Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 98
Filtrar
1.
Nat Cell Biol ; 26(6): 868-877, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38849542

RESUMO

Despite a distinct developmental origin, extraembryonic cells in mice contribute to gut endoderm and converge to transcriptionally resemble their embryonic counterparts. Notably, all extraembryonic progenitors share a non-canonical epigenome, raising several pertinent questions, including whether this landscape is reset to match the embryonic regulation and if extraembryonic cells persist into later development. Here we developed a two-colour lineage-tracing strategy to track and isolate extraembryonic cells over time. We find that extraembryonic gut cells display substantial memory of their developmental origin including retention of the original DNA methylation landscape and resulting transcriptional signatures. Furthermore, we show that extraembryonic gut cells undergo programmed cell death and neighbouring embryonic cells clear their remnants via non-professional phagocytosis. By midgestation, we no longer detect extraembryonic cells in the wild-type gut, whereas they persist and differentiate further in p53-mutant embryos. Our study provides key insights into the molecular and developmental fate of extraembryonic cells inside the embryo.


Assuntos
Apoptose , Linhagem da Célula , Metilação de DNA , Endoderma , Regulação da Expressão Gênica no Desenvolvimento , Animais , Endoderma/citologia , Endoderma/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Proteína Supressora de Tumor p53/genética , Fagocitose , Camundongos Endogâmicos C57BL , Camundongos , Diferenciação Celular , Feminino , Desenvolvimento Embrionário , Embrião de Mamíferos/citologia , Embrião de Mamíferos/metabolismo , Camundongos Transgênicos , Trato Gastrointestinal/citologia , Trato Gastrointestinal/embriologia , Trato Gastrointestinal/metabolismo
2.
PLoS Genet ; 19(11): e1011030, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37948459

RESUMO

Hirschsprung disease (HSCR) is associated with deficiency of the receptor tyrosine kinase RET, resulting in loss of cells of the enteric nervous system (ENS) during fetal gut development. The major contribution to HSCR risk is from common sequence variants in RET enhancers with additional risk from rare coding variants in many genes. Here, we demonstrate that these RET enhancer variants specifically alter the human fetal gut development program through significant decreases in gene expression of RET, members of the RET-EDNRB gene regulatory network (GRN), other HSCR genes, with an altered transcriptome of 2,382 differentially expressed genes across diverse neuronal and mesenchymal functions. A parsimonious hypothesis for these results is that beyond RET's direct effect on its GRN, it also has a major role in enteric neural crest-derived cell (ENCDC) precursor proliferation, its deficiency reducing ENCDCs with relative expansion of non-ENCDC cells. Thus, genes reducing RET proliferative activity can potentially cause HSCR. One such class is the 23 RET-dependent transcription factors enriched in early gut development. We show that their knockdown in human neuroblastoma SK-N-SH cells reduces RET and/or EDNRB gene expression, expanding the RET-EDNRB GRN. The human embryos we studied had major remodeling of the gut transcriptome but were unlikely to have had HSCR: thus, genetic or epigenetic changes in addition to those in RET are required for aganglionosis.


Assuntos
Elementos Facilitadores Genéticos , Trato Gastrointestinal , Proteínas Proto-Oncogênicas c-ret , Haplótipos , Humanos , Proteínas Proto-Oncogênicas c-ret/genética , Neuroblastoma , Linhagem Celular Tumoral , Doença de Hirschsprung/genética , Feto , Trato Gastrointestinal/embriologia , Crista Neural/citologia , Sistema Nervoso Entérico/embriologia , Análise da Expressão Gênica de Célula Única , Regulação da Expressão Gênica no Desenvolvimento
3.
Lancet Gastroenterol Hepatol ; 7(1): 96-106, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34822760

RESUMO

Over the past 5 years, systematic investigation of the mesenteric organ has expanded and shown that the mesentery is the organ in and on which all abdominal digestive organs develop and remain connected to. In turn, this observation has clarified the anatomical foundation of the abdomen and the fundamental order at that level. Findings related to the shape and development of the mesentery have illuminated its function, advancing our understanding of the pathobiology, diagnosis, and treatment of several abdominal and systemic diseases. Inclusion of the mesentery in surgical resections alters the course of benign and malignant diseases. Mesenteric-based scoring systems can enhance the radiological interpretation of abdominal disease. Emerging findings reconcile observations across scientific and clinical fields and have been assimilated into reference curricula and practice guidelines. This Review summarises the developmental, anatomical, and clinical advances made since the mesentery was redesignated as an organ in 2016.


Assuntos
Gastroenteropatias/terapia , Trato Gastrointestinal/embriologia , Mesentério/anatomia & histologia , Mesentério/fisiologia , Gastroenteropatias/diagnóstico por imagem , Gastroenteropatias/etiologia , Humanos , Metástase Linfática , Mesentério/patologia
4.
Toxins (Basel) ; 12(6)2020 06 08.
Artigo em Inglês | MEDLINE | ID: mdl-32521650

RESUMO

Harmful algal blooms (HAB) have become a major health concern worldwide, not just to humans that consume and recreate on contaminated waters, but also to the fauna that inhabit the environments surrounding affected areas. HABs contain heterotrophic bacteria, cyanobacterial lipopolysaccharide, and cyanobacterial toxins such as microcystins, that can cause severe toxicity in many aquatic species as well as bioaccumulation within various organs. Thus, the possibility of trophic transference of this toxin through the food chain has potentially important health implications for other organisms in the related food web. While some species have developed adaptions to attenuate the toxic effects of HAB toxins, there are still numerous species that remain vulnerable, including Lithobates catesbeiana (American bullfrog) tadpoles. In the current study we demonstrate that acute, short-term exposure of tadpoles to HAB toxins containing 1 µg/L (1 nmol/L) of total microcystins for only 7 days results in significant liver and intestinal toxicity within tadpoles. Exposed tadpoles had increased intestinal diameter, decreased intestinal fold heights, and a constant number of intestinal folds, indicating pathological intestinal distension, similar to what is seen in various disease processes, such as toxic megacolon. HAB-toxin-exposed tadpoles also demonstrated hepatocyte hypertrophy with increased hepatocyte binucleation consistent with carcinogenic and oxidative processes within the liver. Both livers and intestines of HAB-toxin-exposed tadpoles demonstrated significant increases in protein carbonylation consistent with oxidative stress and damage. These findings demonstrate that short-term exposure to HAB toxins, including microcystins, can have significant adverse effects in amphibian populations. This acute, short-term toxicity highlights the need to evaluate the influence HAB toxins may have on other vulnerable species within the food web and how those may ultimately also impact human health.


Assuntos
Trato Gastrointestinal/efeitos dos fármacos , Proliferação Nociva de Algas , Fígado/efeitos dos fármacos , Microcistinas/toxicidade , Rana catesbeiana , Microbiologia da Água , Animais , Cadeia Alimentar , Trato Gastrointestinal/embriologia , Trato Gastrointestinal/metabolismo , Larva/efeitos dos fármacos , Fígado/embriologia , Fígado/metabolismo , Carbonilação Proteica/efeitos dos fármacos , Rana catesbeiana/embriologia , Fatores de Tempo , Testes de Toxicidade Aguda
5.
Pediatr Res ; 87(5): 847-852, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-31756731

RESUMO

BACKGROUND: Fetal swallowing of human amniotic fluid (hAF) containing trophic factors (TFs) promotes gastrointestinal tract (GIT) development. Preterm birth interrupts hAF swallowing, which may increase the risk of necrotizing enterocolitis (NEC). Postnatally, it is difficult to replicate fetal swallowing of hAF due to volume. We aimed to evaluate whether hAF lyophilization is feasible and its effect on hAF-borne TFs. METHODS: We collected hAF (n = 16) from uncomplicated pregnancies. hAF was divided into three groups: unprocessed control (C), concentration by microfiltration (F), and by dialysis and lyophilization (L). EGF, HGF, GM-CSF, and TGF-α were measured in each group by multiplex assay. Bioavailability of TFs was measured by proliferation and LPS-induced IL-8 production by intestinal epithelial cells FHs74. RESULTS: After dialysis/lyophilization, GM-CSF and TGF-α were preserved with partial loss of EGF and HGF. hAF increased cell proliferation and reduced LPS-induced IL-8 production compared to medium alone. Compared to control, dialysis/lyophilization and filtration of hAF increased FHs74 cell proliferation (p < 0.001) and decreased LPS-induced IL-8 production (p < 0.01). CONCLUSIONS: Lyophilization and filtration of hAF is feasible with partial loss of TFs but maintains and even improves bioavailability of TFs measured by proliferation and LPS-induced IL-8 production by FHs74.


Assuntos
Líquido Amniótico/metabolismo , Enterocolite Necrosante/metabolismo , Liofilização , Trato Gastrointestinal/embriologia , Líquido Amniótico/química , Proliferação de Células , Criopreservação , Deglutição , Feminino , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Humanos , Inflamação , Interleucina-8/metabolismo , Gravidez , Fator de Crescimento Transformador alfa/metabolismo
6.
Genetics ; 210(2): 357-396, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30287514

RESUMO

The gastrointestinal tract has recently come to the forefront of multiple research fields. It is now recognized as a major source of signals modulating food intake, insulin secretion and energy balance. It is also a key player in immunity and, through its interaction with microbiota, can shape our physiology and behavior in complex and sometimes unexpected ways. The insect intestine had remained, by comparison, relatively unexplored until the identification of adult somatic stem cells in the Drosophila intestine over a decade ago. Since then, a growing scientific community has exploited the genetic amenability of this insect organ in powerful and creative ways. By doing so, we have shed light on a broad range of biological questions revolving around stem cells and their niches, interorgan signaling and immunity. Despite their relatively recent discovery, some of the mechanisms active in the intestine of flies have already been shown to be more widely applicable to other gastrointestinal systems, and may therefore become relevant in the context of human pathologies such as gastrointestinal cancers, aging, or obesity. This review summarizes our current knowledge of both the formation and function of the Drosophila melanogaster digestive tract, with a major focus on its main digestive/absorptive portion: the strikingly adaptable adult midgut.


Assuntos
Drosophila melanogaster/fisiologia , Trato Gastrointestinal/fisiologia , Animais , Drosophila melanogaster/anatomia & histologia , Drosophila melanogaster/embriologia , Trato Gastrointestinal/anatomia & histologia , Trato Gastrointestinal/embriologia , Morfogênese
7.
Nat Cell Biol ; 20(6): 721-734, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29802404

RESUMO

The development of the digestive tract is critical for proper food digestion and nutrient absorption. Here, we analyse the main organs of the digestive tract, including the oesophagus, stomach, small intestine and large intestine, from human embryos between 6 and 25 weeks of gestation as well as the large intestine from adults using single-cell RNA-seq analyses. In total, 5,227 individual cells are analysed and 40 cell types clearly identified. Their crucial biological features, including developmental processes, signalling pathways, cell cycle, nutrient digestion and absorption metabolism, and transcription factor networks, are systematically revealed. Moreover, the differentiation and maturation processes of the large intestine are thoroughly investigated by comparing the corresponding transcriptome profiles between embryonic and adult stages. Our work offers a rich resource for investigating the gene regulation networks of the human fetal digestive tract and adult large intestine at single-cell resolution.


Assuntos
Células Epiteliais/fisiologia , Trato Gastrointestinal/fisiologia , Perfilação da Expressão Gênica/métodos , Regulação da Expressão Gênica no Desenvolvimento , Análise de Sequência de RNA/métodos , Análise de Célula Única/métodos , Transcriptoma , Adulto , Fatores Etários , Diferenciação Celular/genética , Proliferação de Células/genética , Células Epiteliais/metabolismo , Trato Gastrointestinal/embriologia , Trato Gastrointestinal/metabolismo , Marcadores Genéticos , Genótipo , Idade Gestacional , Humanos , Morfogênese , Fenótipo , Fatores de Tempo
8.
Dev Biol ; 435(2): 97-108, 2018 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-29339095

RESUMO

The gastrointestinal (GI) tract, in simplest terms, can be described as an epithelial-lined muscular tube extending along the cephalocaudal axis from the oral cavity to the anus. Although the general architecture of the GI tract organs is conserved from end to end, the presence of different epithelial tissue structures and unique epithelial cell types within each organ enables each to perform the distinct digestive functions required for efficient nutrient assimilation. Spatiotemporal regulation of signaling pathways and downstream transcription factors controls GI epithelial morphogenesis during development to confer essential regional-specific epithelial structures and functions. Here, we discuss the fundamental functions of each GI tract organ and summarize the diversity of epithelial structures present along the cephalocaudal axis of the GI tract. Next, we discuss findings, primarily from genetic mouse models, that have defined the roles of key transcription factors during epithelial morphogenesis, including p63, SOX2, SOX15, GATA4, GATA6, HNF4A, and HNF4G. Additionally, we examine how the Hedgehog, WNT, and BMP signaling pathways contribute to defining unique epithelial features along the cephalocaudal axis of the GI tract. Lastly, we examine the molecular mechanisms controlling regionalized cytodifferentiation of organ-specific epithelial cell types within the GI tract, concentrating on the stomach and small intestine. The delineation of GI epithelial patterning mechanisms in mice has provided fundamental knowledge to guide the development and refinement of three-dimensional GI organotypic culture models such as those derived from directed differentiation of human pluripotent stem cells and those derived directly from human tissue samples. Continued examination of these pathways will undoubtedly provide vital insights into the mechanisms of GI development and disease and may afford new avenues for innovative tissue engineering and personalized medicine approaches to treating GI diseases.


Assuntos
Células Epiteliais/citologia , Trato Gastrointestinal/citologia , Mucosa Intestinal/citologia , Animais , Diferenciação Celular , Células Epiteliais/fisiologia , Trato Gastrointestinal/embriologia , Trato Gastrointestinal/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio/fisiologia , Humanos , Absorção Intestinal , Intestino Delgado/citologia , Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Camundongos , Morfogênese , Família Multigênica , Especificidade de Órgãos , Transdução de Sinais/fisiologia , Estômago/citologia , Fatores de Transcrição/fisiologia
9.
Proc Natl Acad Sci U S A ; 114(45): 11980-11985, 2017 11 07.
Artigo em Inglês | MEDLINE | ID: mdl-29078343

RESUMO

Most of the enteric nervous system derives from the "vagal" neural crest, lying at the level of somites 1-7, which invades the digestive tract rostro-caudally from the foregut to the hindgut. Little is known about the initial phase of this colonization, which brings enteric precursors into the foregut. Here we show that the "vagal crest" subsumes two populations of enteric precursors with contrasted origins, initial modes of migration, and destinations. Crest cells adjacent to somites 1 and 2 produce Schwann cell precursors that colonize the vagus nerve, which in turn guides them into the esophagus and stomach. Crest cells adjacent to somites 3-7 belong to the crest streams contributing to sympathetic chains: they migrate ventrally, seed the sympathetic chains, and colonize the entire digestive tract thence. Accordingly, enteric ganglia, like sympathetic ones, are atrophic when deprived of signaling through the tyrosine kinase receptor ErbB3, while half of the esophageal ganglia require, like parasympathetic ones, the nerve-associated form of the ErbB3 ligand, Neuregulin-1. These dependencies might bear relevance to Hirschsprung disease, with which alleles of Neuregulin-1 are associated.


Assuntos
Sistema Nervoso Entérico/citologia , Gânglios Simpáticos/citologia , Trato Gastrointestinal/embriologia , Crista Neural/citologia , Neuregulina-1/genética , Receptor ErbB-3/genética , Células de Schwann/citologia , Animais , Embrião de Galinha , Trato Gastrointestinal/inervação , Doença de Hirschsprung/genética , Camundongos , Neuregulina-1/metabolismo , Neurogênese/genética , Neurogênese/fisiologia , Receptor ErbB-3/metabolismo , Nervo Vago/citologia
10.
J Histochem Cytochem ; 65(1): 21-32, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-28026654

RESUMO

L1 cell adhesion molecule (L1CAM) is a transmembrane molecule belonging to the L1 protein family. It has shown to be a key player in axonal guidance in the course of neuronal development. Furthermore, L1CAM is also crucial for the establishment of the enteric and urogenital organs and is aberrantly expressed in cancer originating in these organs. Carcinogenesis and embryogenesis follow a lot of similar molecular pathways, but unfortunately, comprehensive data on L1CAM expression and localization in human developing organs are lacking so far. In the present study we, therefore, examined the spatiotemporal distribution of L1CAM in the early human fetal period (weeks 8-12 of gestation) by means of immunohistochemistry and in situ hybridization (ISH). In the epithelia of the gastrointestinal organs, L1CAM localization cannot be observed in the examined stages most likely due to their advanced polarization and differentiation. Despite these results, our ISH data indicate weak L1CAM expression, but only in few epithelial cells. The genital tracts, however, are distinctly L1CAM positive throughout the entire fetal period. We, therefore, conclude that in embryogenesis L1CAM is crucial for further differentiation of epithelia.


Assuntos
Epitélio/embriologia , Trato Gastrointestinal/embriologia , Molécula L1 de Adesão de Célula Nervosa/análise , Sistema Urogenital/embriologia , Adulto , Transição Epitelial-Mesenquimal , Epitélio/metabolismo , Epitélio/ultraestrutura , Trato Gastrointestinal/metabolismo , Trato Gastrointestinal/ultraestrutura , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Imuno-Histoquímica , Hibridização In Situ , Molécula L1 de Adesão de Célula Nervosa/genética , Sistema Urogenital/metabolismo , Sistema Urogenital/ultraestrutura
11.
Cell ; 167(2): 355-368.e10, 2016 Oct 06.
Artigo em Inglês | MEDLINE | ID: mdl-27693352

RESUMO

Common sequence variants in cis-regulatory elements (CREs) are suspected etiological causes of complex disorders. We previously identified an intronic enhancer variant in the RET gene disrupting SOX10 binding and increasing Hirschsprung disease (HSCR) risk 4-fold. We now show that two other functionally independent CRE variants, one binding Gata2 and the other binding Rarb, also reduce Ret expression and increase risk 2- and 1.7-fold. By studying human and mouse fetal gut tissues and cell lines, we demonstrate that reduced RET expression propagates throughout its gene regulatory network, exerting effects on both its positive and negative feedback components. We also provide evidence that the presence of a combination of CRE variants synergistically reduces RET expression and its effects throughout the GRN. These studies show how the effects of functionally independent non-coding variants in a coordinated gene regulatory network amplify their individually small effects, providing a model for complex disorders.


Assuntos
Elementos Facilitadores Genéticos , Regulação da Expressão Gênica , Redes Reguladoras de Genes , Doença de Hirschsprung/genética , Proteínas Proto-Oncogênicas c-ret/genética , Alelos , Animais , Sítios de Ligação , Modelos Animais de Doenças , Fator de Transcrição GATA2/genética , Fator de Transcrição GATA2/metabolismo , Trato Gastrointestinal/embriologia , Humanos , Camundongos , Camundongos Transgênicos , RNA não Traduzido/genética , Receptores do Ácido Retinoico/genética , Receptores do Ácido Retinoico/metabolismo , Fatores de Transcrição SOXE/genética , Fatores de Transcrição SOXE/metabolismo
12.
Nutrition ; 32(6): 620-7, 2016 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-26946974

RESUMO

Gut microbiota establishment and further microbiota shifts are very important for maintaining host health throughout life. There are some factors, including genetics, the mother's health and diet, delivery mode, breast or formula feeding, that may influence the gut microbiota. By the end of approximately the first 3 y of life, the gut microbiota becomes an adult-like stable system. Once established, 60 to 70% of the microbiota composition remains stable throughout life, but 30 to 40% can be altered by changes in the diet and other factors such as physical activity, lifestyle, bacterial infections, and antibiotic or surgical treatment. Diet-related factors that influence the gut microbiota in people of all ages are of great interest. Nutrition may have therapeutic success in gut microbiota correction. This review describes current evidence concerning the links between gut microbiota composition and dietary patterns throughout life.


Assuntos
Dieta/métodos , Microbioma Gastrointestinal/fisiologia , Trato Gastrointestinal/embriologia , Trato Gastrointestinal/microbiologia , Estilo de Vida , Adulto , Criança , Feminino , Humanos , Gravidez
13.
Sci Rep ; 6: 20927, 2016 02 18.
Artigo em Inglês | MEDLINE | ID: mdl-26887292

RESUMO

Neural crest cells (NCCs) are a population of multipotent cells that migrate extensively during vertebrate development. Alterations to neural crest ontogenesis cause several diseases, including cancers and congenital defects, such as Hirschprung disease, which results from incomplete colonization of the colon by enteric NCCs (ENCCs). We investigated the influence of the stiffness and structure of the environment on ENCC migration in vitro and during colonization of the gastrointestinal tract in chicken and mouse embryos. We showed using tensile stretching and atomic force microscopy (AFM) that the mesenchyme of the gut was initially soft but gradually stiffened during the period of ENCC colonization. Second-harmonic generation (SHG) microscopy revealed that this stiffening was associated with a gradual organization and enrichment of collagen fibers in the developing gut. Ex-vivo 2D cell migration assays showed that ENCCs migrated on substrates with very low levels of stiffness. In 3D collagen gels, the speed of the ENCC migratory front decreased with increasing gel stiffness, whereas no correlation was found between porosity and ENCC migration behavior. Metalloprotease inhibition experiments showed that ENCCs actively degraded collagen in order to progress. These results shed light on the role of the mechanical properties of tissues in ENCC migration during development.


Assuntos
Movimento Celular/fisiologia , Embrião de Mamíferos/embriologia , Embrião de Mamíferos/ultraestrutura , Trato Gastrointestinal/embriologia , Trato Gastrointestinal/ultraestrutura , Crista Neural/embriologia , Crista Neural/ultraestrutura , Animais , Embrião de Galinha , Colagenases/metabolismo , Matriz Extracelular/metabolismo , Matriz Extracelular/ultraestrutura , Camundongos , Microscopia de Força Atômica
14.
Cell Mol Life Sci ; 72(20): 3883-96, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26126787

RESUMO

The gastrointestinal tract develops from a simple and uniform tube into a complex organ with specific differentiation patterns along the anterior-posterior and dorso-ventral axes of asymmetry. It is derived from all three germ layers and their cross-talk is important for the regulated development of fetal and adult gastrointestinal structures and organs. Signals from the adjacent mesoderm are essential for the morphogenesis of the overlying epithelium. These mesenchymal-epithelial interactions govern the development and regionalization of the different gastrointestinal epithelia and involve most of the key morphogens and signaling pathways, such as the Hedgehog, BMPs, Notch, WNT, HOX, SOX and FOXF cascades. Moreover, the mechanisms underlying mesenchyme differentiation into smooth muscle cells influence the regionalization of the gastrointestinal epithelium through interactions with the enteric nervous system. In the neonatal and adult gastrointestinal tract, mesenchymal-epithelial interactions are essential for the maintenance of the epithelial regionalization and digestive epithelial homeostasis. Disruption of these interactions is also associated with bowel dysfunction potentially leading to epithelial tumor development. In this review, we will discuss various aspects of the mesenchymal-epithelial interactions observed during digestive epithelium development and differentiation and also during epithelial stem cell regeneration.


Assuntos
Mucosa Intestinal/embriologia , Mesoderma/embriologia , Comunicação Celular , Diferenciação Celular , Trato Gastrointestinal/citologia , Trato Gastrointestinal/embriologia , Trato Gastrointestinal/metabolismo , Humanos , Mucosa Intestinal/citologia , Mucosa Intestinal/metabolismo , Mesoderma/citologia , Mesoderma/metabolismo , Miócitos de Músculo Liso/citologia , Transdução de Sinais , Fatores de Transcrição/metabolismo , Fatores de Transcrição/fisiologia
15.
Rom J Morphol Embryol ; 56(2): 475-80, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26193216

RESUMO

INTRODUCTION: HER2, EGFR, p53 and PTEN are important in organization of the germ layers, in embryonic development and morphogenesis, in the development and differentiation of certain organ systems and in embryonic morphogenesis. Our goal is the comparative examination of the expression of these markers in the digestive tract of 9-24-week-old fetuses. MATERIALS AND METHODS: We studied using immunohistochemical techniques esophagus, stomach, small and large intestine tissue samples collected from 18 post mortem fetuses of 9-24 weeks. RESULTS: HER2 and PTEN expression appears as early as the 9-12 weeks period in the digestive tract, but HER2 expression decreases in the 21-24 weeks period and then disappears. EGFR expression appears only during the 13-16 weeks period. The expression of p53 is strong until week 21, and then it is restricted to the deeper layers of the epithelium. CONCLUSIONS: Our findings suggest that these markers have role also in the fetal period and complete the scarce data found in literature about the expression of the studied markers in the development of the digestive tract.


Assuntos
Receptores ErbB/metabolismo , Feto/metabolismo , Trato Gastrointestinal/embriologia , Trato Gastrointestinal/metabolismo , PTEN Fosfo-Hidrolase/metabolismo , Receptor ErbB-2/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Esôfago/embriologia , Esôfago/metabolismo , Feminino , Mucosa Gástrica/metabolismo , Humanos , Masculino , Gravidez , Estômago/embriologia
16.
PLoS One ; 10(4): e0122820, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25844926

RESUMO

Cytochrome P450 2W1 (CYP2W1) is expressed predominantly in colorectal and also in hepatic tumors, whereas the levels are insignificant in the corresponding normal human adult tissues. CYP2W1 has been proposed as an attractive target for colorectal cancer (CRC) therapy by exploiting its ability to activate duocarmycin prodrugs to cytotoxic metabolites. However, its endogenous function, regulation and developmental pattern of expression remain unexplored. Here we report the CYP2W1 developmental expression in the murine and human gastrointestinal tissues. The gene expression in the colon and small intestine commence at early stages of embryonic life and is completely silenced shortly after the birth. Immunohistochemical analysis of human fetal colon revealed that CYP2W1 expression is restricted to the crypt cells. The silencing of CYP2W1 after birth correlates with the increased methylation of CpG-rich regions in both murine and human CYP2W1 genes. Analysis of CYP2W1 expression in the colon adenocarcinoma cell line HCC2998 revealed that the gene expression can be induced by e.g. the antitumor agent imatinib, linoleic acid and its derivatives. The imatinib mediated induction of CYP2W1 suggests an adjuvant therapy to treatment with duocarmycins that thus would involve induction of tumor CYP2W1 levels followed by the CYP2W1 activated duocarmycin prodrugs. Taken together these data strongly support further exploration of CYP2W1 as a specific drug target in CRC.


Assuntos
Neoplasias Colorretais/genética , Sistema Enzimático do Citocromo P-450/genética , Sistema Enzimático do Citocromo P-450/metabolismo , Trato Gastrointestinal/crescimento & desenvolvimento , Animais , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Família 2 do Citocromo P450 , Epigênese Genética , Trato Gastrointestinal/embriologia , Trato Gastrointestinal/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Células Hep G2 , Humanos , Mesilato de Imatinib/farmacologia , Camundongos
17.
Ann Anat ; 200: 24-9, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25723517

RESUMO

There have been few studies on human embryos describing a specific pattern of hindgut colonization by hematopoietic stem cells (HSCs) and interstitial Cajal cells (ICCs). We aimed to study CD34, CD45 and CD117/c-kit expression in late stage human embryos, to attain observational data that could be related to studies on the aorta-gonad-mesonephros (AGM)-derived HSCs, and data on hindgut ICCs. Antibodies were also applied to identify alpha-smooth muscle actin and neurofilaments. Six human embryos of 48-56 days were used. In the 48 day embryo, the hindgut was sporadically populated by c-kit+ ICCs, but, in all other embryos, a layer of myenteric ICCs had been established. Intraneural c-kit+ cells were found in pelvic nerves and vagal trunks, suggesting that the theory of Ramon y Cajal assuming that ICCs may be primitive neurons may not be so invalid. Also in the 48 day embryo, c-kit+/CD45+ perivascular cells were found along the pelvic neurovascular axes, suggesting that not only liver, but also other organs could be seeded with HSCs from the AGM region. CD45+ cells with dendritic morphologies were found in all hindgut layers, including the epithelium. This last evidence is suggestive of an AGM contribution to the tissue resident macrophages and could be related to processes of sprouting angiogenesis which, in turn, have been found to be guided by filopodia of endothelial tip cells. Further studies on human embryonic and fetal material should be performed to attempt to clarify whether the hindgut colonization with HSCs is a transitory or definitive process.


Assuntos
Células-Tronco Embrionárias/fisiologia , Feto/fisiologia , Trato Gastrointestinal/citologia , Células-Tronco Hematopoéticas/fisiologia , Células Intersticiais de Cajal/fisiologia , Actinas/metabolismo , Adulto , Células Dendríticas/fisiologia , Feminino , Feto/citologia , Trato Gastrointestinal/embriologia , Trato Gastrointestinal/fisiologia , Humanos , Antígenos Comuns de Leucócito/metabolismo , Plexo Mientérico/citologia , Plexo Mientérico/embriologia , Neovascularização Fisiológica/fisiologia , Gravidez , Proteínas Proto-Oncogênicas c-kit/genética , Proteínas Proto-Oncogênicas c-kit/metabolismo
18.
Dev Biol ; 396(1): 1-7, 2014 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-25300580

RESUMO

To feed or breathe, the oral opening must connect with the gut. The foregut and oral tissues converge at the primary mouth, forming the buccopharyngeal membrane (BPM), a bilayer epithelium. Failure to form the opening between gut and mouth has significant ramifications, and many craniofacial disorders have been associated with defects in this process. Oral perforation is characterized by dissolution of the BPM, but little is known about this process. In humans, failure to form a continuous mouth opening is associated with mutations in Hedgehog (Hh) pathway members; however, the role of Hh in primary mouth development is untested. Here, we show, using Xenopus, that Hh signaling is necessary and sufficient to initiate mouth formation, and that Hh activation is required in a dose-dependent fashion to determine the size of the mouth. This activity lies upstream of the previously demonstrated role for Wnt signal inhibition in oral perforation. We then turn to mouse mutants to establish that SHH and Gli3 are indeed necessary for mammalian mouth development. Our data suggest that Hh-mediated BPM persistence may underlie oral defects in human craniofacial syndromes.


Assuntos
Proteínas Hedgehog/metabolismo , Boca/embriologia , Animais , Membrana Basal/embriologia , Epitélio/embriologia , Fibronectinas/metabolismo , Trato Gastrointestinal/embriologia , Regulação da Expressão Gênica no Desenvolvimento , Proteínas Hedgehog/genética , Humanos , Imuno-Histoquímica , Fatores de Transcrição Kruppel-Like/genética , Camundongos , Morfolinas/química , Boca/fisiologia , Proteínas do Tecido Nervoso/genética , Purinas/química , Proteínas Repressoras/genética , Transdução de Sinais , Fatores de Tempo , Proteínas Wnt/metabolismo , Proteínas de Xenopus/genética , Xenopus laevis , Proteína Gli3 com Dedos de Zinco
19.
Dev Growth Differ ; 56(6): 460-8, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25131500

RESUMO

Chemical reagent Ex-527 is widely used as a major inhibitor of Sirtuin enzymes, which are a family of highly conserved protein deacetylases and have been linked with caloric restriction and aging by modulating energy metabolism, genomic stability, and stress resistance. However, the extent to which Ex-527 controls early developmental events of vertebrate embryos remains to be understood. Here, we report an examination of Ex-527 effects during Xenopus early development, followed by a confirmation of expressions of xSirt1 and xSirt2 in embryonic stages and enhancement of acetylation by Ex-527. First, we found that reductions in size of neural plate at neurula stages were induced by Ex-527 treatment. Second, tadpoles with short body length and large edematous swellings in the ventral side were frequently observed. Moreover, Ex-527-treated embryos showed severe gastrointestinal malformations in late tadpole stages. Taken together with these results, we conclude that the Sirtuin family start functioning at early embryonic stages and is required for various developmental events.


Assuntos
Carbazóis/toxicidade , Edema/induzido quimicamente , Embrião não Mamífero/efeitos dos fármacos , Trato Gastrointestinal/anormalidades , Defeitos do Tubo Neural/induzido quimicamente , Sirtuínas/antagonistas & inibidores , Xenopus laevis/embriologia , Animais , Trato Gastrointestinal/embriologia , Defeitos do Tubo Neural/embriologia
20.
Genes Cells ; 19(10): 723-42, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25135772

RESUMO

RE1-silencing transcription factor (REST), also known as NRSF (neuron-restrictive silencer factor), is a well-known transcriptional repressor of neural genes. Rest null mice have embryonic lethality which prevents further investigations of the functions of the Rest gene in vivo. We studied neonatal but not embryonic lethality that was characterized by gastrointestinal tract dilation in the neural crest cell (NCC)-specific Rest conditional knockout (CKO) mice. While no histological abnormalities except the thinning of the digestive tract as a consequence of the gas accumulation were found in the digestive tract of the mutant mice, they do not have proper gastric retention after oral dye administration and the reduction of acetylcholinesterase (AChE) activity in NCC-derived myenteric plexus in the stomach was detected. High CO2 concentration in the dilated digestive tract of the Rest CKO mice indicates a failure of gut function by underdeveloped cholinergic transmission in the enteric nervous system. The observed gastrointestinal distension phenotype provides a model for understanding the genetic and molecular basis of NCC defects in humans.


Assuntos
Gastroenteropatias/patologia , Plexo Mientérico/patologia , Crista Neural/patologia , Proteínas Repressoras/genética , Acetilcolinesterase/metabolismo , Animais , Animais Recém-Nascidos , Dióxido de Carbono/metabolismo , Linhagem da Célula , Dilatação Patológica , Conteúdo Gastrointestinal/química , Gastroenteropatias/genética , Gastroenteropatias/mortalidade , Trato Gastrointestinal/embriologia , Trato Gastrointestinal/crescimento & desenvolvimento , Trato Gastrointestinal/patologia , Camundongos , Camundongos Knockout , Neurônios/patologia , Proteínas Repressoras/metabolismo , Estômago/embriologia , Estômago/crescimento & desenvolvimento , Estômago/inervação , Proteína Wnt1/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA