Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Domest Anim Endocrinol ; 74: 106555, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32947201

RESUMEN

The objective of this experiment was to evaluate the effects of nutrient restriction and melatonin supplementation during mid-to-late gestation on maternal and fetal small intestinal carbohydrase activities in sheep. Ewes were randomly assigned to one of 4 dietary treatments arranged in a 2 × 2 factorial design. Ewes were fed to provide 100% (adequate; ADQ) or 60% (restricted; RES) of nutrient recommendations, and diets were supplemented with either no melatonin (control; CON) or 5 mg melatonin/d (melatonin; MEL). This resulted in 4 treatment groups: CON-ADQ (n = 7), CON-RES (n = 8), MEL-ADQ (n = 8), MEL-RES (n = 8). Treatments began on day 50 of gestation, and ewes were euthanized on day 130 for tissue collection. The maternal and fetal small intestine were collected and assayed for small intestinal carbohydrase activities. Data were analyzed using the GLM procedure of SAS with fetal sex, melatonin, nutrition, and the melatonin by nutrition interaction included in the model statement. There were no melatonin by nutrition interactions for maternal or fetal small intestinal protein concentration or carbohydrase activities (P ≥ 0.11). Dietary melatonin supplementation decreased (P = 0.03) maternal small intestinal protein concentration by 22.7% and increased (P = 0.03) maternal small intestinal glucoamylase, isomaltase, and maltase activity per gram protein by 45.5%, 41.3%, and 40.6%, respectively. Nutrient restriction from mid-to-late gestation did not influence (P ≥ 0.46) maternal small intestinal protein concentration, or maltase, isomaltase, and lactase activity. Maternal glucoamylase activity per gram intestine increased (P = 0.05) with nutrient restriction by 49.1%. Melatonin supplementation and maternal nutrient restriction did not influence (P ≥ 0.15) fetal small intestinal protein concentration, or glucoamylase, isomaltase, and lactase activity. Maternal nutrient restriction from mid-to-late gestation decreased (P = 0.05) fetal maltase activity per gram intestine by 20.5% but did not influence fetal maltase activity per gram protein. These data indicate that some maternal and fetal carbohydrases are influenced by nutrient restriction and melatonin supplementation in sheep. More information is needed to understand how nutritional and hormonal factors regulate digestive enzyme activity in ruminants to design improved maternal nutrition programs to optimize fetal growth and development while maintaining maternal productivity.


Asunto(s)
Alimentación Animal , Dieta , Glicósido Hidrolasas/metabolismo , Intestino Delgado/enzimología , Melatonina/farmacología , Preñez , Animales , Restricción Calórica , Femenino , Desarrollo Fetal , Feto/efectos de los fármacos , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Glicósido Hidrolasas/genética , Intestino Delgado/embriología , Melatonina/administración & dosificación , Embarazo , Ovinos
2.
Animal ; 8(10): 1677-83, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25231282

RESUMEN

Glutamate, which is one of the most important contributors to oxidative metabolism in the intestinal mucosa, is mainly transported by the excitatory amino acids transporters (EAATs) that are expressed in enterocytes. The objective of this study was to evaluate the effects of in ovo administration of l-trans pyrrolidine-2,4-dicarboxylic acid (l-trans-PDC), a potent competitive inhibitor of glutamate uptake by EAATs, on the growth of the small intestine in chicks. Two series of experiments were conducted with hatching eggs; 100 µl of various l-trans-PDC solutions (0, 0.075 or 0.225 mg/egg for the Control group, low-dose l-trans pyrrolidine 2,4-dicarboxylic acid group (L-PDC) or high-dose l-trans pyrrolidine 2,4-dicarboxylic acid group (H-PDC), respectively) was injected into the albumen sac of these hatching eggs before incubation. Hatchlings were sacrificed by cervical dislocation to determine the embryonic development in Experiment I, whereas the birds in Experiment II were raised or sampled at hatching, days 7 and 14 (D7 and D14) for further study. Gene expression in the small intestines was determined by real-time RT-PCR; and serum concentration of free amino acids was determined by an amino acid analyzer. The results showed that the hatchability was decreased by in ovo administration of l-trans-PDC. The small intestinal weights of the H-PDC group were decreased (P<0.05) at hatching and increased (P<0.05) on D7 and D14 compared with those in the Control group. In addition, the gene expression of EAAT2 in the completed or segmental small intestines was not changed (P>0.05); EAAT3 gene expression in the duodenum (P<0.05), jejunum (P=0.084) and ileum (P=0.060) on D14 was lower in the H-PDC group than in the Control group. Furthermore, the serum concentrations of free proline, threonine and phenylalanine but not glutamate or aspartate were increased (P<0.06) in H-PDC group. In conclusion, this paper is the first to report that in ovo administration of l-trans-PDC induces small intestinal growth retardation during the embryonic period and catch-up growth after hatching.


Asunto(s)
Embrión de Pollo/efectos de los fármacos , Pollos/crecimiento & desarrollo , Ácidos Dicarboxílicos/administración & dosificación , Antagonistas de Aminoácidos Excitadores/administración & dosificación , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Ácido Glutámico/metabolismo , Pirrolidinas/administración & dosificación , Animales , Peso Corporal , Embrión de Pollo/embriología , Embrión de Pollo/crecimiento & desarrollo , Pollos/genética , Pollos/metabolismo , Dieta/veterinaria , Intestino Delgado/efectos de los fármacos , Intestino Delgado/embriología , Intestino Delgado/crecimiento & desarrollo , Tamaño de los Órganos
3.
J Anim Physiol Anim Nutr (Berl) ; 98(4): 797-807, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25180375

RESUMEN

To determine how nutrient restriction and melatonin supplementation influence ewe and foetal hepatic and small intestinal energy use, 32 primiparous ewes on d 50 of gestation were fed 60% (RES) or 100% (ADQ) of NRC recommendations with 0 (CON) or 5 mg/d (MEL) of dietary melatonin. On d 130 of gestation, small intestine and liver were weighed and collected. Data were analysed as a completely randomized design with a 2 × 2 factorial arrangement of treatments. Liver weight (g/kg EBW) decreased (p = 0.02) in RES ewes. Jejunum weight (g/kg BW) increased (interaction p = 0.04) in ADQ-MEL ewes compared with all other treatments. Total in vitro O2 consumption (mol/min/tissue) and total citrate synthase activity (mol/min/tissue and mol/min/kg EBW) in liver decreased (p ≤ 0.03) in RES ewes. Oxygen consumption (mol/min/kg EBW) increased (interaction p = 0.02) in jejunum of ADQ-CON versus RES-MEL and ADQ-CON. Citrate synthase activity (mol/min/kg of EBW) increased (interaction p = 0.03) in jejunum of ADQ-MEL compared with RES-MEL and ADQ-CON. Foetal liver weight (g/kg BW) decreased (p = 0.02) in RES versus ADQ. Foetal small intestine weight (g/kg BW) decreased (interaction p = 0.05) in RES-MEL versus ADQ-MEL. Total O2 consumption (mol/min/tissue) and total citrate synthase activity (mol/min/kg of BW) in foetal liver decreased (p ≤ 0.05) in RES versus ADQ. Foetal small intestinal O2 consumption (mol/min/kg of BW) was greater (interaction p = 0.03) in RES-CON and ADQ-MEL than RES-MEL and ADQ-CON. Maternal nutrient restriction had a greater effect than melatonin supplementation on liver and jejunum mass and energy utilization in dams and foetuses. Because intestinal mass and energy utilization were more responsive to melatonin supplementation in ewes fed adequate nutrition compared with restricted ewes, melatonin may have limited use as a therapeutic supplement to help overcome potential negative effects of nutrient restriction.


Asunto(s)
Suplementos Dietéticos , Intestino Delgado/embriología , Hígado/embriología , Melatonina/farmacología , Ovinos/embriología , Ovinos/metabolismo , Alimentación Animal/análisis , Fenómenos Fisiológicos Nutricionales de los Animales , Animales , Dieta/veterinaria , Femenino , Privación de Alimentos , Intestino Delgado/metabolismo , Hígado/metabolismo , Melatonina/administración & dosificación , Consumo de Oxígeno , Embarazo
4.
Am J Physiol Regul Integr Comp Physiol ; 296(4): R1053-62, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19158412

RESUMEN

Transforming growth factor-beta (TGF-beta) plays a role in enterocyte proliferation control, cell differentiation, and immune regulation via binding to specific TGF-beta receptors (TGF-beta R) in the intestinal epithelium. Endogenous TGF-beta production is low in the intestine during the perinatal period, but some exogenous TGF-beta ligands are supplied by amniotic fluid intake in the fetus and by colostrum ingestion in the neonate. It is not clear, however, whether luminal TGF-beta receptors are present and functional at this critical time. We studied intestinal TGF-beta receptors by immunohistochemistry during the last 20% of gestation in pigs and in chronically catheterized fetuses following exposure to colostrum, milk, and amniotic fluid (control). In fetal pigs, the TGF-beta Rs were predominantly localized to the crypt epithelium, but staining intensity increased markedly just before term and shifted to the villous epithelium in newborn pigs, concurrently with marked increases in villous heights and crypt depths (+100-200%, P < 0.05). In contrast to previous observations in term newborn pigs, fetal pigs did not show any milk-induced change in TGF-beta receptor densities or localization, although a moderate increase in villous height was observed, relative to control (+25-50%, P < 0.05). We conclude that intestinal TGF-beta receptor density and localization are immature and unresponsive to TGF-beta containing milk diets in prenatal pigs. Immaturity of TGF-beta-mediated immune regulation may play a role in the increased sensitivity of preterm neonates to diet-induced intestinal inflammatory disorders.


Asunto(s)
Proliferación Celular , Nutrición Enteral , Mucosa Intestinal/metabolismo , Intestino Delgado/metabolismo , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Líquido Amniótico/metabolismo , Animales , Animales Recién Nacidos , Calostro/metabolismo , Femenino , Edad Gestacional , Humanos , Inmunidad Mucosa , Fórmulas Infantiles/metabolismo , Recién Nacido , Mucosa Intestinal/embriología , Mucosa Intestinal/inmunología , Intestino Delgado/embriología , Intestino Delgado/inmunología , Proteínas de la Leche/metabolismo , Embarazo , Porcinos
5.
J Nutr Biochem ; 20(1): 17-25, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18479897

RESUMEN

The current study utilized Ussing chambers to examine the impact of supplementing maternal gestation and/or lactation diets with n-3 polyunsaturated fatty acids (PUFA) provided via a protected fish oil (PFO) product on intestinal fatty acid profiles and ex vivo glucose uptake in the jejunum of weanling piglets. Jejunum tissues were enriched with n-3 PUFA as a result of feeding the sows the PFO during gestation and/or lactation (P<.05). Glucose uptake improved by twofold (P<.042) in intestinal preparations obtained from the offspring of sows fed PFO during gestation or throughout gestation/lactation versus lactation alone. This was also reflected in the jejunum protein expressions of glucose transporter 2 (GLUT2) and sodium-dependent glucose transporter 1 (SGLT1). Furthermore, adding docosahexaenoic acid (DHA) or an AMP-activated protein kinase (AMPK) agonist to the chamber buffer improved glucose uptake (P<.05) in intestinal preparations obtained from the offspring fed the control diet, devoid of the PFO product and containing minimal concentrations of n-3 PUFA. Collectively, these data indicate two important points. First, long-term exposure to n-3 PUFA via the maternal gestation diet effectively enhances glucose uptake in the weanling piglet, and the underlying mechanism may be associated with changes in the intestinal fatty acid profile. Secondly, there is an apparent direct and acute effect of DHA that is achieved within a time frame that precludes substantial changes in the intestinal fatty acid profile. Additionally, both mechanisms may involve activation of AMPK. Thus, n-3 PUFA delivered in utero and postnatally via the maternal diet may help the offspring adapt quickly to rapidly changing diets early in life and allow optimal nutrient uptake.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Ácidos Grasos Insaturados/metabolismo , Glucosa/farmacocinética , Absorción Intestinal/efectos de los fármacos , Intestino Delgado/embriología , Animales , Ácidos Docosahexaenoicos/farmacología , Activación Enzimática , Femenino , Glucosa/metabolismo , Transportador de Glucosa de Tipo 2/metabolismo , Yeyuno/embriología , Microvellosidades/metabolismo , Embarazo , Transportador 1 de Sodio-Glucosa/metabolismo , Porcinos
6.
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
7.
Radiología (Madr., Ed. impr.) ; 47(5): 237-252, sept. 2005. ilus
Artículo en Es | IBECS | ID: ibc-040219

RESUMEN

La malrotación intestinal con su propensión al vólvulo representa un desafío diagnóstico en pacientes pediátricos. El diagnóstico precoz es de suma importancia para conseguir un buen resultado. En este artículo se revisan e ilustran los hallazgos radiológicos de la malrotación y el vólvulo de intestino medio en la infancia y adolescencia, y se discuten la embriología, la presentación clínica, el manejo radiológico y la evolución de este proceso. El diagnóstico de malrotación y vólvulo debe tenerse en cuenta siempre que se evalúe a un lactante o niño con vómitos y dolor abdominal, particularmente si los vómitos son biliosos. Un tránsito baritado generalmente dará el diagnóstico. La ecografía es muy útil, si bien no es suficientemente segura para excluir el diagnóstico. Sin embargo, el «signo del remolino» es un signo altamente sensible para el diagnóstico del vólvulo de intestino medio. El radiólogo siempre debe tener presente que esta complicación puede ocurrir a cualquier edad


Intestinal malrotation with its propensity to volvulus represents a diagnostic challenge in pediatric patients. Early diagnosis is of extreme importance for achieving a good outcome. This article reviews and illustrates the radiologic findings for malrotation and volvulus of the small bowel in childhood and adolescence. The embryology, clinical presentation, radiologic management, and evolution of this process are discussed. The diagnosis of malrotation and volvulus should be considered whenever a nursing baby or child presents with vomiting and abdominal pain, especially if the vomit is bilious. A barium follow-through usually provides the diagnosis. Ultrasound is very useful, although it cannot safely rule out the diagnosis. However, the "whirlpool sign" is a highly sensitive sign for the diagnosis of small bowel volvulus. The radiologist needs to bear in mind that this complication can occur at any age


Asunto(s)
Masculino , Femenino , Niño , Humanos , Vólvulo Intestinal/diagnóstico , Intestino Delgado/anomalías , Diagnóstico por Imagen/métodos , Dolor Abdominal/etiología , Vólvulo Intestinal/etiología , Obstrucción Intestinal/etiología , Intestino Delgado/fisiopatología , Rotación , Enema , Intestino Delgado/embriología , Vómitos/etiología
8.
J Nutr Biochem ; 16(6): 339-46, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15936645

RESUMEN

A 303-bp cDNA of intestinal zinc exporter (ZnT1) was isolated from chicken jejunum by reverse transcriptase-polymerase chain reaction and sequenced, and showed 42% homology to Homo sapiens and Rattus novergicus intestinal ZnT1 genes. This specific probe was used to examine the effect of zinc-methionine (ZnMet) administration on the mRNA expression of ZnT1 and on small intestinal development and functionality. In this study, ZnMet was injected into the naturally consumed amniotic fluid of 17-day-old chicken embryos. The ZnT1 gene showed an approximately 200% increase in its mRNA levels from 48 h post-ZnMet injection, as compared to the control. An analysis of the gene expression of the brush-border enzymes and transporters showed increased mRNA expression of sucrase isomaltase, leucine-aminopeptidase, sodium-glucose cotransporter and Na+K+ATPase transporter (Na+K+ATPase) from 48 h post-ZnMet injection, in comparison to controls. Significant increases (P<.05) in the biochemical activity of the brush-border enzymes and transporters, and in jejunal villus surface area were detected from day of hatch (96 h post-ZnMet injection) as compared to controls. These results suggest that ZnMet administration into prenatal intestine via injection into the amniotic fluid enhances intestinal development and improves its functionality.


Asunto(s)
Pollos/metabolismo , Intestino Delgado/crecimiento & desarrollo , Proteínas de la Membrana/fisiología , Zinc/fisiología , Secuencia de Aminoácidos , Amnios , Animales , Proteínas de Transporte de Catión , Embrión de Pollo/crecimiento & desarrollo , Embrión de Pollo/metabolismo , Pollos/crecimiento & desarrollo , ADN Complementario/análisis , Perfilación de la Expresión Génica , Inyecciones , Intestino Delgado/embriología , Intestino Delgado/enzimología , Intestino Delgado/metabolismo , Proteínas de la Membrana/biosíntesis , Proteínas de la Membrana/genética , Metionina/administración & dosificación , Metionina/análogos & derivados , Metionina/farmacocinética , Microvellosidades/enzimología , Microvellosidades/metabolismo , Datos de Secuencia Molecular , Compuestos Organometálicos/administración & dosificación , Compuestos Organometálicos/farmacocinética , ARN Mensajero/biosíntesis , Ratas , Zinc/metabolismo
9.
J Membr Biol ; 206(1): 9-16, 2005 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16440177

RESUMEN

Oral L-carnitine supplementation is commonly used in sports nutrition and in medicine; however, there is controversy regarding the mechanisms that mediate intestinal L-carnitine transport. We have previously reported that the Na(+)/L-carnitine transporter OCTN2 is present in the small intestinal apical membrane. Herein we aimed to find out if this step of intestinal L-carnitine absorption is ontogenically regulated, and if so, to determine the molecular mechanism(s) involved. L-[(3)H]-Carnitine uptake was measured in the jejunum and ileum of fetuses (E17 and E21), newborn (1 day-old), suckling (15 day-old), weaning (1 month-old) and adult (2 and 6 month-old) Wistar rats. Both, Na(+) -dependent and Na(+) -independent L-carnitine uptake rates, normalized to intestinal weight, significantly increased during the late gestation period, and then declined during the suckling period. After weaning, the rate of Na(+) -dependent L-carnitine uptake is no longer measurable. In E21- fetuses and newborn rats, L-carnitine uptake was higher in the ileum than in the jejunum. The decline in Na(+) -dependent L-carnitine uptake with maturation was mediated via a decrease in the V(max) of the uptake process with no change in its apparent K(m). Semi-quantitative RT-PCR assays showed that OCTN2 mRNA levels were significantly higher in E21-fetuses and newborn rats compared to suckling rats, which were in turn significantly higher than that in adult rats. Neither retardation of weaning nor L-carnitine supplementation prevented the down-regulation of Na(+)/L-carnitine transport activity. The results demonstrate for the first time that intestinal Na(+) -dependent L-carnitine uptake activity is under genetic regulation at the transcriptional level.


Asunto(s)
Envejecimiento/metabolismo , Carnitina/farmacocinética , Intestino Delgado/embriología , Intestino Delgado/metabolismo , Proteínas de Transporte de Catión Orgánico/metabolismo , Sodio/metabolismo , Administración Oral , Animales , Animales Recién Nacidos , Carnitina/administración & dosificación , Tasa de Depuración Metabólica , Ratas , Ratas Wistar , Miembro 5 de la Familia 22 de Transportadores de Solutos , Distribución Tisular
10.
J Pediatr Surg ; 39(6): 891-7; discussion 891-7, 2004 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15185220

RESUMEN

BACKGROUND/PURPOSE: Intrauterine growth retardation (IUGR) infants have impaired gastrointestinal function with resultant feeding difficulties and predisposition to necrotizing enterocolitis. Supplemented amniotic fluid swallowed by the developing fetus is a potential prenatal treatment for IUGR. Rabbits have naturally occurring IUGR fetuses based on uterine position. To determine intestinal response to epidermal growth factor (EGF) infusion, this rabbit model of IUGR was studied. METHODS: Eight pregnant rabbits underwent placement of intraamniotic catheters into 2 normal and 2 IUGR fetuses per mother on gestational day 24 of a 31-day gestation. Miniosmotic pumps infused either EGF (about 300 microg/kg/d) or control solution forming 4 study groups (EGF-Favored [Fav] v. Cont-Fav; EGF-IUGR v. Cont-IUGR). On gestational day 31, the fetal gastrointestinal tracts were harvested for analysis. Intestinal epithelial cell proliferation was studied by 5-bromo-2-deoxyuridine (BrdU) incorporation, villus heights were measured, and EGF mRNA was measured by reverse transcriptase polymerase chain reaction (RT-PCR). Statistical analysis was performed using Students' t test. RESULTS: Fetal survival rate was 87%. EGF-IUGR fetal weights were increased compared with Cont-IUGR fetuses. EGF infusion significantly increased IUGR fetal small intestinal villus height and BrdU-positive small intestinal (SI) crypt cells, all approaching Cont-Fav levels. EGF mRNA was expressed throughout the gastrointestinal tract. CONCLUSIONS: Supplemental amniotic EGF normalizes fetal weight and intestinal proliferation in the IUGR fetal rabbit. The inclusion of EGF in supplemental amniotic feeding solutions is supported.


Asunto(s)
Factor de Crecimiento Epidérmico/uso terapéutico , Retardo del Crecimiento Fetal/prevención & control , Intestino Delgado/embriología , Líquido Amniótico , Animales , Peso al Nacer/efectos de los fármacos , Evaluación Preclínica de Medicamentos , Factor de Crecimiento Epidérmico/administración & dosificación , Factor de Crecimiento Epidérmico/biosíntesis , Factor de Crecimiento Epidérmico/genética , Femenino , Retardo del Crecimiento Fetal/tratamiento farmacológico , Humanos , Bombas de Infusión Implantables , Embarazo , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Conejos , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/uso terapéutico
11.
Dev Growth Differ ; 40(2): 177-88, 1998 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-9572360

RESUMEN

The present study used a molecular approach toward understanding the mechanism of hormone- and region-dependent remodeling of the small intestine during metamorphosis of Xenopus laevis. A protein spot was noticed on a two-dimensional polyacrylamide gel as a protein whose expression was metamorphic stage- and region-dependent. The protein was identified as the Xenopus homolog (Xcalbindin) of chick calbindin D28k. Xcalbindin expression in the intestine was restricted to absorptive cells in the posterior part, being detectable at stages 49-61, not detectable at stages 62-63, detectable again at stages 64-66, and finally becoming undetectable in the adult. During spontaneous metamorphosis, the level of Xcalbindin mRNA was significantly increased between stages 57 and 58, dramatically reduced at stage 59, and the mRNA was undetectable from stages 60-63, after which it was weakly re-expressed until the end of metamorphosis. Such up- and down-regulation of Xcalbindin mRNA was induced precociously by exogenous thyroid hormone. These results indicated that Xcalbindin is a specific marker of the differentiated absorptive cells of the intestine. Immunohistochemistry with specific antibodies against Xcalbindin demonstrated that precursor cells of adult intestinal epithelial cells expressed Xcalbindin. Considering these results, the origin of adult intestinal epithelial cells was discussed.


Asunto(s)
Mucosa Intestinal/embriología , Intestino Delgado/embriología , Metamorfosis Biológica/genética , Proteína G de Unión al Calcio S100/genética , Xenopus laevis/embriología , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Calbindinas , ADN Complementario/genética , Células Epiteliales/metabolismo , Mucosa Intestinal/metabolismo , Larva , Datos de Secuencia Molecular , Peso Molecular , ARN Mensajero/biosíntesis , Proteína G de Unión al Calcio S100/biosíntesis , Triyodotironina/farmacología , Xenopus laevis/genética
12.
Equine Vet J Suppl ; (24): 40-50, 1997 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-9355801

RESUMEN

At birth, the mammalian gastrointestinal tract (GIT) must be able to support a shift from mainly parenteral nutrition in the fetus (via the placenta) to enteral nutrition in the neonate. In the perinatal period the GIT therefore undergoes enhanced growth as well as morphological and functional differentiation, and this maturational programme is influenced by a complex interplay of local, systemic and luminal factors. This review shows how systemic and luminal factors may influence GIT development in the perinatal period of the pig and sheep, two long-gestation species. Adrenocortical hormones play a pivotal role in the prepartum maturation of the GIT in addition to their better known effects on the development of many other tissues and body systems. More particularly, in the fetal pig and sheep, the prenatal development of gastric acid and gastrin secretion, and of GIT hydrolase activities (chymosin, pepsin, amylase, lactase, aminopeptidases) is influenced by cortisol. Additionally, glucocorticoids exert effects throughout the GIT by influencing morphological, cytological, and functional differentiation. Since the GIT epithelial cells comprise a renewing cell population there are also changes in cell kinetics. In addition to systemic factors, the presence of growth factors, hormones and nutrients from swallowed amniotic fluid (fetus) and colostrum (neonate) may influence GIT development. In utero, fetal fluid ingestion has been shown to modulate tissue growth, macromolecule and immunoglobulin transport, enterocyte differentiation, cell turnover and activity of brush-border hydrolases. These effects may be mediated via regulatory peptides (e.g. insulin-like growth factor I, gastrin-releasing peptides, insulin, epidermal growth factor, gastrin). A physiological role of luminally derived growth factors is supported by a number of unique structural and functional adaptations of the GIT in the fetus and neonate (low luminal proteolysis, intestinal macromolecule transport). Thus, in the pig and sheep, both systemic and luminal factors appear to play critical roles in GIT development in the perinatal period.


Asunto(s)
Corticoesteroides/fisiología , Fenómenos Fisiológicos del Sistema Digestivo , Sistema Digestivo/embriología , Sustancias de Crecimiento/fisiología , Ovinos/embriología , Porcinos/embriología , Líquido Amniótico/fisiología , Animales , Animales Recién Nacidos/crecimiento & desarrollo , Animales Recién Nacidos/fisiología , Calostro/fisiología , Sistema Digestivo/crecimiento & desarrollo , Desarrollo Embrionario y Fetal/fisiología , Femenino , Glucocorticoides/fisiología , Intestino Delgado/embriología , Intestino Delgado/crecimiento & desarrollo , Intestino Delgado/fisiología , Páncreas/embriología , Páncreas/crecimiento & desarrollo , Páncreas/fisiología , Embarazo , Ovinos/crecimiento & desarrollo , Ovinos/fisiología , Estómago/embriología , Estómago/crecimiento & desarrollo , Estómago/fisiología , Porcinos/crecimiento & desarrollo , Porcinos/fisiología
13.
Oncogene ; 13(3): 547-59, 1996 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-8760296

RESUMEN

Recently, we isolated a new src family member from a rat small intestinal cDNA library which by RNase protection analysis is selectively expressed in the columnar epithelium of gut. Complete nucleotide sequencing of the gastrointestinal associated tyrosine kinase (gtk) has revealed that it is a rat homologue of frk/rak-a fyn related human tyrosine kinase. Unlike frk/rak, gtk is myristylated, in vivo. Furthermore, by immunohistochemical analysis, the kinase is concentrated in the brush border membranes of epithelial cells, throughout the maturation axis of the adult small intestine. In vitro analysis revealed that gtk kinase activity is present in intestinal cells throughout their maturation, suggesting that the enzyme might influence signal transduction pathways in both mitotic and post-mitotic states. Gtk is expressed in all regions of the gastrointestinal tract which contain columnar epithelium, but is absent in the stratified epithelium of the esophagus. Moreover, during gestation, the kinase dramatically appears at high levels in plasma membranes, at the time of transition of gut cells from an undifferentiated to a simple columnar phenotype. After solubilization of cellular membranes with Triton X-100, sucrose gradient analysis of gtk revealed that it partitions differently than c-yes, demonstrating that the brush border src kinases associate with different components of the plasma membranes. These findings suggest that gtk plays a specialized role in the growth/differentiation of gut columnar epithelial cells.


Asunto(s)
Intestino Delgado/enzimología , Proteínas Tirosina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Células 3T3/fisiología , Adulto , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Fraccionamiento Celular , Membrana Celular/enzimología , ADN Complementario/análisis , Epitelio/enzimología , Humanos , Intestino Delgado/embriología , Intestino Delgado/ultraestructura , Ratones , Microvellosidades/enzimología , Datos de Secuencia Molecular , Octoxinol , Proteínas Tirosina Quinasas/genética , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas c-fyn , Ratas , Homología de Secuencia de Aminoácido , Solubilidad , Acetato de Tetradecanoilforbol/farmacología
15.
Eur J Biochem ; 223(2): 701-9, 1994 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-8055940

RESUMEN

Intestinal genes whose expression is regulated during development and differentiation were identified and cloned from a rat villi cDNA library using a subtracted cDNA probe. The isolated clones are transcribed in the fully differentiated intestinal epithelium 21 days after birth and absent or poorly expressed in the fetal gut at 15 days of gestation. Two of the DRI (differentially-expressed in rat intestine) genes are novel, while the others encode the microvillar protein ezrin and intracellular carrier proteins for retinol and fatty acids. Expression of the newly isolated DRI27 and DRI42 clones parallels epithelial differentiation during development and it is more pronounced in the distal portions of the small intestine. In situ hybridization experiments indicate that the DRI mRNAs are expressed in the differentiated cell types of the gut epithelium. Moreover, the expression of DRI27 and DRI42 is strongly related to the stage of epithelial differentiation during gut development. This relationship holds true also for the expression of DRI42 in other tissues. These clones will be a valuable tool to identify regulatory sequences and factors responsible for confining gene expression to the differentiated epithelial cell types in mammalian small intestine.


Asunto(s)
Regulación de la Expresión Génica/genética , Intestino Delgado/metabolismo , Animales , Northern Blotting , Southern Blotting , Diferenciación Celular , Vellosidades Coriónicas/metabolismo , Clonación Molecular , ADN Complementario/genética , Desarrollo Embrionario y Fetal/genética , Inmunohistoquímica , Hibridación in Situ , Intestino Delgado/citología , Intestino Delgado/embriología , Reacción en Cadena de la Polimerasa , Ratas , Ratas Sprague-Dawley , Distribución Tisular , Transcripción Genética/genética
16.
J Pediatr Surg ; 28(10): 1239-44, 1993 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-8263680

RESUMEN

As fetal swallowing is documented in utero, supplementation of the ingested amniotic fluid with nutrients or hormones has been postulated as a potential prenatal treatment for intrauterine growth retardation (IUGR). To study the effect of epidermal growth factor (EGF) on the developing fetal small intestine, 12 pregnant rabbits underwent operation on day 24 of a normal 31-day gestation. Bilateral ovarian end fetuses underwent catheterization of their respective amniotic cavities with attachment to a miniosmotic pump. Study fetuses received recombinant human EGF at approximately 300 micrograms/kg/d for 1 week; controls received carrier solution only at an equivalent rate. On gestational day 31, fetuses were delivered by cesarean section and somatic measurements were recorded. The small intestine was harvested and proximal, middle, and distal regions were analyzed for lactase and maltase enzyme activity. Additionally, the uptake of radiolabeled glucose and proline was measured by a standard everted mucosal sleeve technique for each segment. Results were analyzed by Student's paired t test and reported as mean +/- SEM. Nine fetal pairs survived (75%). Small intestinal (SI) length was increased in EGF fetuses (54.8 +/- 1.9 cm) versus control (50.4 +/- 2.7 cm) (P = .02). Lactase activity, reported as UE/g protein, was significantly increased in the proximal segments in the EGF-infused fetuses; maltase was significantly increased in both the proximal and middle segments (P < .05).(ABSTRACT TRUNCATED AT 250 WORDS)


Asunto(s)
Disacaridasas/efectos de los fármacos , Factor de Crecimiento Epidérmico/administración & dosificación , Intestino Delgado/efectos de los fármacos , Amnios , Animales , Transporte Biológico/efectos de los fármacos , Disacaridasas/metabolismo , Evaluación Preclínica de Medicamentos , Femenino , Retardo del Crecimiento Fetal/tratamiento farmacológico , Glucosa/metabolismo , Humanos , Bombas de Infusión Implantables , Intestino Delgado/embriología , Intestino Delgado/metabolismo , Embarazo , Prolina/efectos de los fármacos , Prolina/metabolismo , Conejos , Proteínas Recombinantes/administración & dosificación
17.
Clin Chim Acta ; 170(2-3): 219-25, 1987 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-3436056

RESUMEN

Human alkaline phosphatases extracted with butanol from liver, kidney and placenta, and from foetal and adult small intestine each contain fragments with molecular masses within the range of approximately 8 kDa to 20 kDa which can be removed by digestion with bromelain. However, in the case of adult intestine, this fragment (which is presumed to represent a membrane-binding domain) can only be demonstrated in tissue extracted immediately after removal at operation. Similar fragments are also present in foetal intestinal phosphatase in amniotic fluid, and in liver and bone alkaline phosphatases recovered from serum. Again, however, adult intestinal phosphatase from serum differs in the absence of the bromelain-sensitive fragment. These observations indicate differences in the ways in which intestinal and non-intestinal alkaline phosphatases gain access to the circulation, and also have implications for structural studies on intestinal phosphatase extracted post mortem from adult tissue.


Asunto(s)
Fosfatasa Alcalina/metabolismo , Bromelaínas/farmacología , Intestino Delgado/enzimología , Adulto , Huesos/embriología , Huesos/enzimología , Feto , Humanos , Intestino Delgado/embriología , Riñón/embriología , Riñón/enzimología , Hígado/embriología , Hígado/enzimología , Peso Molecular , Placenta/enzimología
18.
Ciba Found Symp ; (70): 201-19, 1979.
Artículo en Inglés | MEDLINE | ID: mdl-261521

RESUMEN

An important adaptation of the gastrointestinal tract to the extrauterine environment is its development of a mucosal barrier against the penetration of harmful substances (bacteria, toxins and antigens) present within the intestinal lumen. At birth, the newborn infant must be prepared to deal with bacterial colonization of the gut, with formation of toxic byproducts of bacteria and viruses (enterotoxins and endotoxins) and with the ingestion of antigens (milk proteins). These potentially noxious substances if allowed to penetrate the mucosal epithelial barrier under pathological conditions can cause inflammatory and allergic reactions which may result in gastrointestinal and systemic disease states. To combat the potential danger of invasion across the mucosal barrier the infant must develop an elaborate system of defence mechanisms within the lumen and on the luminal mucosal surface which act to control and maintain the epithelium as an impermeable barrier to uptake of macromolecular antigens. These defences include a unique immunological system adapted to function in the complicated milieu of the intestine as well as other non-immunological processes such as a gastric barrier, intestinal surface secretions, peristaltic movement and natural antibacterial substances (lysozyme, bile salts) which also help to provide maximum protection for the intestinal surface. Unfortunately, during the immediate postpartum period, particularly for premature and small-for-dates infants, this elaborate local defence system is incompletely developed. As a result of the delay in the maturation of the mucosal barrier newborn infants are particularly vulnerable to pathological penetration by harmful intraluminal substances. The consequences of altered defence are susceptibility to infection and the potential for hypersensitivity reactions and for formation of immune complexes. With these reactions comes the potential for developing life-threatening diseases such as necrotizing enterocolitis, sepsis and hepatitis. Fortunately, 'nature' has provided a means for passively protecting the 'vulnerable' newborn against dangers of a deficient intestinal defence system, namely human milk. It is now increasingly apparent that human milk contains not only antibodies and viable leucocytes but many other substances which can interfere with bacterial colonization and prevent antigen penetration.


Asunto(s)
Absorción Intestinal , Mucosa Intestinal/inmunología , Intestino Delgado/inmunología , Pinocitosis , Animales , Antígenos , Calostro/fisiología , Femenino , Feto , Humanos , Inmunidad Materno-Adquirida , Recién Nacido , Intestino Delgado/embriología , Intestino Delgado/fisiología , Embarazo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA