Etanol/toxicidad , Histidina/farmacología , Humanos , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/patología , Ratones , Canales Catiónicos TRPV/efectos de los fármacos , Canales Catiónicos TRPV/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA