Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 58
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Immunity ; 49(1): 107-119.e4, 2018 07 17.
Artículo en Inglés | MEDLINE | ID: mdl-29958798

RESUMEN

Intestinal macrophages are critical for gastrointestinal (GI) homeostasis, but our understanding of their role in regulating intestinal motility is incomplete. Here, we report that CX3C chemokine receptor 1-expressing muscularis macrophages (MMs) were required to maintain normal GI motility. MMs expressed the transient receptor potential vanilloid 4 (TRPV4) channel, which senses thermal, mechanical, and chemical cues. Selective pharmacologic inhibition of TRPV4 or conditional deletion of TRPV4 from macrophages decreased intestinal motility and was sufficient to reverse the GI hypermotility that is associated with chemotherapy treatment. Mechanistically, stimulation of MMs via TRPV4 promoted the release of prostaglandin E2 and elicited colon contraction in a paracrine manner via prostaglandin E receptor signaling in intestinal smooth muscle cells without input from the enteric nervous system. Collectively, our data identify TRPV4-expressing MMs as an essential component required for maintaining normal GI motility and provide potential drug targets for GI motility disorders.


Asunto(s)
Colon/fisiología , Motilidad Gastrointestinal , Macrófagos/metabolismo , Miocitos del Músculo Liso/metabolismo , Transducción de Señal , Canales Catiónicos TRPV/metabolismo , Animales , Receptor 1 de Quimiocinas CX3C/metabolismo , Colon/fisiopatología , Ciclooxigenasa 1/deficiencia , Ciclooxigenasa 1/metabolismo , Dinoprostona/análisis , Dinoprostona/metabolismo , Femenino , Mucosa Gástrica/citología , Expresión Génica , Masculino , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/metabolismo , Ratones , Ratones Noqueados , Contracción Muscular , Receptores de Prostaglandina E/antagonistas & inhibidores , Receptores de Prostaglandina E/metabolismo , Canales Catiónicos TRPV/antagonistas & inhibidores , Canales Catiónicos TRPV/deficiencia , Canales Catiónicos TRPV/genética
2.
Am J Physiol Gastrointest Liver Physiol ; 319(1): G63-G73, 2020 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-32538139

RESUMEN

Hyaluronic acid (HA), a glycosaminoglycan in the extracellular matrix, binds to CD44 and Toll-like receptor 4 (TLR4). We previously demonstrated that both CD44 and TLR4, but predominately TLR4, mediated HA stimulation of Lgr5+ stem cell proliferation, crypt fission, and intestinal growth in postnatal mice. Here we address the questions of which cell type expresses the relevant TLR4 in driving intestinal growth and what are the downstream events from TLR4 activation. Studies were done in 14-day-old mice: wild type (WT), mice deficient in cyclooxygenase 2 (COX2), mice deficient in myeloid cell TLR4, and mice deficient in epithelial cell epidermal growth factor receptor (EGFR). Biological end points included crypt fission and Lgr5 cell proliferation. In WT mice, treatment with NS-398 (a COX2 inhibitor), clodronate (a macrophage-depleting agent), or tyrphostin (an EGFR inhibitor) resulted in 30% reductions in crypt fission and Lgr5+ stem cell proliferation compared with control mice. Mice deficient in COX2 or myeloid TLR4 or epithelial cell EGFR all had 30% reductions in crypt fission and Lgr5+ stem cell proliferation compared with WT mice. Administration of dimethyl PGE2, a stable PGE2 analog, increased crypt fission and Lgr5+ stem cell proliferation. Administration of dimethyl PGE2 reversed the effects of NS-398, clodronate, COX2 deficiency, and myeloid TLR4 deficiency but had no effect on mice treated with tyrphostin or mice deficient in epithelial cell EGFR. We conclude that, in postnatal mice, ~30% of intestinal growth as manifested by crypt fission and Lgr5+ stem cell proliferation is driven by a novel pathway: Extracellular HA binds TLR4 on pericryptal macrophages, inducing the production of PGE2 through COX2. PGE2 transactivates EGFR in Lgr5+ epithelial stem cells, resulting in Lgr5+ stem cell proliferation and crypt fission.NEW & NOTEWORTHY This study, in newborn mice, describes a novel molecular pathway regulating Lgr5+ epithelial stem cell proliferation and normal intestinal elongation, as assessed by crypt fission. In this pathway, endogenous extracellular hyaluronic acid binds to Toll-like receptor 4 on pericryptal macrophages releasing PGE2 which binds to epidermal growth factor receptor on Lgr5+ stem cells resulting in proliferation. Lgr5+ stem cell proliferation leads to crypt fission and intestinal elongation. The demonstration that normal growth requires microbial-independent Toll-like receptor activation is novel.


Asunto(s)
Dinoprostona/metabolismo , Receptores ErbB/efectos de los fármacos , Ácido Hialurónico/farmacología , Receptor Toll-Like 4/efectos de los fármacos , Animales , Proliferación Celular/efectos de los fármacos , Ciclooxigenasa 2/metabolismo , Receptores ErbB/metabolismo , Ácido Hialurónico/antagonistas & inhibidores , Intestinos/efectos de los fármacos , Ratones Noqueados , Receptor Toll-Like 4/metabolismo , Activación Transcripcional/efectos de los fármacos
3.
Gut ; 68(6): 1003-1013, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-29934438

RESUMEN

OBJECTIVE: Lactobacillus rhamnosus GG (LGG), a probiotic, given by gavage is radioprotective of the mouse intestine. LGG-induced radioprotection is toll-like receptor 2 (TLR2) and cyclooxygenase-2 (COX-2)-dependent and is associated with the migration of COX-2+mesenchymal stem cells (MSCs) from the lamina propria of the villus to the lamina propria near the crypt epithelial stem cells. Our goals were to define the mechanism of LGG radioprotection including identification of the TLR2 agonist, and the mechanism of the MSC migration and to determine the safety and efficacy of this approach in models relevant to clinical radiation therapy. DESIGN: Intestinal radioprotection was modelled in vitro with cell lines and enteroids as well as in vivo by assaying clinical outcomes and crypt survival. Fractionated abdominal and single dose radiation were used along with syngeneic CT26 colon tumour grafts to assess tumour radioprotection. RESULTS: LGG with a mutation in the processing of lipoteichoic acid (LTA), a TLR2 agonist, was not radioprotective, while LTA agonist and native LGG were. An agonist of CXCR4 blocked LGG-induced MSC migration and LGG-induced radioprotection. LGG given by gavage induced expression of CXCL12, a CXCR4 agonist, in pericryptal macrophages and depletion of macrophages by clodronate liposomes blocked LGG-induced MSC migration and radioprotection. LTA effectively protected the normal intestinal crypt, but not tumours in fractionated radiation regimens. CONCLUSIONS: LGG acts as a 'time-release capsule' releasing radioprotective LTA. LTA then primes the epithelial stem cell niche to protect epithelial stem cells by triggering a multicellular, adaptive immune signalling cascade involving macrophages and PGE2 secreting MSCs. TRIAL REGISTRATION NUMBER: NCT01790035; Pre-results.


Asunto(s)
Mucosa Intestinal/metabolismo , Lacticaseibacillus rhamnosus , Lipopolisacáridos/metabolismo , Probióticos/farmacología , Traumatismos por Radiación/prevención & control , Ácidos Teicoicos/metabolismo , Animales , Movimiento Celular/efectos de la radiación , Células Cultivadas , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Inmunohistoquímica , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/efectos de la radiación , Activación de Macrófagos/efectos de la radiación , Células Madre Mesenquimatosas/citología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Protectores contra Radiación , Valores de Referencia , Sensibilidad y Especificidad
4.
Dev Biol ; 409(2): 473-88, 2016 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-26586201

RESUMEN

Hirschsprung Disease (HSCR) is a potentially deadly birth defect characterized by the absence of the enteric nervous system (ENS) in distal bowel. Although HSCR has clear genetic causes, no HSCR-associated mutation is 100% penetrant, suggesting gene-gene and gene-environment interactions determine HSCR occurrence. To test the hypothesis that certain medicines might alter HSCR risk we treated zebrafish with medications commonly used during early human pregnancy and discovered that ibuprofen caused HSCR-like absence of enteric neurons in distal bowel. Using fetal CF-1 mouse gut slice cultures, we found that ibuprofen treated enteric neural crest-derived cells (ENCDC) had reduced migration, fewer lamellipodia and lower levels of active RAC1/CDC42. Additionally, inhibiting ROCK, a RHOA effector and known RAC1 antagonist, reversed ibuprofen effects on migrating mouse ENCDC in culture. Ibuprofen also inhibited colonization of Ret+/- mouse bowel by ENCDC in vivo and dramatically reduced bowel colonization by chick ENCDC in culture. Interestingly, ibuprofen did not affect ENCDC migration until after at least three hours of exposure. Furthermore, mice deficient in Ptgs1 (COX 1) and Ptgs2 (COX 2) had normal bowel colonization by ENCDC and normal ENCDC migration in vitro suggesting COX-independent effects. Consistent with selective and strain specific effects on ENCDC, ibuprofen did not affect migration of gut mesenchymal cells, NIH3T3, or WT C57BL/6 ENCDC, and did not affect dorsal root ganglion cell precursor migration in zebrafish. Thus, ibuprofen inhibits ENCDC migration in vitro and bowel colonization by ENCDC in vivo in zebrafish, mouse and chick, but there are cell type and strain specific responses. These data raise concern that ibuprofen may increase Hirschsprung disease risk in some genetically susceptible children.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Sistema Nervioso Entérico/citología , Ibuprofeno/farmacología , Intestinos/citología , Células-Madre Neurales/citología , Citoesqueleto de Actina/metabolismo , Animales , Caspasa 3/metabolismo , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Pollos , Ciclooxigenasa 1/deficiencia , Ciclooxigenasa 1/metabolismo , Ciclooxigenasa 2/deficiencia , Ciclooxigenasa 2/metabolismo , Activación Enzimática/efectos de los fármacos , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/metabolismo , Mesodermo/citología , Ratones , Modelos Biológicos , Células 3T3 NIH , Células-Madre Neurales/efectos de los fármacos , Neuronas/citología , Neuronas/efectos de los fármacos , Técnicas de Cultivo de Órganos , PPAR gamma/metabolismo , Seudópodos/efectos de los fármacos , Seudópodos/metabolismo , Pez Cebra , Proteína de Unión al GTP rac1/metabolismo , Quinasas Asociadas a rho/antagonistas & inhibidores , Quinasas Asociadas a rho/metabolismo
5.
Am J Physiol Gastrointest Liver Physiol ; 309(11): G874-87, 2015 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-26505972

RESUMEN

Hyaluronic acid, a glycosaminoglycan in the extracellular matrix, binds to CD44 and Toll-like receptor 4 (TLR4). We previously addressed the role of hyaluronic acid in small intestinal and colonic growth in mice. We addressed the role of exogenous hyaluronic acid by giving hyaluronic acid intraperitoneally and the role of endogenous hyaluronic acid by giving PEP-1, a peptide that blocks hyaluronic acid binding to its receptors. Exogenous hyaluronic acid increased epithelial proliferation but had no effect on intestinal length. PEP-1 resulted in a shortened small intestine and colon and diminished epithelial proliferation. In the current study, we sought to determine whether the effects of hyaluronic acid on growth were mediated by signaling through CD44 or TLR4 by giving exogenous hyaluronic acid or PEP-1 twice a week from 3-8 wk of age to wild-type, CD44(-/-), and TLR4(-/-) mice. These studies demonstrated that signaling through both CD44 and TLR4 were important in mediating the effects of hyaluronic acid on growth in the small intestine and colon. Extending our studies to early postnatal life, we assessed the effects of exogenous hyaluronic acid and PEP-1 on Lgr5(+) stem cell proliferation and crypt fission. Administration of PEP-1 to Lgr5(+) reporter mice from postnatal day 7 to day 14 decreased Lgr5(+) cell proliferation and decreased crypt fission. These studies indicate that endogenous hyaluronic acid increases Lgr5(+) stem cell proliferation, crypt fission, and intestinal lengthening and that these effects are dependent on signaling through CD44 and TLR4.


Asunto(s)
Proliferación Celular , Colon/metabolismo , Receptores de Hialuranos/metabolismo , Ácido Hialurónico/metabolismo , Mucosa Intestinal/metabolismo , Intestino Delgado/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Células Madre/efectos de los fármacos , Receptor Toll-Like 4/metabolismo , Animales , Biomarcadores/metabolismo , Proliferación Celular/efectos de los fármacos , Colon/efectos de los fármacos , Colon/crecimiento & desarrollo , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Genotipo , Receptores de Hialuranos/genética , Ácido Hialurónico/administración & dosificación , Ácido Hialurónico/antagonistas & inhibidores , Inyecciones Intraperitoneales , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/crecimiento & desarrollo , Intestino Delgado/efectos de los fármacos , Intestino Delgado/crecimiento & desarrollo , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Péptidos/farmacología , Fenotipo , Transducción de Señal/efectos de los fármacos , Células Madre/metabolismo , Receptor Toll-Like 4/deficiencia , Receptor Toll-Like 4/genética
6.
Clin Gastroenterol Hepatol ; 13(6): 1197-200, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25460565

RESUMEN

We performed a prospective study of patients with inflammatory bowel diseases to examine variations in treatment among medical centers. In a prospective cohort study of 1659 patients with Crohn's disease and 946 patients with ulcerative colitis seen at 7 high-volume referral centers, we collected data on demographics, disease characteristics, and medical and surgical treatments. We used logistic regression to determine differences in treatment among centers, controlling for potential confounders. We found significant variations among centers in the treatment of Crohn's disease with immunomodulators (odds ratio [OR], 3.34; 95% confidence interval [CI], 2.09-5.32) but not anti-tumor necrosis factor agents (OR, 1.64; 95% CI, 0.97-2.77). There was less variation in the treatment of ulcerative colitis; we found no difference in use of immunomodulators (OR, 1.83; 95% CI, 1.00-3.36) or anti-tumor necrosis factor therapy (OR, 0.81; 95% CI, 0.40-1.65). The development and implementation of evidence-based standards of care for inflammatory bowel disease may help reduce variation and improve outcomes.


Asunto(s)
Colitis Ulcerosa/tratamiento farmacológico , Colitis Ulcerosa/cirugía , Enfermedad de Crohn/tratamiento farmacológico , Enfermedad de Crohn/cirugía , Quimioterapia/métodos , Quimioterapia/normas , Investigación sobre Servicios de Salud , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Femenino , Humanos , Factores Inmunológicos/uso terapéutico , Masculino , Persona de Mediana Edad , Estudios Prospectivos , Estados Unidos , Adulto Joven
7.
J Biol Chem ; 288(22): 16085-97, 2013 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-23589310

RESUMEN

The stem cell in the isthmus of gastric units continually replenishes the epithelium. Atrophy of acid-secreting parietal cells (PCs) frequently occurs during infection with Helicobacter pylori, predisposing patients to cancer. Atrophy causes increased proliferation of stem cells, yet little is known about how this process is regulated. Here we show that CD44 labels a population of small, undifferentiated cells in the gastric unit isthmus where stem cells are known to reside. Loss of CD44 in vivo results in decreased proliferation of the gastric epithelium. When we induce PC atrophy by Helicobacter infection or tamoxifen treatment, this CD44(+) population expands from the isthmus toward the base of the unit. CD44 blockade during PC atrophy abrogates the expansion. We find that CD44 binds STAT3, and inhibition of either CD44 or STAT3 signaling causes decreased proliferation. Atrophy-induced CD44 expansion depends on pERK, which labels isthmal cells in mice and humans. Our studies delineate an in vivo signaling pathway, ERK → CD44 → STAT3, that regulates normal and atrophy-induced gastric stem/progenitor-cell proliferation. We further show that we can intervene pharmacologically at each signaling step in vivo to modulate proliferation.


Asunto(s)
Proliferación Celular , Infecciones por Helicobacter/metabolismo , Helicobacter pylori/metabolismo , Receptores de Hialuranos/metabolismo , Células Parietales Gástricas/metabolismo , Células Madre/metabolismo , Animales , Femenino , Infecciones por Helicobacter/genética , Infecciones por Helicobacter/patología , Humanos , Receptores de Hialuranos/genética , Sistema de Señalización de MAP Quinasas/genética , Masculino , Metaplasia/genética , Metaplasia/metabolismo , Metaplasia/patología , Ratones , Células Parietales Gástricas/patología , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/metabolismo , Células Madre/patología
8.
Gastroenterology ; 145(2): 416-25.e1-4, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23669411

RESUMEN

BACKGROUND & AIMS: Indoleamine 2,3 dioxygenase-1 (IDO1) catabolizes tryptophan along the kynurenine pathway. Although IDO1 is expressed in inflamed and neoplastic epithelial cells of the colon, its role in colon tumorigenesis is not well understood. We used genetic and pharmacologic approaches to manipulate IDO1 activity in mice with colitis-associated cancer and human colon cancer cell lines. METHODS: C57Bl6 wild-type (control), IDO1-/-, Rag1-/-, and Rag1/IDO1 double-knockout mice were exposed to azoxymethane and dextran sodium sulfate to induce colitis and tumorigenesis. Colitis severity was assessed by measurements of disease activity, cytokine levels, and histologic analysis. In vitro experiments were conducted using HCT 116 and HT-29 human colon cancer cells. 1-methyl tryptophan and small interfering RNA were used to inhibit IDO1. Kynurenine pathway metabolites were used to simulate IDO1 activity. RESULTS: C57Bl6 mice given pharmacologic inhibitors of IDO1 and IDO1-/- mice had lower tumor burdens and reduced proliferation in the neoplastic epithelium after administration of dextran sodium sulfate and azoxymethane than control mice. These reductions also were observed in Rag1/IDO1 double-knockout mice compared with Rag1-/- mice (which lack mature adaptive immunity). In human colon cancer cells, blockade of IDO1 activity reduced nuclear and activated ß-catenin, transcription of its target genes (cyclin D1 and Axin2), and, ultimately, proliferation. Exogenous administration of IDO1 pathway metabolites kynurenine and quinolinic acid led to activation of ß-catenin and proliferation of human colon cancer cells, and increased tumor growth in mice. CONCLUSIONS: IDO1, which catabolizes tryptophan, promotes colitis-associated tumorigenesis in mice, independent of its ability to limit T-cell-mediated immune surveillance. The epithelial cell-autonomous survival advantage provided by IDO1 to colon epithelial cells indicate its potential as a therapeutic target.


Asunto(s)
Colitis/fisiopatología , Neoplasias del Colon/etiología , Células Epiteliales/fisiología , Indolamina-Pirrol 2,3,-Dioxigenasa/fisiología , Mucosa Intestinal/fisiopatología , beta Catenina/efectos de los fármacos , Animales , Azoximetano , Carcinógenos , Proliferación Celular/efectos de los fármacos , Colitis/metabolismo , Colitis Ulcerosa , Neoplasias del Colon/inducido químicamente , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Células HCT116 , Células HT29 , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/fisiología , Humanos , Indolamina-Pirrol 2,3,-Dioxigenasa/antagonistas & inhibidores , Indolamina-Pirrol 2,3,-Dioxigenasa/genética , Mucosa Intestinal/metabolismo , Quinurenina/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ácido Quinolínico/farmacología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , beta Catenina/fisiología
9.
FASEB J ; 27(3): 1191-202, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23233532

RESUMEN

Genetic variants in the fatty acid (FA) translocase FAT/CD36 associate with abnormal postprandial lipids and influence risk for the metabolic syndrome. CD36 is abundant on apical enterocyte membranes in the proximal small intestine, where it facilitates FA uptake and FA-initiated signaling. We explored whether CD36 signaling influences FA-mediated secretion of cholecystokinin (CCK) and secretin, peptides released by enteroendocrine cells (EECs) in the duodenum/jejunum, which regulate events important for fat digestion and homeostasis. CD36 was immunodetected on apical membranes of secretin- and CCK-positive EECs and colocalized with cytosolic granules. Intragastric lipid administration to CD36 mice released less secretin (-60%) and CCK (-50%) compared with wild-type mice. Likewise, diminished secretin and CCK responses to FA were observed with CD36 intestinal segments in vitro, arguing against influence of alterations in fat absorption. Signaling mechanisms underlying peptide release were examined in STC-1 cells stably expressing human CD36 or a signaling-impaired mutant (CD36K/A). FA stimulation of cells expressing CD36 (vs. vector or CD36K/A) released more secretin (3.5- to 4-fold) and CCK (2- to 3-fold), generated more cAMP (2- to 2.5-fold), and enhanced protein kinase A activation. Protein kinase A inhibition (H-89) blunted secretin (80%) but not CCK release, which was reduced (50%) by blocking of calmodulin kinase II (KN-62). Coculture of STC-1 cells with Caco-2 cells stably expressing CD36 did not alter secretin or CCK release, consistent with a minimal effect of adjacent enterocytes. In summary, CD36 is a major mediator of FA-induced release of CCK and secretin. These peptides contribute to the role of CD36 in fat absorption and to its pleiotropic metabolic effects.


Asunto(s)
Antígenos CD36/metabolismo , Colecistoquinina/metabolismo , Duodeno/metabolismo , Células Enteroendocrinas/metabolismo , Ácidos Grasos/metabolismo , Yeyuno/metabolismo , Secretina/metabolismo , Animales , Antígenos CD36/genética , Células CACO-2 , Colecistoquinina/genética , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Duodeno/citología , Células Enteroendocrinas/citología , Activación Enzimática , Ácidos Grasos/genética , Humanos , Yeyuno/citología , Ratones , Ratones Noqueados , Secretina/genética , Transducción de Señal/fisiología
10.
Curr Opin Gastroenterol ; 30(4): 347-51, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24837228

RESUMEN

PURPOSE OF REVIEW: This review summarizes recent developments in the role of soluble mediators of inflammation, particularly arachidonic acid metabolites, in inflammatory bowel disease (IBD). RECENT FINDINGS: The role of prostaglandin E2 in immune regulation has been better defined. Prostaglandin E2 promotes not only immune tolerance and epithelial homeostasis but also the proinflammatory Th17 pathway. Prostaglandin D2 has been established as promoting the resolution of inflammation in the gastrointestinal mucosa. The 12-lipoxygenase product hepoxilin A3 mediates the migration of neutrophils from the mucosa into the lumen. SUMMARY: Recent studies of soluble mediators, especially arachidonic acid metabolites, have defined their proinflammatory and anti-inflammatory roles in IBD.


Asunto(s)
Antiinflamatorios/uso terapéutico , Ácido Araquidónico/inmunología , Inflamación/inmunología , Enfermedades Inflamatorias del Intestino/inmunología , Mucosa Intestinal/inmunología , Células Th17/metabolismo , Ácido 8,11,14-Eicosatrienoico/análogos & derivados , Ácido 8,11,14-Eicosatrienoico/metabolismo , Antiinflamatorios/efectos adversos , Antiinflamatorios no Esteroideos/efectos adversos , Movimiento Celular , Dinoprostona/inmunología , Humanos , Inflamación/fisiopatología , Inflamación/terapia , Enfermedades Inflamatorias del Intestino/fisiopatología , Enfermedades Inflamatorias del Intestino/terapia , Neutrófilos , Prostaglandina D2/inmunología , Transducción de Señal , Células Th17/inmunología
11.
Gut ; 61(6): 829-38, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22027478

RESUMEN

BACKGROUND: The small intestinal epithelium is highly sensitive to radiation and is a major site of injury during radiation therapy and environmental overexposure. OBJECTIVE: To examine probiotic bacteria as potential radioprotective agents in the intestine. METHODS: 8-week-old C57BL/6 wild-type or knockout mice were administered probiotic by gavage for 3 days before 12 Gy whole body radiation. The intestine was evaluated for cell-positional apoptosis (6 h) and crypt survival (84 h). RESULTS: Gavage of 5×107 Lactobacillus rhamnosus GG (LGG) improved crypt survival about twofold (p<0.01); the effect was observed when administered before, but not after, radiation. Conditioned medium (CM) from LGG improved crypt survival (1.95-fold, p<0.01), and both LGG and LGG-CM reduced epithelial apoptosis particularly at the crypt base (33% to 18%, p<0.01). LGG was detected in the distal ileal contents after the gavage cycle, but did not lead to a detectable shift in bacterial family composition. The reduction in epithelial apoptosis and improved crypt survival offered by LGG was lost in MyD88⁻/⁻, TLR-2⁻/⁻ and cyclo-oxygenase-2⁻/⁻ (COX-2) mice but not TLR-4⁻/⁻ mice. LGG administration did not lead to increased jejunal COX-2 mRNA or prostaglandin E2 levels or a change in number of COX-2-expressing cells. However, a location shift was observed in constitutively COX-2-expressing cells of the lamina propria from the villi to a position near the crypt base (villi to crypt ratio 80:20 for control and 62:38 for LGG; p<0.001). Co-staining revealed these COX-2-expressing small intestinal lamina propria cells to be mesenchymal stem cells. CONCLUSIONS: LGG or its CM reduce radiation-induced epithelial injury and improve crypt survival. A TLR-2/MyD88 signalling mechanism leading to repositioning of constitutive COX-2-expressing mesenchymal stem cells to the crypt base is invoked.


Asunto(s)
Ciclooxigenasa 2/fisiología , Mucosa Intestinal/efectos de la radiación , Lacticaseibacillus rhamnosus/metabolismo , Probióticos/uso terapéutico , Traumatismos Experimentales por Radiación/prevención & control , Receptor Toll-Like 2/fisiología , Irradiación Corporal Total/efectos adversos , Animales , Apoptosis/efectos de la radiación , Femenino , Mucosa Intestinal/microbiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/fisiología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
12.
Am J Physiol Gastrointest Liver Physiol ; 302(3): G309-16, 2012 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-22038822

RESUMEN

The intestinal epithelium is sensitive to radiation injury. Damage to the intestinal epithelium is dose limiting in radiation therapy of abdominal cancers. There is a need for agents that can be given before radiation therapy to protect the intestinal epithelium. C57BL6 mice were subjected to 12 Gy of total body radiation. Some mice received intraperitoneal hyaluronic acid (HA) before radiation. Mice were killed 6 h after radiation to assess radiation-induced apoptosis in the intestine; other mice were killed at 84 h to assess crypt survival. Total body radiation (12 Gy) resulted in increased expression of HA synthases and HA in the intestine and increased plasma HA (5-fold). Intraperitoneal injection of HA (30 mg/kg) before radiation resulted in a 1.8-fold increase in intestinal crypt survival and a decrease in radiation-induced apoptosis. The radioprotective effects of HA were not seen in Toll-like receptor 4 (TLR4)- or cyclooxygenase-2 (COX-2)-deficient mice. Intraperitoneal injection of HA induced a 1.5-fold increase in intestinal COX-2 expression, a 1.5-fold increase in intestinal PGE2, and the migration of COX-2-expressing mesenchymal stem cells from the lamina propria in the villi to the lamina propria near the crypt. We conclude that 1) radiation induces increased HA expression through inducing HA synthases, 2) intraperitoneal HA given before radiation reduces radiation-induced apoptosis and increases crypt survival, and 3) these radioprotective effects are mediated through TLR4, COX-2, and the migration of COX-2-expressing mesenchymal stem cells.


Asunto(s)
Ciclooxigenasa 2/metabolismo , Ácido Hialurónico/farmacología , Intestinos/efectos de los fármacos , Intestinos/efectos de la radiación , Protectores contra Radiación/farmacología , Receptor Toll-Like 4/metabolismo , Animales , Antígenos CD/metabolismo , Apoptosis/efectos de los fármacos , Apoptosis/efectos de la radiación , Movimiento Celular/efectos de los fármacos , Ciclooxigenasa 2/genética , Dinoprostona/metabolismo , Expresión Génica/genética , Expresión Génica/efectos de la radiación , Glucuronosiltransferasa/genética , Receptores de Hialuranos/genética , Hialuronano Sintasas , Ácido Hialurónico/antagonistas & inhibidores , Ácido Hialurónico/sangre , Ácido Hialurónico/metabolismo , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Mucosa Intestinal/efectos de la radiación , Intestinos/patología , Yeyuno/efectos de los fármacos , Yeyuno/metabolismo , Yeyuno/patología , Yeyuno/efectos de la radiación , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Protectores contra Radiación/metabolismo , Receptor Toll-Like 4/agonistas , Receptor Toll-Like 4/genética
13.
Am J Physiol Gastrointest Liver Physiol ; 303(3): G377-88, 2012 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-22556141

RESUMEN

Hyaluronic acid (HA), a component of the extracellular matrix, affects gastrointestinal epithelial proliferation in injury models, but its role in normal growth is unknown. We sought to determine the effects of exogenous HA on intestinal and colonic growth by intraperitoneal injection of HA twice a week into C57BL/6 mice from 3 to 8 wk of age. Similarly, to determine the effects of endogenous HA on intestinal and colonic growth, we administered PEP-1, a peptide that blocks the binding of HA to its receptors, on the same schedule. In mice treated with exogenous HA, villus height and crypt depth in the intestine, crypt depth in the colon, and epithelial proliferation in the intestine and colon were increased. In mice treated with PEP-1, intestinal and colonic length were markedly decreased and crypt depth and villus height in the intestine, crypt depth in the colon, and epithelial proliferation in the intestine and colon were decreased. Administration of HA was associated with increased levels of EGF (intestine) and IGF-I (colon), whereas administration of PEP-1 was associated with decreased levels of IGF-I (intestine) and epiregulin (colon). Exogenous HA increases intestinal and colonic epithelial proliferation, resulting in hyperplasia. Blocking the binding of endogenous HA to its receptors results in decreased intestinal and colonic length and a mucosal picture of hypoplasia, suggesting that endogenous HA contributes to the regulation of normal intestinal and colonic growth.


Asunto(s)
Colon/crecimiento & desarrollo , Ácido Hialurónico/farmacología , Intestinos/crecimiento & desarrollo , Animales , Proliferación Celular/efectos de los fármacos , Colon/efectos de los fármacos , Factor de Crecimiento Epidérmico/metabolismo , Epirregulina , Células Epiteliales/citología , Glucuronosiltransferasa/biosíntesis , Receptores de Hialuranos/efectos de los fármacos , Hialuronano Sintasas , Hialuronoglucosaminidasa/biosíntesis , Factor I del Crecimiento Similar a la Insulina/metabolismo , Intestinos/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Péptidos/farmacología , ARN Mensajero/metabolismo
14.
J Immunol ; 184(7): 3907-16, 2010 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-20181893

RESUMEN

The chronic inflammatory bowel diseases are characterized by aberrant innate and adaptive immune responses to commensal luminal bacteria. In both human inflammatory bowel disease and in experimental models of colitis, there is an increased expression of the enzyme IDO. IDO expression has the capacity to exert antimicrobial effects and dampen adaptive immune responses. In the murine trinitrobenzene sulfonic acid model of colitis, inhibition of this enzyme leads to worsened disease severity, suggesting that IDO acts as a natural break in limiting colitis. In this investigation, we show that induction of IDO-1 by a TLR-9 agonist, immunostimulatory (ISS) DNA, critically contributes to its colitis limiting capacities. ISS DNA induces intestinal expression of IDO-1 but not the recently described paralog enzyme IDO-2. This induction occurred in both epithelial cells and in subsets of CD11c(+) and CD11b(+) cells of the lamina propria, which also increase after ISS-oligodeoxynucleotide. Signaling required for intestinal IDO-1 induction involves IFN-dependent pathways, as IDO-1 was not induced in STAT-1 knockout mice. Using both the trinitrobenzene sulfonic acid and dextran sodium sulfate models of colitis, we show the importance of IDO-1s induction in limiting colitis severity. The clinical parameters and histological correlates of colitis in these models were improved by administration of the TLR-9 agonist; however, when the function of IDO is inhibited, the colitis limiting effects of ISS-oligodeoxynucleotide were abrogated. These findings support the possibility that targeted induction of IDO-1 is an approach deserving further investigation as a therapeutic strategy for diseases of intestinal inflammation.


Asunto(s)
Adyuvantes Inmunológicos/farmacología , Colitis/enzimología , Colitis/inmunología , Indolamina-Pirrol 2,3,-Dioxigenasa/metabolismo , Oligodesoxirribonucleótidos/farmacología , Transducción de Señal/inmunología , Animales , Western Blotting , Cromatografía Líquida de Alta Presión , Colitis/patología , ADN/farmacología , Sulfato de Dextran/toxicidad , Femenino , Técnica del Anticuerpo Fluorescente , Mucosa Intestinal/enzimología , Isoenzimas/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Membrana Mucosa/enzimología , Oligodesoxirribonucleótidos/inmunología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Receptor Toll-Like 9/agonistas , Ácido Trinitrobencenosulfónico/toxicidad
15.
J Biol Chem ; 285(7): 5026-39, 2010 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-20018844

RESUMEN

We previously found that a population of colonic stromal cells that constitutively express high levels of prostaglandin-endoperoxide synthase 2 (Ptgs2, also known as Cox-2) altered their location in the lamina propria in response to injury in a Myd88-dependent manner (Brown, S. L., Riehl, T. E., Walker, M. R., Geske, M. J., Doherty, J. M., Stenson, W. F., and Stappenbeck, T. S. (2007) J. Clin. Invest. 117, 258-269). At the time of this study, the identity of these cells and the mechanism by which they expressed high levels of Ptgs2 were unknown. Here we found that these colonic stromal cells were mesenchymal stem cells (MSCs). These colonic MSCs expressed high Ptgs2 levels not through interaction with bacterial products but instead as a consequence of mRNA stabilization downstream of Fgf9 (fibroblast growth factor 9), a growth factor that is constitutively expressed by the intestinal epithelium. This stabilization was mediated partially through a mechanism involving endogenous CUG-binding protein 2 (CUGbp2). These studies suggest that Fgf9 is an important factor in the regulation of Ptgs2 in colonic MSCs and may be a factor involved in its constitutive expression in vivo.


Asunto(s)
Colon/citología , Ciclooxigenasa 2/metabolismo , Células Madre Mesenquimatosas/metabolismo , Animales , Proteínas CELF , Línea Celular , Células Cultivadas , Ciclooxigenasa 2/genética , Factor 9 de Crecimiento de Fibroblastos/farmacología , Flavonoides/farmacología , Citometría de Flujo , Humanos , Immunoblotting , Inmunohistoquímica , Lipopolisacáridos/farmacología , Células Madre Mesenquimatosas/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Modelos Biológicos , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Células del Estroma/metabolismo , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/metabolismo
16.
J Clin Invest ; 117(1): 258-69, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17200722

RESUMEN

We identified cellular and molecular mechanisms within the stem cell niche that control the activity of colonic epithelial progenitors (ColEPs) during injury. Here, we show that while WT mice maintained ColEP proliferation in the rectum following injury with dextran sodium sulfate, similarly treated Myd88(-/-) (TLR signaling-deficient) and prostaglandin-endoperoxide synthase 2(-/-) (Ptgs2(-/-)) mice exhibited a profound inhibition of epithelial proliferation and cellular organization within rectal crypts. Exogenous addition of 16,16-dimethyl PGE(2) (dmPGE(2)) rescued the effects of this injury in both knockout mouse strains, indicating that Myd88 signaling is upstream of Ptgs2 and PGE(2). In WT and Myd88(-/-) mice, Ptgs2 was expressed in scattered mesenchymal cells. Surprisingly, Ptgs2 expression was not regulated by injury. Rather, in WT mice, the combination of injury and Myd88 signaling led to the repositioning of a subset of the Ptgs2-expressing stromal cells from the mesenchyme surrounding the middle and upper crypts to an area surrounding the crypt base adjacent to ColEPs. These findings demonstrate that Myd88 and prostaglandin signaling pathways interact to preserve epithelial proliferation during injury using what we believe to be a previously undescribed mechanism requiring proper cellular mobilization within the crypt niche.


Asunto(s)
Ciclooxigenasa 2/genética , Mucosa Intestinal/patología , Factor 88 de Diferenciación Mieloide/fisiología , Células del Estroma/fisiología , Animales , División Celular , Ciclooxigenasa 2/deficiencia , Mucosa Intestinal/fisiopatología , Ratones , Ratones Noqueados , Modelos Genéticos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células del Estroma/enzimología , Transcripción Genética , Heridas y Lesiones/fisiopatología
17.
Gastroenterology ; 137(6): 2041-51, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19732774

RESUMEN

BACKGROUND & AIMS: The protective component of the host response to dextran sodium sulfate (DSS)-induced colitis in the mouse is mediated through the activation of Toll-like receptor (TLR) 4, the induction of cyclooxygenase (COX)-2, and prostaglandin E(2) production. TLR4 ligands include bacterial lipopolysaccharide and hyaluronic acid, a component of the extracellular matrix. Our hypothesis is that hyaluronic acid, through TLRs, plays a protective role in the host response to DSS-induced colitis. METHODS: DSS (2.5%) was administered for 7 days in wild-type and MyD88(-/-) mice. The mice also received intraperitoneal hyaluronic acid. The expression of hyaluronic acid, COX-2, and macrophage inflammatory protein (MIP)-2 was evaluated by immunohistochemistry. RESULTS: DSS induced a marked increase in hyaluronic acid in the lamina propria of wild-type but not MyD88(-/-) mice. Treatment with DSS also induced the MyD88-dependent expression of hyaluronic acid synthases 2 and 3, enzymes involved in hyaluronic acid synthesis, in lamina propria macrophages. Exogenous hyaluronic acid induced the expression of tumor necrosis factor alpha, MIP-2, and COX-2 in the colon in a MyD88-dependent manner. In wild-type but not MyD88(-/-), TLR4(-/-), COX-2(-/-) mice, hyaluronic acid was protective against DSS-induced colitis. In wild-type mice, hyaluronic acid was therapeutic in established DSS-induced colitis. CONCLUSIONS: Endogenous hyaluronic acid expression is markedly increased in DSS-induced colitis and preserves the epithelium through TLR activation and COX-2 expression. Furthermore, exogenous hyaluronic acid, through the activation of TLRs and the production of prostaglandin E(2) through COX-2, has protective effects in DSS-induced colitis.


Asunto(s)
Antiinflamatorios/metabolismo , Proliferación Celular , Colitis/patología , Colon/patología , Ácido Hialurónico/metabolismo , Mucosa Intestinal/patología , Receptor Toll-Like 4/metabolismo , Animales , Antiinflamatorios/administración & dosificación , Quimiocina CXCL2/metabolismo , Colitis/inducido químicamente , Colitis/inmunología , Colitis/metabolismo , Colitis/prevención & control , Colon/inmunología , Colon/metabolismo , Ciclooxigenasa 2/genética , Ciclooxigenasa 2/metabolismo , Sulfato de Dextran , Modelos Animales de Enfermedad , Femenino , Glucuronosiltransferasa/metabolismo , Receptores de Hialuranos/genética , Receptores de Hialuranos/metabolismo , Hialuronano Sintasas , Ácido Hialurónico/administración & dosificación , Inyecciones Intraperitoneales , Mucosa Intestinal/inmunología , Mucosa Intestinal/metabolismo , Ligandos , Macrófagos Peritoneales/inmunología , Macrófagos Peritoneales/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/genética , Factor 88 de Diferenciación Mieloide/metabolismo , Regeneración , Factores de Tiempo , Receptor Toll-Like 4/deficiencia , Receptor Toll-Like 4/genética , Factor de Necrosis Tumoral alfa/metabolismo , Regulación hacia Arriba
18.
J Immunol ; 181(4): 2544-55, 2008 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-18684945

RESUMEN

Tissue homing of activated T cells is typically mediated through their specific integrin and chemokine receptor repertoire. Activation of human primary CD4(+) T cells in the presence of CD46 cross-linking induces the development of a distinct immunomodulatory T cell population characterized by high IL-10/granzyme B production. How these regulatory T cells (Tregs) migrate/home to specific tissue sites is not understood. In this study, we determined the adhesion protein and chemokine receptor expression pattern on human CD3/CD46-activated peripheral blood CD4(+) T cells. CD3/CD46-activated, but not CD3/CD28-activated, T cells up-regulate the integrin alpha(4)beta(7). The interaction of alpha(4)beta(7) with its ligand mucosal addressin cell adhesion molecule 1 (MAdCAM-1) mediates homing or retention of T cells to the intestine. CD3/CD46-activated Tregs adhere to/roll on MAdCAM-1-expressing HeLa cells, similar to T cells isolated from the human lamina propria (LP). This interaction is inhibited by silencing MAdCAM-1 expression in HeLa cells or by the addition of blocking Abs to beta(7). CD46 activation of T cells also induced the expression of the surface-bound cytokine LIGHT and the chemokine receptor CCR9, both marker constitutively expressed by gut LP-resident T cells. In addition, we found that approximately 10% of the CD4(+) T lymphocytes isolated from the LP of patients undergoing bariatric surgery contain T cells that spontaneously secrete a cytokine pattern consistent with that from CD46-activated T cells. These data suggest that CD46-induced Tregs might play a role in intestinal immune homeostasis where they could dampen unwanted effector T cell responses through local IL-10/granzyme B production.


Asunto(s)
Mucosa Intestinal/inmunología , Mucosa Intestinal/metabolismo , Activación de Linfocitos/inmunología , Proteína Cofactora de Membrana/fisiología , Receptores de Quimiocina/biosíntesis , Receptores Mensajeros de Linfocitos/biosíntesis , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Moléculas de Adhesión Celular , Células Cultivadas , Quimiotaxis de Leucocito/inmunología , Granzimas/biosíntesis , Granzimas/fisiología , Células HeLa , Humanos , Inmunoglobulinas/biosíntesis , Inmunoglobulinas/fisiología , Integrinas/biosíntesis , Integrinas/fisiología , Interleucina-10/biosíntesis , Interleucina-10/metabolismo , Mucosa Intestinal/citología , Mucoproteínas/biosíntesis , Mucoproteínas/fisiología , Receptores de Quimiocina/genética , Receptores Mensajeros de Linfocitos/fisiología , Regulación hacia Arriba/inmunología
20.
Front Immunol ; 11: 617510, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33552081

RESUMEN

TLRs, key components of the innate immune system, recognize microbial molecules. However, TLRs also recognize some nonmicrobial molecules. In particular, TLR2 and TLR4 recognize hyaluronic acid, a glycosaminoglycan in the extracellular matrix. In neonatal mice endogenous hyaluronic acid binding to TLR4 drives normal intestinal growth. Hyaluronic acid binding to TLR4 in pericryptal macrophages results in cyclooxygenase2- dependent PGE2 production, which transactivates EGFR in LGR5+ crypt epithelial stem cells leading to increased proliferation. The expanded population of LGR5+ stem cells leads to crypt fission and lengthening of the intestine and colon. Blocking this pathway at any point (TLR4 activation, PGE2 production, EGFR transactivation) results in diminished intestinal and colonic growth. A similar pathway leads to epithelial proliferation in wound repair. The repair phase of dextran sodium sulfate colitis is marked by increased epithelial proliferation. In this model, TLR2 and TLR4 in pericryptal macrophages are activated by microbial products or by host hyaluronic acid, resulting in production of CXCL12, a chemokine. CXCL12 induces the migration of cyclooxygenase2-expressing mesenchymal stem cells from the lamina propria of the upper colonic crypts to a site adjacent to LGR5+ epithelial stem cells. PGE2 released by these mesenchymal stem cells transactivates EGFR in LGR5+ epithelial stem cells leading to increased proliferation. Several TLR2 and TLR4 agonists, including hyaluronic acid, are radioprotective in the intestine through the inhibition of radiation-induced apoptosis in LGR5+ epithelial stem cells. Administration of exogenous TLR2 or TLR4 agonists activates TLR2/TLR4 on pericryptal macrophages inducing CXCL12 production with migration of cyclooxygenase2-expressing mesenchymal stem cells from the lamina propria of the villi to a site adjacent to LGR5+ epithelial stem cells. PGE2 produced by these mesenchymal stem cells, blocks radiation-induced apoptosis in LGR5+ epithelial stem cells by an EGFR mediated pathway.


Asunto(s)
Intestinos/crecimiento & desarrollo , Efectos de la Radiación , Cicatrización de Heridas/fisiología , Animales , Proliferación Celular/fisiología , Humanos , Mucosa Intestinal/metabolismo , Receptores Toll-Like/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA