Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Dev Biol ; 445(2): 256-270, 2019 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-30472119

RESUMO

The enteric nervous system is thought to originate solely from the neural crest. Transgenic lineage tracing revealed a novel population of clonal pancreatic duodenal homeobox-1 (Pdx1)-Cre lineage progenitor cells in the tunica muscularis of the gut that produced pancreatic descendants as well as neurons upon differentiation in vitro. Additionally, an in vivo subpopulation of endoderm lineage enteric neurons, but not glial cells, was seen especially in the proximal gut. Analysis of early transgenic embryos revealed Pdx1-Cre progeny (as well as Sox-17-Cre and Foxa2-Cre progeny) migrating from the developing pancreas and duodenum at E11.5 and contributing to the enteric nervous system. These results show that the mammalian enteric nervous system arises from both the neural crest and the endoderm. Moreover, in adult mice there are separate Wnt1-Cre neural crest stem cells and Pdx1-Cre pancreatic progenitors within the muscle layer of the gut.


Assuntos
Sistema Nervoso Entérico/embriologia , Animais , Linhagem da Célula/genética , Duodeno/embriologia , Duodeno/inervação , Duodeno/metabolismo , Endoderma/citologia , Endoderma/embriologia , Endoderma/metabolismo , Sistema Nervoso Entérico/citologia , Sistema Nervoso Entérico/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Proteínas HMGB/genética , Proteínas HMGB/metabolismo , Fator 3-beta Nuclear de Hepatócito/genética , Fator 3-beta Nuclear de Hepatócito/metabolismo , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Camundongos , Camundongos Transgênicos , Crista Neural/citologia , Crista Neural/embriologia , Crista Neural/metabolismo , Células-Tronco Neurais/citologia , Células-Tronco Neurais/metabolismo , Pâncreas/embriologia , Pâncreas/inervação , Pâncreas/metabolismo , Fatores de Transcrição SOXF/genética , Fatores de Transcrição SOXF/metabolismo , Transativadores/genética , Transativadores/metabolismo , Proteína Wnt1/genética , Proteína Wnt1/metabolismo
2.
PLoS One ; 12(12): e0189894, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29281691

RESUMO

BACKGROUND: Airway mucociliary clearance (MCC) is an important defense mechanism against pulmonary infections and is compromised in cystic fibrosis (CF). Cl- and HCO3- epithelial transport are integral to MCC. During pulmonary infections prostaglandin E2 (PGE2) production is abundant. AIM: To determine the effect of PGE2 on airway Cl- and HCO3- secretion and MCC in normal and CF airways. METHODS: We examined PGE2 stimulated MCC, Cl- and HCO3- secretion using ferret trachea, human bronchial epithelial cell cultures (CFBE41o- with wildtype CFTR (CFBE41 WT) or homozygous F508del CFTR (CFBE41 CF) and human normal bronchial submucosal gland cell line (Calu-3) in Ussing chambers with or without pH-stat. RESULTS: PGE2 stimulated MCC in a dose-dependent manner and was partially impaired by CFTRinh-172. PGE2-stimulated Cl- current in ferret trachea was partially inhibited by CFTRinh-172, with niflumic acid eliminating the residual current. CFBE41 WT cell monolayers produced a robust Cl- and HCO3- secretory response to PGE2, both of which were completely inhibited by CFTRinh-172. CFBE41 CF cells exhibited no response to PGE2. In Calu-3 cells, PGE2 stimulated Cl- and HCO3- secretion. Cl- secretion was partially inhibited by CFTRinh-172, with additional inhibition by niflumic acid. HCO3- secretion was completely inhibited by CFTRinh-172. CONCLUSIONS: PGE2 stimulates bronchotracheal MCC and this response is decreased in CF. In CF airway, PGE2-stimulated Cl- and HCO3- conductance is impaired and may contribute to decreased MCC. There remains a CFTR-independent Cl- current in submucosal glands, which if exploited, could represent a means of improving airway Cl- secretion and MCC in CF.


Assuntos
Bicarbonatos/metabolismo , Brônquios/efeitos dos fármacos , Cloretos/metabolismo , Fibrose Cística/metabolismo , Dinoprostona/farmacologia , Traqueia/efeitos dos fármacos , Animais , Brônquios/metabolismo , Brônquios/patologia , Células Cultivadas , Humanos , Técnicas In Vitro , Traqueia/metabolismo
3.
Biochem Biophys Res Commun ; 424(3): 549-53, 2012 Aug 03.
Artigo em Inglês | MEDLINE | ID: mdl-22771330

RESUMO

The homeodomain transcription factor Pdx1 is essential for pancreas formation and functions in pancreatic islets cells to regulate genes involved in maintenance of glucose homeostasis. In order to investigate a role for Pdx1 in intestinal cells, we analyzed the functions and networks associated with genes differentially expressed by Pdx1 overexpression in human Caco-2 cells. In agreement with previous results for intestine isolated from mice with Pdx1 inactivation, functional analysis of genes differentially expressed with Pdx1 overexpression revealed functions significantly associated with nutrient metabolism. Similarly, network analysis examining the interactions among the differentially expressed genes revealed gene networks involved in lipid metabolism. Consistent with defects in maternal nutrient metabolism, mouse pups born to dams with intestine-specific Pdx1 inactivation are underweight and fail to thrive in the neonatal period compared to pups born to control dams. We conclude that Pdx1 mediates lipid metabolism gene networks in intestinal cells and that maternal expression is essential for perinatal growth in mice.


Assuntos
Regulação da Expressão Gênica , Redes Reguladoras de Genes , Proteínas de Homeodomínio/fisiologia , Mucosa Intestinal/metabolismo , Metabolismo dos Lipídeos/genética , Fenômenos Fisiológicos da Nutrição Materna/genética , Troca Materno-Fetal/genética , Magreza/genética , Transativadores/fisiologia , Animais , Células CACO-2 , Feminino , Peso Fetal/genética , Proteínas de Homeodomínio/genética , Humanos , Intestinos/crescimento & desenvolvimento , Camundongos , Análise de Sequência com Séries de Oligonucleotídeos , Gravidez , Transativadores/genética
4.
Biochem Biophys Res Commun ; 423(1): 183-7, 2012 Jun 22.
Artigo em Inglês | MEDLINE | ID: mdl-22640736

RESUMO

The transcription factor pancreatic and duodenal homeobox 1 (PDX1) plays an essential role in pancreatic development and in maintaining proper islet function via target gene regulation. Few intestinal PDX1 targets, however, have been described. We sought to define novel PDX1-regulated intestinal genes. Caco-2 human intestinal epithelial cells were engineered to overexpress PDX1 and gene expression profiles relative to control cells were assessed. Expression of 80 genes significantly increased while that of 49 genes significantly decreased more than 4-fold following PDX1 overexpression in differentiated Caco-2 cells. Analysis of the differentially regulated genes with known functional annotations revealed genes encoding transcription factors, growth factors, kinases, digestive glycosidases, nutrient transporters, nutrient binding proteins, and structural components. The gene for fatty acid binding protein 1, liver, FABP1, is repressed by PDX1 in Caco-2 cells. PDX1 overexpression in Caco-2 cells also results in repression of promoter activity driven by the 0.6kb FABP1 promoter. PDX1 regulation of promoter activity is consistent with the decrease in FABP1 RNA abundance resulting from PDX1 overexpression and identifies FABP1 as a candidate PDX1 target. PDX1 repression of FABP1, LCT, and SI suggests a role for PDX1 in patterning anterior intestinal development.


Assuntos
Proteínas de Ligação a Ácido Graxo/genética , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio/metabolismo , Mucosa Intestinal/metabolismo , Intestinos/crescimento & desenvolvimento , Transativadores/metabolismo , Células CACO-2 , Perfilação da Expressão Gênica , Proteínas de Homeodomínio/genética , Humanos , Intestinos/citologia , Regiões Promotoras Genéticas , Transativadores/genética , Transcrição Gênica
5.
Hum Genet ; 131(7): 1153-9, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22258180

RESUMO

Lactase is the intestinal enzyme responsible for digestion of the milk sugar lactose. Lactase gene expression declines dramatically upon weaning in mammals and during early childhood in humans (lactase nonpersistence). In various ethnic groups, however, lactase persists in high levels throughout adulthood (lactase persistence). Genetic association studies have identified that lactase persistence in northern Europeans is strongly associated with a single nucleotide polymorphism (SNP) located 14 kb upstream of the lactase gene: -13910*C/T. To determine whether the -13910*T SNP can function in vivo to mediate lactase persistence, we generated transgenic mice harboring human DNA fragments with the -13910*T SNP or the ancestral -13910*C SNP cloned upstream of a 2-kb rat lactase gene promoter in a luciferase reporter construct. We previously reported that the 2-kb rat lactase promoter directs a post-weaning decline of luciferase transgene expression similar to that of the endogenous lactase gene. In the present study, the post-weaning decline directed by the rat lactase promoter is impeded by addition of the -13910*T SNP human DNA fragment, but not by addition of the -13910*C ancestral SNP fragment. Persistence of transgene expression associated with the -13910*T SNP represents the first in vivo data in support of a functional role for the -13910*T SNP in mediating the human lactase persistence phenotype.


Assuntos
Expressão Gênica , Genes Reporter , Lactase/genética , Polimorfismo de Nucleotídeo Único , Transgenes , Animais , Linhagem Celular , Humanos , Lactase/metabolismo , Camundongos , Camundongos Transgênicos , Fenótipo , Regiões Promotoras Genéticas , Ratos
6.
Am J Physiol Gastrointest Liver Physiol ; 302(4): G407-19, 2012 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-22135308

RESUMO

Transcription factor pancreatic and duodenal homeobox 1 (Pdx1) plays an essential role in the pancreas to regulate its development and maintain proper islet function. However, the functions of Pdx1 in mature small intestine are less known. We aimed to investigate the intestinal role of Pdx1 by profiling the expression of genes differentially regulated in response to inactivation of Pdx1 specifically in the intestinal epithelium. Pdx1 was conditionally inactivated in the intestinal epithelium of Pdx1(flox/flox);VilCre mice. Total RNA was isolated from the first 5 cm of the small intestine from mature Pdx1(flox/flox);VilCre and littermate control mice. Microarray analysis identified 86 probe sets representing 68 genes significantly upregulated or downregulated 1.5-fold or greater in Pdx(flox/flox);VilCre mice maintained under standard conditions. Ingenuity Pathway Analysis revealed that functions of the differentially expressed genes are significantly associated with metabolism of nutrients including lipids and iron. Network analysis examining the interactions among the differentially expressed genes further supports the notion that Pdx1 may modulate metabolism of lipids and iron from mature intestinal epithelium. Following forced oil feeding, Pdx1(flox/flox);VilCre mice showed diminished lipid staining in the duodenal epithelium and decreased serum triglyceride levels, indicating reduced lipid absorption compared with control duodenal epithelium. Blood samples from Pdx1(flox/flox);VilCre mice have significantly lower mean values for mean corpuscular volume and mean corpuscular hemoglobin, consistent with iron deficiency. The absence of nonheme iron in the villous epithelium and lamina propria of Pdx1(flox/flox);VilCre duodenum indicates that the duodenal epithelium lacking Pdx1 may have defects in importing iron through enterocytes, resulting in iron deficiency in Pdx1(flox/flox);VilCre mice.


Assuntos
Duodeno/metabolismo , Proteínas de Homeodomínio/genética , Mucosa Intestinal/metabolismo , Ferro/metabolismo , Metabolismo dos Lipídeos/fisiologia , Transativadores/genética , Animais , Enterócitos/metabolismo , Perfilação da Expressão Gênica , Proteínas de Homeodomínio/metabolismo , Camundongos , Camundongos Transgênicos , Transativadores/metabolismo
7.
Trans Am Clin Climatol Assoc ; 122: 155-65, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21686221

RESUMO

Lactase-phlorizin hydrolase, lactase, is the intestinal enzyme responsible for the digestion of the milk sugar lactose. The majority of the world's human population experiences a decline in expression of the lactase gene by late childhood (lactase non-persistence). Individuals with lactase persistence, however, continue to express high levels of the lactase gene throughout adulthood. Lactase persistence is a heritable autosomal dominant condition and has been strongly correlated with several single nucleotide polymorphisms (SNPs) located ∼14 kb upstream of the lactase gene in different ethnic populations: -13910*T in Europeans and -13907*G, -13915*G, and -14010*C in several African populations. The coincidence of the four SNPs clustering within 100 bp strongly suggests that this region mediates the lactase non-persistence/persistence phenotype. Having previously characterized the European SNP, we aimed to determine whether the African SNPs similarly mediate a functional role in regulating the lactase promoter. Human intestinal Caco-2 cells were transfected with lactase SNP/promoter-reporter constructs and assayed for promoter activity. The -13907*G and -13915*G SNPs result in a significant enhancement of lactase promoter activity relative to the ancestral lactase non-persistence genotype. Such differential regulation by the SNPs is consistent with a causative role in the mechanism specifying the lactase persistence phenotype.


Assuntos
População Negra/genética , Erros Inatos do Metabolismo dos Carboidratos/genética , Mucosa Intestinal/enzimologia , Lactase-Florizina Hidrolase/genética , Lactase/genética , Polimorfismo de Nucleotídeo Único , Regiões Promotoras Genéticas , Distinções e Prêmios , Células CACO-2 , Erros Inatos do Metabolismo dos Carboidratos/enzimologia , Erros Inatos do Metabolismo dos Carboidratos/etnologia , Regulação Enzimológica da Expressão Gênica , Predisposição Genética para Doença , Humanos , Hidrólise , Lactase/metabolismo , Lactase-Florizina Hidrolase/metabolismo , Lactose/metabolismo , Fenótipo , Transfecção
8.
Hum Genet ; 129(1): 111-3, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20960210

RESUMO

Lactase gene expression declines with aging (lactase non-persistence) in the majority of humans worldwide. Lactase persistence is a heritable autosomal dominant condition and has been strongly correlated with several single nucleotide polymorphisms (SNPs) located ~14-kb upstream (-13907, -13910 and -13915) of the lactase gene in different ethnic populations. In contrast to the -13907*G and -13910*T SNPs, the -13915*G SNP was previously believed not to interact with Oct-1. In the present study, however, Oct-1 is shown to interact with the -13915*G SNP region DNA sequence by EMSAs and gel supershift. In addition, Oct-1 is capable of enhancing promoter activity of a lactase promoter-reporter construct harboring the 13915*G SNP sequence in cell culture. Oct-1 binding to the -13907 to -13915 SNP region therefore remains a candidate interaction involved in lactase persistence.


Assuntos
Regulação Enzimológica da Expressão Gênica , Lactase/genética , Intolerância à Lactose/genética , Lactose/metabolismo , Fator 1 de Transcrição de Octâmero/genética , Polimorfismo de Nucleotídeo Único , África , Sequência de Bases , Ensaio de Desvio de Mobilidade Eletroforética , Epistasia Genética , Humanos , Fator 1 de Transcrição de Octâmero/metabolismo , Regiões Promotoras Genéticas
9.
Allergy Asthma Clin Immunol ; 6(1): 25, 2010 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-20815913

RESUMO

BACKGROUND: Eosinophilic esophagitis (EoE) is characterized by the inflammation of the esophagus and the infiltration of eosinophils into the esophagus, leading to symptoms such as dysphagia and stricture formation. Systemic immune indicators like eotaxin and fibroblast growth factor were evaluated for possible synergistic pathological effects. Moreover, blood cells, local tissue, and plasma from EoE and control subjects were studied to determine if the localized disease was associated with a systemic effect that correlated with presence of EoE disease. METHOD: Real-time polymerase chain reaction from peripheral blood mononuclear cells (PBMC), immunohistochemistry from local esophageal biopsies, fluid assays on plasma, and fluorescence-activated cell sorting on peripheral blood cells from subjects were used to study the systemic immune indicators in newly diagnosed EoE (n = 35), treated EoE (n = 9), Gastroesophageal reflux disease (GERD) (n = 8), ulcerative colitis (n = 5), Crohn's disease (n = 5), and healthy controls (n = 8). RESULT: Of the transcripts tested for possible immune indicators, we found extracellular signal-regulated kinase (ERK), Bcl-2, bFGF (basic fibroblast growth factor), and eotaxin levels were highly upregulated in PBMC and associated with disease presence of EoE. Increased FGF detected by immunohistochemistry in esophageal tissues and in PBMC was correlated with low levels of pro-apoptotic factors (Fas, Caspase 8) in PBMC from EoE subjects. Plasma-derived bFGF was shown to be the most elevated and most specific in EoE subjects in comparison to healthy controls and disease control subjects. CONCLUSION: We describe for the first time a possible mechanism by which increased FGF is associated with inhibiting apoptosis in local esophageal tissues of EoE subjects as compared to controls. Eotaxin and FGF signaling pathways share activation through the ERK pathway; together, they could act to increase eosinophil activation and prolong the half-life of eosinophils in local tissues of the esophagus in EoE subjects.

10.
J Biomed Opt ; 15(4): 046018, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20799820

RESUMO

Microscopic visualization of islets of Langerhans under normal and diabetic conditions is essential for understanding the pathophysiology of the disease. The intrinsic opacity of pancreata, however, limits optical accessibility for high-resolution light microscopy of islets in situ. Because the standard microtome-based, 2-D tissue analysis confines visualization of the islet architecture at a specific cut plane, 3-D representation of image data is preferable for islet assessment. We applied optical clearing to minimize the random light scattering in the mouse pancreatic tissue. The optical-cleared pancreas allowed penetrative, 3-D microscopic imaging of the islet microstructure and vasculature. Specifically, the islet vasculature was revealed by vessel painting-lipophilic dye labeling of blood vessels-for confocal microscopy. The voxel-based confocal micrographs were digitally processed with projection algorithms for 3-D visualization. Unlike the microtome-based tissue imaging, this optical method for penetrative imaging of mouse islets yielded clear, continuous optical sections for an integrated visualization of the islet microstructure and vasculature with subcellular-level resolution. We thus provide a useful imaging approach to change our conventional planar view of the islet structure into a 3-D panorama for better understanding of the islet physiology.


Assuntos
Interpretação de Imagem Assistida por Computador/métodos , Imageamento Tridimensional/métodos , Ilhotas Pancreáticas/irrigação sanguínea , Ilhotas Pancreáticas/citologia , Microscopia Confocal/métodos , Microvasos/ultraestrutura , Frações Subcelulares/ultraestrutura , Algoritmos , Animais , Desenho de Equipamento , Análise de Falha de Equipamento , Aumento da Imagem/métodos , Camundongos , Reprodutibilidade dos Testes , Sensibilidade e Especificidade , Integração de Sistemas
11.
J Pediatr Gastroenterol Nutr ; 51(3): 290-4, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20639774

RESUMO

OBJECTIVE: The aim of the study was to investigate whether eosinophils have increased human leukocyte antigen (HLA)-DR expression in subjects with eosinophilic esophagitis (EoE) compared with controls. PATIENTS AND METHODS: Patients who were undergoing an upper endoscopy with biopsies for suspected gastroesophageal reflux disease (GERD) or EoE at Lucile Packard Children's Hospital were enrolled. In total, the blood and tissue samples of 10 healthy controls (HC), 11 subjects with GERD, and 10 with EoE were studied. Multiple tissue staining to identify eosinophils (via eosinophil cationic protein-clone EG2) and major histocompatibility complex class II cell surface receptors (via HLA-DR) was performed via immunohistochemistry. The peripheral blood was analyzed using flow cytometry to detect eosinophil HLA-DR expression among these subjects. RESULTS: In the tissue, a greater proportion of eosinophils expressed HLA-DR among the subjects with EoE (mean 0.83 +/- 0.14, n = 9) relative to those with GERD (mean 0.18 +/- 0.19, n = 8, P < 0.01) and HC (mean 0.18 +/- 0.13, n = 6, P < 0.01). In total, 6 participants (4 HC subjects and 2 subjects with GERD) did not have any eosinophils identified on tissue staining and were unable to be included in the present statistical analysis. In the blood, there was no statistically significant difference in eosinophil HLA-DR expression among HC subjects (mean 415 +/- 217, n = 6), subjects with GERD (mean 507 +/- 429, n = 2), and those with EoE (mean 334 +/- 181, n = 6). CONCLUSIONS: These data demonstrate that the eosinophils from the esophagus of subjects with EoE have increased HLA-DR expression within this tissue.


Assuntos
Esofagite Eosinofílica/imunologia , Eosinófilos/metabolismo , Esôfago/imunologia , Refluxo Gastroesofágico/imunologia , Antígenos HLA-DR/metabolismo , Adolescente , Adulto , Criança , Pré-Escolar , Esofagite Eosinofílica/metabolismo , Eosinófilos/imunologia , Esôfago/metabolismo , Feminino , Refluxo Gastroesofágico/metabolismo , Humanos , Lactente , Masculino , Adulto Jovem
12.
Am J Physiol Gastrointest Liver Physiol ; 297(6): G1126-37, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19808654

RESUMO

Null mutant mice lacking the transcription factor pancreatic and duodenal homeobox 1 (Pdx1) are apancreatic and survive only a few days after birth. The role of Pdx1 in regulating intestinal gene expression has therefore yet to be determined in viable mice with normal pancreatic development. We hypothesized that conditional inactivation of Pdx1 restricted to the intestinal epithelium would alter intestinal gene expression and cell differentiation. Pdx1(flox/flox);VilCre mice with intestine-specific Pdx1 inactivation were generated by crossing a transgenic mouse strain expressing Cre recombinase, driven by a mouse villin 1 gene promoter fragment, with a mutant mouse strain homozygous for loxP site-flanked Pdx1. Pdx1 protein is undetectable in all epithelial cells in the intestinal epithelium of Pdx1(flox/flox);VilCre mice. Goblet cell number and mRNA abundance for mucin 3 and mucin 13 genes in the proximal small intestine are comparable between Pdx1(flox/flox);VilCre and control mice. Similarly, Paneth cell number and expression of Paneth cell-related genes Defa1, Defcr-rs1, and Mmp7 in the proximal small intestine remain statistically unchanged by Pdx1 inactivation. Although the number of enteroendocrine cells expressing chromogranin A/B, gastric inhibitory polypeptide (Gip), or somatostatin (Sst) is unaffected in the Pdx1(flox/flox);VilCre mice, mRNA abundance for Gip and Sst is significantly reduced in the proximal small intestine. Conditional Pdx1 inactivation attenuates intestinal alkaline phosphatase (IAP) activity in the duodenal epithelium, consistent with an average 91% decrease in expression of the mouse enterocyte IAP gene, alkaline phosphatase 3 (a novel Pdx1 target candidate), in the proximal small intestine following Pdx1 inactivation. We conclude that Pdx1 is necessary for patterning appropriate gene expression in enterocytes and enteroendocrine cells of the proximal small intestine.


Assuntos
Duodeno/metabolismo , Enterócitos/metabolismo , Células Enteroendócrinas/metabolismo , Regulação da Expressão Gênica , Células Caliciformes/metabolismo , Mucosa Intestinal/metabolismo , Celulas de Paneth/metabolismo , Transativadores/deficiência , Animais , Diferenciação Celular , Proteínas de Homeodomínio/genética , Integrases/genética , Camundongos , Camundongos Mutantes , Camundongos Transgênicos , Proteínas dos Microfilamentos/genética , Regiões Promotoras Genéticas , RNA Mensageiro/metabolismo , Transativadores/genética
13.
Gastroenterology ; 137(2): 453-65, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19447107

RESUMO

BACKGROUND & AIMS: The intrinsic opacity of mouse intestinal tissue prevents its evaluation by high-resolution, in-depth optical microscopy. Instead, intestinal tissue is usually sectioned to expose the interior domains of the mucosa and submucosa for microscopic examination. However, microtome sectioning can cause distortions and artifacts that prevent acquisition of an accurate view of the sample. We therefore attempted to develop a microtome-free 3-dimensional (3D) confocal imaging method for characterization of mouse intestine. METHODS: We applied an optical-clearing solution, FocusClear, to permeate and reduce the opacity of mouse colon and ileum. Tissues were labeled with fluorescent probes and examined by confocal microscopy with efficient fluorescence excitation and emission in the FocusClear solution. The voxel-based confocal micrographs were processed with Amira software for 3D visualization and analysis. RESULTS: Treatment of tissues with the optical-clearing solution improved photon penetration, resulting in the acquisition of images with subcellular-level resolution across the mucosa, submucosa, and muscle layers. Collectively, the acquired image stacks were processed by projection algorithms for 3D analysis of the spatial relations in villi, crypts, and connective tissues. These imaging technologies allowed for identification of spatiotemporal changes in crypt morphology of colon tissues from mice with dextran sulfate sodium-induced colitis as well as detection of transgenic fluorescent proteins expressed in the colon and ileum. CONCLUSIONS: This new optical method for penetrative imaging of mouse intestine does not require tissue sectioning and provides a useful tool for 3D presentation and analysis of diseased and transgenic intestine in an integrated fashion.


Assuntos
Colite Ulcerativa/patologia , Colo/ultraestrutura , Íleo/ultraestrutura , Interpretação de Imagem Assistida por Computador , Imageamento Tridimensional/métodos , Microscopia Confocal/instrumentação , Animais , Colo/patologia , Modelos Animais de Doenças , Feminino , Tecnologia de Fibra Óptica , Íleo/patologia , Mucosa Intestinal/patologia , Mucosa Intestinal/ultraestrutura , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Transgênicos , Microscopia Confocal/métodos , Probabilidade , Distribuição Aleatória , Valores de Referência , Sensibilidade e Especificidade
14.
J Gen Virol ; 89(Pt 12): 3004-3008, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19008386

RESUMO

Enteropathogens are known to disrupt apical actin filaments and/or tight-junction barriers of intestinal epithelial cells to promote infection. In this study, we show that a controlled, cytochalasin-D (Cyto-D)-mediated disruption of actin filaments and tight junctions enhanced the apical delivery of the gene-therapy vector recombinant adeno-associated virus serotype 2 (rAAV2). This increase in transduction efficiency can be attributed to the enhanced delivery of rAAV2 across the Cyto-D disrupted tight junctions, allowing basolateral entry of rAAV2. Previously, we have shown that MG101 and doxorubicin are capable of overcoming proteasome-mediated transduction barriers of rAAV2 in enterocytes. In this study, when Cyto-D was combined with MG101 and doxorubicin in apical delivery of rAAV2 to transduce the differentiated Caco-2 enterocytes, a synergistic >2300-fold increase in transgene expression was achieved. We conclude that Cyto-D is capable of permeating the polarized enterocytes for rAAV2 transduction, which may potentially be a useful device to facilitate intestinal gene transfer via the gut lumen.


Assuntos
Citocalasina D/farmacologia , Dependovirus/fisiologia , Enterócitos , Vetores Genéticos , Recombinação Genética , Junções Íntimas , Transdução Genética , Citoesqueleto de Actina/efeitos dos fármacos , Citoesqueleto de Actina/virologia , Células CACO-2 , Citocalasina D/metabolismo , Dependovirus/genética , Enterócitos/efeitos dos fármacos , Enterócitos/virologia , Terapia Genética/métodos , Humanos , Junções Íntimas/efeitos dos fármacos , Junções Íntimas/virologia
15.
Cancer Biol Ther ; 7(2): 303-9, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18059187

RESUMO

p53 gene transfer has been proposed as a potential therapeutic option for treatment of hepatocellular carcinoma (HCC). Compared to other commonly used gene transfer vectors such as adenovirus and retrovirus, recombinant adeno-associated virus serotype 2 (rAAV2) has shown promising results in human clinical trials. Significant enhancement in the gene transfer efficiency is needed, however, for HCC applications. In the present study, we applied chemotherapy drug Doxorubicin (DOX) to induce rAAV2 transduction of hepatomas. Using reporter assays, we showed that the DOX-treated hepatomas became more susceptible to rAAV2 infection in comparison to untreated controls: the permissiveness increased >350-fold and >120-fold for HepG2 (p53 wild-type) and Hep3B (p53 null) hepatomas, respectively. Using the induced permissiveness, we applied rAAV2-p53 transduction to restore p53 expression in the p53-null Hep3B hepatomas. Compared to rAAV2-p53 transduction alone, rAAV2-p53 transduction with DOX resulted in a >16-fold induction of p53 expression. The transduced Hep3B expressed as much as 380% more immunoreactive p53 in comparison to the wild-type p53 expression in the HepG2 hepatomas. Significantly, when Hep3B cells were treated with 0.5 muM of DOX and rAAV2-p53 (MOI = 10) for twelve hours, the cell viability dropped to 66% four days after the administration. This decrease in cell viability was similar to that of treatment with 1 microM of DOX alone in the absence of rAAV2. The 50% reduction in DOX administration--from 1 microM to 0.5 microM--revealed the antitumor property of the rAAV2-p53 transduction as well as the joint cytotoxicity of DOX and rAAV2-p53 against the p53-null hepatomas. We conclude that DOX mediates the enhancement effect on rAAV2 transduction of human hepatomas. Combined DOX and rAAV2-p53 administration may facilitate more efficient treatment for the HCC caused by p53 mutations.


Assuntos
Carcinoma Hepatocelular/terapia , Dependovirus/genética , Doxorrubicina/uso terapêutico , Técnicas de Transferência de Genes , Genes p53 , Neoplasias Hepáticas/terapia , Proteína Supressora de Tumor p53/genética , Antibióticos Antineoplásicos/uso terapêutico , Carcinoma Hepatocelular/genética , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Ensaios Clínicos Fase I como Assunto , Ensaios Clínicos Fase II como Assunto , Terapia Combinada , Relação Dose-Resposta a Droga , Corantes Fluorescentes/metabolismo , Genes Reporter , Terapia Genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Neoplasias Hepáticas/genética , Luciferases/metabolismo , Proteína Supressora de Tumor p53/metabolismo
17.
DNA Cell Biol ; 25(4): 215-22, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16629594

RESUMO

Lactase gene expression is spatiotemporally regulated during mammalian gut development. We hypothesize that distinct DNA control regions specify appropriate spatial and temporal patterning of lactase gene expression. In order to define regions of the lactase promoter involved in mediating intestine-specific and spatiotemporal restricted expression, transgenic mice harboring 100 bp, 1.3- and 2.0- kb fragments of the 5' flanking region of the rat lactase gene cloned upstream of a luciferase reporter were characterized. The 100-bp lactase promoter-reporter transgenic mouse line expressed maximal luciferase activity in the intestine with a posterior shift in spatial restriction and ectopic expression in the stomach and lung. The temporal pattern of expression mediated by the 1.3-kb promoter?reporter transgene increases with postnatal maturation in contrast with the postnatal decline mediated by the 2.0-kb promoter-reporter transgene and the endogenous lactase gene. The differential transgene expression patterns mediated by the lactase promoter fragments suggests that intestine-specific spatial and temporal control elements reside in distinct regions of the DNA sequences upstream of the lactase gene transcription start-site.


Assuntos
Envelhecimento/fisiologia , Regulação Enzimológica da Expressão Gênica , Intestino Delgado/enzimologia , Lactase/genética , Regiões Promotoras Genéticas , Região 5'-Flanqueadora , Animais , Genes Reporter , Luciferases/metabolismo , Camundongos , Camundongos Transgênicos , Modelos Genéticos , Especificidade de Órgãos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transcrição Gênica , Transgenes
18.
Gene Expr Patterns ; 6(4): 426-32, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16377257

RESUMO

The small intestine matures from a primitive tube into morphologically and functionally distinct regions during gut development. Maximal expression of the genes encoding the digestive enzymes lactase-phlorizin hydrolase and sucrase-isomaltase is spatially restricted to distinct segments along the anterior-posterior axis of the small intestine and is temporally regulated during postnatal maturation. Transcription factors capable of interacting with the intestinal lactase and sucrase gene promoters are candidate regulators of spatio-temporal patterning during gut development and maturation. We aimed to quantitatively examine and compare the relative expression levels of a set of intestine-specific transcription factors along the anterior-posterior gut axis during postnatal maturation. Our analysis was focused on the transcription factors capable of regulating the intestinal lactase and sucrase-isomaltase genes. A real-time PCR protocol was used to quantitatively examine and compare spatially and temporally the relative transcript abundance levels for intestine-specific factors during postnatal intestinal maturation. Distinct spatial expressions patterns were detected along the length of the small intestine for PDX-1, Cdx-2, GATA-4, GATA-5, GATA-6, HNF-1alpha, HNF-1beta and CDP transcription factor genes. There is a general decline in transcript abundance for the factor genes during postnatal maturation. Defining the spatio-temporal expression patterns for intestine-specific transcription factor genes contributes to investigation of the roles that factor gradients play in mediating gut development and differentiation.


Assuntos
Padronização Corporal , Regulação da Expressão Gênica , Intestino Delgado/crescimento & desenvolvimento , Intestino Delgado/metabolismo , Fatores de Transcrição/metabolismo , Animais , Fator de Transcrição CDX2 , Fator de Transcrição GATA4/genética , Fator de Transcrição GATA4/metabolismo , Fator de Transcrição GATA5/genética , Fator de Transcrição GATA5/metabolismo , Fator de Transcrição GATA6/genética , Fator de Transcrição GATA6/metabolismo , Fator 1-alfa Nuclear de Hepatócito/genética , Fator 1-alfa Nuclear de Hepatócito/metabolismo , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Intestino Delgado/enzimologia , Camundongos , Camundongos Endogâmicos BALB C , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , RNA Mensageiro/análise , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Complexo Sacarase-Isomaltase/genética , Complexo Sacarase-Isomaltase/metabolismo , Fatores de Tempo , Transativadores/genética , Transativadores/metabolismo , Fatores de Transcrição/genética
19.
Biochem Biophys Res Commun ; 331(4): 1392-400, 2005 Jun 17.
Artigo em Inglês | MEDLINE | ID: mdl-15883029

RESUMO

Intestinal gene transfer offers promise as a therapeutic option for treatment of both intestinal and non-intestinal diseases. Recombinant adeno-associated virus serotype 2, rAAV2, based vectors have been utilized to transduce lung epithelial cells in culture and in human subjects. rAAV2 transduction of intestinal epithelial cells, however, is limited both in culture and in vivo. Proteasome-inhibiting agents have recently been shown to enhance rAAV2-mediated transgene expression in airway epithelial cells. We hypothesized that similar inhibition of proteasome-related cellular processes can function to induce rAAV2 transduction of intestinal epithelial cells. Our results demonstrate that combined treatment with proteasome-modulating agents MG101 (N-acetyl-L-leucyl-L-leucyl-L-norleucine) and Doxorubicin synergistically induces rAAV2-mediated luciferase transgene expression by >400-fold in undifferentiated Caco-2 cells. In differentiated Caco-2 monolayers, treatment with MG101 and Doxorubicin induces transduction preferentially from the basolateral cell surface. In addition to Caco-2 cells, treatment with MG101 and Doxorubicin also results in enhanced rAAV2 transduction of HT-29, T84, and HCT-116 human intestinal epithelial cell lines. We conclude that MG101 and Doxorubicin mediate generic effects on intestinal epithelial cells that result in enhanced rAAV2 transduction. Use of proteasome-modulating agents to enhance viral transduction may facilitate the development of more efficient intestinal gene transfer protocols.


Assuntos
Inibidores de Cisteína Proteinase/farmacologia , Dependovirus/genética , Doxorrubicina/farmacologia , Mucosa Intestinal/efeitos dos fármacos , Oligopeptídeos/farmacologia , Complexo de Endopeptidases do Proteassoma/efeitos dos fármacos , Transgenes , Células CACO-2 , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Humanos , Mucosa Intestinal/metabolismo , Transdução Genética
20.
Genet Test ; 8(2): 190-3, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15345119

RESUMO

The majority of the world's human population experiences a decline of lactase gene expression during maturation, so-called lactase nonpersistence. Thus, adults with lactase nonpersistence are susceptible to developing symptoms of lactose intolerance. By contrast, lactase persistence is an autosomal dominant heritable condition that results in a high level of lactase gene expression throughout adulthood and sustained lactose tolerance. Lactase persistence has recently been correlated with a single nucleotide genetic variant (a C --> T mutation) located 13,910 bases upstream from the lactase structural gene. We aimed to develop a restriction fragment length polymorphism (RFLP) method of detecting the C/T variants as a means of identifying individuals genetically inclined toward lactase persistence or nonpersistence. Genomic DNA in a 210-bp region surrounding the -13,910-bp variant site was PCR amplified with unique primers designed to avoid or mutate adjacent restriction sites. The amplified DNA was digested with a restriction enzyme, CviJI, that recognizes the base pair sequence generated by the lactase nonpersistence variant. Restriction digest gel analysis yielded DNA fragments of the expected diagnostic molecular weight sizes for individuals that were homozygote or heterozygote for the lactase persistence and nonpersistence variants. The genotypes predicted by the RFLP-based method were confirmed by DNA sequence analysis. The RFLP-based method provides a quick and noninvasive means of molecular detection of the presence or absence of the lactase persistence variant.


Assuntos
Lactase-Florizina Hidrolase/genética , Lactase/genética , Reação em Cadeia da Polimerase/métodos , Polimorfismo Genético , Adulto , Sequência de Bases , Humanos , Intolerância à Lactose/genética , Polimorfismo de Nucleotídeo Único
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...