Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 114
Filtrar
1.
Funct Integr Genomics ; 20(2): 277-291, 2020 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-31586277

RESUMO

The alterations of the intestinal proteome were observed in intrauterine growth restriction (IUGR) piglets during early life by gel-based approaches. Nevertheless, how IUGR affects the intestinal membrane proteome during neonatal development remains unclear. Here, we applied the iTRAQ-based proteomics technology and biochemical analysis to investigate the impact of IUGR on the membrane proteome of the jejunal mucosa in the piglets. Three hundred sixty-one membrane proteins were screened by functional prediction. Among them, eight, five, and one differentially expressed membrane proteins were identified between IUGR and NBW piglets at day 0, day 7, and day 21 after birth, respectively. Differentially expressed membrane proteins (DEMPs) including F1SBL3, F1RRW8, F1S539, F1S2Z2, F1RIR2, F1RUF2 I3LP60, Q2EN79, and F1SIH8 were reduced while the relative abundance of I3L6A2, F1SCJ1, F1RI18, I3LRJ7, and F1RNN0 were increased in IUGR piglets than NBW piglets. From the aspects of function, F1RRW8, F1S539, F1S2Z2, and F1RIR2 are mainly associated with D2 dopamine receptor binding, transmembrane transport of small molecules, signal transduction, and translocation of GLUT4, respectively, and F1SIH8, I3LRJ7, and F1RNN0 are related to autophagy, metabolism of vitamins, and intracellular protein transport. Additionally, IUGR decreased the level of proteins (F1RRW8, Q2EN79, and F1RI18) that are involved in response to oxidative stress.


Assuntos
Retardo do Crescimento Fetal/patologia , Jejuno/embriologia , Jejuno/crescimento & desenvolvimento , Proteoma/análise , Animais , Animais Recém-Nascidos , Peso ao Nascer , Feminino , Perfilação da Expressão Gênica , Mucosa Intestinal/patologia , Espectrometria de Massas , Estresse Oxidativo , Gravidez , Prenhez , Proteômica , Suínos
2.
J Med Case Rep ; 12(1): 261, 2018 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-30213270

RESUMO

BACKGROUND: Reversed rotation of the midgut is the rarest variation of midgut malrotations, which are congenital disorders that result from aberrant rotation and fixation of the midgut during embryological development. Common complications of these disorders are small bowel obstruction by volvulus or peritoneal bands, usually occurring in early infancy. CASE PRESENTATION: A 23-year-old Caucasian woman presented with recurrent abdominal pain. A contrast-enhanced multidetector computed tomography study revealed a novel variant of reversed rotation of the midgut. Besides the specific finding of a retro-arterial transverse colon, we also found the proximal jejunum to cross posterior to the mesenteric root, a variation that has not been reported in the literature so far. In this case, substantial symptomatic relief was achieved with conservative management. CONCLUSIONS: The hypothesis of a double reversed rotation of the pre-arterial segment of the umbilical loop around the superior mesenteric artery axis provides a possible explanation for this anomaly. There is no evidence-based consensus on the management of patients presenting with non-symptomatic or mildly symptomatic intestinal malrotations. In this case, radiologic and clinical presentations excluded acute small bowel obstruction, and surgical intervention was avoided.


Assuntos
Dor Abdominal/etiologia , Colo Transverso/diagnóstico por imagem , Anormalidades do Sistema Digestório/diagnóstico por imagem , Volvo Intestinal/diagnóstico por imagem , Jejuno/diagnóstico por imagem , Dor Abdominal/terapia , Colo Transverso/anormalidades , Colo Transverso/enzimologia , Anormalidades do Sistema Digestório/complicações , Anormalidades do Sistema Digestório/embriologia , Feminino , Humanos , Volvo Intestinal/congênito , Jejuno/anormalidades , Jejuno/embriologia , Tomografia Computadorizada Multidetectores , Recidiva , Rotação , Adulto Jovem
3.
Mol Genet Genomics ; 293(4): 807-818, 2018 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-29392408

RESUMO

Intrauterine growth restriction (IUGR) may elicit a series of postnatal body developmental and metabolic diseases due to their impaired growth and development in the mammalian embryo/fetus during pregnancy. In the present study, we hypothesized that IUGR may lead to abnormally regulated DNA methylation in the intestine, causing intestinal dysfunctions. We applied reduced representation bisulfite sequencing (RRBS) technology to study the jejunum tissues from four newborn IUGR piglets and their normal body weight (NBW) littermates. The results revealed extensively regional DNA methylation changes between IUGR/NBW pairs from different gilts, affecting dozens of genes. Hiseq-based bisulfite sequencing PCR (Hiseq-BSP) was used for validations of 19 genes with epigenetic abnormality, confirming three genes (AIFM1, MTMR1, and TWIST2) in extra samples. Furthermore, integrated analysis of these 19 genes with proteome data indicated that there were three main genes (BCAP31, IRAK1, and AIFM1) interacting with important immunity- or metabolism-related proteins, which could explain the potential intestinal dysfunctions of IUGR piglets. We conclude that IUGR can lead to disparate DNA methylation in the intestine and these changes may affect several important biological processes such as cell apoptosis, cell differentiation, and immunity, which provides more clues linking IUGR and its long-term complications.


Assuntos
Metilação de DNA , Retardo do Crescimento Fetal/metabolismo , Jejuno/embriologia , Animais , Feminino , Retardo do Crescimento Fetal/patologia , Jejuno/patologia , Gravidez , Sus scrofa
4.
J Tissue Eng Regen Med ; 12(1): 252-264, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-28084682

RESUMO

An ex vivo experimental strategy that replicates in vivo intestinal development would in theory provide an accessible setting with which to study normal and dysmorphic gut biology. The current authors recently described a system in which mouse embryonic jejunal segments were explanted onto semipermeable platforms and fed with chemically defined serum-free media. Over 3 days in organ culture, explants formed villi and they began to undergo spontaneous peristalsis. As defined in the current study, the wall of the explanted gut failed to form a robust longitudinal smooth muscle (SM) layer as it would do in vivo over the same time period. Given the role of transforming growth factor ß1 (TGFß1) in SM differentiation in other organs, it was hypothesized that exogenous TGFß1 would enhance SM differentiation in these explants. In vivo, TGFß receptors I and II were both detected in embryonic longitudinal jejunal SM cells and, in organ culture, exogenous TGFß1 induced robust differentiation of longitudinal SM. Microarray profiling showed that TGFß1 increased SM specific transcripts in a dose dependent manner. TGFß1 proteins were detected in amniotic fluid at a time when the intestine was physiologically herniated. By analogy with the requirement for exogenous TGFß1 for SM differentiation in organ culture, the TGFß1 protein that was demonstrated to be present in the amniotic fluid may enhance intestinal development when it is physiologically herniated in early gestation. Future studies of embryonic intestinal cultures should include TGFß1 in the defined media to produce a more faithful model of in vivo muscle differentiation. Copyright © 2017 The Authors Journal of Tissue Engineering and Regenerative Medicine Published by John Wiley & Sons, Ltd.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Jejuno/embriologia , Miócitos de Músculo Liso/citologia , Fator de Crescimento Transformador beta1/farmacologia , Animais , Proliferação de Células/efeitos dos fármacos , Embrião de Mamíferos/efeitos dos fármacos , Humanos , Camundongos , Miócitos de Músculo Liso/efeitos dos fármacos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Transcriptoma/genética
5.
Am J Physiol Gastrointest Liver Physiol ; 312(5): G434-G442, 2017 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-28280141

RESUMO

An important characteristic of intrauterine growth restricted (IUGR) neonate is the impaired intestinal barrier function. With the use of a pig model, this study was conducted to identify the responsible microRNA (miRNA) for the intestinal damage in IUGR neonates through comparing the miRNA profile of IUGR and normal porcine neonates and to investigate the regulation mechanism. Compared with the normal ones, we identified 83 upregulated and 76 downregulated miRNAs in the jejunum of IUGR pigs. Notably, IUGR is associated with profoundly increasesd miR-29 family and decreased expression of extracellular matrix (ECM) and tight junction (TJ) proteins in the jejunum. Furthermore, in vitro study using theporcine intestinal epithelial cell line (IPEC-1) showed that inhibition of miR-29a expression could improve the monolayer integrity by increasing cell proliferation and transepithelial resistance. Also, overexpression/inhibition of miR-29a in IPEC-1 cells can suppress/increase the expression of integrin-ß1, collagen I, collagen IV, fibronectin, and claudin 1, both at transcriptional and translational levels. Subsequent luciferase reporter assay confirmed a direct interaction between miR-29a and the 3'-untranslated regions of these genes. In conclusion, this study reveals that IUGR-impaired intestinal barrier function is associated with downregulated ECM and TJ protein expression mediated by the upregulation of miR-29a.NEW & NOTEWORTHY Intrauterine growth restricted (IUGR) remains a major problem for both human health and animal production due to its association with high rates of preweaning morbidity and mortality. We have identified the abnormal expression of microRNA-29a (miR-29a) in the small intestine of IUGR neonates, as well as its targets and mechanisms. These results provide new information about biological characteristics of IUGR-affected intestinal dysfunction and can lead to the development of potentially solution for preventing and treating IUGR in the future.


Assuntos
Retardo do Crescimento Fetal/metabolismo , Absorção Intestinal , Mucosa Intestinal/metabolismo , Jejuno/metabolismo , Animais , Retardo do Crescimento Fetal/patologia , Mucosa Intestinal/embriologia , Mucosa Intestinal/patologia , Jejuno/embriologia , Jejuno/patologia , MicroRNAs/metabolismo , Suínos
6.
Cell Tissue Res ; 365(2): 367-79, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27053245

RESUMO

Although gut flexures characterize gut morphology, the mechanisms underlying flexure formation remain obscure. Previously, we analyzed the mouse duodenojejunal flexure (DJF) as a model for its formation and reported asymmetric morphologies between the inner and outer bending sides of the fetal mouse DJF, implying their contribution to DJF formation. We now present the extracellular matrix (ECM) as an important factor for gut morphogenesis. We investigate ECM distribution during mouse DJF formation by histological techniques. In the intercellular space of the gut wall, high Alcian-Blue positivity for proteoglycans shifted from the outer to the inner side of the gut wall during DJF formation. Immunopositivity for fibronectin, collagen I, or pan-tenascin was higher at the inner than at the outer side. Collagen IV and laminins localized to the epithelial basement membrane. Beneath the mesothelium at the pre-formation stage, collagen IV and laminin immunopositivity showed inverse results, corresponding to the different cellular characteristics at this site. At the post-formation stage, however, laminin positivity beneath the mesothelium was the reverse of that observed during the pre-formation stage. High immunopositivity for collagen IV and laminins at the inner gut wall mesenchyme of the post-formation DJF implied a different blood vessel distribution. We conclude that ECM distribution changes spatiotemporally during mouse DJF formation, indicating ECM association with the establishment of asymmetric morphologies during this process.


Assuntos
Duodeno/embriologia , Matriz Extracelular/metabolismo , Jejuno/embriologia , Morfogênese , Animais , Colágeno Tipo I/metabolismo , Duodeno/metabolismo , Feminino , Fibronectinas , Jejuno/metabolismo , Laminina/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Modelos Biológicos , Proteoglicanas/metabolismo , Tenascina/metabolismo
7.
Poult Sci ; 95(1): 90-8, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26574034

RESUMO

Nutrients are absorbed in the small intestine through a variety of transporter proteins, which have not been as well characterized in turkeys as in chickens. The objective of this study was to profile the mRNA expression of amino acid and monosaccharide transporters in the small intestine of male and female turkeys. Jejunum was collected during embryonic development (embryonic d 21 and 24, and d of hatch (DOH)) and duodenum, jejunum, and ileum were collected in a separate experiment during posthatch development (DOH, d 7, 14, 21, and 28). Real-time PCR was used to determine expression of aminopeptidase N (APN), one peptide (PepT1), 6 amino acid (ASCT1, b(o,+)AT, CAT1, EAAT3, LAT1, y(+)LAT2) and 3 monosaccharide (GLUT2, GLUT5, SGLT1) transporters. Data were analyzed by ANOVA using JMP Pro 11.0. APN, b(o,+)AT, PepT1, y(+)LAT2, GLUT5, and SGLT1 showed increased expression from embryonic d 21 and 24 to DOH. During posthatch, all genes except GLUT2 and SGLT1 were expressed greater in females than males. GLUT2 was expressed the same in males as females and SGLT1 was expressed greater in males than females. All basolateral membrane transporters were expressed greater during early development then decreased with age, while the brush border membrane transporters EAAT3, GLUT5, and SGLT1 showed increased expression later in development. Because turkeys showed high-level expression of the anionic amino acid transporter EAAT3, a direct comparison of tissue-specific expression of EAAT3 between chicken and turkey was conducted. The anionic amino acid transporter EAAT3 showed 6-fold greater expression in the ileum of turkeys at d 14 compared to chickens. This new knowledge can be used not only to better formulate turkey diets to accommodate increased glutamate transport, but also to optimize nutrition for both sexes.


Assuntos
Duodeno/metabolismo , Íleo/metabolismo , Jejuno/metabolismo , Proteínas de Membrana Transportadoras/genética , Perus/metabolismo , Animais , Dieta/veterinária , Duodeno/enzimologia , Duodeno/crescimento & desenvolvimento , Feminino , Íleo/enzimologia , Íleo/crescimento & desenvolvimento , Jejuno/embriologia , Jejuno/enzimologia , Jejuno/crescimento & desenvolvimento , Masculino , Proteínas de Membrana Transportadoras/metabolismo , Reação em Cadeia da Polimerase em Tempo Real/veterinária , Perus/embriologia , Perus/crescimento & desenvolvimento
8.
Cell Tissue Res ; 360(2): 273-85, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25616556

RESUMO

The asymmetric shape of component cells determines the asymmetric features of developing organs. Here, we focused on the murine duodenojejunal flexure (DJF), which bends without affecting the mesentery, and analyzed the morphological asymmetries of the mucosal epithelium and gut wall cells between the inner and outer bending sides at embryonic days 10.75-11.75. In the mucosal epithelium, the cell shape and the expression of epithelial markers (Cdx2, E-cadherin) showed no differences between the two DJF sides. In contrast, the gut wall cells comprising the inner and outer sides of the DJF were elongated along the inner-outer axis and perpendicular to this axis, respectively. Furthermore, the gut wall cells in the outer side possessed cytoplasmic processes connecting cells via adherens junctions, but those in the inner side were attached via adherens junctions of juxtaposed cell bodies and were relatively more crowded. In immunohistochemistry experiments, there was no remarkable difference in the positive reactions of markers for mesenchyme (vimentin), smooth muscle cells (αSMA), endothelial cells (LYVE-1, CD34), and undifferentiated neurons (Sox10) between the DJF sides. Interestingly, Tuj1-positive cells, indicating differentiated neurons, were observed in the middle layer of the gut wall, and these cells were significantly more abundant and tended to be larger in the inner side than in the outer side of the DJF. In conclusion, we clarified the asymmetries of gut wall cell morphology and neural differentiation between the inner and outer sides of the DJF. These characteristics of the developing murine DJF indicate its asymmetric formation.


Assuntos
Antígenos de Diferenciação/metabolismo , Duodeno/citologia , Duodeno/embriologia , Jejuno/citologia , Jejuno/embriologia , Animais , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Mucosa Intestinal/citologia , Mucosa Intestinal/embriologia , Camundongos , Miócitos de Músculo Liso/citologia , Miócitos de Músculo Liso/metabolismo , Neurônios/citologia , Neurônios/metabolismo
9.
Ultrasound Obstet Gynecol ; 45(5): 523-9, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25157626

RESUMO

OBJECTIVE: The accuracy of prenatal ultrasound examination in detecting jejunal and ileal atresia has been reported in the literature to be highly variable, at 25-90%. The aim of this systematic review was to evaluate the accuracy of prenatal ultrasound in detecting non-duodenal small bowel atresia (ND-SBA). METHODS: MEDLINE, EMBASE and The Cochrane Library, including The Cochrane Database of Systematic Reviews (CDSR), Database of Abstracts of Reviews of Effects (DARE) and The Cochrane Central Register of Controlled Trials (CENTRAL), were searched electronically. The overall detection rate of jejunal or ileal atresia using ultrasound was reported. The accuracy of using polyhydramnios and dilated loops of bowel as diagnostic signs was also explored. RESULTS: Sixteen studies involving 640 fetuses were included in this review. The detection rate of ND-SBA by prenatal ultrasound was highly variable, with values ranging from 10 to 100%, with an overall prediction of 50.6% (95% CI, 38.0-63.2%). When analyzed separately, the detection rates of jejunal and ileal atresia were 66.3%, (95% CI, 33.9-91.8%) and 25.9% (95% CI, 4.0-58.0%), respectively. Both dilated loops of bowel and polyhydramnios as diagnostic signs for ND-SBA provided a low overall detection rate. CONCLUSIONS: The diagnostic performance of prenatal ultrasound in identifying ND-SBA is extremely variable. Large studies are needed in order to identify objective and combined criteria for the diagnosis of these anomalies.


Assuntos
Íleo/diagnóstico por imagem , Atresia Intestinal/diagnóstico por imagem , Intestino Delgado/anormalidades , Jejuno/diagnóstico por imagem , Ultrassonografia Pré-Natal , Feminino , Humanos , Íleo/embriologia , Recém-Nascido , Atresia Intestinal/embriologia , Atresia Intestinal/patologia , Intestino Delgado/diagnóstico por imagem , Intestino Delgado/embriologia , Intestino Delgado/patologia , Jejuno/embriologia , Gravidez , Reprodutibilidade dos Testes , Sensibilidade e Especificidade
10.
J Biosci ; 39(5): 835-48, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25431412

RESUMO

The enteric nervous system (ENS) arises from the enteric neural crest-derived cells (ENCCs), and many molecules and biochemical processes may be involved in its development. This study examined the effects of modulating embryonic nitric oxide (NO) activity on the intestinal motility induced by ENS. One-hundred-and-twenty fertilized chicken eggs were assigned to three main groups and incubated at 37 degrees Centigrade and 60 percent humidity. The eggs were treated with NG-nitro-Larginine methyl ester (L-NAME), sodium nitroprusside (SNP), L-arginine (L-Arg) or vehicle from days 3 (1st group), 7 (2nd group) and 10 (3rd group) of incubation and continued up to day 18. On day 19, the embryos were sacrificed, the jejunal and colorectal segments were taken and the intestinal motility was assessed using isolated organ system. The intestinal motility was recorded normally and following cholinergic, adrenergic and non-adrenergic non-cholinergic (NANC) stimulations. The ENS structure was assessed by immunohistochemistry (IHC) using glial fibrillary acidic protein (GFAP). Rhythmic intestinal contractions were seen in all treatment groups, but inhibition of NO in the LNAME- treated embryos caused significant decrease (p less than 0.01) in the frequency and amplitude of the contraction. The responsiveness to adrenergic, cholinergic and NANC stimulations was also significantly decreased (p less than 0.05). The GFAP expression was significantly (p less than 0.05) reduced in the L-NAME-treated embryos. This study showed that the inhibition of NO caused a deficient development of the ENS, leading to a decrease in the frequency and amplitude of the intestinal contractions and reduced the responsiveness to adrenergic, cholinergic and NANC signalling.


Assuntos
Sistema Nervoso Entérico/efeitos dos fármacos , Motilidade Gastrointestinal/efeitos dos fármacos , Óxido Nítrico/metabolismo , Animais , Arginina/farmacologia , Embrião de Galinha , Sistema Nervoso Entérico/embriologia , Intestinos/efeitos dos fármacos , Intestinos/embriologia , Jejuno/efeitos dos fármacos , Jejuno/embriologia , NG-Nitroarginina Metil Éster/farmacologia , Nitroprussiato/farmacologia , Transmissão Sináptica
11.
J Anat ; 223(4): 385-98, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23961897

RESUMO

The mammalian gut undergoes morphological changes during development. We studied the developing mouse duodenojejunal flexure (DJF) to elucidate the mechanism of formation. During embryonic days 10.75-13.75, DJF formation was morphologically classified into three stages: the expansion stage, flexure formation stage, and flexure elongation stage. From the expansion to the flexure formation stages, the DJF wall showed asymmetric morphology and proliferation along the left-right intestinal axis. From the flexure formation to the flexure elongation stage, the DJF started to bend dorsally with counterclockwise rotation along the antero-caudal intestinal axis, indicating that the original right side of the duodenum was rotated towards the dorsal body wall during development of the DJF. The direction of attachment of the dorsal mesentery to the DJF did not correspond to the bending direction of the DJF during flexure formation, and this finding indicated that the dorsal mesentery contributed very little to DJF formation. During DJF formation, Aldh1a2 and hedgehog mRNAs were detected at the DJF, and their expression levels differed along the bending axis. In conclusion, DJF formation might be triggered by asymmetric morphology and proliferation along the left-right intestinal axis under the control of retinoic acid and hedgehog signaling.


Assuntos
Duodeno/embriologia , Jejuno/embriologia , Animais , Proliferação de Células , Duodeno/citologia , Duodeno/fisiologia , Perfilação da Expressão Gênica , Jejuno/citologia , Jejuno/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Modelos Anatômicos , Modelos Animais , RNA Mensageiro/metabolismo , Transdução de Sinais/genética
12.
Am J Surg ; 204(5): 598-601, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22906252

RESUMO

BACKGROUND: Understanding gut development may illuminate the adaptive response to massive small-bowel resection and facilitate enteral nutrition. We reported that Schlafen-3 (Slfn3) mediates differentiation in vitro in rat intestinal epithelial. We hypothesized that Slfn3 is involved in intestinal development in vivo. METHODS: We removed fetal intestines, liver, and lungs on day 20 of gestation, at birth, and on postnatal days 1 and 5. Expression of Slfn3, markers of intestinal differentiation, and Slfn5, to address specificity, were determined by quantitative reverse-transcription polymerase chain reaction. RESULTS: Villin expression increased on days 1 and 5 (8.7 ± .6 and 5.4 ± .4, respectively; P < .01). Intestinal Slfn3 expression was increased substantially after birth (2.1- ± .5-fold) and on days 1 and 5 (P < .02). Slfn3 was higher after birth in liver and lung but decreased sharply thereafter. Slfn5 expression was mostly unchanged. CONCLUSIONS: The data suggest that the developmental/maturation effects we observed correlate with Slfn3 but not Slfn5 and are more relevant to the intestines. A better understanding of how Slfn3 promotes intestinal differentiation could help promote intestinal maturation, improving outcomes in children or adults with short-gut syndrome.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Mucosa Intestinal/metabolismo , Jejuno/metabolismo , Proteínas/metabolismo , Animais , Biomarcadores/metabolismo , Dipeptidil Peptidase 4/metabolismo , Mucosa Intestinal/embriologia , Mucosa Intestinal/crescimento & desenvolvimento , Jejuno/embriologia , Jejuno/crescimento & desenvolvimento , Fígado/embriologia , Fígado/crescimento & desenvolvimento , Fígado/metabolismo , Pulmão/embriologia , Pulmão/crescimento & desenvolvimento , Pulmão/metabolismo , Proteínas dos Microfilamentos/metabolismo , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa
14.
Poult Sci ; 90(4): 842-55, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21406371

RESUMO

The potential for growth and feed efficiency in turkey poults directly correlates with the early development of the intestinal epithelium. Although the metabolic aspects of enteric maturation have been studied, little is known about the ultrastructural development of the enteric epithelium in the turkey embryo and poult. Hence, the objective of this study was to document the morphological and ultrastructural development of the jejunum mucosa in turkeys, from 15 d of incubation (embryonic day; E) to 12 d posthatch. Intestinal samples from 4 embryos or poults were collected and analyzed by light and electron microscopy (transmission and scanning). In addition, amniotic fluid volume was determined in 6 eggs from E15 to E25. Longitudinal previllus ridges at E15 gradually formed zigzag patterns that led to the formation of 2 parallel lines of mature villi by E25. The volume of amniotic fluid was rapidly depleted as the embryo swallowed it between E19 and E25. During this period, a major increase occurs in villus height, the apical end of epithelial cells is gradually tightened by the junctional complex, and mature goblet cells are visible at the apical end of villi. Villus height steadily increases until reaching a plateau at 8 d. Villi morphology shifts gradually from finger-like projections before hatch to leaf-like projections by 12 d. At this age, the enteric epithelium is in intimate association with microbes such as segmented filamentous bacteria. The profound morphological adaptations of the turkey gut epithelium in response to amniotic fluid swallowing before hatch, and dietary factors and bacteria after hatch, demonstrate the plasticity of the enteric epithelium at this time. Hence, the supplementation of enteric modulators before hatch (in ovo feeding) and after hatch has the potential to shape gut maturation and enhance the growth performance of turkey poults.


Assuntos
Mucosa Intestinal/fisiologia , Jejuno/fisiologia , Perus/embriologia , Animais , Embrião não Mamífero/fisiologia , Embrião não Mamífero/ultraestrutura , Células Caliciformes/citologia , Células Caliciformes/ultraestrutura , Histocitoquímica/veterinária , Mucosa Intestinal/citologia , Mucosa Intestinal/embriologia , Mucosa Intestinal/ultraestrutura , Jejuno/citologia , Jejuno/embriologia , Jejuno/ultraestrutura , Microscopia Eletrônica de Varredura/veterinária , Microscopia Eletrônica de Transmissão/veterinária
15.
J Cell Biochem ; 112(1): 354-61, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21080337

RESUMO

Disabled-2 (Dab2) is an intracellular adaptor protein proposed to function in endocytosis. Here, we investigate the intestinal and renal Dab2 expression versus maturation. Dab2 mRNA levels measured by RT-PCR are greater in the small than in the large intestine. Immunological studies localize Dab2 to the terminal web domain of the enterocytes and reveal the presence of a 96-kDa Dab2 isoform in the apical membrane of the jejunum, ileum, and renal cortex of the suckling and adult rat. A 69-kDa Dab2 isoform is only observed in the apical membranes of the suckling ileum. During the suckling period, the Dab2 mRNA levels measured in the enterocytes and crypts and those of the 96-kDa Dab2 isoform are greater in the ileum than in the jejunum. No segmental differences are observed in the adult intestine. In the intestine, the levels of Dab2 mRNA and those of the 96-kDa Dab2 isoform decrease to adult values at weaning, whereas in the kidney they increase with development. Weaning the pups on a commercial milk diet slows the periweaning decline in the levels of Dab2 mRNA in the crypts and of those of the 96-kDa isoform. This is the first report showing that the 96-kDa Dab2 isoform is expressed at the apical domain of rat small intestine, that ontogeny regulates Dab2 gene expression in intestine and kidney and that retarding weaning affects intestinal Dab2 gene expression.


Assuntos
Proteínas Adaptadoras de Transporte Vesicular/genética , Epitélio/crescimento & desenvolvimento , Regulação da Expressão Gênica no Desenvolvimento , Intestino Grosso/crescimento & desenvolvimento , Rim/crescimento & desenvolvimento , Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Animais , Epitélio/embriologia , Epitélio/metabolismo , Íleo/embriologia , Íleo/crescimento & desenvolvimento , Íleo/metabolismo , Intestino Grosso/embriologia , Intestino Grosso/metabolismo , Jejuno/embriologia , Jejuno/crescimento & desenvolvimento , Jejuno/metabolismo , Rim/embriologia , Rim/metabolismo , RNA Mensageiro/metabolismo , Ratos , Ratos Wistar , Reação em Cadeia da Polimerase Via Transcriptase Reversa
16.
J Anim Sci ; 88(7): 2410-24, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20382875

RESUMO

The objectives were to evaluate effects of maternal nutrient restriction and stage of gestation on maternal and fetal visceral organ mass and indices of jejunal growth and vascularity in beef cows. Thirty multiparous beef cows (BW = 571 +/- 63 kg; BCS = 5.4 +/- 0.7) carrying female fetuses (d 30 of gestation) were allocated to receive a diet of native grass hay (CON; 12.1% CP, 70.7% IVDMD, DM basis) to meet NRC recommendations for BW gain during early gestation or a nutrient-restricted diet of millet straw (NR; 9.9% CP, 54.5% IVDMD, DM basis) to provide 68.1% of NE(m) and 86.7% of MP estimated requirements. On d 125 of gestation, 10 CON and 10 NR cows were killed and necropsied. Five remaining CON cows received the CON diet, and 5 NR cows were realimented with a concentrate supplement (13.2% CP, 77.6% IVDMD, DM basis) and the CON hay to achieve a BCS similar to CON cows by d 220 of gestation. Remaining cows were necropsied on d 245 of gestation. Cow BW and eviscerated BW (EBW) were less (P < 0.01) for NR than CON at d 125 but did not differ (P > 0.63) at d 245. Cows fed the CON diet had greater (P < 0.09) total gastrointestinal (GI) tract, omasal, and pancreatic weights. Stomach complex, ruminal, and liver weights were greater for CON than NR cows (P < 0.09) on d 125. Total GI, stomach complex, and pancreatic weights increased (P < 0.001) with day of gestation. Restricted cows had decreased (P = 0.09) duodenal RNA:DNA compared with CON. Duodenal DNA was less (P = 0.01) and jejunal RNA:DNA (P = 0.09) was greater for cows at d 125 vs. 245. Cow jejunal capillary area density increased with day of gestation (P = 0.02). Fetal BW and EBW were unaffected by dietary treatment (P > or = 0.32). Total GI tract and all components increased in mass with day of gestation (P < 0.001). Fetuses from NR dams had greater (P = 0.003) reticular mass at d 245 than CON fetuses. Fetuses from NR cows had greater (P = 0.02) percent jejunal proliferation at d 125 and greater (P = 0.03) total intestinal vascularity (mL) at d 245. Fetal jejunal DNA decreased (P = 0.09), RNA:DNA increased (P = 0.05), and total jejunal proliferating cells increased (P < 0.001) with day of gestation. Jejunal capillary area density, number density, and surface density were greater (P < 0.008) during late gestation. Results indicate that maternal and fetal intestines undergo changes during gestation, which can be affected by nutrient restriction and may partially explain differences observed in fetal development and postnatal performance.


Assuntos
Jejuno/embriologia , Fenômenos Fisiológicos da Nutrição Materna/fisiologia , Vísceras/embriologia , Animais , Bovinos , Feminino , Desenvolvimento Fetal , Privação de Alimentos/fisiologia , Trato Gastrointestinal/anatomia & histologia , Idade Gestacional , Intestinos/irrigação sanguínea , Intestinos/embriologia , Jejuno/irrigação sanguínea , Omaso/anatomia & histologia , Tamanho do Órgão/fisiologia , Pâncreas/anatomia & histologia , Gravidez , Vísceras/anatomia & histologia
17.
J Anim Sci ; 88(8): 2692-702, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20407071

RESUMO

The objectives of this study were to evaluate effects of maternal dietary restriction and Se supply on angiogenic factor mRNA expression in intestinal and mammary tissues, and jejunal crypt cell proliferation and vascularity in late-term fetal intestines. In Exp. 1, pregnant ewe lambs (n = 32; initial BW = 45.6 +/- 2.3 kg) were allotted randomly to 1 of 4 treatments. Treatments (initiated d 50 +/- 5 of gestation) were control (3.5 microg of Se.kg of BW(-1).d(-1)), Se-wheat (75 microg of Se.kg of BW(-1).d(-1)), selenate (Se3; providing 75 microg of Se.kg of BW(-1).d(-1)), selenate (Se15; providing 375 microg of Se.kg of BW(-1).d(-1)). Diets (DM basis) were similar in CP (15.5%) and ME (2.68 Mcal/kg). In Exp. 2, pregnant ewe lambs (n = 36; initial BW 53.8 +/- 1.3 kg) were allotted randomly to treatments in a 2 x 2 factorial arrangement. Factors were nutrition (control, 100% of requirements vs. restricted nutrition, 60% of controls) and dietary Se (adequate Se; 6 microg of Se.kg of BW(-1).d(-1) vs. high Se; 80 microg of Se.kg of BW(-1).d(-1)). Selenium treatments were initiated 21 d before breeding, and nutritional treatments were initiated on d 64 of gestation. Diets (DM basis) were 16% CP and 2.12 Mcal/kg of ME. In Exp. 1, Se15 increased (P = 0.07) vascular endothelial growth factor (VEGF) mRNA expression, whereas Se supplementation decreased (P = 0.06) kinase insert domain receptor (KDR) mRNA in maternal mucosal scrape on d 134 of gestation. Expression of VEGF mRNA was decreased by Se (P = 0.10) in fetal jejunum. In mammary tissue, fms-related tyrosine kinase 1 and KDR mRNA were greater in Se-wheat compared with Se3, and KDR expression was increased (P = 0.10) in Se15 compared with Se3. In Exp. 2, dietary restriction increased (P < or = 0.07) expression of mRNA for VEGF, fms-related tyrosine kinase 1, KDR, neuropilin 1, neuropilin 2, and hypoxia-inducible factor 1, alpha subunit in mucosal scrapes from maternal jejunum. In fetal jejunum, soluble guanylate cyclase, was decreased (P = 0.01) by maternal dietary restriction from d 64 to 135 of gestation. Total microvascularity in fetal jejunum was reduced (P = 0.002) by maternal dietary restriction. Mammary gland expression of VEGF, neuropilin 1, angiopoietin receptor (endothelial tyrosine kinase), and endothelial nitric oxide synthase 3 increased (P < or = 0.09), whereas angiopoietin 1 decreased (P = 0.05) due to nutrient restriction. Data indicate that expression of angiogenic factors and receptors in maternal intestine, mammary gland, and fetal jejunum are responsive to maternal nutrition and likely explain observed changes in tissue vascularity.


Assuntos
Intestinos/efeitos dos fármacos , Jejuno/embriologia , Glândulas Mamárias Animais/efeitos dos fármacos , Selênio/farmacologia , Fator A de Crescimento do Endotélio Vascular/biossíntese , Angiopoietina-1/análise , Angiopoietina-1/biossíntese , Animais , Dieta/veterinária , Feminino , Mucosa Intestinal/metabolismo , Intestinos/química , Jejuno/química , Jejuno/efeitos dos fármacos , Glândulas Mamárias Animais/química , Glândulas Mamárias Animais/metabolismo , Neuropilina-1/análise , Neuropilina-1/biossíntese , Neuropilina-2/análise , Neuropilina-2/biossíntese , Gravidez , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa/veterinária , Selênio/deficiência , Ovinos , Fator A de Crescimento do Endotélio Vascular/análise , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/análise , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/biossíntese
18.
J Proteome Res ; 9(2): 924-35, 2010 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-19938879

RESUMO

Efficiency of nutrient utilization is reduced in neonates with intrauterine growth restriction (IUGR) compared with those with a normal birth weight (NBW). However, the underlying mechanisms are largely unknown. In this study, we applied temporal proteomic approach, coupled with histological and biochemical analyses, to study dynamic changes of the proteome in the small intestinal mucosa of IUGR piglets during the nursing period (Days 1, 7 and 21). We identified 56 differentially expressed protein spots between IUGR and NBW piglets. These proteins participate in key biological processes, including (1) absorption, digestion and transport of nutrients; (2) cell structure and motility; (3) glucose and energy metabolism; (4) lipid metabolism; (5) amino acid metabolism; (6) mineral and vitamin metabolism; (7) cellular redox homeostasis; (8) stress response; and (9) apoptosis. The results of our temporal proteomics analysis reveal continuous impairment of intestinal development in neonatal piglets with IUGR. The findings have important implications for understanding metabolic defects in the small intestine of IUGR neonates and are expected to provide new strategies to improve their survival and growth.


Assuntos
Animais Recém-Nascidos , Retardo do Crescimento Fetal/fisiopatologia , Jejuno/embriologia , Proteoma , Aminoácidos/metabolismo , Animais , Apoptose , Metabolismo Energético , Homeostase , Metabolismo dos Lipídeos , Espectrometria de Massas , Minerais/metabolismo , Oxirredução , Suínos , Vitaminas/metabolismo
19.
Histochem Cell Biol ; 132(3): 351-67, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19499240

RESUMO

Although intestinal (I) and liver (L) fatty acid binding proteins (FABP) have been widely studied, the physiological significance of the presence of the two FABP forms (I- and L-FABP) in absorptive cells remains unknown as do the differences related to their distribution along the crypt-villus axis, regional expression, ontogeny and regulation in the human intestine. Our morphological experiments supported the expression of I- and L-FABP as early as 13 weeks of gestation. Whereas cytoplasmic immunofluorescence staining of L-FABP was barely detectable in the lower half of the villus and in the crypt epithelial cells, I-FABP was visualized in epithelial cells of the crypt-villus axis in all intestinal segments until the adult period in which the staining was maximized in the upper part of the villus. Immunoelectron microscopy revealed more intense labeling of L-FABP compared with I-FABP, accompanied with a heterogeneous distribution in the cytoplasm, microvilli and basolateral membranes. By western blot analysis, I- and L-FABP at 15 weeks of gestation appeared predominant in jejunum compared with duodenum, ileum, proximal and distal colon. Exploration of the maturation aspect documented a rise in L-FABP in adult tissues. Permanent transfections of Caco-2 cells with I-FABP cDNA resulted in decreased lipid export, apolipoprotein (apo) biogenesis and chylomicron secretion. Additionally, supplementation of Caco-2 with insulin, hydrocortisone and epidermal growth factor differentially modulated the expression of I- and L-FABP, apo B-48 and microsomal triglyceride transfer protein (MTP), emphasizing that these key proteins do not exhibit a parallel modulation. Overall, our findings indicate that the two FABPs display differences in localization, regulation and developmental pattern.


Assuntos
Colo/metabolismo , Proteínas de Ligação a Ácido Graxo/metabolismo , Jejuno/metabolismo , Lipoproteínas/metabolismo , Células CACO-2 , Colo/embriologia , Colo/crescimento & desenvolvimento , Humanos , Lactente , Jejuno/embriologia , Jejuno/crescimento & desenvolvimento , Especificidade de Órgãos
20.
Dev Comp Immunol ; 33(11): 1156-64, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19527747

RESUMO

Jejunal development occurs in a spatio-temporal pattern and is characterized by morphological and functional changes. To investigate jejunal development at the transcriptomic level, we performed microarray studies in 1-21-day-old chickens. Nine gene clusters were identified, each with a specific gene expression pattern. Subsequently, groups of genes with similar functions could be identified. Genes involved in morphological and functional development were highly expressed immediately after hatch with declining expression patterns afterwards. Immunological development can be roughly divided based on expression patterns into three processes over time; first innate response and immigration of immune cells, secondly differentiation and specialization, and thirdly maturation and immune regulation. We conclude that specific gene expression patterns coincide with the immunological, morphological, and functional development as measured by other methods. Our data show that transcriptomic approaches provide more detailed information on the biological processes underlying jejunal development.


Assuntos
Galinhas/genética , Regulação da Expressão Gênica no Desenvolvimento , Jejuno/embriologia , Jejuno/metabolismo , Organogênese/genética , Animais , Diferenciação Celular , Movimento Celular , Galinhas/crescimento & desenvolvimento , Galinhas/imunologia , Perfilação da Expressão Gênica , Imunidade Inata/genética , Jejuno/crescimento & desenvolvimento , Modelos Biológicos , Análise de Sequência com Séries de Oligonucleotídeos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...