Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Pediatr Surg Int ; 40(1): 35, 2024 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-38216767

RESUMEN

PURPOSE: Necrotizing enterocolitis (NEC) is a severe intestinal disease primarily affecting premature infants, marked by impaired epithelial regeneration. Breastfed infants are less susceptible to NEC than formula-fed ones, and human milk oligosaccharides (HMO) found in breast milk have prebiotic properties that can protect against NEC. However, it is unclear how HMOs influence intestinal epithelium regeneration in relation to the gut microbiota. METHODS: Broad-spectrum antibiotics were administered to pregnant dams to reduce the microbiota in offspring. NEC was induced through administration of hyperosmolar formula, lipopolysaccharide, and hypoxia from postnatal days (p) 5-9. Intestinal epithelial organoids were derived from p9 mice. HMOs were isolated from human donor breast milk and then solubilized in the formula for each feed or culture media for organoids. RESULTS: HMOs did not alter the microbiota profile in the presence of a normal or reduced microbiota. In the reduced microbiota, HMO treatment decreased NEC intestinal injury, and increased proliferation and stem cell activity. Additionally, in the complete absence of the microbiota, HMOs stimulated intestinal organoid growth. CONCLUSION: This study demonstrates that HMOs promoted intestinal epithelial regeneration independent of the gut microbiota. These findings provide further insight into the various benefits HMOs may have in the protection against NEC.


Asunto(s)
Enterocolitis Necrotizante , Enfermedades del Recién Nacido , Microbiota , Lactante , Femenino , Embarazo , Recién Nacido , Animales , Humanos , Ratones , Leche Humana , Enterocolitis Necrotizante/prevención & control , Mucosa Intestinal , Oligosacáridos/farmacología , Regeneración
2.
Front Cell Infect Microbiol ; 12: 883163, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35774405

RESUMEN

Compositional analysis of the intestinal microbiome in pre-schoolers is understudied. Effects of probiotics on the gut microbiota were evaluated in children under 4-years-old presenting to an emergency department with acute gastroenteritis. Included were 70 study participants (n=32 placebo, n=38 probiotics) with stool specimens at baseline (day 0), day 5, and after a washout period (day 28). Microbiota composition and deduced functions were profiled using 16S ribosomal RNA sequencing and predictive metagenomics, respectively. Probiotics were detected at day 5 of administration but otherwise had no discernable effects, whereas detection of bacterial infection (P<0.001) and participant age (P<0.001) had the largest effects on microbiota composition, microbial diversity, and deduced bacterial functions. Participants under 1 year had lower bacterial diversity than older aged pre-schoolers; compositional changes of individual bacterial taxa were associated with maturation of the gut microbiota. Advances in age were associated with differences in gut microbiota composition and deduced microbial functions, which have the potential to impact health later in life. Clinical Trial Registration: www.ClinicalTrials.gov, identifier: NCT01853124.


Asunto(s)
Gastroenteritis , Microbioma Gastrointestinal , Microbiota , Probióticos , Niño , Preescolar , Heces/microbiología , Gastroenteritis/tratamiento farmacológico , Humanos , Intestinos , Probióticos/uso terapéutico , ARN Ribosómico 16S/genética
3.
Nutrients ; 14(5)2022 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-35267989

RESUMEN

BACKGROUND: Human milk oligosaccharides are complex, non-digestible carbohydrates that directly interact with intestinal epithelial cells to alter barrier function and host inflammation. Oligosaccharide composition varies widely between individual mothers, but it is unclear if this inter-individual variation has any impact on intestinal epithelial barrier function and gut inflammation. METHODS: Human milk oligosaccharides were extracted from the mature human milk of four individual donors. Using an in vitro model of intestinal injury, the effects of the oligosaccharides on the intestinal epithelial barrier and select innate and adaptive immune functions were assessed. RESULTS: Individual oligosaccharide compositions shared comparable effects on increasing transepithelial electrical resistance and reducing the macromolecular permeability of polarized (Caco-2Bbe1) monolayers but exerted distinct effects on the localization of the intercellular tight junction protein zona occludins-1 in response to injury induced by a human enteric bacterial pathogen Escherichia coli, serotype O157:H7. Immunoblots showed the differential effects of oligosaccharide compositions in reducing host chemokine interleukin 8 expression and inhibiting of p38 MAP kinase activation. CONCLUSIONS: These results provide evidence of both shared and distinct effects on the host intestinal epithelial function that are attributable to inter-individual differences in the composition of human milk oligosaccharides.


Asunto(s)
Mucosa Intestinal , Leche Humana , Humanos , Inflamación/metabolismo , Mucosa Intestinal/metabolismo , Oligosacáridos/farmacología , Proyectos Piloto
4.
Pediatr Res ; 91(1): 101-106, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34561550

RESUMEN

BACKGROUND: Stem cell therapy has been proven to rescue intestinal injury and stimulate intestinal regeneration in necrotizing enterocolitis (NEC). Specifically, stem cells derived from amniotic fluid (AFSCs) and mesenchymal stem cells (MSCs) derived from bone marrow have shown promising results in the treatment of experimental NEC. This study aims to examine the effects of AFSCs and MSCs on the prevention of intestinal injury during experimental NEC. METHODS: Supernatants from AFSC and MSC cultures were collected to perform proteomic analysis. Prior to NEC induction, mice received intraperitoneal injections of phosphate-buffered saline (PBS), 2 × 106 AFSCs, or 2 × 106 MSCs. RESULTS: We found that AFSCs grew faster than MSCs. Proteomic analysis indicated that AFSCs are primarily involved in cell development and growth, while MSCs are involved in immune regulation. Administering AFSCs before NEC induction decreased NEC severity and mucosal inflammation. Intestinal proliferation and endogenous stem cell activation were increased after AFSC administration. However, administering MSCs before NEC induction had no beneficial effects. CONCLUSIONS: This study demonstrated that AFSCs and MSCs have different protein release profiles. AFSCs can potentially be used as a preventative strategy for neonates at risk of NEC, while MSCs cannot be used. IMPACT: AFSCs and MSCs have distinct protein secretory profiles, and AFSCs are primarily involved in cell development and growth, while MSCs are involved in immune regulation. AFSCs are unique in transiently enhancing healthy intestinal epithelial cell growth, which offers protection against the development of experimental NEC. The prevention of NEC via the administration of AFSCs should be evaluated in infants at great risk of developing NEC or in infants with early signs of NEC.


Asunto(s)
Líquido Amniótico/citología , Trasplante de Células Madre , Animales , Enterocolitis Necrotizante , Humanos , Recién Nacido , Ratones
5.
Mol Nutr Food Res ; 66(4): e2100893, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34921749

RESUMEN

SCOPE: Necrotizing enterocolitis (NEC) is a devastating gastrointestinal emergency affecting preterm infants. Breastmilk protects against NEC, partly due to human milk oligosaccharides (HMOs). HMO compositions are highly diverse, and it is unclear if anti-NEC properties are specific to carbohydrate motifs. Here, this study compares intestinal epithelial transcriptomes of five synthetic HMOs (sHMOs) and examines structure-function relationships of HMOs on intestinal signaling. METHODS AND RESULTS: This study interrogates the transcriptome of Caco-2Bbe1 cells in response to five synthetic HMOs (sHMOs) using RNA sequencing: 2'-fucosyllactose (2'-FL), 3-fucosyllactose (3FL), 6'-siallyllactose (6'-SL), lacto-N-tetraose (LNT), lacto-N-neotetraose (LNnT). Protection against intestinal barrier dysfunction and inflammation occurred in an HMO-dependent manner. Each sHMO exerts a unique set of host transcriptome changes and modulated unique signaling pathways. There is clustering between HMOs bearing similar side chains, with little overlap in gene regulation which is shared by all sHMOs. Interestingly, most sHMOs protect pups against NEC, exerting divergent mechanisms on intestinal cell morphology and inflammation. CONCLUSIONS: These results demonstrate that while structurally distinct HMOs impact intestinal physiology, their mechanisms of action differ. This finding establishes the first structure-function relationship of HMOs in the context of intestinal cell signaling responses and offers a functional framework by which to screen and design HMO-like compounds.


Asunto(s)
Enterocolitis Necrotizante , Leche Humana , Animales , Células CACO-2 , Modelos Animales de Enfermedad , Enterocolitis Necrotizante/prevención & control , Humanos , Lactante , Recién Nacido , Recien Nacido Prematuro , Ratones , Leche Humana/química , Oligosacáridos/química , Relación Estructura-Actividad , Transcriptoma
6.
Mol Nutr Food Res ; 64(21): e2000519, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32926533

RESUMEN

SCOPE: Necrotizing enterocolitis (NEC) is a devastating gastrointestinal emergency and currently the leading cause of mortality in preterm infants. Recent studies show that human milk oligosaccharides (HMOs) reduce the frequency and incidence of NEC; however, the molecular mechanisms for their protection are largely unexplored. METHODS AND RESULTS: To address this gap, a genome-wide profiling of the intestinal epithelial transcriptome in response to HMOs using RNA-sequencing is performed. It is found that HMOs alter the host transcriptome in 225 unique target genes pertaining to cell proliferation and differentiation, including upregulation of stem cell differentiation marker HMGCS2. To validate these results, differentiation in Caco-2Bbe1 (Caco-2) intestinal cells is verified by Alcian Blue staining and transepithelial electrical resistance (TER) recordings. Furthermore, an in vivo model of NEC is also employed whereby neonatal pups are gavage fed HMOs. Interestingly, HMOs-fed pups show enhanced cell MUC2 differentiation and HMGCS2 expression. CONCLUSIONS: These findings demonstrate HMOs protect against NEC in part by altering the differentiation of the crypt-villus axis. In addition, this study suggests that pooled HMOs directly induce a series of biological processes, which provide mechanistic insights to how HMOs protect the host intestine.


Asunto(s)
Enterocolitis Necrotizante/patología , Enterocolitis Necrotizante/prevención & control , Leche Humana/química , Oligosacáridos/farmacología , Animales , Células CACO-2 , Ciclo Celular/efectos de los fármacos , Ciclo Celular/genética , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Perros , Enterocolitis Necrotizante/genética , Femenino , Perfilación de la Expresión Génica , Humanos , Hidroximetilglutaril-CoA Sintasa/metabolismo , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/metabolismo , Células de Riñón Canino Madin Darby , Masculino , Ratones Endogámicos C57BL , Receptores Activados del Proliferador del Peroxisoma/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Serina-Treonina Quinasas TOR/metabolismo
7.
Cell Death Dis ; 11(9): 750, 2020 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-32929076

RESUMEN

Necrotizing enterocolitis (NEC) is a devastating intestinal disease primarily affecting preterm neonates and causing high morbidity, high mortality, and huge costs for the family and society. The treatment and the outcome of the disease have not changed in recent decades. Emerging evidence has shown that stimulating the Wnt/ß-catenin pathway and enhancing intestinal regeneration are beneficial in experimental NEC, and that they could potentially be used as a novel treatment. Amniotic fluid stem cells (AFSC) and AFSC-derived extracellular vesicles (EV) can be used to improve intestinal injury in experimental NEC. However, the mechanisms by which they affect the Wnt/ß-catenin pathway and intestinal regeneration are unknown. In our current study, we demonstrated that AFSC and EV attenuate NEC intestinal injury by activating the Wnt signaling pathway. AFSC and EV stimulate intestinal recovery from NEC by increasing cellular proliferation, reducing inflammation and ultimately regenerating a normal intestinal epithelium. EV administration has a rescuing effect on intestinal injury when given during NEC induction; however, it failed to prevent injury when given prior to NEC induction. AFSC-derived EV administration is thus a potential emergent novel treatment strategy for NEC.


Asunto(s)
Enterocolitis Necrotizante/genética , Vesículas Extracelulares/metabolismo , Intestinos/lesiones , Vía de Señalización Wnt/genética , Animales , Modelos Animales de Enfermedad , Humanos , Ratones , Ratas
8.
Front Nutr ; 7: 83, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32582756

RESUMEN

Purpose: Inflammatory bowel disease (IBD) refers to a spectrum of autoimmune diseases, which result in chronic intestinal inflammation. Previous findings suggest a role for diet, nutrition and dysbiosis of the gut microbiota in both the development and progression of the condition. Vitamin B12 is a key cofactor of methionine synthase and is produced solely by microbes. Previous work links increased levels of homocysteine, a substrate of methionine synthase, MetH, to IBD indicating a potential role for vitamin B12 deficiency in intestinal injury and inflammation. This study assessed the role of vitamin B12 in shaping the gut microbiota and determining responses to intestinal injury using a reproducible murine model of colitis. Methods: The effects of vitamin B12 supplementation and deficiency were assessed in vivo; 3-week-old post-weanling C57Bl/6 mice were divided into three dietary treatment groups: (1) sufficient vitamin B12 (50 mg/Kg), (2) deficient vitamin B12 (0 mg/Kg) and (3) supplemented vitamin B12 (200 mg/Kg) for a period of 4 weeks. Intestinal injury was induced with 2% dextran sodium sulphate (DSS) via drinking water for 5 days. The impact of varying levels of dietary vitamin B12 on gut microbiota composition was assessed using 16S rRNA gene sequencing from fecal samples collected at day 0 and day 28 of the dietary intervention, and 7 days following induction of colitis on day 38, when blood and colonic tissues were also collected. Results: No significant alterations were found in the gut microbiota composition of disease-free animals in response to dietary interventions. By contrast, after DSS-induced colitis, >30 genera were significantly altered in vitamin B12 deficient mice. Altered B12 levels produced no significant effect on composite disease-activity scores; however, administration of a B12 deficient diet resulted in reduced DSS-induced epithelial tissue damage. Conclusions: Vitamin B12 supplementation does not alter the gut microbiota composition under healthy conditions, but does contribute to differential microbial responses and intestinal dysbiosis following the induction of experimental colitis.

9.
Mol Nutr Food Res ; 64(6): e1900873, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31945799

RESUMEN

SCOPE: Marine-derived n-3 PUFAs may ameliorate inflammation associated with inflammatory bowel diseases. Plant-derived n-3 PUFAs are thought to be inferior owing to shorter chain lengths. The aim of this study is to compare the impact of plant- and fish-derived PUFAs on murine colitis. METHODS AND RESULTS: C57BL/6 mice are fed high fat (36% kcal) diets with either 2.5% w/w sunflower oil (SO), flaxseed oil (FSO), ahiflower oil (AO), or fish oil (FO). After 4 weeks, mice are orogastrically challenged with Citrobacter rodentium (108 CFU) or sham gavaged. Fecal shedding is assayed at 2, 7, 10, and 14 days post infection (PI), and fecal microbiota at 14 days PI. Colonic inflammation and lipid mediators are measured. Supplementation regulates intestinal inflammation with crypt lengths being 66, 73, and 62 ±17 µm shorter (compared to SO) for FSO, AO, and FO respectively, p < 0.01. FSO blunts pathogen shedding at the peak of infection and FSO and AO both enhance fecal microbial diversity. FO attenuates levels of lipoxin and leukotriene B4 while plant oils increase pro-resolving mediator concentrations including D, E, and T-series resolvins. CONCLUSION: Plant and fish n-3 PUFAs attenuate colitis-induced inflammation while exhibiting characteristic pro-resolving lipid mediator metabolomes. Plant oils additionally promote microbial diversity.


Asunto(s)
Citrobacter rodentium/patogenicidad , Colitis/dietoterapia , Ácidos Grasos Omega-3/farmacología , Aceites de Pescado/farmacología , Aceites de Plantas/farmacología , Animales , Derrame de Bacterias/efectos de los fármacos , Colitis/microbiología , Colitis/patología , Colon/efectos de los fármacos , Colon/metabolismo , Suplementos Dietéticos , Infecciones por Enterobacteriaceae/dietoterapia , Mediadores de Inflamación/metabolismo , Aceite de Linaza/química , Aceite de Linaza/farmacología , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Ratones Endogámicos C57BL , Aceite de Girasol/farmacología
10.
Cell Death Dis ; 10(10): 743, 2019 10 03.
Artículo en Inglés | MEDLINE | ID: mdl-31582728

RESUMEN

Necrotizing enterocolitis (NEC) is a devastating neonatal disease characterized by acute intestinal injury. Intestinal stem cell (ISC) renewal is required for gut regeneration in response to acute injury. The Wnt/ß-catenin pathway is essential for intestinal renewal and ISC maintenance. We found that ISC expression, Wnt activity and intestinal regeneration were all decreased in both mice with experimental NEC and in infants with acute active NEC. Moreover, intestinal organoids derived from NEC-injured intestine of both mice and humans failed to maintain proliferation and presented more differentiation. Administration of Wnt7b reversed these changes and promoted growth of intestinal organoids. Additionally, administration of exogenous Wnt7b rescued intestinal injury, restored ISC, and reestablished intestinal epithelial homeostasis in mice with NEC. Our findings demonstrate that during NEC, Wnt/ß-catenin signaling is decreased, ISC activity is impaired, and intestinal regeneration is defective. Administration of Wnt resulted in the maintenance of intestinal epithelial homeostasis and avoidance of NEC intestinal injury.


Asunto(s)
Enterocolitis Necrotizante/fisiopatología , Intestinos/fisiopatología , Regeneración/fisiología , Vía de Señalización Wnt , Animales , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Enterocolitis Necrotizante/genética , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Intestinos/efectos de los fármacos , Intestinos/patología , Ratones Endogámicos C57BL , Modelos Biológicos , Organoides/efectos de los fármacos , Organoides/metabolismo , Proteínas Proto-Oncogénicas/administración & dosificación , Proteínas Proto-Oncogénicas/farmacología , Regeneración/efectos de los fármacos , Células Madre/efectos de los fármacos , Células Madre/metabolismo , Análisis de Supervivencia , Proteínas Wnt/administración & dosificación , Proteínas Wnt/farmacología , Vía de Señalización Wnt/efectos de los fármacos , Vía de Señalización Wnt/genética
11.
Mol Nutr Food Res ; 63(3): e1800658, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30407734

RESUMEN

SCOPE: Necrotizing enterocolitis (NEC) is a leading cause of morbidity and death in preterm infants, occurring more often in formula-fed than breastfed infants. Studies in both rats and humans show that human milk oligosaccharides (HMOs) lower the incidence of NEC, but the mechanism underlying such protection is currently unclear. METHODS AND RESULTS: By extracting HMOs from pooled human breastmilk, the impact of HMOs on the intestinal mucin levels in a murine model of NEC are investigated. To confirm the results, the findings are validated by exposing human intestinal epithelial cells and intestinal organoids to HMOs and evaluated for mucin expression. HMO-gavage to pups increases Muc2 levels and decreases intestinal permeability to macromolecular dextran. HMO-treated cells have increased Muc2 expression, decreased bacterial attachment and dextran permeability during challenge by enteric pathogens. To identify the mediators involved in HMO induction of mucins, it is demonstrated that HMOs directly induce the expression of chaperone proteins including protein disulfide isomerase (PDI). Suppression of PDI activity removes the protective effects of HMOs on barrier function in vitro as well as NEC protection in vivo. CONCLUSIONS: Taken together, the results provide insights to the possible mechanisms by which HMOs protect the neonatal intestine through upregulation of mucins.


Asunto(s)
Enterocolitis Necrotizante/prevención & control , Leche Humana/química , Mucina 2/genética , Oligosacáridos/farmacología , Animales , Animales Recién Nacidos , Células CACO-2 , Estrés del Retículo Endoplásmico/efectos de los fármacos , Enterocolitis Necrotizante/metabolismo , Células Caliciformes/efectos de los fármacos , Humanos , Mucosa Intestinal/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Mucina 2/análisis , Proteína Disulfuro Isomerasas/fisiología
12.
Am J Physiol Gastrointest Liver Physiol ; 315(5): G788-G798, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-30095298

RESUMEN

Flaxseed is high in ω-3 polyunsaturated fatty acids, fiber, and lignans known to lower cholesterol levels. However, its use for prevention or treatment of inflammatory bowel diseases has yielded mixed results, perhaps related to dietary interactions. In this study, we evaluated the impact of ground flaxseed supplementation on the severity of Citrobacter rodentium-induced colitis in the setting of either a high-fat (HF, ~36%kcal) or reduced-fat (RF, ~12%kcal) diet. After weaning, C57BL/6 mice ( n = 8-15/treatment) were fed ground flaxseed (7 g/100 g diet) with either HF (HF Flx) or RF (RF Flx) diets for 4 wk before infection with C. rodentium or sham gavage. Weight changes, mucosal inflammation, pathogen burden, gut microbiota composition, tissue polyunsaturated fatty acids, and cecal short-chain fatty acids were compared over a 14-day infection period. The RF diet protected against C. rodentium-induced colitis, whereas the RF Flx diet increased pathogen burden, exacerbated gut inflammation, and promoted gut dysbiosis. When compared with the RF diet, both HF and HF Flx diets resulted in more severe pathology in response to C. rodentium infection. Our findings demonstrate that although an RF diet protected against C. rodentium-induced colitis and associated gut dysbiosis in mice, beneficial effects were diminished with ground flaxseed supplementation. NEW & NOTEWORTHY Our results demonstrate a strong protective effect of a reduced-fat diet against intestinal inflammation, dysbiosis, and pathogen burden during Citrobacter rodentium-induced colitis. However, ground flaxseed supplementation in the setting of a reduced-fat diet exacerbated colitis despite higher levels of intestinal n-3 polyunsaturated fatty acids and cecal short-chain fatty acids.


Asunto(s)
Colitis Ulcerosa/dietoterapia , Dieta con Restricción de Grasas , Infecciones por Enterobacteriaceae/dietoterapia , Ácidos Grasos Insaturados/efectos adversos , Lino/química , Animales , Citrobacter rodentium/efectos de los fármacos , Colitis Ulcerosa/microbiología , Infecciones por Enterobacteriaceae/microbiología , Ácidos Grasos Insaturados/farmacología , Microbioma Gastrointestinal/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL
13.
Microbiome ; 5(1): 135, 2017 10 10.
Artículo en Inglés | MEDLINE | ID: mdl-29017607

RESUMEN

BACKGROUND: Prebiotics are non-digestible food ingredients that enhance the growth of certain microbes within the gut microbiota. Prebiotic consumption generates immune-modulatory effects that are traditionally thought to reflect microbial interactions within the gut. However, recent evidence suggests they may also impart direct microbe-independent effects on the host, though the mechanisms of which are currently unclear. METHODS: Kinome arrays were used to profile the host intestinal signaling responses to prebiotic exposures in the absence of microbes. Identified pathways were functionally validated in Caco-2Bbe1 intestinal cell line and in vivo model of murine endotoxemia. RESULTS: We found that prebiotics directly regulate host mucosal signaling to alter response to bacterial infection. Intestinal epithelial cells (IECs) exposed to prebiotics are hyporesponsive to pathogen-induced mitogen-activated protein kinase (MAPK) and nuclear factor kappa B (NF-κB) activations, and have a kinome profile distinct from non-treated cells pertaining to multiple innate immune signaling pathways. Consistent with this finding, mice orally gavaged with prebiotics showed dampened inflammatory response to lipopolysaccharide (LPS) without alterations in the gut microbiota. CONCLUSIONS: These findings provide molecular mechanisms of direct host-prebiotic interactions to support prebiotics as potent modulators of host inflammation.


Asunto(s)
Microbioma Gastrointestinal , Inflamación , Oligosacáridos/metabolismo , Prebióticos , Proteínas Quinasas/inmunología , Animales , Células CACO-2 , Endotoxemia , Microbioma Gastrointestinal/inmunología , Microbioma Gastrointestinal/fisiología , Humanos , Inmunidad Innata , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiología , Lipopolisacáridos/inmunología , Ratones , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Oligosacáridos/química , Oligosacáridos/genética , Oligosacáridos/farmacología , Análisis por Matrices de Proteínas , Proteínas Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Proteómica , Transducción de Señal/efectos de los fármacos , Quinasa de Factor Nuclear kappa B
14.
Sci Rep ; 7: 40820, 2017 01 18.
Artículo en Inglés | MEDLINE | ID: mdl-28098206

RESUMEN

Prebiotics are non-digestible oligosaccharides that promote the growth of beneficial gut microbes, but it is unclear whether they also have direct effects on the intestinal mucosal barrier. Here we demonstrate two commercial prebiotics, inulin and short-chain fructo-oligosaccharide (scFOS), when applied onto intestinal epithelia in the absence of microbes, directly promote barrier integrity to prevent pathogen-induced barrier disruptions. We further show that these effects involve the induction of select tight junction (TJ) proteins through a protein kinase C (PKC) δ-dependent mechanism. These results suggest that in the absence of microbiota, prebiotics can directly exert barrier protective effects by activating host cell signaling in the intestinal epithelium, which represents a novel alternative mechanism of action of prebiotics.


Asunto(s)
Mucosa Intestinal/metabolismo , Prebióticos , Proteína Quinasa C-delta/metabolismo , Células CACO-2 , Células Cultivadas , Suplementos Dietéticos , Humanos , Mucosa Intestinal/citología , Mucosa Intestinal/microbiología , Inulina/farmacología , Microbiota , Oligosacáridos/farmacología , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Proteína Quinasa C-delta/antagonistas & inhibidores , Proteína Quinasa C-delta/genética , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Transducción de Señal/efectos de los fármacos , Proteínas de Uniones Estrechas/genética , Proteínas de Uniones Estrechas/metabolismo , Receptores Toll-Like/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Proteína de la Zonula Occludens-1/genética , Proteína de la Zonula Occludens-1/metabolismo
15.
Adv Nutr ; 7(5): 928-37, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27633108

RESUMEN

Necrotizing enterocolitis (NEC) is a devastating intestinal disease in preterm infants characterized by barrier disruption, intestinal microbial dysbiosis, and persistent inflammation of the colon, which results in high mortality rates. Current strategies used to manage this disease are not sufficient, although the use of human breast milk reduces the risk of NEC. Mother's milk is regarded as a fundamental nutritional source for neonates, but pasteurization of donor breast milk affects the composition of bioactive compounds. Current research is evaluating the benefits and potential pitfalls of adding probiotics and prebiotics to pasteurized milk so as to improve the functionality of the milk and thereby reduce the burden of illness caused by NEC. Probiotics (live micro-organisms that confer health to the host) and prebiotics (nondigestible oligosaccharides that stimulate the growth of healthy bacteria) are functional foods known to mediate immune responses and modulate microbial populations in the gut. Clinical research shows strain- and compound-specific responses when probiotics or prebiotics are administered in conjunction with donor breast milk for the prevention of NEC. Despite ongoing controversy surrounding optimal treatment strategies, randomized controlled studies are now investigating the use of synbiotics to reduce the incidence and severity of NEC. Synbiotics, a combination of probiotics and prebiotics, have been proposed to enhance beneficial health effects in the intestinal tract more than either agent administered alone. This review considers the implications of using probiotic-, prebiotic-, and synbiotic-supplemented breast milk as a strategy to prevent NEC and issues that could be encountered with the preparations.


Asunto(s)
Enterocolitis Necrotizante/prevención & control , Leche Humana , Prebióticos , Probióticos , Simbióticos , Bacterias , Humanos , Leche Humana/química , Leche Humana/microbiología , Neonatología , Oligosacáridos
16.
Diabetes ; 64(7): 2537-49, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25735732

RESUMEN

Obesity and diabetes are characterized by increased inflammation reflecting disordered control of innate immunity. We reveal a local intestinal intraepithelial lymphocyte (IEL)-GLP-1 receptor (GLP-1R) signaling network that controls mucosal immune responses. Glp1r expression was enriched in intestinal IEL preparations and copurified with markers of Tαß and Tγδ IELs, the two main subsets of intestinal IELs. Exendin-4 increased cAMP accumulation in purified IELs and reduced the production of cytokines from activated IELs but not from splenocytes ex vivo. These actions were mimicked by forskolin, absent in IELs from Glp1r(-/-) mice, and attenuated by the GLP-1R agonist exendin (9-39) consistent with a GLP-1R-dependent mechanism of action. Furthermore, Glp1r(-/-) mice exhibited dysregulated intestinal gene expression, an abnormal representation of microbial species in feces, and enhanced sensitivity to intestinal injury following administration of dextran sodium sulfate. Bone marrow transplantation using wild-type C57BL/6 donors normalized expression of multiple genes regulating immune function and epithelial integrity in Glp1r(-/-) recipient mice, whereas acute exendin-4 administration robustly induced the expression of genes encoding cytokines and chemokines in normal and injured intestine. Taken together, these findings define a local enteroendocrine-IEL axis linking energy availability, host microbial responses, and mucosal integrity to the control of innate immunity.


Asunto(s)
Mucosa Intestinal/inmunología , Linfocitos/inmunología , Receptores de Glucagón/agonistas , Receptores de Glucagón/fisiología , Animales , Citocinas/análisis , Exenatida , Femenino , Péptido 1 Similar al Glucagón/metabolismo , Péptido 1 Similar al Glucagón/farmacología , Receptor del Péptido 1 Similar al Glucagón , Inmunidad Innata , Masculino , Ratones , Ratones Endogámicos C57BL , Péptidos/farmacología , Transducción de Señal , Ponzoñas/farmacología
17.
Shock ; 43(5): 483-9, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25608140

RESUMEN

Intestinal epithelia regulate barrier integrity when challenged by inflammation, oxidative stress, and microbes. Transforming growth factor-ß1 (TGF-ß1) is a cytokine with known beneficial effects on intestinal epithelia, including barrier enhancement, after exposure to proinflammatory cytokines and infectious agents. The aim of this study was to determine whether TGF-ß1 directly protects intestinal epithelia during hypoxia-reoxygenation (HR). Intestinal epithelial monolayers (T84, Caco-2) were exposed to either hypoxia (1% O2, 1 h) or oxidative stress (hydrogen peroxide, 1 mM), followed by normoxic atmosphere for different time points in the absence and presence of varying concentrations of TGF-ß1. Transepithelial electrical resistance (TER) assessed barrier function, with RNA extracted for reverse transcription polymerase chain reaction analysis of GPx-1, HIF-1, heme-oxygenase-1 (HO-1), and NOX-1. In some experiments, intestinal epithelia were exposed to enterohemorrhagic Escherichia coli (EHEC) O157:H7 during the reoxygenation period and TER recorded 7 h after the infectious challenge. Hypoxia-reoxygenation significantly decreased TER in intestinal epithelia compared with normoxic controls. Transforming growth factor-ß1 pretreatment ameliorated HR-induced epithelial barrier dysfunction in T84 (at 1 - 3 h) and Caco-2 (1 h) monolayers. Transforming growth factor-ß1 preserved barrier integrity for up to 16 h after challenge with hydrogen peroxide. In TGF-ß1-treated epithelial monolayers, only HO-1 mRNA significantly increased after HR (P < 0.05 vs. normoxic controls). The EHEC-induced epithelial barrier dysfunction was significantly worsened by intestinal HR (P < 0.05 vs. normoxia-EHEC-infected cells), but this was not protected by TGF-ß1 pretreatment. Transforming growth factor-ß1 preserves loss of epithelial barrier integrity caused by the stress of HR via a mechanism that may involve the upregulation of HO-1 transcription. Targeted treatment with TGF-ß could lead to novel therapies in enteric diseases characterized by HR injury.


Asunto(s)
Mucosa Intestinal/efectos de los fármacos , Estrés Oxidativo , Factor de Crecimiento Transformador beta1/farmacología , Células CACO-2 , Hipoxia de la Célula , Citocinas/metabolismo , Impedancia Eléctrica , Escherichia coli Enterohemorrágica/patogenicidad , Células Epiteliales/efectos de los fármacos , Epitelio/metabolismo , Expresión Génica , Hemo-Oxigenasa 1/metabolismo , Humanos , Peróxido de Hidrógeno/química , Enfermedades Intestinales/metabolismo , Mucosa Intestinal/patología , Oxígeno/química , ARN Mensajero/metabolismo , Proteínas Recombinantes/farmacología , Factores de Tiempo
18.
Inflamm Bowel Dis ; 21(2): 297-306, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25590952

RESUMEN

BACKGROUND: Adherent-invasive Escherichia coli (AIEC) colonization has been strongly implicated in the pathogenesis of Crohn's disease. Environmental triggers such as vitamin D deficiency have emerged as key factors in the pathogenesis of inflammatory bowel diseases. The aim of this study was to investigate the effects of 1,25(OH)2D3 on AIEC infection-induced changes in vivo and in vitro. METHODS: Barrier function was assessed in polarized epithelial Caco-2-bbe cells grown in medium with or without vitamin D and challenged with AIEC strain LF82. Weaned C57BL/6 mice were fed either a vitamin D-sufficient or -deficient diet for 5 weeks and then infected with AIEC, in the absence and presence of low-dose dextran sodium sulphate. Disease severity was assessed by histological analysis and in vivo intestinal permeability assay. Presence of invasive bacteria was assessed by transmission electron microscopy. RESULTS: Caco-2-bbe cells incubated with 1,25(OH)2D3 were protected against AIEC-induced disruption of transepithelial electrical resistance and tight-junction protein redistribution. Vitamin D-deficient C57BL/6 mice given a course of 2% dextran sodium sulphate exhibited pronounced epithelial barrier dysfunction, were more susceptible to AIEC colonization, and showed exacerbated colonic injury. Transmission electron microscopy of colonic tissue from infected mice demonstrated invasion of AIEC and fecal microbiome analysis revealed shifts in microbial communities. CONCLUSIONS: These data show that vitamin D is able to mitigate the deleterious effects of AIEC on the intestinal mucosa, by maintaining intestinal epithelial barrier homeostasis and preserving tight-junction architecture. This study highlights the association between vitamin D status, dysbiosis, and Crohn's disease.


Asunto(s)
Calcitriol/farmacología , Colitis/etiología , Enfermedad de Crohn/etiología , Modelos Animales de Enfermedad , Infecciones por Escherichia coli/etiología , Escherichia coli/patogenicidad , Mucosa Intestinal/patología , Deficiencia de Vitamina D/fisiopatología , Animales , Apoptosis , Adhesión Bacteriana/efectos de los fármacos , Western Blotting , Células CACO-2 , Permeabilidad de la Membrana Celular/efectos de los fármacos , Proliferación Celular , Células Cultivadas , Colitis/metabolismo , Colitis/patología , Enfermedad de Crohn/metabolismo , Enfermedad de Crohn/patología , Sulfato de Dextran/farmacología , Conductividad Eléctrica , Infecciones por Escherichia coli/metabolismo , Infecciones por Escherichia coli/patología , Femenino , Técnica del Anticuerpo Fluorescente , Tracto Gastrointestinal/metabolismo , Tracto Gastrointestinal/microbiología , Tracto Gastrointestinal/patología , Humanos , Técnicas para Inmunoenzimas , Inflamación/etiología , Inflamación/metabolismo , Inflamación/patología , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiología , Ratones , Ratones Endogámicos C57BL , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Deficiencia de Vitamina D/complicaciones
19.
J Nutr ; 144(11): 1725-33, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25143376

RESUMEN

BACKGROUND: Few studies have focused on the ability of prebiotics to prevent pathogen-induced cellular changes or alter the composition of the intestinal microbiota in complimentary relevant cell and animal models of inflammatory bowel disease. OBJECTIVE: The objective of this study was to determine if pretreatment with inulin and a short-chain fructo-oligosaccharide (sc-FOS) prevents enterohemorrhagic Escherichia coli (EHEC) O157:H7 infection in Caco2-bbe epithelial cells and what effect 10% wt:v sc-FOS or inulin has on C57BL/6 mice under sham conditions or pretreatment with prebiotics before Citrobacter rodentium infection (10(8) colony-forming units). METHODS: Actin rearrangement and tight junction protein (zona occludin-1) were examined with immunofluorescence. Barrier function was assessed by a fluorescent probe and by measuring transepithelial electrical resistance (TER). Alterations in cytokine gene expression and microbiome were assessed with quantitative reverse transcriptase-polymerase chain reaction and fluorescence in situ hybridization. Short-chain fatty acids (SCFAs) were measured by GC. RESULTS: sc-FOS added to monolayers altered actin polymerization without affecting TER or permeability to a fluorescein isothiocyanate (FITC) probe, whereas inulin increased TER (P < 0.005) and altered actin arrangement without affecting FITC permeability. Neither prebiotic attenuated EHEC-induced decreases in barrier function. Prebiotics increased interleukin 10 (Il10) and transforming growth factor-ß (Tgfß) cytokine responses alone (P < 0.05) or with EHEC O157:H7 infection (P < 0.05) in vitro. Increases in tumor necrosis factor-α (Tnfα) (P < 0.05) and decreases in chemokine CXC motif ligand 8 (Cxcl8) (P < 0.05) expression were observed with prebiotic treatment prior to EHEC infection. No differences were noted in barrier function or cytokine responses in the absence or presence of C. rodentium in vivo. Alterations in microbiome were evident at 6 d and 10 d postinfection in treatment groups, but a change in C. rodentium load was not observed. Inulin and sc-FOS (P < 0.05) increased fecal SCFAs in the absence of infection. CONCLUSION: This study provides new insights as to how prebiotics act in complementary in vitro and in vivo models of intestinal injury.


Asunto(s)
Infecciones por Enterobacteriaceae/complicaciones , Escherichia coli O157 , Inflamación/tratamiento farmacológico , Inulina/farmacología , Oligosacáridos/farmacología , Animales , Antiinflamatorios/química , Antiinflamatorios/farmacología , Antiinflamatorios/uso terapéutico , Células CACO-2 , Citrobacter rodentium , Colitis/tratamiento farmacológico , Colitis/microbiología , Heces/microbiología , Femenino , Humanos , Inulina/química , Ratones , Ratones Endogámicos C57BL , Oligosacáridos/química , Prebióticos
20.
J Infect Dis ; 210(8): 1296-305, 2014 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-24755435

RESUMEN

BACKGROUND: Vitamin D, an important modulator of the immune system, has been shown to protect mucosal barrier homeostasis. This study investigates the effects of vitamin D deficiency on infection-induced changes in intestinal epithelial barrier function in vitro and on Citrobacter rodentium-induced colitis in mice. METHODS: Polarized epithelial Caco2-bbe cells were grown in medium with or without vitamin D and challenged with enterohemorrhagic Escherichia coli O157:H7. Barrier function and tight junction protein expression were assessed. Weaned C57BL/6 mice were fed either a vitamin D-sufficient or vitamin D-deficient diet and then infected with C. rodentium. Disease severity was assessed by histological analysis, intestinal permeability assay, measurement of inflammatory cytokine levels, and microbiome analysis. RESULTS: 1,25(OH)2D3 altered E. coli O157:H7-induced reductions in transepithelial electrical resistance (P < .01), decreased permeability (P < .05), and preserved barrier integrity. Vitamin D-deficient mice challenged with C. rodentium demonstrated increased colonic hyperplasia and epithelial barrier dysfunction (P < .0001 and P < .05, respectively). Vitamin D deficiency resulted in an altered composition of the fecal microbiome both in the absence and presence of C. rodentium infection. CONCLUSIONS: This study demonstrates that vitamin D is an important mediator of intestinal epithelial defenses against infectious agents. Vitamin D deficiency predisposes to more-severe intestinal injury in an infectious model of colitis.


Asunto(s)
Calcitriol/farmacología , Inflamación/metabolismo , Enfermedades Intestinales/metabolismo , Mucosa Intestinal/fisiopatología , Deficiencia de Vitamina D/patología , Animales , Células CACO-2 , Citrobacter rodentium , Colitis/etiología , Infecciones por Enterobacteriaceae/microbiología , Escherichia coli Enterohemorrágica , Infecciones por Escherichia coli , Heces/microbiología , Humanos , Mucosa Intestinal/patología , Ratones , Ratones Endogámicos C57BL , Deficiencia de Vitamina D/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...