Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 82
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Clin Gastroenterol Hepatol ; 22(1): 22-33.e6, 2024 01.
Artículo en Inglés | MEDLINE | ID: mdl-37716619

RESUMEN

BACKGROUND & AIMS: Withdrawal of immunomodulators (IMMs) or tumor necrosis factor (TNF) antagonists in patients with inflammatory bowel diseases (IBDs) in remission on combination therapy is attractive. We evaluated the efficacy and safety of (1) IMM, or (2) TNF antagonist withdrawal in patients with IBD in sustained remission on combination therapy. METHODS: Through a systematic review till March 31, 2023, we identified randomized controlled trials (RCTs) that compared the efficacy and safety of IMM or TNF antagonist withdrawal vs continued combination therapy, in patients with IBD in sustained corticosteroid-free clinical remission for >6 months on combination therapy. Primary outcome was risk of relapse and serious adverse events at 12 months. We conducted meta-analysis to calculate relative risk (RR) and 95% confidence interval (CI) and used Grading of Recommendations Assessment, Development and Evaluation (GRADE) to appraise certainty of evidence. RESULTS: We identified 8 RCTs with 733 patients (77% with Crohn's disease, 91% on infliximab-based combination therapy). On meta-analysis of 5 RCTs, there was no difference in the risk of relapse between patients with IMM withdrawal (continued TNF antagonist monotherapy) vs continued combination therapy (16.8% vs 14.9%; RR, 1.15; 95% CI, 0.75-1.76) without heterogeneity (low certainty of evidence). TNF antagonist withdrawal (continued IMM monotherapy) was associated with 2.4-times higher risk of relapse compared with continuing combination therapy (31.5% vs 11.2%; RR, 2.35; 95% CI, 1.38-4.01), with minimal heterogeneity (low certainty of evidence). There was no difference in the risk of serious adverse events with IMM or TNF antagonist withdrawal vs continued combination therapy. CONCLUSIONS: In patients with IBD in sustained corticosteroid-free clinical remission for >6 months on combination therapy, de-escalation with TNF antagonist withdrawal, but not IMM withdrawal, was associated with an increased risk of relapse.


Asunto(s)
Enfermedad de Crohn , Enfermedades Inflamatorias del Intestino , Humanos , Inmunosupresores/uso terapéutico , Inhibidores del Factor de Necrosis Tumoral/efectos adversos , Factores Inmunológicos/efectos adversos , Enfermedad de Crohn/tratamiento farmacológico , Recurrencia , Inducción de Remisión , Enfermedades Inflamatorias del Intestino/tratamiento farmacológico
2.
Gastroenterology ; 162(7): 1975-1989, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35227778

RESUMEN

BACKGROUND & AIMS: Epithelial wound healing is compromised and represents an unleveraged therapeutic target in inflammatory bowel disease (IBD). Intestinal epithelial cells exhibit plasticity that facilitates dedifferentiation and repair during the response to injury. However, it is not known whether epithelial cells of a neighboring organ can be activated to mediate re-epithelialization in acute colitis. Histological findings of a permanent squamous tissue structure in the distal colon in human IBD could suggest diverse cellular origins of repair-associated epithelium. Here, we tested whether skin-like cells from the anus mediate colonic re-epithelialization in murine colitis. METHODS: We studied dextran sulfate sodium-induced colitis and interleukin 10-deficient colitis in transgenic mice. We performed lineage tracing, 3-dimensional (3D) imaging, single-cell transcriptomics, and biophysical modeling to map squamous cell fates and to identify squamous cell types involved in colonic repair. RESULTS: In acute and chronic colitis, we found a large squamous epithelium, called squamous neo-epithelium of the colon (SNEC), near the anorectal junction. Neighboring squamous cells of the anus rapidly migrate into the ulcerated colon and establish this permanent epithelium of crypt-like morphology. These squamous cells derive from a small unique transition zone, distal to the border of colonic and anal epithelium, that resists colitic injury. The cells of this zone have a pre-loaded program of colonic differentiation and further upregulate key aspects of colonic epithelium during repair. CONCLUSION: Transitional anal cells represent unique reserve cells capable of rebuilding epithelial structures in the colon after colitis. Further study of these cells could reveal novel approaches to direct mucosal healing in inflammation and disease.


Asunto(s)
Carcinoma de Células Escamosas , Colitis , Enfermedades Inflamatorias del Intestino , Canal Anal/patología , Animales , Carcinoma de Células Escamosas/patología , Colitis/metabolismo , Colon/patología , Sulfato de Dextran/toxicidad , Modelos Animales de Enfermedad , Células Epiteliales/patología , Humanos , Enfermedades Inflamatorias del Intestino/patología , Mucosa Intestinal/patología , Ratones , Ratones Endogámicos C57BL , Repitelización
3.
Am J Physiol Gastrointest Liver Physiol ; 321(3): G308-G324, 2021 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-34260310

RESUMEN

Intestinal mucosal healing is the primary therapeutic goal of medical treatments for inflammatory bowel disease (IBD). Epithelial stem cells are key players in the healing process. Lgr5+ stem cells maintain cellular turnover during homeostasis in the colonic crypt. However, they are lost and dispensable for repair in a wide variety of injury models, including dextran sulfate sodium (DSS) colitis, radiation, helminth infection, and T-cell activation. The direct loss of Lgr5+ cells activates a plasticity response in the epithelium in which other cell types can serve as stem cells. Whether this paradigm applies to mouse models of IBD remains unknown. In contrast to previously tested models, IBD models involve an inflammatory response rooted in the loss of immunologic tolerance to intestinal luminal contents including the microbiome. Here, we show the persistence of Lgr5+ cells in oxazolone, 2,4,6-trinitrobenzene sulfonic acid (TNBS), and Il10-/-, and Il10-/- Tnfr1-/- IBD models. This contrasts with results obtained from DSS-induced injury. Through high-throughput expression profiling, we find that these colitis models were associated with distinct patterns of cytokine expression. Direct exposure of colonic epithelial organoids to DSS, oxazolone, or TNBS resulted in increased apoptosis and loss of Lgr5+ cells. Targeted ablation of Lgr5+ cells resulted in severe exacerbation of chronic, antibody-induced IL-10-deficient colitis, but had only modest effects in TNBS-induced colitis. These results show that distinct mouse models of IBD-like colitis induce different patterns of Lgr5+ stem cell retention and function.NEW & NOTEWORTHY Acute intestinal injury and epithelial repair are associated with the loss of fast-cycling Lgr5+ stem cells and plasticity in the activation of formerly quiescent cell populations. In contrast, here we show in murine inflammatory bowel disease the persistence of the Lgr5+ stem cell population and its essential role in restricting the severity of chronic colitis. This demonstrates a diversity of stem cell responses to colitis.


Asunto(s)
Colitis/metabolismo , Enfermedades Inflamatorias del Intestino/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Células Madre/citología , Animales , Proliferación Celular/fisiología , Modelos Animales de Enfermedad , Epitelio/metabolismo , Homeostasis/fisiología , Mucosa Intestinal/metabolismo , Ratones , Regeneración/fisiología , Células Madre/metabolismo
4.
Am J Physiol Gastrointest Liver Physiol ; 320(6): G990-G1001, 2021 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-33826403

RESUMEN

Proinflammatory macrophages are essential drivers of colitis and express the growth factor receptor ErbB4. This study tested the role of ErbB4 and its specific ligand, NRG4, in regulating macrophage function. We show that endogenous NRG4-ErbB4 signaling limits macrophage production of proinflammatory cytokines in vitro and limits colitis severity in vivo and thus is a potential target for therapeutic intervention.


Asunto(s)
Inflamación/metabolismo , Macrófagos/metabolismo , Neurregulinas/metabolismo , Receptor ErbB-4/metabolismo , Transducción de Señal/fisiología , Animales , Colitis/metabolismo , Colon/metabolismo , Citocinas/metabolismo , Inflamación/genética , Interleucina-10/genética , Interleucina-10/metabolismo , Activación de Macrófagos/fisiología , Ratones , Ratones Noqueados
5.
Am J Physiol Gastrointest Liver Physiol ; 319(1): G1-G10, 2020 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-32421359

RESUMEN

The development of modern methods to induce optical transparency ("clearing") in biological tissues has enabled the three-dimensional (3D) reconstruction of intact organs at cellular resolution. New capabilities in visualization of rare cellular events, long-range interactions, and irregular structures will facilitate novel studies in the alimentary tract and gastrointestinal systems. The tubular geometry of the alimentary tract facilitates large-scale cellular reconstruction of cleared tissue without specialized microscopy setups. However, with the rapid pace of development of clearing agents and current relative paucity of research groups in the gastrointestinal field using these techniques, it can be daunting to incorporate tissue clearing into experimental workflows. Here, we give some advice and describe our own experience bringing tissue clearing and whole mount reconstruction into our laboratory's investigations. We present a brief overview of the chemical concepts that underpin tissue clearing, what sorts of questions whole mount imaging can answer, how to choose a clearing agent, an example of how to clear and image alimentary tissue, and what to do after obtaining the image. This short review will encourage other gastrointestinal researchers to consider how utilizing tissue clearing and creating 3D "maps" of tissue might deepen the impact of their studies.


Asunto(s)
Tracto Gastrointestinal/patología , Técnicas de Cultivo de Tejidos , Animales , Microambiente Celular/fisiología , Humanos , Imagenología Tridimensional/métodos , Investigación
6.
Infect Immun ; 87(7)2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31010817

RESUMEN

The symbiotic relationship between the gut microbiome and the host provides a nutrient-rich environment for gut microbes and has beneficial effects on host health. Although the composition of the gut microbiome is known to be influenced by both host genetics and environmental factors, host effects on the activities and functions of the gut microbial communities remain poorly understood. Intestinal epithelial cells exert front-line responses to gut microbes and contribute to maintaining a healthy intestinal homeostasis. Here, seeking to elucidate whether intestinal epithelial cells modulate Lactobacillus rhamnosus GG (LGG) functions, we examined the production of p40, an LGG-derived secretory protein that protects intestinal epithelial cells against inflammation. We found that growth medium conditioned with colonic epithelial cell-derived components promotes p40 protein synthesis and secretion by LGG and enhances LGG-stimulated protective responses in intestinal epithelial cells. Furthermore, when LGG was cultured with the colonic luminal contents from healthy mice, p40 production was upregulated but was attenuated with luminal contents from mice with intestinal inflammation. Importantly, the colonic epithelial cell-derived components potentiated LGG-produced p40 levels in a mouse model of colitis and enhanced LGG-mediated amelioration of intestinal inflammation in this model. Notably, we found that colonic epithelial cell-secreted extracellular vesicles participate in communicating with LGG and that heat shock protein 90 (HSP90) in these vesicles might mediate the promotion of p40 production. These results reveal a previously unrecognized mechanism by which the anti-inflammatory effect of LGG is reinforced by intestinal epithelial cells and thereby maintains intestinal health.


Asunto(s)
Proteínas Bacterianas/metabolismo , Células Epiteliales/microbiología , Mucosa Intestinal/microbiología , Lacticaseibacillus rhamnosus/metabolismo , Vesículas Secretoras/microbiología , Animales , Proteínas Bacterianas/genética , Células Epiteliales/metabolismo , Proteínas HSP90 de Choque Térmico/metabolismo , Mucosa Intestinal/metabolismo , Lacticaseibacillus rhamnosus/genética , Ratones , Ratones Endogámicos C57BL , Vesículas Secretoras/metabolismo
7.
J Cell Sci ; 130(1): 90-96, 2017 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-27026527

RESUMEN

Cell shedding from the intestinal villus is a key element of tissue turnover that is essential to maintain health and homeostasis. However, the signals regulating this process are not well understood. We asked whether shedding is controlled by epidermal growth factor receptor (EGFR), an important driver of intestinal growth and differentiation. In 3D ileal enteroid culture and cell culture models (MDCK, IEC-6 and IPEC-J2 cells), extrusion events were suppressed by EGF, as determined by direct counting of released cells or rhodamine-phalloidin labeling of condensed actin rings. Blockade of the MEK-ERK pathway, but not other downstream pathways such as phosphoinositide 3-kinase (PI3K) or protein kinase C (PKC), reversed EGF inhibition of shedding. These effects were not due to a change in cell viability. Furthermore, EGF-driven MAPK signaling inhibited both caspase-independent and -dependent shedding pathways. Similar results were found in vivo, in a novel zebrafish model for intestinal epithelial shedding. Taken together, the data show that EGF suppresses cell shedding in the intestinal epithelium through a selective MAPK-dependent pathway affecting multiple extrusion mechanisms. EGFR signaling might be a therapeutic target for disorders featuring excessive cell turnover, such as inflammatory bowel diseases.


Asunto(s)
Factor de Crecimiento Epidérmico/farmacología , Células Epiteliales/metabolismo , Intestinos/citología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Animales , Inhibidores de Caspasas/farmacología , Caspasas/metabolismo , Perros , Células Epiteliales/efectos de los fármacos , Células de Riñón Canino Madin Darby , Ratones , Quinasas de Proteína Quinasa Activadas por Mitógenos/antagonistas & inhibidores , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Pez Cebra , Proteínas de Unión al GTP rho/metabolismo
8.
J Biol Chem ; 291(39): 20462-72, 2016 09 23.
Artículo en Inglés | MEDLINE | ID: mdl-27507810

RESUMEN

EGF receptor (EGFR) in tumor cells serves as a tumor promoter. However, information about EGFR activation in macrophages in regulating M2 polarization and tumor development is limited. This study aimed to investigate the effects of EGFR activation in macrophages on M2 polarization and development of gastrointestinal tumors. IL-4, a cytokine to elicit M2 polarization, stimulated release of an EGFR ligand, HB-EGF, and transactivation and down-regulation of EGFR in Raw 264.7 cells and peritoneal macrophages from WT mice. Knockdown of HB-EGF in macrophages inhibited EGFR transactivation by IL-4. IL-4-stimulated STAT6 activation, Arg1 and YM1 gene expression, and HB-EGF production were further enhanced by inhibition of EGFR activity in Raw 264.7 cells using an EGFR kinase inhibitor and in peritoneal macrophages from Egfr(wa5) mice with kinase inactive EGFR and by knockdown of EGFR in peritoneal macrophages from Egfr(fl/fl) LysM-Cre mice with myeloid cell-specific EGFR deletion. Chitin induced a higher level of M2 polarization in peritoneal macrophages in Egfr(fl/fl) LysM-Cre mice than that in Egfr(fl/fl) mice. Accordingly, IL-4-conditioned medium stimulated growth and epithelial-to-mesenchymal transition in gastric epithelial and colonic tumor cells, which were suppressed by that from Raw 264.7 cells with HB-EGF knockdown but promoted by that from Egfr(wa5) and Egfr(fl/fl) LysM-Cre peritoneal macrophages. Clinical assessment revealed that the number of macrophages with EGFR expression became less, indicating decreased inhibitory effects on M2 polarization, in late stage of human gastric cancers. Thus, IL-4-stimulated HB-EGF-dependent transactivation of EGFR in macrophages may mediate inhibitory feedback for M2 polarization and HB-EGF production, thereby inhibiting gastrointestinal tumor growth.


Asunto(s)
Receptores ErbB/biosíntesis , Neoplasias Gastrointestinales/metabolismo , Regulación Neoplásica de la Expresión Génica , Macrófagos Peritoneales/metabolismo , Activación Transcripcional , Animales , Línea Celular Tumoral , Receptores ErbB/genética , Neoplasias Gastrointestinales/genética , Neoplasias Gastrointestinales/patología , Factor de Crecimiento Similar a EGF de Unión a Heparina/genética , Factor de Crecimiento Similar a EGF de Unión a Heparina/metabolismo , Humanos , Interleucina-4/genética , Interleucina-4/metabolismo , Macrófagos Peritoneales/patología , Ratones , Ratones Noqueados , Células RAW 264.7
9.
Gastroenterology ; 149(4): 993-1005.e2, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26072395

RESUMEN

BACKGROUND & AIMS: Tumor necrosis factor receptor 2 (TNFR2, Tnfrsf1b) regulates multiple aspects of immune function, but little is known about its role in the immunopathogenesis of inflammatory bowel disease (IBD). We investigated whether TNFR2 restricts the activity of specific immune cell subtypes to protect against the development of colitis in mice. METHODS: Tnfr2(-/-) mice were crossed with interleukin (Il) 10(-/-) mice, which spontaneously develop colitis, to generate Il10(-/-)Tnfr2(-/-) mice. Colonic tissues were collected from Il10(-/-)Tnfr2(-/-) mice along with Il10(-/-) mice (controls) and analyzed by flow cytometry and histology. Bone marrow was transplanted into Il10(-/-) and Il10(-/-)Tnfr2(-/-) mice from Il10(-/-) or Il10(-/-)Tnfr2(-/-) donors by intravenous injection. CD8(+) T cells were neutralized in Il10(-/-)Tnfr2(-/-) mice by intraperitoneal injection of anti-CD8 or isotype control antibodies. Colitis was induced in Rag2(-/-) mice by intravenous injections of naïve CD8(+) T cells isolated from C57BL/6 or Tnfr2(-/-) mice. RESULTS: Il10(-/-)Tnfr2(-/-) mice spontaneously developed more severe colitis compared with Il10(-/-) controls, characterized by selective expansion of colonic CD8(+) T cells. Transplantation of TNFR2-deficient bone marrow resulted in significantly increased incidence and severity of colitis. Transcriptome analyses showed that the expression of genes regulated by TNFR2 were specific to CD8(+) T cells and included genes associated with risk for IBD. Depletion of CD8(+) T cells from Il10(-/-)Tnfr2(-/-) mice prevented colonic inflammation. Adoptive transfer of TNFR2-null naïve CD8(+) T cells compared with CD8(+) T cells from control mice increased the severity of colitis that developed in Rag2(-/-) mice. CONCLUSIONS: TNFR2 protects mice from colitis by inhibiting the expansion of colonic CD8(+) T cells. TNFR2 regulates expression of genes that regulate CD8(+) T cells and have been associated with susceptibility to IBD. Disruption in TNFR2 signaling might therefore be associated with pathogenesis. Strategies to increase levels or activity of TNFR2 and thereby reduce the activity of CD8(+) T cells might be developed to treat IBD patients with CD8(+) T cell dysfunction.


Asunto(s)
Linfocitos T CD8-positivos/metabolismo , Colitis/prevención & control , Colon/metabolismo , Inmunidad Celular , Receptores Tipo II del Factor de Necrosis Tumoral/metabolismo , Traslado Adoptivo , Animales , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/trasplante , Proliferación Celular , Colitis/genética , Colitis/inmunología , Colitis/metabolismo , Colitis/patología , Colon/inmunología , Colon/patología , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Interleucina-10/genética , Interleucina-10/metabolismo , Activación de Linfocitos , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores Tipo II del Factor de Necrosis Tumoral/deficiencia , Receptores Tipo II del Factor de Necrosis Tumoral/genética , Receptores Tipo II del Factor de Necrosis Tumoral/inmunología , Transducción de Señal , Factores de Tiempo
10.
J Immunol ; 192(3): 1013-23, 2014 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-24391216

RESUMEN

Macrophages regulate innate immunity to maintain intestinal homeostasis and play pathological roles in intestinal inflammation. Activation of the epidermal growth factor receptor (EGFR) promotes cellular proliferation, differentiation, survival, and wound closure in several cell types. However, the impact of EGFR in macrophages remains unclear. This study was to investigate whether EGFR activation in macrophages regulates cytokine production and intestinal inflammation. We found that EGFR was activated in colonic macrophages in mice with dextran sulfate sodium (DSS)-induced colitis and in patients with ulcerative colitis. DSS-induced acute colitis was ameliorated, and recovery from colitis was promoted in Egfr(fl/fl)LysM-Cre mice with myeloid cell-specific deletion of EGFR, compared with LysM-Cre mice. DSS treatment increased IL-10 and TNF levels during the acute phase of colitis, and increased IL-10 but reduced TNF levels during the recovery phase in Egfr(fl/fl)LysM-Cre mice. An anti-IL-10 neutralizing Ab abolished these effects of macrophage-specific EGFR deletion on DSS-induced colitis in Egfr(fl/fl)LysM-Cre mice. LPS stimulated EGFR activation and inhibition of EGFR kinase activity enhanced LPS-stimulated NF-κB activation in RAW 264.7 macrophages. Furthermore, induction of IL-10 production by EGFR kinase-blocked RAW 264.7 cells, in response to LPS plus IFN-γ, correlated with decreased TNF production. Thus, although selective deletion of EGFR in macrophages leads to increases in both pro- and anti-inflammatory cytokines in response to inflammatory stimuli, the increase in the IL-10 level plays a role in suppressing proinflammatory cytokine production, resulting in protection of mice from intestinal inflammation. These results reveal an integrated response of macrophages regulated by EGFR in intestinal inflammatory disorders.


Asunto(s)
Colitis/inmunología , Citocinas/biosíntesis , Receptores ErbB/fisiología , Macrófagos/inmunología , Adolescente , Adulto , Anciano , Animales , Línea Celular , Colitis/inducido químicamente , Colitis/metabolismo , Colitis Ulcerosa/metabolismo , Colon/inmunología , Colon/patología , Colon/fisiología , Ciclooxigenasa 2/biosíntesis , Ciclooxigenasa 2/genética , Citocinas/genética , Sulfato de Dextran/toxicidad , Receptores ErbB/deficiencia , Receptores ErbB/genética , Femenino , Regulación de la Expresión Génica/inmunología , Humanos , Inmunidad Innata , Inflamación , Interferón gamma/farmacología , Interleucina-10/antagonistas & inhibidores , Interleucina-10/biosíntesis , Interleucina-10/inmunología , Lipopolisacáridos/farmacología , Activación de Macrófagos , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Células Mieloides/metabolismo , FN-kappa B/metabolismo , Neutrófilos/metabolismo , Quinazolinas/farmacología , Regeneración , Transducción de Señal/inmunología , Bazo/inmunología , Bazo/patología , Factor de Necrosis Tumoral alfa/biosíntesis , Factor de Necrosis Tumoral alfa/genética , Tirfostinos/farmacología , Adulto Joven
11.
Am J Physiol Gastrointest Liver Physiol ; 308(3): G161-70, 2015 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-25477373

RESUMEN

Tumor necrosis factor (TNF) and its receptors TNFR1 and TNFR2 are major therapeutic targets for inflammatory bowel disease. Research advances have demonstrated that TNF produces pleiotropic responses in the gastrointestinal (GI) tract. Although in excess TNF can contribute to GI pathology, TNF is also a critical protective factor to promote GI homeostasis following injury and inflammation. Genetic studies using candidate and genome-wide association study approaches have identified variants in TNF or its receptors that are associated with Crohn's disease or ulcerative colitis in multiple populations, although the basis for these associations remains unclear. This review considers the efficacy and mechanism of anti-TNF therapies for inflammatory bowel disease to reconcile the many disparate aspects of TNF research and to consider the potential protective effects of TNF signaling in GI health.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Enfermedades Inflamatorias del Intestino/tratamiento farmacológico , Factores de Necrosis Tumoral/inmunología , Animales , Anticuerpos Monoclonales/inmunología , Fármacos Gastrointestinales/uso terapéutico , Tracto Gastrointestinal/inmunología , Tracto Gastrointestinal/metabolismo , Humanos , Enfermedades Inflamatorias del Intestino/inmunología , Receptores del Factor de Necrosis Tumoral/inmunología , Factores de Necrosis Tumoral/metabolismo
12.
Am J Physiol Gastrointest Liver Physiol ; 308(9): G721-35, 2015 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-25721303

RESUMEN

The mucosal layer of the colon is a unique and dynamic site where host cells interface with one another and the microbiome, with major implications for physiology and disease. However, the cellular mechanisms mediating colonic regeneration, inflammation, dysplasia, and dysbiosis remain undercharacterized, partly because the use of thin tissue sections in many studies removes important volumetric context. To address these challenges in visualization, we have developed the deep mucosal imaging (DMI) method to reconstruct continuous extended volumes of mouse colorectal mucosa at cellular resolution. Use of ScaleA2 and SeeDB clearing agents enabled full visualization of the colonic crypt, the fundamental unit of adult colon. Confocal imaging of large colorectal expanses revealed epithelial structures involved in repair, inflammation, tumorigenesis, and stem cell function, in fluorescent protein-labeled, immunostained, paraffin-embedded, or human biopsy samples. We provide freely available software to reconstruct and explore on computers with standard memory allocations the large DMI datasets containing in toto representations of distal colonic mucosal volume. Extended-volume imaging of colonic mucosa through the novel, extensible, and readily adopted DMI approach will expedite mechanistic investigations of intestinal physiology and pathophysiology at intracrypt to multicrypt length scales.


Asunto(s)
Colitis/patología , Colon/patología , Mucosa Intestinal/patología , Microscopía Confocal/métodos , Recto/patología , Animales , Colitis/inducido químicamente , Colitis/genética , Colitis/metabolismo , Colon/metabolismo , Sulfato de Dextran , Modelos Animales de Enfermedad , Fructosa , Genes Reporteros , Glicerol/análogos & derivados , Humanos , Procesamiento de Imagen Asistido por Computador , Mucosa Intestinal/metabolismo , Ratones Endogámicos C57BL , Ratones Transgénicos , Recto/metabolismo , Programas Informáticos , Soluciones
13.
Gastroenterology ; 146(7): 1739-51.e14, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24530706

RESUMEN

BACKGROUND & AIMS: The gastric cancer-causing pathogen Helicobacter pylori up-regulates spermine oxidase (SMOX) in gastric epithelial cells, causing oxidative stress-induced apoptosis and DNA damage. A subpopulation of SMOX(high) cells are resistant to apoptosis, despite their high levels of DNA damage. Because epidermal growth factor receptor (EGFR) activation can regulate apoptosis, we determined its role in SMOX-mediated effects. METHODS: SMOX, apoptosis, and DNA damage were measured in gastric epithelial cells from H. pylori-infected Egfr(wa5) mice (which have attenuated EGFR activity), Egfr wild-type mice, or in infected cells incubated with EGFR inhibitors or deficient in EGFR. A phosphoproteomic analysis was performed. Two independent tissue microarrays containing each stage of disease, from gastritis to carcinoma, and gastric biopsy specimens from Colombian and Honduran cohorts were analyzed by immunohistochemistry. RESULTS: SMOX expression and DNA damage were decreased, and apoptosis increased in H. pylori-infected Egfr(wa5) mice. H. pylori-infected cells with deletion or inhibition of EGFR had reduced levels of SMOX, DNA damage, and DNA damage(high) apoptosis(low) cells. Phosphoproteomic analysis showed increased EGFR and erythroblastic leukemia-associated viral oncogene B (ERBB)2 signaling. Immunoblot analysis showed the presence of a phosphorylated (p)EGFR-ERBB2 heterodimer and pERBB2; knockdown of ErbB2 facilitated apoptosis of DNA damage(high) apoptosis(low) cells. SMOX was increased in all stages of gastric disease, peaking in tissues with intestinal metaplasia, whereas pEGFR, pEGFR-ERBB2, and pERBB2 were increased predominantly in tissues showing gastritis or atrophic gastritis. Principal component analysis separated gastritis tissues from patients with cancer vs those without cancer. pEGFR, pEGFR-ERBB2, pERBB2, and SMOX were increased in gastric samples from patients whose disease progressed to intestinal metaplasia or dysplasia, compared with patients whose disease did not progress. CONCLUSIONS: In an analysis of gastric tissues from mice and patients, we identified a molecular signature (based on levels of pEGFR, pERBB2, and SMOX) for the initiation of gastric carcinogenesis.


Asunto(s)
Daño del ADN , Células Epiteliales/enzimología , Receptores ErbB/metabolismo , Mucosa Gástrica/enzimología , Infecciones por Helicobacter/enzimología , Helicobacter pylori/metabolismo , Receptor ErbB-2/metabolismo , Animales , Apoptosis , Línea Celular , Supervivencia Celular , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Técnicas de Cocultivo , Colombia , Progresión de la Enfermedad , Activación Enzimática , Células Epiteliales/microbiología , Células Epiteliales/patología , Receptores ErbB/deficiencia , Receptores ErbB/genética , Mucosa Gástrica/microbiología , Mucosa Gástrica/patología , Gastritis/enzimología , Gastritis/microbiología , Gastritis/patología , Infecciones por Helicobacter/genética , Infecciones por Helicobacter/microbiología , Infecciones por Helicobacter/patología , Helicobacter pylori/patogenicidad , Honduras , Humanos , Metaplasia , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Oxidorreductasas actuantes sobre Donantes de Grupo CH-NH/metabolismo , Fosforilación , Lesiones Precancerosas/enzimología , Lesiones Precancerosas/microbiología , Lesiones Precancerosas/patología , Análisis de Componente Principal , Multimerización de Proteína , Receptor ErbB-2/genética , Transducción de Señal , Neoplasias Gástricas/enzimología , Neoplasias Gástricas/microbiología , Neoplasias Gástricas/patología , Tennessee , Poliamino Oxidasa
14.
J Biol Chem ; 288(42): 30742-30751, 2013 Oct 18.
Artículo en Inglés | MEDLINE | ID: mdl-24043629

RESUMEN

p40, a Lactobacillus rhamnosus GG (LGG)-derived soluble protein, ameliorates intestinal injury and colitis, reduces apoptosis, and preserves barrier function by transactivation of the EGF receptor (EGFR) in intestinal epithelial cells. The aim of this study is to determine the mechanisms by which p40 transactivates the EGFR in intestinal epithelial cells. Here we show that p40-conditioned medium activates EGFR in young adult mouse colon epithelial cells and human colonic epithelial cell line, T84 cells. p40 up-regulates a disintegrin and metalloproteinase domain-containing protein 17 (ADAM17) catalytic activity, and broad spectrum metalloproteinase inhibitors block EGFR transactivation by p40 in these two cell lines. In ADAM17-deficient mouse colonic epithelial (ADAM17(-/-) MCE) cells, p40 transactivation of EGFR is blocked, but can be rescued by re-expression with WT ADAM17. Furthermore, p40 stimulates release of heparin binding (HB)-EGF, but not transforming growth factor (TGF)α or amphiregulin, in young adult mouse colon cells and ADAM17(-/-) MCE cells overexpressing WT ADAM17. Knockdown of HB-EGF expression by siRNA suppresses p40 effects on transactivating EGFR and Akt, preventing apoptosis, and preserving tight junction function. The effects of p40 on HB-EGF release and ADAM17 activation in vivo are examined after administration of p40-containing pectin/zein hydrogel beads to mice. p40 stimulates ADAM17 activity and EGFR activation in colonic epithelial cells and increases HB-EGF levels in blood from WT mice, but not from mice with intestinal epithelial cell-specific ADAM17 deletion. Thus, these data define a mechanism of a probiotic-derived soluble protein in modulating intestinal epithelial cell homeostasis through ADAM17-mediated HB-EGF release, leading to transactivation of EGFR.


Asunto(s)
Proteínas Bacterianas/metabolismo , Células Epiteliales/metabolismo , Receptores ErbB/biosíntesis , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Mucosa Intestinal/metabolismo , Lacticaseibacillus rhamnosus/metabolismo , Probióticos/metabolismo , Activación Transcripcional , Proteínas ADAM/biosíntesis , Proteínas ADAM/genética , Proteína ADAM17 , Animales , Línea Celular Tumoral , Activación Enzimática/genética , Células Epiteliales/citología , Células Epiteliales/microbiología , Receptores ErbB/genética , Regulación Enzimológica de la Expresión Génica/genética , Técnicas de Silenciamiento del Gen , Factor de Crecimiento Similar a EGF de Unión a Heparina , Humanos , Péptidos y Proteínas de Señalización Intercelular/genética , Mucosa Intestinal/citología , Mucosa Intestinal/microbiología , Ratones , Ratones Noqueados , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Factor de Crecimiento Transformador alfa/genética , Factor de Crecimiento Transformador alfa/metabolismo , Regulación hacia Arriba/genética
15.
Clin Gastroenterol Hepatol ; 12(12): 2026-32, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24732284

RESUMEN

BACKGROUND & AIMS: Pediatric functional abdominal pain has been linked to functional gastrointestinal disorders (FGIDs) in adulthood, but little is known about patient characteristics in childhood that increase the risk for FGID in young adulthood. We investigated the contribution of gastrointestinal symptoms, extraintestinal somatic symptoms, and depressive symptoms in pediatric patients with functional abdominal pain and whether these predicted FGIDs later in life. METHODS: In a longitudinal study, consecutive new pediatric patients, diagnosed with functional abdominal pain in a subspecialty clinic, completed a comprehensive baseline evaluation of the severity of their physical and emotional symptoms. They were contacted 5 to 15 years later and evaluated, based on Rome III symptom criteria, for abdominal pain-related FGIDs, including irritable bowel syndrome, functional dyspepsia, functional abdominal pain syndrome, and abdominal migraine. Controlling for age, sex, baseline severity of abdominal pain, and time to follow-up evaluation, multivariable logistic regression was used to evaluate the association of baseline gastrointestinal, extraintestinal somatic, and depressive symptoms in childhood with FGID in adolescence and young adulthood. RESULTS: Of 392 patients interviewed an average of 9.2 years after their initial evaluation, 41% (n = 162) met symptom criteria for FGID; most met the criteria for irritable bowel syndrome. Extraintestinal somatic and depressive symptoms at the initial pediatric evaluation were significant predictors of FGID later in life, after controlling for initial levels of GI symptoms. Age, sex, and abdominal pain severity at initial presentation were not significant predictors of FGID later in life. CONCLUSIONS: In pediatric patients with functional abdominal pain, assessment of extraintestinal and depressive symptoms may be useful in identifying those at risk for FGID in adolescence and young adulthood.


Asunto(s)
Dolor Abdominal/diagnóstico , Dolor Abdominal/epidemiología , Dolor Abdominal/fisiopatología , Adolescente , Niño , Comorbilidad , Depresión/diagnóstico , Depresión/epidemiología , Femenino , Enfermedades Gastrointestinales/diagnóstico , Enfermedades Gastrointestinales/epidemiología , Humanos , Estudios Longitudinales , Masculino , Pronóstico , Adulto Joven
16.
Gut ; 62(1): 73-82, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22267598

RESUMEN

BACKGROUND: Necrotising enterocolitis (NEC) is the most common gastrointestinal emergency in premature infants. Immaturity of gastrointestinal immune regulation may predispose preterm infants to NEC as FOXP3 T regulatory cells (Treg) are critical for intestinal immune homoeostasis. OBJECTIVE: To investigate the hypothesis that abnormal developmental regulation of lamina propria Treg would define premature infants with NEC. DESIGN: Lamina propria mononuclear cell populations from surgically resected ileum from 18 patients with NEC and 30 gestational age-matched non-NEC surgical controls were prospectively isolated. Polychromatic flow cytometry was performed to phenotype and analyse lamina propria T cell populations. The cytokine gene expression profile in NEC tissue was compared with that of non-NEC controls. RESULTS: The total number of Treg, CD4, or CD8 T cells in each ileum section was independent of gestational age, age or postmenstrual age and similar between patients with NEC and controls. In contrast, the ratio of Treg to CD4 T cells or Treg to CD8 T cells was significantly lower in NEC ileum than in infants without NEC (medians 2.9% vs 6.6%, p=0.001 and medians 6.6% vs 25.9%, p<0.001, respectively). For any given number of CD4 or CD8 T cells, Treg were, on average, 60% lower in NEC ileum than in controls. NEC tissue cytokine gene expression profiles were characteristic of inhibited Treg development or function. Treg/CD4 and Treg/CD8 ratios recovered between initial resection for NEC and reanastomosis. CONCLUSION: The proportion of lamina propria Treg is significantly reduced in the ileum of premature infants with NEC and may contribute to the excessive inflammatory state of this disease.


Asunto(s)
Linfocitos T CD8-positivos/metabolismo , Enterocolitis Necrotizante/inmunología , Factores de Transcripción Forkhead/metabolismo , Enfermedades del Prematuro/inmunología , Mucosa Intestinal/inmunología , Linfocitos T Reguladores/metabolismo , Biomarcadores/metabolismo , Linfocitos T CD4-Positivos/metabolismo , Estudios de Casos y Controles , Femenino , Citometría de Flujo , Perfilación de la Expresión Génica , Humanos , Recién Nacido , Recien Nacido Prematuro , Recuento de Linfocitos , Masculino , Estudios Prospectivos
17.
Inflamm Bowel Dis ; 30(3): 402-409, 2024 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-37229656

RESUMEN

BACKGROUND: Intestinal ultrasound (IUS) is a noninvasive tool to assess bowel inflammation. There is a paucity of data on its accuracy in pediatric patients. AIM: The aim of this study is to evaluate the diagnostic performance of bowel wall thickness (BWT) measured using IUS compared with endoscopic disease activity in children suspected of having inflammatory bowel disease (IBD). METHODS: We conducted a single-center cross-sectional pilot study of pediatric patients suspected to have previously undiagnosed IBD. Endoscopic inflammation was graded using segmental scores of the Simple Endoscopic Score for Crohn's Disease (SES-CD) and the Ulcerative Colitis Endoscopic Index of Severity (UCEIS) and classified as having healthy, mild, or moderate/severe disease activity. Association between BWT and endoscopic severity was assessed using the Kruskal-Wallis test. The diagnostic performance of BWT to detect active disease at endoscopy was evaluated using the area under the receiver operating characteristic curve; sensitivity and specificity were calculated. RESULTS: In all, 174 bowel segments in 33 children were assessed by IUS and ileocolonoscopy. An elevated median BWT was associated with increased bowel segment disease severity, classified by the SES-CD (P < .001) and the UCEIS (P < .01). Using a cutoff value of 1.9 mm, we found that the BWT had an area under the receiver operating characteristic curve of 0.743 (95% CI, 0.67-0.82), a sensitivity of 64% (95% CI, 53%-73%), and a specificity of 76% (95% CI, 65%-85%) to detect inflamed bowel. CONCLUSION: Increasing BWT is associated with increasing endoscopic activity in pediatric IBD. Our study suggests that the optimal BWT cutoff value for detecting active disease may be less than that seen in adults. Additional pediatric studies are needed.


Increasing bowel wall thickness (BWT) is associated with increasing IBD endoscopic scores on colonoscopy. There is moderate to fair agreement between the prediction of IBD diagnosis and Paris classification using intestinal ultrasound (IUS). Bowel wall thickness cutoff values to detect inflamed bowel segments are likely lower for children with IBD than for adults, although further studies with wider age ranges are needed to confirm this finding.


Asunto(s)
Colitis Ulcerosa , Enfermedades Inflamatorias del Intestino , Adulto , Humanos , Niño , Estudios Transversales , Proyectos Piloto , Colitis Ulcerosa/diagnóstico por imagen , Inflamación , Gravedad del Paciente
18.
Adv Ther ; 41(3): 901-914, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38286962

RESUMEN

Dysbiosis corresponds to the disruption of a formerly stable, functionally complete microbiota. In the gut, this imbalance can lead to adverse health outcomes in both the short and long terms, with a potential increase in the lifetime risks of various noncommunicable diseases and disorders such as atopy (like asthma), inflammatory bowel disease, neurological disorders, and even behavioural and psychological disorders. Although antibiotics are highly effective in reducing morbidity and mortality in infectious diseases, antibiotic-associated diarrhoea is a common, non-negligible clinical sign of gut dysbiosis (and the only visible one). Re-establishment of a normal (functional) gut microbiota is promoted by completion of the clinically indicated course of antibiotics, the removal of any other perturbing external factors, the passage of time (i.e. recovery through the microbiota's natural resilience), appropriate nutritional support, and-in selected cases-the addition of probiotics. Systematic reviews and meta-analyses of clinical trials have confirmed the strain-specific efficacy of some probiotics (notably the yeast Saccharomyces boulardii CNCM I-745 and the bacterium Lactobacillus rhamnosus GG) in the treatment and/or prevention of antibiotic-associated diarrhoea in children and in adults. Unusually for a probiotic, S. boulardii is a eukaryote and is not therefore directly affected by antibiotics-making it suitable for administration in cases of antibiotic-associated diarrhoea. A robust body of evidence from clinical trials and meta-analyses shows that the timely administration of an adequately dosed probiotic (upon initiation of antibiotic treatment or within 48 h) can help to prevent or resolve the consequences of antibiotic-associated dysbiosis (such as diarrhoea) and promote the resilience of the gut microbiota and a return to the pre-antibiotic state. A focus on the prescription of evidence-based, adequately dosed probiotics should help to limit unjustified and potentially ineffective self-medication.


Asunto(s)
Lacticaseibacillus rhamnosus , Probióticos , Saccharomyces boulardii , Adulto , Niño , Humanos , Antibacterianos/efectos adversos , Diarrea/inducido químicamente , Diarrea/prevención & control , Disbiosis/inducido químicamente , Disbiosis/terapia , Probióticos/uso terapéutico , Saccharomyces cerevisiae , Metaanálisis como Asunto , Revisiones Sistemáticas como Asunto
19.
Am J Physiol Gastrointest Liver Physiol ; 305(2): G196-203, 2013 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-23681474

RESUMEN

Chronic infection of the gastric mucosa by Helicobacter pylori is associated with an increased risk of developing gastric cancer; however, the vast majority of infected individuals never develop this disease. One H. pylori virulence factor that increases gastric cancer risk is the cag pathogenicity island, which encodes a bacterial type IV secretion system. Cyclooxygenase-2 (COX-2) expression is induced by proinflammatory stimuli, leading to increased prostaglandin E2 (PGE2) secretion by gastric epithelial cells. COX-2 expression is increased in gastric tissue from H. pylori-infected persons. H. pylori also activates the epidermal growth factor receptor (EGFR) in gastric epithelial cells. We now demonstrate that H. pylori-induced activation of COX-2 in gastric cells is dependent upon EGFR activation, and that a functional cag type IV secretion system and direct bacterial contact are necessary for full induction of COX-2 by gastric epithelial cells. PGE2 secretion is increased in cells infected with H. pylori, and this induction is dependent on a functional EGFR. Increased apoptosis in response to H. pylori occurs in cells treated with a COX-2 inhibitor, as well as COX-2-/- cells, indicating that COX-2 expression promotes cell survival. In vivo, COX-2 induction by H. pylori is significantly reduced in mice deficient for EGFR activation compared with wild-type mice with a fully functional receptor. Collectively, these findings indicate that aberrant activation of the EGFR-COX-2 axis may lower the threshold for carcinogenesis associated with chronic H. pylori infection.


Asunto(s)
Ciclooxigenasa 2/metabolismo , Células Epiteliales/metabolismo , Receptores ErbB/metabolismo , Infecciones por Helicobacter/metabolismo , Helicobacter pylori , Estómago/citología , Animales , Línea Celular , Ciclooxigenasa 2/genética , Dinoprostona , Receptores ErbB/genética , Regulación Enzimológica de la Expresión Génica/fisiología , Infecciones por Helicobacter/microbiología , Ratones , Ratones Noqueados
20.
Dev Cell ; 58(21): 2309-2325.e7, 2023 11 06.
Artículo en Inglés | MEDLINE | ID: mdl-37652012

RESUMEN

Chronic colonic injury and inflammation pose high risks for field cancerization, wherein injury-associated mutations promote stem cell fitness and gradual clonal expansion. However, the long-term stability of some colitis-associated mutational fields could suggest alternate origins. Here, studies of acute murine colitis reveal a punctuated mechanism of massive, neutral clonal expansion during normal wound healing. Through three-dimensional (3D) imaging, quantitative fate mapping, and single-cell transcriptomics, we show that epithelial wound repair begins with the loss of structural constraints on regeneration, forming fused labyrinthine channels containing epithelial cells reprogrammed to a non-proliferative plastic state. A small but highly proliferative set of epithelial founder progenitor cells (FPCs) subsequently emerges and undergoes extensive cell division, enabling fluid-like lineage mixing and spreading across the colonic surface. Crypt budding restores the glandular organization, imprinting the pattern of clonal expansion. The emergence and functions of FPCs within a critical window of plasticity represent regenerative targets with implications for preneoplasia.


Asunto(s)
Colitis , Ratones , Animales , Colitis/genética , Células Epiteliales , Células Madre , Cicatrización de Heridas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA