Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Proc Natl Acad Sci U S A ; 109(28): 11330-5, 2012 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-22733781

RESUMEN

The fetal intestinal mucosa is characterized by elevated Toll-like receptor 4 (TLR4) expression, which can lead to the development of necrotizing enterocolitis (NEC)--a devastating inflammatory disease of the premature intestine--upon exposure to microbes. To define endogenous strategies that could reduce TLR4 signaling, we hypothesized that amniotic fluid can inhibit TLR4 signaling within the fetal intestine and attenuate experimental NEC, and we sought to determine the mechanisms involved. We show here that microinjection of amniotic fluid into the fetal (embryonic day 18.5) gastrointestinal tract reduced LPS-mediated signaling within the fetal intestinal mucosa. Amniotic fluid is abundant in EGF, which we show is required for its inhibitory effects on TLR4 signaling via peroxisome proliferator-activated receptor, because inhibition of EGF receptor (EGFR) with cetuximab or EGF-depleted amniotic fluid blocked the inhibitory effects of amniotic fluid on TLR4, whereas amniotic fluid did not prevent TLR4 signaling in EGFR- or peroxisome proliferator-activated receptor γ-deficient enterocytes or in mice deficient in intestinal epithelial EGFR, and purified EGF attenuated the exaggerated intestinal mucosal TLR4 signaling in wild-type mice. Moreover, amniotic fluid-mediated TLR4 inhibition reduced the severity of NEC in mice through EGFR activation. Strikingly, NEC development in both mice and humans was associated with reduced EGFR expression that was restored upon the administration of amniotic fluid in mice or recovery from NEC in humans, suggesting that a lack of amniotic fluid-mediated EGFR signaling could predispose to NEC. These findings may explain the unique susceptibility of premature infants to the development of NEC and offer therapeutic approaches to this devastating disease.


Asunto(s)
Líquido Amniótico/metabolismo , Regulación del Desarrollo de la Expresión Génica , Mucosa Intestinal/metabolismo , Receptor Toll-Like 4/metabolismo , Animales , Línea Celular , Enterocolitis Necrotizante/metabolismo , Enterocitos/metabolismo , Receptores ErbB/metabolismo , Humanos , Recién Nacido , Mucosa Intestinal/embriología , Intestinos/embriología , Ratones , Microscopía Confocal/métodos , Transducción de Señal , Factores de Tiempo
2.
Am J Physiol Gastrointest Liver Physiol ; 304(10): G864-75, 2013 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-23518680

RESUMEN

Preterm neonates are susceptible to gastrointestinal disorders such as necrotizing enterocolitis (NEC). Maternal milk and colostrum protects against NEC via growth promoting, immunomodulatory, and antimicrobial factors. The fetal enteral diet amniotic fluid (AF), contains similar components, and we hypothesized that postnatal AF administration reduces inflammatory responses and NEC in preterm neonates. Preterm pigs (92% gestation) were delivered by caesarean section and fed parental nutrition (2 days) followed by enteral (2 days) porcine colostrum (COLOS, n = 7), infant formula (FORM, n = 13), or AF supplied before and after introduction of formula (AF, n = 10) in experiment 1, and supplied only during the enteral feeding period in experiment 2 (FORM, n = 16; AF, n = 14). The NEC score was reduced in both AF and COLOS pigs, relative to FORM, when AF was provided prior to full enteral feeding (9.9 and 7.7 compared with 17.3, P < 0.05). There was no effect of AF when provided only during enteral feeding. AF pigs showed decreased bacterial abundance in colon and intestinal inflammation-related genes (e.g., TNF-α, IL-1α, IL-6, NOS) were downregulated, relative to FORM pigs with NEC. Anti-inflammatory properties of AF were supported by delayed maturation and decreased TNF-α production in murine dendritic cells, as well as increased proliferation and migration, and downregulation of IL-6 expression in intestinal cells (IEC-6, IPEC-J2). Like colostrum, AF may reduce NEC development in preterm neonates by suppressing the proinflammatory responses to enteral formula feeding and gut colonization when provided before the onset of NEC.


Asunto(s)
Líquido Amniótico/fisiología , Calostro/fisiología , Enterocolitis Necrotizante/terapia , Gastroenteritis/terapia , Animales , Animales Recién Nacidos , Citocinas/metabolismo , Células Dendríticas/metabolismo , Nutrición Enteral , Enterocolitis Necrotizante/microbiología , Enterocolitis Necrotizante/patología , Enterocitos/metabolismo , Femenino , Gastroenteritis/microbiología , Gastroenteritis/patología , Humanos , Recién Nacido , Recien Nacido Prematuro , Absorción Intestinal , Intestinos/microbiología , Análisis por Micromatrices , Nutrición Parenteral Total , Permeabilidad , Embarazo , ARN/biosíntesis , ARN/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Porcinos
3.
Cell Mol Life Sci ; 68(22): 3623-34, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21986983

RESUMEN

The maintenance of mucosal barrier equilibrium in the intestine requires a delicate and dynamic balance between enterocyte loss by apoptosis and the generation of new cells by proliferation from stem cell precursors at the base of the intestinal crypts. When the balance shifts towards either excessive or insufficient apoptosis, a broad range of gastrointestinal diseases can manifest. Recent work from a variety of laboratories has provided evidence in support of a role for receptors of the innate immune system, including Toll-like receptors 2, 4, and 9 as well as the intracellular pathogen recognition receptor NOD2/CARD15, in the initiation of enterocyte apoptosis. The subsequent induction of enterocyte apoptosis in response to the activation of these innate immune receptors plays a key role in the development of various intestinal diseases, including necrotizing enterocolitis, Crohn's disease, ulcerative colitis, and intestinal cancer. This review will detail the regulatory pathways that govern enterocyte apoptosis, and will explore the role of the innate immune system in the induction of enterocyte apoptosis in gastrointestinal disease.


Asunto(s)
Apoptosis/inmunología , Enterocitos/fisiología , Enfermedades Gastrointestinales/patología , Inmunidad Innata/inmunología , Inflamación/patología , Mucosa Intestinal/inmunología , Animales , Enterocitos/citología , Enfermedades Gastrointestinales/inmunología , Humanos , Inflamación/inmunología , Mucosa Intestinal/citología , Mucosa Intestinal/patología , FN-kappa B/inmunología , Proteínas Adaptadoras de Señalización NOD/inmunología , Transducción de Señal/inmunología , Receptores Toll-Like/inmunología
4.
Am J Physiol Gastrointest Liver Physiol ; 301(3): G435-45, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21700903

RESUMEN

Necrotizing enterocolitis (NEC) in preterm infants develops very rapidly from a mild intolerance to enteral feeding into intestinal mucosal hemorrhage, inflammation, and necrosis. We hypothesized that immediate feeding-induced gut responses precede later clinical NEC symptoms in preterm pigs. Fifty-six preterm pigs were fed total parenteral nutrition (TPN) for 48 h followed by enteral feeding for 0, 8, 17, or 34 h with either colostrum (Colos, n = 20) or formula (Form, n = 31). Macroscopic NEC lesions were detected in Form pigs throughout the enteral feeding period (20/31, 65%), whereas most Colos pigs remained protected (1/20, 5%). Just 8 h of formula feeding induced histopathological lesions, as evidenced by capillary stasis and necrosis, epithelial degeneration, edema, and mucosal hemorrhage. These immediate formula-induced changes were paralleled by decreased digestive enzyme activities (lactase and dipeptidylpeptidase IV), increased nutrient fermentation, and altered expression of innate immune defense genes such as interleukins (IL-1α, IL-6, IL-18), nitric oxide synthetase, tight junction proteins (claudins), Toll-like receptors (TLR-4), and TNF-α. In contrast, the first hours of colostrum feeding induced no histopathological lesions, increased maltase activity, and induced changes in gene expressions related to tissue development. Total bacterial density was high after 2 days of parenteral feeding and was not significantly affected by diet (colostrum, formula) or length of enteral feeding (8-34 h), except that a few bacterial groups (Clostridium, Enterococcus, Streptococcus species) increased with time. We conclude that a switch from parenteral to enteral nutrition rapidly induces diet-dependent histopathological, functional, and proinflammatory insults to the immature intestine. Great care is required when introducing enteral feeds to TPN-fed preterm infants, particularly when using formula, because early feeding-induced insults may predispose to NEC lesions that are difficult to revert by later dietary or medical interventions.


Asunto(s)
Nutrición Enteral , Enterocolitis Necrotizante/patología , Nutrición Parenteral Total , Animales , Calostro , Enterocolitis Necrotizante/etiología , Humanos , Fórmulas Infantiles/farmacología , Recién Nacido , Recien Nacido Prematuro , Porcinos
5.
Gastroenterology ; 139(3): 904-17, 917.e1-6, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20580721

RESUMEN

BACKGROUND & AIMS: Factors that regulate enterocyte apoptosis in necrotizing enterocolitis (NEC) remain incompletely understood, although Toll-like receptor-4 (TLR4) signaling in enterocytes plays a major role. Nucleotide-binding oligomerization domain-2 (NOD2) is an immune receptor that regulates other branches of the immune system, although its effects on TLR4 in enterocytes and its role in NEC remain unknown. We now hypothesize that activation of NOD2 in the newborn intestine inhibits TLR4, and that failure of NOD2 signaling leads to NEC through increased TLR4-mediated enterocyte apoptosis. METHODS: The effects of NOD2 on enterocyte TLR4 signaling and intestinal injury and repair were assessed in enterocytes lacking TLR4 or NOD2, in mice with intestinal-specific wild-type or dominant-negative TLR4 or NOD2, and in mice with NEC. A protein array was performed on NOD2-activated enterocytes to identify novel effector molecules involved. RESULTS: TLR4 activation caused apoptosis in newborn but not adult small intestine or colon, and its intestinal expression was influenced by NOD2. NOD2 activation inhibited TLR4 in enterocytes, but not macrophages, and reversed the effects of TLR4 on intestinal mucosal injury and repair. Protection from TLR4-induced enterocyte apoptosis by NOD2 required a novel pathway linking NOD2 with the apoptosis mediator second mitochondria-derived activator of caspase/direct inhibitor of apoptosis-binding protein with low PI (SMAC-DIABLO), both in vitro and in vivo. Strikingly, activation of NOD2 reduced SMAC-DIABLO expression, attenuated the extent of enterocyte apoptosis, and reduced the severity of NEC. CONCLUSIONS: These findings reveal a novel inhibitory interaction between TLR4 and NOD2 signaling in enterocytes leading to the regulation of enterocyte apoptosis and suggest a therapeutic role for NOD2 in the protection of intestinal diseases such as NEC.


Asunto(s)
Endotoxemia/metabolismo , Enterocolitis Necrotizante/metabolismo , Mucosa Intestinal/metabolismo , Proteína Adaptadora de Señalización NOD2/metabolismo , Transducción de Señal , Receptor Toll-Like 4/metabolismo , Acetilmuramil-Alanil-Isoglutamina/farmacología , Factores de Edad , Animales , Animales Recién Nacidos , Apoptosis , Proteínas Reguladoras de la Apoptosis , Proteínas Portadoras/metabolismo , Línea Celular , Movimiento Celular , Modelos Animales de Enfermedad , Endotoxemia/genética , Endotoxemia/patología , Enterocolitis Necrotizante/genética , Enterocolitis Necrotizante/patología , Enterocolitis Necrotizante/prevención & control , Enterocitos/metabolismo , Humanos , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Mitocondriales/metabolismo , FN-kappa B/metabolismo , Proteína Adaptadora de Señalización NOD2/agonistas , Proteína Adaptadora de Señalización NOD2/deficiencia , Proteína Adaptadora de Señalización NOD2/genética , Análisis por Matrices de Proteínas , Ratas , Índice de Severidad de la Enfermedad , Transducción de Señal/efectos de los fármacos , Receptor Toll-Like 4/genética , Transducción Genética
6.
Am J Physiol Gastrointest Liver Physiol ; 297(6): G1115-25, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19808655

RESUMEN

Necrotizing enterocolitis (NEC) remains the most severe gastrointestinal disorder in preterm infants. It is associated with the initiation of enteral nutrition and may be related to immature carbohydrate digestive capacity. We tested the hypothesis that a formula containing maltodextrin vs. a formula containing lactose as the principal source of carbohydrate would predispose preterm pigs to a higher NEC incidence. Cesarean-derived preterm pigs were given total parenteral nutrition for 48 h followed by total enteral nutrition with a lactose-based (n = 11) or maltodextrin-based (n = 11) formula for 36 h. A higher incidence (91% vs. 27%) and severity (score of 3.3 vs. 1.8) of NEC were observed in the maltodextrin than in the lactose group. This higher incidence of NEC in the maltodextrin group was associated with significantly lower activities of lactase, maltase, and aminopeptidase; reduced villus height; transiently reduced in vivo aldohexose uptake; and reduced ex vivo aldohexose uptake capacity in the middle region of the small intestine. Bacterial diversity was low for both diets, but alterations in bacterial composition and luminal concentrations of short-chain fatty acids were observed in the maltodextrin group. In a second study, we quantified net portal absorption of aldohexoses (glucose and galactose) during acute jejunal infusion of a maltodextrin- or a lactose-based formula (n = 8) into preterm pigs. We found lower net portal aldohexose absorption (4% vs. 42%) and greater intestinal recovery of undigested carbohydrate (68% vs. 27%) in pigs acutely perfused with the maltodextrin-based formula than those perfused with the lactose-based formula. The higher digestibility of the lactose than the maltodextrin in the formulas can be attributed to a 5- to 20-fold higher hydrolytic activity of tissue-specific lactase than maltases. We conclude that carbohydrate maldigestion is sufficient to increase the incidence and severity of NEC in preterm pigs.


Asunto(s)
Fenómenos Fisiológicos Nutricionales de los Animales , Digestión , Enterocolitis Necrotizante/fisiopatología , Intestinos/fisiopatología , Aminopeptidasas/metabolismo , Animales , Animales Recién Nacidos , Cesárea , Modelos Animales de Enfermedad , Nutrición Enteral , Enterocolitis Necrotizante/inducido químicamente , Enterocolitis Necrotizante/enzimología , Enterocolitis Necrotizante/microbiología , Galactosa/metabolismo , Glucosa/metabolismo , Humanos , Hidrólisis , Fórmulas Infantiles/administración & dosificación , Recién Nacido , Absorción Intestinal , Intestinos/enzimología , Intestinos/crecimiento & desarrollo , Intestinos/microbiología , Lactasa/metabolismo , Lactosa/administración & dosificación , Nutrición Parenteral , Polisacáridos/administración & dosificación , Nacimiento Prematuro , Índice de Severidad de la Enfermedad , Porcinos , Factores de Tiempo , alfa-Glucosidasas/metabolismo
7.
J Nutr ; 138(8): 1437-44, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18641188

RESUMEN

Following preterm birth, bacterial colonization and enteral formula feeding predispose neonates to gut dysfunction and necrotizing enterocolitis (NEC), a serious gastrointestinal inflammatory disease. We hypothesized that administration of probiotics would beneficially influence early bacterial colonization, thereby reducing the susceptibility to formula-induced gut atrophy, dysfunction, and NEC. Caesarean-delivered preterm pigs were provided total parenteral nutrition (1.5 d) followed by enteral feeding (2 d) with porcine colostrum (COLOS; n = 5), formula (FORM; n = 9), or formula with probiotics (FORM-P; Bifidobacterium animalis and Lactobacillus: L. acidophilus, L. casei, L. pentosus, L. plantarum; n = 13). Clinical NEC scores were reduced (P < 0.05) in FORM-P (2.0 +/- 0.2) and COLOS groups (1.7 +/- 0.5) compared with FORM pigs (3.4 +/- 0.6). Lower NEC scores were associated with elevated intestinal weight, mucosa proportion, villus height, RNA integrity, and brush border aminopeptidase A and N activities, and lower gastric organic acid concentration in the FORM-P and COLOS groups (P < 0.05). Diversity of the mucosa-associated bacteria in the distal small intestine was similar among formula-fed pigs, yet the abundance of specific bacterial groups differed between FORM-P and FORM pigs. FORM-P pigs had lower colonization density of a potential pathogen, Clostridium perfringens, and had commensal Lactobacillus bacteria more closely associated with enterocytes along the villus-crypt axis relative to FORM pigs. These results suggest that probiotic administration immediately after birth promotes the colonization of a beneficial commensal microbiota capable of limiting the formula-induced mucosal atrophy, dysfunction, and pathogen load in preterm neonates, thereby reducing the incidence and severity of NEC.


Asunto(s)
Enterocolitis Necrotizante/prevención & control , Enfermedades Gastrointestinales/prevención & control , Intestinos/microbiología , Probióticos/uso terapéutico , Animales , Animales Recién Nacidos , Bifidobacterium/fisiología , Citocinas/genética , Citocinas/metabolismo , Dieta/efectos adversos , Modelos Animales de Enfermedad , Esquema de Medicación , Enterocolitis Necrotizante/tratamiento farmacológico , Femenino , Enfermedades Gastrointestinales/tratamiento farmacológico , Intestinos/citología , Intestinos/efectos de los fármacos , Intestinos/enzimología , Lactobacillus/fisiología , Masculino , Embarazo , Nacimiento Prematuro , Probióticos/administración & dosificación , Distribución Aleatoria , Porcinos
8.
J Nutr Biochem ; 22(6): 511-21, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21193301

RESUMEN

The gastrointestinal inflammatory disorder, necrotizing enterocolitis (NEC), is among the most serious diseases for preterm neonates. Nutritional, microbiological and immunological dysfunctions all play a role in disease progression but the relationship among these determinants is not understood. The preterm gut is very sensitive to enteral feeding which may either promote gut adaptation and health, or induce gut dysfunction, bacterial overgrowth and inflammation. Uncontrolled inflammatory reactions may be initiated by maldigestion and impaired mucosal protection, leading to bacterial overgrowth and excessive nutrient fermentation. Tumor necrosis factor alpha, toll-like receptors and heat-shock proteins are identified among the immunological components of the early mucosal dysfunction. It remains difficult, however, to distinguish the early initiators of NEC from the later consequences of the disease pathology. To elucidate the mechanisms and identify clinical interventions, animal models showing spontaneous NEC development after preterm birth coupled with different forms of feeding may help. In this review, we summarize the literature and some recent results from studies on preterm pigs on the nutritional, microbial and immunological interactions during the early feeding-induced mucosal dysfunction and later NEC development. We show that introduction of suboptimal enteral formula diets, coupled with parenteral nutrition, predispose to disease, while advancing amounts of mother's milk from birth (particularly colostrum) protects against disease. Hence, the transition from parenteral to enteral nutrition shortly after birth plays a pivotal role to secure gut growth, digestive maturation and an appropriate response to bacterial colonization in the sensitive gut of preterm neonates.


Asunto(s)
Enterocolitis Necrotizante/etiología , Tracto Gastrointestinal/inmunología , Tracto Gastrointestinal/microbiología , Fenómenos Fisiológicos Nutricionales del Lactante , Enfermedades del Prematuro/etiología , Animales , Animales Recién Nacidos , Enterocolitis Necrotizante/inmunología , Enterocolitis Necrotizante/microbiología , Tracto Gastrointestinal/crecimiento & desarrollo , Proteínas de Choque Térmico/metabolismo , Humanos , Sistema Inmunológico/inmunología , Recién Nacido , Recien Nacido Prematuro , Enfermedades del Prematuro/inmunología , Enfermedades del Prematuro/microbiología , Mucosa Intestinal/inmunología , Mucosa Intestinal/microbiología , Intestino Delgado/metabolismo , Metagenoma/fisiología
9.
Neonatology ; 99(4): 280-8, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21135563

RESUMEN

BACKGROUND: In newborns, colonizing bacteria and enteral nutrition are important for early gut development and immunity. However, in preterm newborns, bacterial colonization, coupled with enteral feeding, can lead to marked intestinal inflammation and disease such as necrotizing enterocolitis (NEC). We hypothesized that the initial bacterial colonization of the gut affects the intestinal proteome independently of enteral feeding. OBJECTIVE: To identify the intestinal proteins affected by the first colonizing bacteria by comparing the intestinal proteome in formula-fed preterm pigs reared under germ free (GF) or conventional conditions. METHODS: Gel-based proteomics of the small intestine to detect proteins that may play a part in the response of the immature intestine to bacterial colonization after birth. RESULTS: Fourteen proteins involved in stress response and detoxification (e.g. heat-shock proteins, peroxiredoxin 1), tissue metabolism and apoptosis (e.g. annexin 2), and some signal transduction pathways were differentially expressed between GF and conventionally reared pigs. CONCLUSION: The premature intestine is highly responsive to initial bacterial colonization and the specific bacteria-related proteome changes may contribute to the stress response that makes the immature intestine sensitive to the pro-inflammatory effects of enteral feeding.


Asunto(s)
Bacterias/crecimiento & desarrollo , Enterocolitis Necrotizante/metabolismo , Mucosa Intestinal/metabolismo , Nacimiento Prematuro/veterinaria , Proteoma/metabolismo , Animales , Animales Recién Nacidos , Carga Bacteriana/fisiología , Susceptibilidad a Enfermedades , Electroforesis en Gel Bidimensional , Enterocolitis Necrotizante/etiología , Enterocolitis Necrotizante/microbiología , Enterocolitis Necrotizante/veterinaria , Femenino , Intestinos/microbiología , Embarazo , Proteoma/análisis , Porcinos , Enfermedades de los Porcinos/etiología , Enfermedades de los Porcinos/metabolismo , Enfermedades de los Porcinos/microbiología
10.
J Proteome Res ; 6(7): 2596-604, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17542629

RESUMEN

The gastrointestinal tract harbors a complex community of bacteria, of which many may be beneficial. Studies of germ-free animal models have shown that the gastrointestinal microbiota not only assists in making nutrients available for the host but also contributes to intestinal health and development. We studied small intestinal protein expression patterns in gnotobiotic pigs maintained germ-free, or monoassociated with either Lactobacillus fermentum or non-pathogenic Escherichia coli. A common reference design in combination with labeling with stable isobaric tags allowed the individual comparison of 12 animals. Our results showed that bacterial colonization differentially affected mechanisms such as proteolysis, epithelial proliferation, and lipid metabolism, which is in good agreement with previous studies of other germ-free animal models. We have also found that E. coli has a profound effect on actin remodeling and intestinal proliferation, which may be related to stimulated migration and turnover of enterocytes. Regulations related to L. fermentum colonization involved individual markers for immunoregulatory mechanisms.


Asunto(s)
Escherichia coli/fisiología , Mucosa Intestinal/metabolismo , Intestinos/microbiología , Limosilactobacillus fermentum/fisiología , Proteoma/metabolismo , Actinas/metabolismo , Animales , Proliferación Celular , Proteínas/análisis , Proteínas/metabolismo , Proteoma/análisis , Sus scrofa/metabolismo , Sus scrofa/microbiología
11.
PLoS One ; 2(7): e677, 2007 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-17653288

RESUMEN

BACKGROUND: CCL25/TECK and CCL28/MEC are CC chemokines primarily expressed in thymic dendritic cells and mucosal epithelial cells. Their receptors, CCR9 and CCR10, are mainly expressed on T and B lymphocytes. In human, mouse, pig and sheep CCL25 and CCL28 play an important role in the segregation and the compartmentalization of the mucosal immune system. As evidenced by early comparisons of germ-free and conventional animals, the intestinal bacterial microflora has a marked effect on host intestinal immune functions. However, little is known about the impact of bacterial colonization on constitutive and induced chemokine expressions as well as on the generation of anti-inflammatory mechanisms. METHODOLOGY/PRINCIPAL FINDINGS: Therefore, we decided to focus by qPCR on the mRNA expression of two main gut chemokines, CCL25 and CCL28, their receptors CCR9 and CCR10, the Tregs marker Foxp3 and anti-inflammatory cytokines TGF-beta and IL-10 following colonization with different bacterial species within the small intestine. To accomplish this we used an original germ-free neonatal pig model and monoassociated pigs with a representative Gram-negative (Escherichia coli) or Gram-positive (Lactobacillus fermentum) commensal bacteria commonly isolated from the neonatal pig intestine. Our results show a consistent and marked effect of microbial colonization on the mRNA expression of intestinal chemokines, chemokine receptors, Foxp3 and TGF-beta. Moreover, as evidenced by in vitro experiments using two different cell lines, the pattern of regulation of CCL25 and CCL28 expression in the gut appears complex and suggests an additional role for in vivo factors. CONCLUSIONS/SIGNIFICANCE: Taken together, the results highlight the key role of bacterial microflora in the development of a functional intestinal immune system in an elegant and relevant model for human immune system development.


Asunto(s)
Quimiocinas CC/genética , Receptores CCR10/genética , Receptores CCR/genética , Animales , Quimiocinas/genética , Escherichia coli/genética , Factores de Transcripción Forkhead/genética , Regulación de la Expresión Génica , Humanos , Intestino Delgado/fisiología , Ratones , ARN Mensajero/genética , Ovinos , Porcinos , Linfocitos T Reguladores/fisiología , Transcripción Genética
12.
Gastroenterology ; 130(6): 1776-92, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16697741

RESUMEN

BACKGROUND & AIMS: Preterm birth and formula feeding are key risk factors associated with necrotizing enterocolitis (NEC) in infants, but little is known about intestinal conditions that predispose to disease. Thus, structural, functional, and microbiologic indices were used to investigate the etiology of spontaneous NEC development in preterm pigs. METHODS: Piglets were delivered by cesarean section at 92% gestation, reared in infant incubators, and fed infant formula or colostrum every 3 hours (n = 120) until tissue collection at 1-2 days of age. RESULTS: Clinical and histopathologic signs of NEC were observed in 57% of pigs fed FORMULA (26/46) and in 5% of pigs fed COLOSTRUM (2/38) (P < .05). Relative to COLOSTRUM, both healthy and sick FORMULA pigs had reduced intestinal villous heights, enzyme activities, nutrient absorption, and antioxidant levels and higher inducible nitric oxide synthetase activity (P < .05). In healthy pigs, mucosal microbial diversity remained low and diet independent. NEC pigs showed bacterial overgrowth, and a high mucosal density of Clostridium perfringens was detected in some but not all pigs. Germ-free conditions and antiserum against Clostridium perfringens toxin prevented intestinal dysfunction and NEC in formula-fed pigs, whereas the gut trophic factors, epidermal growth factor, and glucagon-like peptide 2 had limited effects. CONCLUSIONS: A subclinical, formula-induced mucosal atrophy and dysfunction predispose to NEC and bacterial overgrowth. The adverse feeding effects are colonization dependent and may be reduced by factors in colostrum that include antibodies against aggressive toxins such as those of Clostridium perfringens.


Asunto(s)
Enterocolitis Necrotizante/epidemiología , Enterocolitis Necrotizante/prevención & control , Fórmulas Infantiles/administración & dosificación , Intestino Delgado/metabolismo , Preñez , Animales , Animales Recién Nacidos , Secuencia de Bases , Biopsia con Aguja , Causalidad , Recuento de Colonia Microbiana , Calostro , ADN Bacteriano/análisis , Enterocolitis Necrotizante/patología , Femenino , Tracto Gastrointestinal/microbiología , Inmunohistoquímica , Absorción Intestinal/fisiología , Mucosa Intestinal/microbiología , Mucosa Intestinal/patología , Intestino Delgado/embriología , Intestino Delgado/patología , Datos de Secuencia Molecular , Reacción en Cadena de la Polimerasa/métodos , Embarazo , Nacimiento Prematuro , Probabilidad , Factores de Riesgo , Sensibilidad y Especificidad , Porcinos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA