Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 88
Filtrar
1.
Nature ; 579(7800): 575-580, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32050257

RESUMEN

The intestinal mucosa serves both as a conduit for the uptake of food-derived nutrients and microbiome-derived metabolites, and as a barrier that prevents tissue invasion by microorganisms and tempers inflammatory responses to the myriad contents of the lumen. How the intestine coordinates physiological and immune responses to food consumption to optimize nutrient uptake while maintaining barrier functions remains unclear. Here we show in mice how a gut neuronal signal triggered by food intake is integrated with intestinal antimicrobial and metabolic responses that are controlled by type-3 innate lymphoid cells (ILC3)1-3. Food consumption rapidly activates a population of enteric neurons that express vasoactive intestinal peptide (VIP)4. Projections of VIP-producing neurons (VIPergic neurons) in the lamina propria are in close proximity to clusters of ILC3 that selectively express VIP receptor type 2 (VIPR2; also known as VPAC2). Production of interleukin (IL)-22 by ILC3, which is upregulated by the presence of commensal microorganisms such as segmented filamentous bacteria5-7, is inhibited upon engagement of VIPR2. As a consequence, levels of antimicrobial peptide derived from epithelial cells are reduced but the expression of lipid-binding proteins and transporters is increased8. During food consumption, the activation of VIPergic neurons thus enhances the growth of segmented filamentous bacteria associated with the epithelium, and increases lipid absorption. Our results reveal a feeding- and circadian-regulated dynamic neuroimmune circuit in the intestine that promotes a trade-off between innate immune protection mediated by IL-22 and the efficiency of nutrient absorption. Modulation of this pathway may therefore be effective for enhancing resistance to enteropathogens2,3,9 and for the treatment of metabolic diseases.


Asunto(s)
Ingestión de Alimentos/fisiología , Inmunidad Innata/inmunología , Absorción Intestinal/fisiología , Intestinos/inmunología , Intestinos/fisiología , Linfocitos/inmunología , Neuronas/metabolismo , Péptido Intestinal Vasoactivo/metabolismo , Animales , Ritmo Circadiano/fisiología , Ingestión de Alimentos/inmunología , Femenino , Interleucinas/biosíntesis , Interleucinas/inmunología , Absorción Intestinal/inmunología , Intestinos/citología , Intestinos/microbiología , Linfocitos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Periodo Posprandial/fisiología , Receptores CCR6/metabolismo , Receptores de Tipo II del Péptido Intestinal Vasoactivo/metabolismo , Simbiosis , Interleucina-22
2.
Am J Gastroenterol ; 114(10): 1587-1592, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31274511

RESUMEN

Celiac disease is a common inflammatory disease triggered by dietary gluten in genetically susceptible individuals. The strongest and best-characterized genetic susceptibilities in celiac disease are class II human leukocyte antigen (HLA) genes known as HLA-DQ2 and DQ8. HLA genetic testing is available through a number of commercial and academic laboratories and is used in the evaluation of celiac disease and to identify at-risk family members. Importantly, HLA genetic testing has a high negative predictive value for celiac disease, but a low positive predictive value. Therefore, for a practicing clinician, it is important to understand when to order HLA genetic testing, what test to order, and how to interpret the result. This review provides a practical primer on HLA genetics in celiac disease.


Asunto(s)
Enfermedad Celíaca/diagnóstico , Pruebas Genéticas/normas , Antígenos HLA-DQ/genética , Guías de Práctica Clínica como Asunto , Biomarcadores/sangre , Biopsia , Enfermedad Celíaca/sangre , Enfermedad Celíaca/genética , Enfermedad Celíaca/inmunología , Duodeno/inmunología , Duodeno/metabolismo , Duodeno/patología , Gastroenterología/normas , Predisposición Genética a la Enfermedad , Glútenes/inmunología , Glútenes/metabolismo , Antígenos HLA-DQ/inmunología , Humanos , Absorción Intestinal/genética , Absorción Intestinal/inmunología , Mucosa Intestinal/inmunología , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Valor Predictivo de las Pruebas
3.
Am J Physiol Cell Physiol ; 312(4): C438-C445, 2017 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-28249988

RESUMEN

Association between circulating lipopolysaccharide (LPS) and metabolic diseases (such as type 2 diabetes and atherosclerosis) has shifted the focus from high-fat high-cholesterol containing Western-type diet (WD)-induced changes in gut microbiota per se to release of gut bacteria-derived products (e.g., LPS) into circulation due to intestinal barrier dysfunction as the possible mechanism for the chronic inflammatory state underlying the development of these diseases. We demonstrated earlier that oral supplementation with curcumin attenuates WD-induced development of type 2 diabetes and atherosclerosis. Poor bioavailability of curcumin has precluded the establishment of a causal relationship between oral supplementation and it is in vivo effects. We hypothesized that curcumin attenuates WD-induced chronic inflammation and associated metabolic diseases by modulating the function of intestinal epithelial cells (IECs) and the intestinal barrier function. The objective of the present study was to delineate the underlying mechanisms. The human IEC lines Caco-2 and HT-29 were used for these studies and modulation of direct as well as indirect effects of LPS on intracellular signaling as well as tight junctions were examined. Pretreatment with curcumin significantly attenuated LPS-induced secretion of master cytokine IL-1ß from IECs and macrophages. Furthermore, curcumin also reduced IL-1ß-induced activation of p38 MAPK in IECs and subsequent increase in expression of myosin light chain kinase involved in the phosphorylation of tight junction proteins and ensuing disruption of their normal arrangement. The major site of action of curcumin is, therefore, likely the IECs and the intestinal barrier, and by reducing intestinal barrier dysfunction, curcumin modulates chronic inflammatory diseases despite poor bioavailability.


Asunto(s)
Comunicación Celular/inmunología , Colitis/inmunología , Curcumina/administración & dosificación , Absorción Intestinal/inmunología , Mucosa Intestinal/inmunología , Uniones Estrechas/inmunología , Células CACO-2 , Comunicación Celular/efectos de los fármacos , Colitis/inducido químicamente , Colitis/tratamiento farmacológico , Colon/efectos de los fármacos , Colon/inmunología , Relación Dosis-Respuesta a Droga , Células Epiteliales , Células HT29 , Humanos , Absorción Intestinal/efectos de los fármacos , Mucosa Intestinal/efectos de los fármacos , Lipopolisacáridos , Uniones Estrechas/efectos de los fármacos
4.
Gastroenterology ; 151(5): 923-932, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27436071

RESUMEN

BACKGROUND & AIMS: The gut microbiota affects intestinal permeability and mucosal mast cells (MMCs) responses. Activation of MMCs has been associated with absorption of dietary fat. We investigated whether the gut microbiota contributes to the fat-induced activation of MMCs in rats, and how antibiotics might affect this process. METHODS: Adult male Sprague-Dawley rats were given streptomycin and penicillin for 4 days (n = 6-8) to reduce the abundance of their gut flora, or normal drinking water (controls, n = 6-8). They underwent lymph fistula surgery and after an overnight recovery were given an intraduodenal bolus of intralipid. We collected intestinal tissues and lymph fluid and assessed activation of MMCs, intestinal permeability, and fat transport parameters. RESULTS: Compared with controls, intestinal lymph from rats given antibiotics had reduced levels of mucosal mast cell protease II (produced by MMCs) and decreased activity of diamine oxidase (produced by enterocytes) (P < .05). Rats given antibiotics had reduced intestinal permeability in response to dietary lipid compared with controls (P < .01). Unexpectedly, antibiotics also reduced lymphatic transport of triacylglycerol and phospholipid (P < .01), concomitant with decreased levels of mucosal apolipoproteins B, A-I, and A-IV (P < .01). No differences were found in intestinal motility or luminal pancreatic lipase activity between rats given antibiotics and controls. These effects were not seen with an acute dose of antibiotics or 4 weeks after the antibiotic regimen ended. CONCLUSIONS: The intestinal microbiota appears to activate MMCs after the ingestion of fat in rats; this contributes to fat-induced intestinal permeability. We found that the gut microbiome promotes absorption of lipid, probably by intestinal production of apolipoproteins and secretion of chylomicrons.


Asunto(s)
Antibacterianos/farmacología , Grasas de la Dieta/metabolismo , Absorción Intestinal/efectos de los fármacos , Mucosa Intestinal/efectos de los fármacos , Mastocitos/efectos de los fármacos , Penicilinas/farmacología , Estreptomicina/farmacología , Animales , Antibacterianos/administración & dosificación , Microbioma Gastrointestinal/efectos de los fármacos , Microbioma Gastrointestinal/fisiología , Absorción Intestinal/inmunología , Mucosa Intestinal/inmunología , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiología , Masculino , Mastocitos/metabolismo , Mastocitos/microbiología , Penicilinas/administración & dosificación , Permeabilidad , Ratas , Ratas Sprague-Dawley , Estreptomicina/administración & dosificación
5.
Biochem Biophys Res Commun ; 473(4): 1090-1093, 2016 05 13.
Artículo en Inglés | MEDLINE | ID: mdl-27060548

RESUMEN

A lectin like protein designated as LSMT is recently discovered in Agaricus bisporus. The protein adopts very similar structure to Ricin-B like lectin from Clitocybe nebularis (CNL) and HA-33 from Clostridium botulinum (HA-33), which both recognize sugar molecules that decorate the surface of the epithelial cells of the intestine. A preliminary study in silico pointed out potential capability of LSMT to perform such biological activity. Following that hypothesis, we demonstrated that LSMT is indeed capable of penetrating out from a dialysis tube of the mice intestine origin. Furthermore, the protein appeared not to evoke the immune response upon introduction into mice, unlike its structural homologs. This is the first report on the biological implication of LSMT that might lead to its application.


Asunto(s)
Tolerancia Inmunológica/inmunología , Absorción Intestinal/inmunología , Lectinas/química , Lectinas/inmunología , Modelos Inmunológicos , Animales , Simulación por Computador , Femenino , Lectinas/clasificación , Ratones , Modelos Químicos , Permeabilidad , Análisis de Secuencia de Proteína , Homología de Secuencia de Aminoácido
6.
Lipids Health Dis ; 15: 108, 2016 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-27317359

RESUMEN

Obesity and its associated complications like type 2 diabetes (T2D) are reaching epidemic stages. Increased food intake and lack of exercise are two main contributing factors. Recent work has been highlighting an increasingly more important role of gut microbiota in metabolic disorders. It's well known that gut microbiota plays a major role in the development of food absorption and low grade inflammation, two key processes in obesity and diabetes. This review summarizes key discoveries during the past decade that established the role of gut microbiota in the development of obesity and diabetes. It will look at the role of key metabolites mainly the short chain fatty acids (SCFA) that are produced by gut microbiota and how they impact key metabolic pathways such as insulin signalling, incretin production as well as inflammation. It will further look at the possible ways to harness the beneficial aspects of the gut microbiota to combat these metabolic disorders and reduce their impact.


Asunto(s)
Diabetes Mellitus Tipo 2/microbiología , Microbioma Gastrointestinal/inmunología , Tracto Gastrointestinal/microbiología , Metabolismo de los Lípidos/inmunología , Obesidad/microbiología , Diabetes Mellitus Tipo 2/complicaciones , Diabetes Mellitus Tipo 2/inmunología , Diabetes Mellitus Tipo 2/patología , Ácidos Grasos Volátiles/biosíntesis , Ácidos Grasos Volátiles/metabolismo , Tracto Gastrointestinal/inmunología , Tracto Gastrointestinal/metabolismo , Tracto Gastrointestinal/patología , Regulación de la Expresión Génica , Humanos , Incretinas/genética , Incretinas/metabolismo , Inflamación , Insulina/genética , Insulina/metabolismo , Absorción Intestinal/inmunología , Obesidad/complicaciones , Obesidad/inmunología , Obesidad/patología , Transducción de Señal
7.
Curr Oncol Rep ; 16(10): 406, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25123079

RESUMEN

Humans depend on our commensal bacteria for nutritive, immune-modulating, and metabolic contributions to maintenance of health. However, this commensal community exists in careful balance that, if disrupted, enters dysbiosis; this has been shown to contribute to the pathogenesis of colon, gastric, esophageal, pancreatic, laryngeal, breast, and gallbladder carcinomas. This development is closely tied to host inflammation, which causes and is aggravated by microbial dysbiosis and increases vulnerability to pathogens. Advances in sequencing technology have increased our ability to catalog microbial species associated with various cancer types across the body. However, defining microbial biomarkers as cancer predictors presents multiple challenges, and existing studies identifying cancer-associated bacteria have reported inconsistent outcomes. Combining metabolites and microbiome analyses can help elucidate interactions between gut microbiota, metabolism, and the host. Ultimately, understanding how gut dysbiosis impacts host response and inflammation will be critical to creating an accurate picture of the role of the microbiome in cancer.


Asunto(s)
Neoplasias Colorrectales/microbiología , Neoplasias Colorrectales/patología , Disbiosis/patología , Infecciones por Bacterias Gramnegativas/patología , Inmunidad Adaptativa/inmunología , Carga Bacteriana , Neoplasias Colorrectales/inmunología , Progresión de la Enfermedad , Disbiosis/inmunología , Disbiosis/microbiología , Regulación Bacteriana de la Expresión Génica , Infecciones por Bacterias Gramnegativas/inmunología , Interacciones Huésped-Patógeno/inmunología , Humanos , Inmunidad Innata , Absorción Intestinal/inmunología , Microbiota/inmunología , Estadificación de Neoplasias
8.
J Allergy Clin Immunol ; 131(2): 442-50, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23182172

RESUMEN

BACKGROUND: The prevalence of peanut allergies is increasing. Peanuts and many other allergen sources contain significant amounts of triglycerides, which affect absorption of antigens but have unknown effects on sensitization and anaphylaxis. We recently reported that dietary medium-chain triglycerides (MCTs), which bypass mesenteric lymph and directly enter portal blood, reduce intestinal antigen absorption into blood compared with long-chain triglycerides (LCTs), which stimulate mesenteric lymph flow and are absorbed in chylomicrons through mesenteric lymph. OBJECTIVE: We sought to test how dietary MCTs affect food allergy. METHODS: C3H/HeJ mice were fed peanut butter protein in MCT, LCT (peanut oil), or LCT plus an inhibitor of chylomicron formation (Pluronic L81). Peanut-specific antibodies in plasma, responses of the mice to antigen challenges, and intestinal epithelial cytokine expression were subsequently measured. RESULTS: MCT suppressed antigen absorption into blood but stimulated absorption into Peyer patches. A single gavage of peanut protein with MCT, as well as prolonged feeding in MCT-based diets, caused spontaneous allergic sensitization. MCT-sensitized mice experienced IgG-dependent anaphylaxis on systemic challenge and IgE-dependent anaphylaxis on oral challenge. MCT feeding stimulated jejunal-epithelial thymic stromal lymphopoietin, Il25, and Il33 expression compared with that seen after LCT feeding and promoted T(H)2 cytokine responses in splenocytes. Moreover, oral challenges of sensitized mice with antigen in MCT significantly aggravated anaphylaxis compared with challenges with the LCT. Importantly, the effects of MCTs could be mimicked by adding Pluronic L81 to LCTs, and in vitro assays indicated that chylomicrons prevent basophil activation. CONCLUSION: Dietary MCTs promote allergic sensitization and anaphylaxis by affecting antigen absorption and availability and by stimulating T(H)2 responses.


Asunto(s)
Anafilaxia/inmunología , Arachis/inmunología , Hipersensibilidad al Cacahuete/inmunología , Triglicéridos/inmunología , Alérgenos/inmunología , Anafilaxia/metabolismo , Animales , Anticuerpos/inmunología , Antígenos/inmunología , Arachis/química , Basófilos/inmunología , Basófilos/metabolismo , Quilomicrones/inmunología , Citocinas/inmunología , Dieta/métodos , Femenino , Inmunoglobulina E/inmunología , Inmunoglobulina G/inmunología , Interleucinas/inmunología , Absorción Intestinal/inmunología , Mucosa Intestinal/inmunología , Mucosa Intestinal/metabolismo , Yeyuno/inmunología , Yeyuno/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C3H , Ratas , Ratas Sprague-Dawley , Células Th2/inmunología , Células Th2/metabolismo , Triglicéridos/administración & dosificación , Triglicéridos/metabolismo
9.
Internist (Berl) ; 55(8): 883-8, 2014 Aug.
Artículo en Alemán | MEDLINE | ID: mdl-24831679

RESUMEN

The challenge of the mucosal immune system is to develop tolerance toward intestinal antigens. Considering the quantity of bacteria that continuously attack the intestinal barrier, one can only imagine the complexity involved. To master this task, a tight network between the intestinal microbiota, the barrier, immune cells of the lamina propria as well as the adjacent mesenteric fat tissue is required. The key pathways involved have been revealed by the genome-wide association studies as well as functional data from experimental models. However, although knowledge with regard to the pathogenesis of chronic inflammatory bowel disease has been increasing continuously over recent decades, the current therapeutic strategies are limited to controlling the pro-inflammatory effector phase rather than achieving cure. The best example is cytokine neutralizing antibodies. The present review aims to describe the role of the various cell populations within the intestinal wall for disease pathogenesis and, thus, identify possible therapeutic strategies.


Asunto(s)
Citocinas/inmunología , Enfermedades Inflamatorias del Intestino/inmunología , Enfermedades Inflamatorias del Intestino/microbiología , Mucosa Intestinal/inmunología , Mucosa Intestinal/microbiología , Intestinos/inmunología , Microbiota/inmunología , Enfermedad Crónica , Humanos , Absorción Intestinal/inmunología , Modelos Inmunológicos
10.
Clin Exp Allergy ; 42(12): 1791-800, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23181795

RESUMEN

BACKGROUND: The mechanism(s) responsible for the acquisition of maternal antibody isotypes other than IgG are not fully understood. OBJECTIVE: To define the ability of the neonatal Fc receptor for IgG uptake (FcRn) to mediate intestinal absorption of IgG(1) anti-IgE/IgE immune complexes. METHODS: C57BL/6 allergic ovalbumin (OVA)-immune foster mothers were generated to nurse naïve FcRn(+/-) or FcRn(-/-) progeny. At the time of weaning, serum levels of OVA-specific antibodies and IgG(1) anti-IgE/IgE immune complexes were determined in allergic foster mothers and FcRn(+/+), FcRn(+/-), or FcRn(-/-) breastfed offspring. In separate experiments, FcRn(+/-) or FcRn(-/-) neonatal mice were gavage fed TNP-specific IgE as IgG(1) anti-IgE/IgE immune complexes, IgG(1) isotype control and IgE, or IgE alone. Mice were killed 2 h after feeding to determine serum levels and biological activity of absorbed TNP-specific IgE. RESULTS: As expected, the absorption of maternal OVA-specific IgG(1) in FcRn(-/-) offspring was at levels 10(3) -10(4) less than observed in FcRn(+/+) or FcRn(+/-) offspring. Surprisingly, FcRn expression also influenced the absorption of maternal IgE. OVA-specific IgE was detected in FcRn(+/+) and FcRn(+/-) offspring, but not in FcRn(-/-) offspring. IgG(1) anti-IgE/IgE immune complexes were detected in allergic foster mothers and correlated strongly with levels in FcRn(+/+) and FcRn(+/-) offspring (ρ = 0.88, P < 0.0001). Furthermore, FcRn expression was required for neonatal mice to absorb TNP-specific IgE when fed as IgG(1) anti-IgE/IgE immune complexes. When immune complexes were generated with IgG(1) anti-IgE directed against the Cε4 domain, the absorbed IgE was able to function in antigen-dependent basophil degranulation. CONCLUSIONS AND CLINICAL RELEVANCE: These data demonstrate a novel mechanism by which FcRn may facilitate absorption of maternal antibodies other than IgG. These findings are clinically relevant because FcRn mediates the transplacental passage of maternal IgG to the fetus. This raises the possibility that FcRn could mediate the transplacental passage of maternal IgE as IgG anti-IgE/IgE immune complexes.


Asunto(s)
Anticuerpos Antiidiotipos/metabolismo , Complejo Antígeno-Anticuerpo/metabolismo , Antígenos de Histocompatibilidad Clase I/metabolismo , Inmunoglobulina E/metabolismo , Inmunoglobulina G/metabolismo , Absorción Intestinal/inmunología , Receptores Fc/metabolismo , Alérgenos/inmunología , Animales , Femenino , Hipersensibilidad/inmunología , Inmunoglobulina E/inmunología , Inmunoglobulina G/inmunología , Ratones , Ratones Endogámicos C57BL
11.
Pharmacol Rep ; 74(1): 111-123, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-34757518

RESUMEN

BACKGROUND: Obesity is still a worldwide public health problem, requiring the development of adjuvant therapies to combat it. In this context, modulation of the intestinal microbiota seems prominent, given that the composition of the intestinal microbiota contributes to the outcome of this disease. The aim of this work is to investigate the treatment with an antimicrobial and/or a potential probiotic against overweight. METHODS: Male C57BL/6J mice were subjected to a 12-week overweight induction protocol. After that, 4-week treatment was started, with mice divided into four groups: control, treated with distilled water; potential probiotic, with Lactobacillus gasseri LG-G12; antimicrobial, with ceftriaxone; and antimicrobial + potential probiotic with ceftriaxone in the first 2 weeks and L. gasseri LG-G12 in the subsequent weeks. RESULTS: The treatment with ceftriaxone in isolated form or in combination with the potential probiotic provided a reduction in body fat. However, such effect is supposed to be a consequence of the negative action of ceftriaxone on the intestinal microbiota composition, and this intestinal dysbiosis may have contributed to the destruction of the intestinal villi structure, which led to a reduction in the absorptive surface. Also, the effects of L. gasseri LG-G12 apparently have been masked by the consumption of the high-fat diet. CONCLUSIONS: The results indicate that the use of a ceftriaxone in the adjuvant treatment of overweight is not recommended due to the potential risk of developing inflammatory bowel disease.


Asunto(s)
Ceftriaxona/farmacología , Disbiosis , Microbioma Gastrointestinal , Absorción Intestinal , Obesidad , Adyuvantes Farmacéuticos/farmacología , Animales , Antibacterianos/farmacología , Disbiosis/inducido químicamente , Disbiosis/inmunología , Microbioma Gastrointestinal/efectos de los fármacos , Microbioma Gastrointestinal/fisiología , Enfermedades Inflamatorias del Intestino/inmunología , Absorción Intestinal/efectos de los fármacos , Absorción Intestinal/inmunología , Lactobacillus gasseri/fisiología , Ratones , Ratones Endogámicos C57BL , Obesidad/tratamiento farmacológico , Obesidad/microbiología , Probióticos/farmacología , Medición de Riesgo
12.
J Dairy Sci ; 94(11): 5656-60, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22032388

RESUMEN

Fifty-two dairy calves were blocked by birth date and, within each block, randomly assigned to 1 of 4 treatments to investigate the effects of incremental levels of sodium bicarbonate (NaHCO(3)) on IgG metabolism. Treatments were (1) colostrum replacer (CR)+0 g of NaHCO(3) (control); (2) CR+15 g of NaHCO(3); (3) CR+30 g of NaHCO(3); or (4) CR+45 g of NaHCO(3). Calves were fed colostrum replacer (>200 g of IgG) in one feeding within 45 min of birth (0 h) and 2 L of milk replacer at 12, 24, 36, and 48 h. Only calves born in calving pens from multiparous cows with no dystocia were used in this study. Blood samples were taken at 0, 6, 12, 24, and 48 h postpartum, and serum was analyzed for IgG using radial immunoassay and bicarbonate using spectrophotometry. Feeding increasing levels of sodium bicarbonate had negative linear effects on IgG concentration, IgG apparent efficiency of absorption, and IgG area under the curve, primarily due to the effect of the highest dose of NaHCO(3) (45 g). Sodium bicarbonate treatments had no effect on serum bicarbonate concentration. However, area under the curve of serum bicarbonate increased linearly with the amount of NaHCO(3) fed.


Asunto(s)
Bovinos/fisiología , Inmunoglobulina G/metabolismo , Absorción Intestinal/efectos de los fármacos , Sustitutos de la Leche/química , Bicarbonato de Sodio/sangre , Bicarbonato de Sodio/farmacología , Animales , Animales Recién Nacidos , Bovinos/inmunología , Calostro/química , Calostro/inmunología , Absorción Intestinal/inmunología , Distribución Aleatoria , Bicarbonato de Sodio/administración & dosificación , Factores de Tiempo
13.
Gut ; 59(2): 207-17, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19837677

RESUMEN

Nucleotide oligomerisation domain 2 (NOD2) mutations are associated with susceptibility to Crohn's disease and graft-versus-host disease, two human disorders related with dysfunctions of Peyer's patches (PPs). In Nod2(-/-) mice transcellular permeability and bacterial translocation are increased in PPs. In this study, we show that both anti-CD4(+) and anti-interferon gamma (anti-IFNgamma) monoclonal antibodies abrogate this phenotype and reduce the expression of tumour necrosis factor (TNF) receptor 2 and the long isoform of myosin light chain kinase, thus demonstrating that immune T cells influence the epithelial functions. In turn, intraperitoneal injection of ML-7 (a myosin light chain kinase inhibitor) normalises the values of CD4(+) T cells, IFNgamma and TNFalpha. This reciprocal cross-talk is under the control of the gut microflora as shown by the normalisation of all parameters after antibiotic treatment. Toll-like receptor 2 (TLR2) and TLR4 expression were increased in Nod2(-/-) mice under basal conditions and TLR2 and TLR4 agonists induced an increased transcellular permeability in Nod2(+/+) mice. Muramyldipeptide (a Nod2 agonist) or ML-7 was able to reverse this phenomenon. It thus appears that Nod2 modulates the cross-talk between CD4(+) T cells and the epithelium recovering PP and that it downregulates the pro-inflammatory effect driven by the ileal microflora, likely by inhibiting the TLR pathways.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Absorción Intestinal/inmunología , Proteína Adaptadora de Señalización NOD2/inmunología , Ganglios Linfáticos Agregados/inmunología , Ganglios Linfáticos Agregados/microbiología , Animales , Antibacterianos/farmacología , Traslocación Bacteriana , Linfocitos T CD4-Positivos/efectos de los fármacos , Cámaras de Difusión de Cultivos , Íleon/microbiología , Interferón gamma/inmunología , Mucosa Intestinal/inmunología , Mucosa Intestinal/fisiopatología , Ratones , Ratones Endogámicos C57BL , Permeabilidad , Ganglios Linfáticos Agregados/fisiopatología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos , Receptor Toll-Like 2/agonistas , Receptor Toll-Like 2/fisiología , Receptor Toll-Like 4/agonistas , Receptor Toll-Like 4/fisiología
14.
Clin Transl Sci ; 14(2): 729-736, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33278326

RESUMEN

Changes in absorptive capacity and first-pass metabolism in the small intestine affect oral drug bioavailability. Characterization of such changes as a consequence of inflammation is important for developing physiologically-based pharmacokinetic (PBPK) models for inflammatory bowel disease. We sought to elucidate the impact of small intestinal Crohn's disease (CD) on villous length and CYP3A4 expression in children. Freshly frozen duodenal and terminal ileum (TI) biopsies from 107 children (1-19 years) with and without CD were evaluated for active inflammation. Villous length and CYP3A4 mRNA/protein expression were compared among regions of active and inactive inflammation in CD and controls. A twofold reduction in villous length was observed in inflamed duodena and ilia of children with CD, but in the absence of regional inflammation, villi in CD were comparable in length to controls. Expression of CYP3A4 mRNA correlated significantly with villous length in the TI (P = 0.0003), with a trend observed in the duodenum that did not reach statistical significance. In the presence of active inflammation, a significant decrease in CYP3A protein expression was confirmed in the duodenum, where protein expression also correlated significantly with villous length across diagnoses (P < 0.0001). Our findings suggest that previous observations of decreased CYP3A4 expression and function in inflamed intestine may not be due solely to downregulation by inflammatory cytokines, but also to villous blunting and subsequent loss of surface area for protein expression. This information is relevant for PBPK model development and could aid with dose adjustment decisions for oral CYP3A4 substrates administered during CD flare (e.g., budesonide).


Asunto(s)
Antiinflamatorios/farmacocinética , Enfermedad de Crohn/tratamiento farmacológico , Citocromo P-450 CYP3A/metabolismo , Mucosa Intestinal/metabolismo , Administración Oral , Adolescente , Antiinflamatorios/administración & dosificación , Disponibilidad Biológica , Biopsia , Budesonida/administración & dosificación , Budesonida/farmacocinética , Estudios de Casos y Controles , Niño , Preescolar , Enfermedad de Crohn/inmunología , Enfermedad de Crohn/patología , Relación Dosis-Respuesta a Droga , Duodeno/citología , Duodeno/inmunología , Duodeno/metabolismo , Duodeno/patología , Femenino , Humanos , Íleon/citología , Íleon/inmunología , Íleon/metabolismo , Íleon/patología , Lactante , Absorción Intestinal/inmunología , Mucosa Intestinal/citología , Mucosa Intestinal/patología , Masculino , Modelos Biológicos , Adulto Joven
15.
Front Immunol ; 11: 599637, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33542716

RESUMEN

Food allergy is a potentially fatal disease affecting 8% of children and has become increasingly common in the past two decades. Despite the prevalence and severe nature of the disease, the mechanisms underlying sensitization remain to be further elucidated. The Collaborative Cross is a genetically diverse panel of inbred mice that were specifically developed to study the influence of genetics on complex diseases. Using this panel of mouse strains, we previously demonstrated CC027/GeniUnc mice, but not C3H/HeJ mice, develop peanut allergy after oral exposure to peanut in the absence of a Th2-skewing adjuvant. Here, we investigated factors associated with sensitization in CC027/GeniUnc mice following oral exposure to peanut, walnut, milk, or egg. CC027/GeniUnc mice mounted antigen-specific IgE responses to peanut, walnut and egg, but not milk, while C3H/HeJ mice were not sensitized to any antigen. Naïve CC027/GeniUnc mice had markedly lower total fecal IgA compared to C3H/HeJ, which was accompanied by stark differences in gut microbiome composition. Sensitized CC027/GeniUnc mice had significantly fewer CD3+ T cells but higher numbers of CXCR5+ B cells and T follicular helper cells in the mesenteric lymph nodes compared to C3H/HeJ mice, which is consistent with their relative immunoglobulin production. After oral challenge to the corresponding food, peanut- and walnut-sensitized CC027/GeniUnc mice experienced anaphylaxis, whereas mice exposed to milk and egg did not. Ara h 2 was detected in serum collected post-challenge from peanut-sensitized mice, indicating increased absorption of this allergen, while Bos d 5 and Gal d 2 were not detected in mice exposed to milk and egg, respectively. Machine learning on the change in gut microbiome composition as a result of food protein exposure identified a unique signature in CC027/GeniUnc mice that experienced anaphylaxis, including the depletion of Akkermansia. Overall, these results demonstrate several factors associated with enteral sensitization in CC027/GeniUnc mice, including diminished total fecal IgA, increased allergen absorption and altered gut microbiome composition. Furthermore, peanuts and tree nuts may have inherent properties distinct from milk and eggs that contribute to allergy.


Asunto(s)
Alérgenos/inmunología , Heces/microbiología , Microbioma Gastrointestinal/inmunología , Inmunoglobulina A/inmunología , Absorción Intestinal/inmunología , Hipersensibilidad al Cacahuete , Alérgenos/genética , Animales , Microbioma Gastrointestinal/genética , Predisposición Genética a la Enfermedad , Inmunoglobulina A/genética , Absorción Intestinal/genética , Ratones , Ratones Transgénicos , Hipersensibilidad al Cacahuete/genética , Hipersensibilidad al Cacahuete/inmunología , Hipersensibilidad al Cacahuete/microbiología
16.
Curr Pharm Biotechnol ; 21(15): 1603-1615, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32410561

RESUMEN

In recent decades, there has been a very rapid increase in the prevalence of diabetes globally, with serious health and economic implications. Although today there are several therapeutic treatments for this disease, these do not address the causes of the disease and have serious side effects, so it is necessary to seek new treatments to replace or complement the existing ones. Among these complementary treatments, a strong link between the intestinal microbiota and diabetes has been demonstrated, which has focused attention on the use of biotherapy to regulate the function of the intestinal microbiota and, thus, treat diabetes. In this way, the main objective of this work is to provide a review of the latest scientific evidence on diabetes, gathering information about new trends in its management, and especially, the influence of the intestinal microbiota and microbiome on this pathology. It is possible to conclude that the relationship between the intestinal microbiota and diabetes is carried out through alterations in energy metabolism, the immune system, changes in intestinal permeability, and a state of low-intensity systemic inflammation. Although, currently, most of the experimental work, using probiotics for diabetes management, has been done on experimental animals, the results obtained are promising. Thus, the modification of the microbiota through biotherapy has shown to improve the symptoms and severity of diabetes through various mechanisms related to these alterations.


Asunto(s)
Diabetes Mellitus/tratamiento farmacológico , Microbioma Gastrointestinal/efectos de los fármacos , Prebióticos/administración & dosificación , Probióticos/uso terapéutico , Animales , Diabetes Mellitus/inmunología , Diabetes Mellitus/metabolismo , Diabetes Mellitus/microbiología , Metabolismo Energético/efectos de los fármacos , Humanos , Inflamación , Absorción Intestinal/efectos de los fármacos , Absorción Intestinal/inmunología , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/inmunología , Mucosa Intestinal/microbiología , Permeabilidad
17.
J Crohns Colitis ; 14(3): 316-322, 2020 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-31665264

RESUMEN

BACKGROUND AND AIMS: Anaemia is common in patients with inflammatory bowel disease [IBD], its two main aetiologies being iron deficiency anaemia [IDA] and anaemia of chronic inflammation [ACI]. Impaired intestinal iron absorption due to inflammatory cytokines is thought to play a role in ACI. We undertook for the first time a controlled prospective study investigating effects of differing underlying diseases, disease locations, and types of iron deficiency or anaemia on oral iron absorption in adult IBD patients with and without inflammation. METHODS: This study was a comparative, single-centred open clinical trial in adults with IBD [n = 73] and healthy controls [n = 22]. Baseline parameters included blood count, iron status [ferritin, transferrin, transferrin saturation, soluble transferrin receptor, hepcidin, serum iron], high-sensitivity C-reactive protein [hsCRP] and interleukin-6. Iron absorption was tested using one oral, enteric-coated capsule containing 567.7 mg iron[II]-glycine-sulphate complex. Serum iron was determined 60/90/120/180/240 min after ingestion. RESULTS: Iron absorption capacity was shown to be influenced by inflammation and anaemia or iron deficiency [ID] type but not by underlying disease type or localisation. The ACI group showed a significantly lower iron absorption capacity than all others. Whereas hsCRP levels [-0.387, p < 0.001], IL-6 [-0.331, p = 0.006], ferritin [-0.531, p < 0.001], and serum hepcidin [-0.353, p = 0.003] correlated negatively with serum iron change at 2 h, transferrin showed a positive correlation at the same time point [0.379, p < 0.001]. CONCLUSIONS: Underlying disease type and localisation appear to have little effect on iron absorption capacity, whereas lack of response to oral iron correlates well with serum markers of inflammation. Iron absorption capacity is thus significantly reduced in the presence of inflammation.


Asunto(s)
Anemia Ferropénica , Ferritinas/sangre , Inflamación/sangre , Enfermedades Inflamatorias del Intestino , Interleucina-6/sangre , Hierro , Absorción por la Mucosa Oral/inmunología , Transferrina/análisis , Adulto , Anemia Ferropénica/diagnóstico , Anemia Ferropénica/etiología , Anemia Ferropénica/inmunología , Biomarcadores/sangre , Proteína C-Reactiva/análisis , Correlación de Datos , Femenino , Alemania/epidemiología , Hepcidinas/sangre , Humanos , Enfermedades Inflamatorias del Intestino/sangre , Enfermedades Inflamatorias del Intestino/diagnóstico , Enfermedades Inflamatorias del Intestino/epidemiología , Absorción Intestinal/inmunología , Hierro/sangre , Deficiencias de Hierro , Masculino , Persona de Mediana Edad
18.
Curr Protein Pept Sci ; 21(8): 777-784, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31889482

RESUMEN

Medium-chain fatty acids (MCFAs) are the main form of Medium Chain Triglycerides (MCTs) utilized by monogastric animals. MCFAs can be directly absorbed and supply rapid energy to promote the renewal and repair of intestinal epithelial cells, maintain the integrity of intestinal mucosal barrier function, and reduce inflammation and stress. In our review, we pay more attention to the role of MCFAs on intestinal microbiota and mucosa immunity to explore MCFA's positive effect. It was found that MCFAs and their esterified forms can decrease pathogens while increasing probiotics. In addition, being recognized via specific receptors, MCFAs are capable of alleviating inflammation to a certain extent by regulating inflammation and immune-related pathways. MCFAs may also have a certain value to relieve intestinal allergy and inflammatory bowel disease (IBD). Unknown mechanism of various MCFA characteristics still causes dilemmas in the application, thus MCFAs are used generally in limited dosages and combined with short-chain organic acids (SOAs) to attain ideal results. We hope that further studies will provide guidance for the practical use of MCFAs in animal feed.


Asunto(s)
Caprilatos/inmunología , Colitis Ulcerosa/dietoterapia , Enfermedad de Crohn/dietoterapia , Ácidos Decanoicos/inmunología , Síndrome del Colon Irritable/dietoterapia , Ácidos Láuricos/inmunología , Alimentación Animal/análisis , Animales , Caprilatos/administración & dosificación , Caprilatos/metabolismo , Colitis Ulcerosa/inmunología , Colitis Ulcerosa/microbiología , Colitis Ulcerosa/patología , Enfermedad de Crohn/inmunología , Enfermedad de Crohn/microbiología , Enfermedad de Crohn/patología , Citocinas/genética , Citocinas/inmunología , Ácidos Decanoicos/administración & dosificación , Ácidos Decanoicos/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Inmunidad Mucosa/efectos de los fármacos , Absorción Intestinal/efectos de los fármacos , Absorción Intestinal/inmunología , Intestinos/efectos de los fármacos , Intestinos/inmunología , Intestinos/microbiología , Síndrome del Colon Irritable/inmunología , Síndrome del Colon Irritable/microbiología , Síndrome del Colon Irritable/patología , Ácidos Láuricos/administración & dosificación , Ácidos Láuricos/metabolismo , FN-kappa B/genética , FN-kappa B/inmunología , Estómago/efectos de los fármacos , Estómago/inmunología , Estómago/microbiología , Triglicéridos/inmunología , Triglicéridos/metabolismo
19.
J Pediatr ; 155(5): S61-70, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19840609

RESUMEN

Initial bacterial colonization, including colonization with health-positive bacteria, such as bifidobacteria and lactobacilli, is necessary for the normal development of intestinal innate and adaptive immune defenses. The predominance of beneficial bacteria in the gut microflora of breast-fed infants is thought to be, at least in part, supported by the metabolism of the complex mixture of oligosaccharides present in human breast milk, and a more adult-type intestinal microbiota is found in formula-fed infants. Inadequate gut colonization, dysbiosis, may lead to an increased risk of infectious, allergic, and autoimmune disorders later in life. The addition of appropriate amounts of selected prebiotics to infant formulas can enhance the growth of bifidobacteria or lactobacilli in the colonic microbiota and, thereby, might produce beneficial effects. Among the substrates considered as prebiotics are the oligosaccharides inulin, fructo-oligosaccharides, galacto-oligosaccharides, and lactulose. There are some reports that such prebiotics have beneficial effects on various markers of health. For example, primary prevention trials in infants have provided promising data on prevention of infections and atopic dermatitis. Additional well-designed prospective clinical trials and mechanistic studies are needed to advance knowledge further in this promising field.


Asunto(s)
Bifidobacterium/inmunología , Inmunidad Mucosa/fisiología , Intestinos/efectos de los fármacos , Intestinos/inmunología , Probióticos/administración & dosificación , Factores de Edad , Bifidobacterium/fisiología , Lactancia Materna , Preescolar , Femenino , Estudios de Seguimiento , Humanos , Lactante , Alimentos Infantiles/microbiología , Recién Nacido , Recien Nacido Prematuro , Absorción Intestinal/inmunología , Absorción Intestinal/fisiología , Mucosa Intestinal/inmunología , Mucosa Intestinal/microbiología , Intestinos/microbiología , Lactobacillus/inmunología , Masculino , Factores de Riesgo , Nacimiento a Término
20.
Pediatr Diabetes ; 10(8): 563-72, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19622083

RESUMEN

AIMS: To determine whether Glo-3A, (formerly referred to as homologue of Glb1 or Glb1) antibodies are associated with islet autoimmunity (IA) in children at increased risk for type 1 diabetes (T1D) and to investigate their relation with environmental correlates of T1D. METHODS: We selected a sample from the Diabetes Autoimmunity Study in the Young (DAISY), a prospective study of children at increased risk for T1D. Cases were positive for insulin, glutamic acid decarboxylase (GAD), or insulinoma-associated antigen-2 (IA-2) autoantibodies on two consecutive visits and either diagnosed with diabetes mellitus or still autoantibody positive when selected. Controls were from the same increased risk group, of similar age as the cases but negative for autoantibodies. Sera from 91 IA cases and 82 controls were analyzed in a blinded manner for immunoglobulin G (IgG) antibodies to Glo-3A by ELISA. RESULTS: Adjusting for family history of T1D and human leukocyte antigen (HLA)-DR4 positivity, Glo-3A antibodies were not associated with IA case status (OR: 1.01, 95% CI: 0.99-1.03). Adjusting for age, family history of T1D, and HLA-DR4 positivity, Glo-3A antibody levels were inversely associated with breast-feeding duration (beta = -0.08, p = 0.001) and directly associated with current intake of foods containing gluten (beta = 0.24, p = 0.007) in IA cases but not in controls. Zonulin, a biomarker of gut permeability, was directly associated with Glo-3A antibody levels in cases (beta = 0.73, p = 0.003) but not in controls. CONCLUSION: Differing correlates of Glo-3A antibodies in IA cases and controls suggest an underlying difference in mucosal immune response.


Asunto(s)
Autoanticuerpos/sangre , Enfermedad Celíaca/epidemiología , Enfermedad Celíaca/inmunología , Diabetes Mellitus Tipo 1/epidemiología , Diabetes Mellitus Tipo 1/inmunología , Niño , Preescolar , Toxina del Cólera/metabolismo , Femenino , Gliadina/inmunología , Glutamato Descarboxilasa/inmunología , Haptoglobinas , Humanos , Inmunoglobulina G/sangre , Lactante , Absorción Intestinal/inmunología , Islotes Pancreáticos/inmunología , Masculino , Estudios Prospectivos , Precursores de Proteínas , Factores de Riesgo , Estudios Seroepidemiológicos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA