Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Autophagy ; 15(11): 1954-1969, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-30894069

RESUMO

Impaired macroautophagy/autophagy has been implicated in experimental and human pancreatitis. However, the transcriptional control governing the autophagy-lysosomal process in pancreatitis is largely unknown. We investigated the role and mechanisms of TFEB (transcription factor EB), a master regulator of lysosomal biogenesis, in the pathogenesis of experimental pancreatitis. We analyzed autophagic flux, TFEB nuclear translocation, lysosomal biogenesis, inflammation and fibrosis in GFP-LC3 transgenic mice, acinar cell-specific tfeb knockout (KO) and tfeb and tfe3 double-knockout (DKO) mice as well as human pancreatitis samples. We found that cerulein activated MTOR (mechanistic target of rapamycin kinase) and increased the levels of phosphorylated TFEB as well as pancreatic proteasome activities that led to rapid TFEB degradation. As a result, cerulein decreased the number of lysosomes resulting in insufficient autophagy in mouse pancreas. Pharmacological inhibition of MTOR or proteasome partially rescued cerulein-induced TFEB degradation and pancreatic damage. Furthermore, genetic deletion of tfeb specifically in mouse pancreatic acinar cells increased pancreatic edema, necrotic cell death, infiltration of inflammatory cells and fibrosis in pancreas after cerulein treatment. tfeb and tfe3 DKO mice also developed spontaneous pancreatitis with increased pancreatic trypsin activities, edema and infiltration of inflammatory cells. Finally, decreased TFEB nuclear staining was associated with human pancreatitis. In conclusion, our results indicate a critical role of impaired TFEB-mediated lysosomal biogenesis in promoting the pathogenesis of pancreatitis. Abbreviations: AC: acinar cell; AMY: amylase; ATP6V1A: ATPase, H+ transporting, lysosomal V1 subunit A; ATP6V1B2: ATPase, H+ transporting, lysosomal V1 subunit B2; ATP6V1D: ATPase, H+ transporting, lysosomal V1 subunit D; ATP6V1H: ATPase, H+ transporting, lysosomal V1 subunit H; AV: autophagic vacuole; CDE: choline-deficient, ethionine-supplemented; CLEAR: coordinated lysosomal expression and regulation; CQ: chloroquine; EIF4EBP1: eukaryotic translation initiation factor 4E binding protein 1; EM: electron microscopy; GAPDH: glyceraldehyde-3-phosphate dehydrogenase; GFP: green fluorescent protein; H & E: hematoxylin and eosin; KO: knockout; LAMP1: lysosomal-associated membrane protein 1; MAP1LC3/LC3: microtubule associated protein 1 light chain 3; MAPK1/ERK2: mitogen-activated protein kinase 1; MTORC1: mechanistic target of rapamycin kinase complex 1; ND: normal donor; NEU: neutrophil; PPARGC1A/PGC1α: peroxisome proliferator-activated receptor, gamma, coactivator 1 alpha; RIPA: radio-immunoprecipitation; RPS6: ribosomal protein S6; SQSTM1/p62: sequestosome 1; TFEB: transcription factor EB; TM: tamoxifen; WT: wild-type; ZG: zymogen granule.


Assuntos
Células Acinares/metabolismo , Autofagossomos/metabolismo , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/metabolismo , Lisossomos/metabolismo , Pancreatite/metabolismo , Células Acinares/efeitos dos fármacos , Células Acinares/enzimologia , Animais , Autofagossomos/efeitos dos fármacos , Autofagossomos/ultraestrutura , Autofagia/efeitos dos fármacos , Autofagia/genética , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/química , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/genética , Núcleo Celular/metabolismo , Ceruletídeo/toxicidade , Modelos Animais de Doenças , Humanos , Inflamação/metabolismo , Lisossomos/efeitos dos fármacos , Lisossomos/genética , Lisossomos/ultraestrutura , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Pâncreas/efeitos dos fármacos , Pâncreas/enzimologia , Pâncreas/metabolismo , Pâncreas/patologia , Pancreatite/induzido quimicamente , Pancreatite/enzimologia , Pancreatite/genética , Fosforilação , Complexo de Endopeptidases do Proteassoma/efeitos dos fármacos , Complexo de Endopeptidases do Proteassoma/genética , Complexo de Endopeptidases do Proteassoma/metabolismo , Serina-Treonina Quinases TOR/antagonistas & inibidores , Serina-Treonina Quinases TOR/genética , Serina-Treonina Quinases TOR/metabolismo
2.
Alcohol Clin Exp Res ; 40(6): 1215-23, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27130888

RESUMO

Alcoholic liver disease (ALD) is a major health problem in the United States and worldwide without successful treatments. Chronic alcohol consumption can lead to ALD, which is characterized by steatosis, inflammation, fibrosis, cirrhosis, and even liver cancer. Recent studies suggest that alcohol induces both cell death and adaptive cell survival pathways in the liver, and the balance of cell death and cell survival ultimately decides the pathogenesis of ALD. This review summarizes the recent progress on the role and mechanisms of apoptosis, necroptosis, and autophagy in the pathogenesis of ALD. Understanding the complex regulation of apoptosis, necrosis, and autophagy may help to develop novel therapeutic strategies by targeting all 3 pathways simultaneously.


Assuntos
Apoptose/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Etanol/efeitos adversos , Hepatopatias Alcoólicas/fisiopatologia , Necrose , Animais , Humanos
3.
J Pediatr Gastroenterol Nutr ; 62(4): 627-34, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26551319

RESUMO

OBJECTIVES: Cystic fibrosis (CF) has major effects on the intestinal tract with potential consequences on nutrition, but these are not fully understood. I investigated the possibility of altered enterocyte maturation in CF, as suggested by decreased enterocyte nutrient assimilation gene expression in published transcriptome analysis of the small intestine of CF mouse. METHODS: In CF and wild-type (WT) mice, enterocyte gene/protein expression was analyzed by quantitative realtime polymerase chain reaction (qRT-PCR), enzyme histochemistry, immunohistochemistry, and Western blot. One group of mice was maintained on a control liquid diet; to manipulate the gut microbiota, a second group was treated with oral antibiotics; to improve hydration of the gut lumen, a third group was given a laxative drinking solution. RESULTS: On the control diet in the CF intestine, there were decreased levels (67%-85% reduction of WT levels) of enterocyte genes/proteins. Antibiotics did not normalize the expression of enterocyte markers in the CF mouse. In contrast, the laxative treatment of CF mice significantly increased expression to near WT levels. CONCLUSIONS: These studies suggest that the environment of the CF intestinal lumen plays a role in reduced maturation of enterocytes. Because changing the gut lumen environment can affect enterocyte maturation, this is not a cell-autonomous effect of loss of CF transmembrane conductance regulator.


Assuntos
Fibrose Cística/metabolismo , Regulação para Baixo , Enterócitos/metabolismo , Regulação da Expressão Gênica , Mucosa Intestinal/metabolismo , Intestino Delgado/metabolismo , Animais , Antibacterianos/uso terapêutico , Diferenciação Celular/efeitos dos fármacos , Terapia Combinada , Fibrose Cística/microbiologia , Fibrose Cística/patologia , Fibrose Cística/terapia , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Regulação para Baixo/efeitos dos fármacos , Disbiose/etiologia , Disbiose/prevenção & controle , Metabolismo Energético/efeitos dos fármacos , Enterócitos/efeitos dos fármacos , Enterócitos/microbiologia , Enterócitos/patologia , Feminino , Hidratação , Microbioma Gastrointestinal/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/microbiologia , Mucosa Intestinal/patologia , Intestino Delgado/efeitos dos fármacos , Intestino Delgado/microbiologia , Intestino Delgado/patologia , Laxantes/uso terapêutico , Masculino , Camundongos Knockout
4.
Cell Tissue Res ; 355(1): 131-42, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24169862

RESUMO

The tight junction (TJ) is the major determinant of paracellular permeability, which in the gut protects the body from entry of harmful substances such as microbial components. In cystic fibrosis (CF), there is increased permeability of the small intestine both in humans and in CF mice. To gain insight into the mechanisms of increased intestinal permeability in CF, I analyze the composition of the TJ in a cystic fibrosis transmembrane conductance regulator (Cftr) knockout mouse model. Significant changes in TJ gene expression in the CF intestine were found for Cldn1, Cldn7, Cldn8 and Pmp22, which were expressed at lower levels and Cldn2 that was expressed at a higher level. Protein levels of claudin-2 were increased in the CF intestine as compared to wild-type, while other TJ proteins were not significantly different. In the villus epithelium of the CF intestine, all TJ components analyzed were mislocalized to the basal cytoplasm and showed varying degrees of loss from the TJ and apico-lateral surfaces. The pore-forming claudin-2 in the CF intestine showed more intense staining but was correctly localized to the TJ, principally in the crypts that are enlarged in CF. The cytokine TNFα, known to affect TJ, was elevated to 160% of wild-type in the CF intestine. In summary, there is a dramatic redistribution of claudin proteins from the TJ/lateral membrane to the basal cytoplasm of the villus epithelium in the CF intestine. These changes in TJ protein localization in CF are likely to be involved in the increased permeability of the CF small intestine to macromolecules and TNFα may be a causative factor.


Assuntos
Fibrose Cística/patologia , Intestino Delgado/patologia , Junções Íntimas/patologia , Animais , Claudinas/análise , Claudinas/genética , Fibrose Cística/genética , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Citocinas/análise , Regulação da Expressão Gênica , Intestino Delgado/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Permeabilidade , Junções Íntimas/metabolismo
5.
PLoS One ; 8(11): e82149, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24303081

RESUMO

BACKGROUND: ZIP5 localizes to the baso-lateral membranes of intestinal enterocytes and pancreatic acinar cells and is internalized and degraded coordinately in these cell-types during periods of dietary zinc deficiency. These cell-types are thought to control zinc excretion from the body. The baso-lateral localization and zinc-regulation of ZIP5 in these cells are unique among the 14 members of the Slc39a family and suggest that ZIP5 plays a role in zinc excretion. METHODS/PRINCIPAL FINDINGS: We created mice with floxed Zip5 genes and deleted this gene in the entire mouse or specifically in enterocytes or acinar cells and then examined the effects on zinc homeostasis. We found that ZIP5 is not essential for growth and viability but total knockout of ZIP5 led to increased zinc in the liver in mice fed a zinc-adequate (ZnA) diet but impaired accumulation of pancreatic zinc in mice fed a zinc-excess (ZnE) diet. Loss-of-function of enterocyte ZIP5, in contrast, led to increased pancreatic zinc in mice fed a ZnA diet and increased abundance of intestinal Zip4 mRNA. Finally, loss-of-function of acinar cell ZIP5 modestly reduced pancreatic zinc in mice fed a ZnA diet but did not impair zinc uptake as measured by the rapid accumulation of (67)zinc. Retention of pancreatic (67)zinc was impaired in these mice but the absence of pancreatic ZIP5 sensitized them to zinc-induced pancreatitis and exacerbated the formation of large cytoplasmic vacuoles containing secretory protein in acinar cells. CONCLUSIONS: These studies demonstrate that ZIP5 participates in the control of zinc excretion in mice. Specifically, they reveal a paramount function of intestinal ZIP5 in zinc excretion but suggest a role for pancreatic ZIP5 in zinc accumulation/retention in acinar cells. ZIP5 functions in acinar cells to protect against zinc-induced acute pancreatitis and attenuate the process of zymophagy. This suggests that it may play a role in autophagy.


Assuntos
Proteínas de Transporte de Cátions/metabolismo , Mucosa Intestinal/metabolismo , Pâncreas/metabolismo , Zinco/metabolismo , Células Acinares/metabolismo , Animais , Proteínas de Transporte de Cátions/genética , Feminino , Técnicas de Inativação de Genes , Ordem dos Genes , Marcação de Genes , Fígado/metabolismo , Camundongos , Camundongos Transgênicos , Pâncreas/efeitos dos fármacos , Pancreatina , Pancreatite/induzido quimicamente , Pancreatite/genética , Pancreatite/metabolismo , Pancreatite/patologia , Zinco/toxicidade
6.
PLoS One ; 8(8): e72543, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24015258

RESUMO

BACKGROUND: Zinc deficiency due to poor nutrition or genetic mutations in zinc transporters is a global health problem and approaches to providing effective dietary zinc supplementation while avoiding potential toxic side effects are needed. METHODS/PRINCIPAL FINDINGS: Conditional knockout of the intestinal zinc transporter Zip4 (Slc39a4) in mice creates a model of the lethal human genetic disease acrodermatitis enteropathica (AE). This knockout leads to acute zinc deficiency resulting in rapid weight loss, disrupted intestine integrity and eventually lethality, and therefore provides a model system in which to examine novel approaches to zinc supplementation. We examined the efficacy of dietary clioquinol (CQ), a well characterized zinc chelator/ionophore, in rescuing the Zip4 (intest KO) phenotype. By 8 days after initiation of the knockout neither dietary CQ nor zinc supplementation in the drinking water was found to be effective at improving this phenotype. In contrast, dietary CQ in conjunction with zinc supplementation was highly effective. Dietary CQ with zinc supplementation rapidly restored intestine stem cell division and differentiation of secretory and the absorptive cells. These changes were accompanied by rapid growth and dramatically increased longevity in the majority of mice, as well as the apparent restoration of the homeostasis of several essential metals in the liver. CONCLUSIONS: These studies suggest that oral CQ (or other 8-hydroxyquinolines) coupled with zinc supplementation could provide a facile approach toward treating zinc deficiency in humans by stimulating stem cell proliferation and differentiation of intestinal epithelial cells.


Assuntos
Acrodermatite/tratamento farmacológico , Clioquinol/farmacologia , Suplementos Nutricionais , Zinco/deficiência , Zinco/farmacologia , Acrodermatite/genética , Acrodermatite/metabolismo , Acrodermatite/patologia , Animais , Proteínas de Transporte de Cátions/genética , Proteínas de Transporte de Cátions/metabolismo , Clioquinol/agonistas , Modelos Animais de Doenças , Sinergismo Farmacológico , Humanos , Mucosa Intestinal/metabolismo , Intestinos/patologia , Fígado/metabolismo , Fígado/patologia , Camundongos , Camundongos Knockout , Zinco/agonistas , Zinco/metabolismo
7.
Cold Spring Harb Perspect Med ; 3(9): a009753, 2013 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-23788646

RESUMO

The clinical manifestations of cystic fibrosis (CF) result from dysfunction of the cystic fibrosis transmembrane regulator protein (CFTR). The majority of people with CF have a limited life span as a consequence of CFTR dysfunction in the respiratory tract. However, CFTR dysfunction in the gastrointestinal (GI) tract occurs earlier in ontogeny and is present in all patients, regardless of genotype. The same pathophysiologic triad of obstruction, infection, and inflammation that causes disease in the airways also causes disease in the intestines. This article describes the effects of CFTR dysfunction on the intestinal tissues and the intraluminal environment. Mouse models of CF have greatly advanced our understanding of the GI manifestations of CF, which can be directly applied to understanding CF disease in humans.


Assuntos
Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Fibrose Cística/complicações , Disbiose/etiologia , Gastroenterite/etiologia , Obstrução Intestinal/etiologia , Animais , Bicarbonatos/metabolismo , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Modelos Animais de Doenças , Insuficiência de Crescimento/etiologia , Humanos , Concentração de Íons de Hidrogênio , Mucosa Intestinal/metabolismo , Desnutrição/etiologia , Camundongos , RNA Mensageiro/metabolismo
8.
BMC Physiol ; 13: 6, 2013 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-23537040

RESUMO

BACKGROUND: The vestibular system controls the ion composition of its luminal fluid through several epithelial cell transport mechanisms under hormonal regulation. The semicircular canal duct (SCCD) epithelium has been shown to secrete Cl- under ß2-adrenergic stimulation. In the current study, we sought to determine the ion transporters involved in Cl- secretion and whether secretion is regulated by PKA and glucocorticoids. RESULTS: Short circuit current (Isc) from rat SCCD epithelia demonstrated stimulation by forskolin (EC50: 0.8 µM), 8-Br-cAMP (EC50: 180 µM), 8-pCPT-cAMP (100 µM), IBMX (250 µM), and RO-20-1724 (100 µM). The PKA activator N6-BNZ-cAMP (0.1, 0.3 & 1 mM) also stimulated Isc. Partial inhibition of stimulated Isc individually by bumetanide (10 & 50 µM), and [(dihydroindenyl)oxy]alkanoic acid (DIOA, 100 µM) were additive and complete. Stimulated Isc was also partially inhibited by CFTRinh-172 (5 & 30 µM), flufenamic acid (5 µM) and diphenylamine-2,2'-dicarboxylic acid (DPC; 1 mM). Native canals of CFTR+/- mice showed a stimulation of Isc from isoproterenol and forskolin+IBMX but not in the presence of both bumetanide and DIOA, while canals from CFTR-/- mice had no responses. Nonetheless, CFTR-/- mice showed no difference from CFTR+/- mice in their ability to balance (rota-rod). Stimulated Isc was greater after chronic incubation (24 hr) with the glucocorticoids dexamethasone (0.1 & 0.3 µM), prednisolone (0.3, 1 & 3 µM), hydrocortisone (0.01, 0.1 & 1 µM), and corticosterone (0.1 & 1 µM) and mineralocorticoid aldosterone (1 µM). Steroid action was blocked by mifepristone but not by spironolactone, indicating all the steroids activated the glucocorticoid, but not mineralocorticoid, receptor. Expression of transcripts for CFTR; for KCC1, KCC3a, KCC3b and KCC4, but not KCC2; for NKCC1 but not NKCC2 and for WNK1 but only very low WNK4 was determined. CONCLUSIONS: These results are consistent with a model of Cl- secretion whereby Cl- is taken up across the basolateral membrane by a Na+-K+-2Cl- cotransporter (NKCC) and potentially another transporter, is secreted across the apical membrane via a Cl- channel, likely CFTR, and demonstrate the regulation of Cl- secretion by protein kinase A and glucocorticoids.


Assuntos
Bumetanida/farmacologia , Cloretos/metabolismo , AMP Cíclico/metabolismo , Epitélio/efeitos dos fármacos , Glucocorticoides/metabolismo , Adenilil Ciclases/metabolismo , Animais , Colforsina/farmacologia , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Epitélio/metabolismo , Transporte de Íons/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Inibidores de Fosfodiesterase/farmacologia , Potássio/metabolismo , Ratos , Ratos Wistar , Receptores de Glucocorticoides/metabolismo , Canais Semicirculares , Sódio/metabolismo
9.
Gut Microbes ; 4(1): 41-7, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23060053

RESUMO

The composition of the gastrointestinal microbiome is increasingly recognized as a crucial contributor to immune and metabolic homeostasis-deficiencies in which are characteristic of cystic fibrosis (CF) patients. The murine model (CFTR (-/-) , CF), has, in previous studies, demonstrated characteristic CF gastrointestinal (GI) manifestations including slowed transit and significant upregulation of genes associated with inflammation. To determine if characteristics of the microbiome are associated with these phenotypes we used a phylogenetic microarray to compare small intestine bacterial communities of wild type and congenic CF mice. Loss of functional CFTR is associated with significant decreases in GI bacterial community richness, evenness and diversity and reduced relative abundance of putative protective species such as Acinetobacter lwoffii and a multitude of Lactobacilliales members. CF mice exhibited significant enrichment of Mycobacteria species and Bacteroides fragilis, previously associated with GI infection and immunomodulation. Antibiotic administration to WT and CF animals resulted in convergence of their microbiome composition and significant increases in community diversity in CF mice. These communities were characterized by enrichment of members of the Lactobacillaceae and Bifidobacteriaceae and reduced abundance of Enterobacteriaceae and Clostridiaceae. These data suggest that Enterobacteria and Clostridia species, long associated with small intestinal overgrowth and inflammatory bowel disease, may suppress both ileal bacterial diversity and the particular species which maintain motility and immune homeostasis in this niche. Thus, these data provide the first indications that GI bacterial colonization is strongly impacted by the loss of functional CFTR and opens up avenues for alternative therapeutic approaches to improve CF disease management.


Assuntos
Biota , Regulador de Condutância Transmembrana em Fibrose Cística/deficiência , Trato Gastrointestinal/microbiologia , Animais , Bactérias/classificação , Bactérias/genética , Carga Bacteriana , Biodiversidade , Fibrose Cística/microbiologia , Fibrose Cística/patologia , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Modelos Animais de Doenças , Feminino , Masculino , Camundongos , Camundongos Knockout , Análise em Microsséries
10.
BMC Gastroenterol ; 12: 156, 2012 Nov 06.
Artigo em Inglês | MEDLINE | ID: mdl-23130661

RESUMO

BACKGROUND: Lubiprostone is a synthetic bicyclic fatty acid derivative of prostaglandin E1 (PGE1) used for chronic constipation. The best known action of lubiprostone is simulation of Cl- dependent fluid secretion. In a mouse model of the genetic disease cystic fibrosis, we previously showed that in vivo administration of lubiprostone resulted in greater mucus accumulation in the small intestine. The aim of this study was to directly test whether lubiprostone stimulates intestinal mucin release. METHODS: Mucin release was measured by mounting segments (4-5 cm) of mouse proximal-mid small intestine in an organ bath, allowing access to the perfusate (luminal) and the bath (serosal) solutions. Nifedipine (10-6 M) and indomethacin (10-5 M) were included in all solutions to inhibit smooth muscle activity and endogenous prostaglandin production, respectively. The tissue was equilibrated under flow for 30 min, using the perfusate collected during the final 10 min of the equilibration period to measure unstimulated release rate. Stimulus was then added to either the perfusate or the bath and the perfusate was collected for another 30 min to measure the stimulated mucin release rate. Mucin in perfusates was quantified by periodic acid-Schiff's base dot-blot assay, using purified pig gastric mucin as a standard. RESULTS: When applied luminally at 1 µM lubiprostone was ineffective at stimulating mucin release. When added to the serosal solution, 1 µM lubiprostone stimulated mucin release to ~300% of the unstimulated rate. As a positive control, serosal 1 µM prostaglandin E2 increased mucin release to ~400% of the unstimulated rate. CONCLUSIONS: These results support the idea that lubiprostone has prostaglandin-like actions on the intestine, which includes stimulation of mucin release. Stimulation of mucin release by lubiprostone may be protective in gastrointestinal conditions where loss of mucus is believed to contribute to pathogenesis. Thus, in addition to chronic constipation, there is greater potential for the therapeutic applications of lubiprostone.


Assuntos
Alprostadil/análogos & derivados , Intestino Delgado/efeitos dos fármacos , Intestino Delgado/metabolismo , Mucinas/metabolismo , Alprostadil/farmacologia , Animais , Indometacina/farmacologia , Lubiprostona , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Mucinas/análise , Músculo Liso/efeitos dos fármacos , Nifedipino/farmacologia
11.
PLoS Genet ; 8(6): e1002766, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22737083

RESUMO

Mutations in the human Zip4 gene cause acrodermatitis enteropathica, a rare, pseudo-dominant, lethal genetic disorder. We created a tamoxifen-inducible, enterocyte-specific knockout of this gene in mice which mimics this human disorder. We found that the enterocyte Zip4 gene in mice is essential throughout life, and loss-of-function of this gene rapidly leads to wasting and death unless mice are nursed or provided excess dietary zinc. An initial effect of the knockout was the reprogramming of Paneth cells, which contribute to the intestinal stem cell niche in the crypts. Labile zinc in Paneth cells was lost, followed by diminished Sox9 (sex determining region Y-box 9) and lysozyme expression, and accumulation of mucin, which is normally found in goblet cells. This was accompanied by dysplasia of the intestinal crypts and significantly diminished small intestine cell division, and attenuated mTOR1 activity in villus enterocytes, indicative of increased catabolic metabolism, and diminished protein synthesis. This was followed by disorganization of the absorptive epithelium. Elemental analyses of small intestine, liver, and pancreas from Zip4-intestine knockout mice revealed that total zinc was dramatically and rapidly decreased in these organs whereas iron, manganese, and copper slowly accumulated to high levels in the liver as the disease progressed. These studies strongly suggest that wasting and lethality in acrodermatitis enteropathica patients reflects the loss-of-function of the intestine zinc transporter ZIP4, which leads to abnormal Paneth cell gene expression, disruption of the intestinal stem cell niche, and diminished function of the intestinal mucosa. These changes, in turn, cause a switch from anabolic to catabolic metabolism and altered homeostasis of several essential metals, which, if untreated by excess dietary zinc, leads to dramatic weight loss and death.


Assuntos
Acrodermatite/genética , Proteínas de Transporte de Cátions/genética , Mucosa Intestinal , Intestinos , Nicho de Células-Tronco , Zinco , Acrodermatite/patologia , Animais , Proteínas de Transporte de Cátions/metabolismo , Modelos Animais de Doenças , Enterócitos/metabolismo , Regulação da Expressão Gênica , Humanos , Mucosa Intestinal/metabolismo , Intestinos/patologia , Metais/metabolismo , Camundongos , Camundongos Knockout , Celulas de Paneth/metabolismo , Fatores de Transcrição SOX9/metabolismo , Nicho de Células-Tronco/genética , Serina-Treonina Quinases TOR/metabolismo , Zinco/deficiência , Zinco/metabolismo
12.
J Pediatr Gastroenterol Nutr ; 55(6): 689-94, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22699839

RESUMO

OBJECTIVES: Intestinal dysmotility is one of the effects of cystic fibrosis (CF), but when and how this develops is not well understood. The goal of the present study was to use the Cftr knockout mouse to determine when in development circular smooth muscle of the small intestine becomes dysfunctional. METHODS: Wild-type (WT) and CF mice were used at postnatal day 5 (P5) through adult. Pieces of small intestine were used to measure contractile activity of the circular muscle. Bacterial overgrowth was measured by quantitative polymerase chain reaction (PCR) of the bacterial 16S gene. Intestinal gene expression was determined by quantitative reverse transcription polymerase chain reaction (RT-PCR). Prostaglandin E2 (PGE2) and its metabolites were measured by enzyme immunoassay. RESULTS: CF circular muscle response to cholinergic stimulation was similar to WT at P5, became somewhat impaired at P7, and was severely impaired by P14. In the CF intestine, bacterial overgrowth occurred by P4 and was maintained into adulthood. Eicosanoid metabolic gene expression in the CF intestine did not differ from WT shortly after birth. The phospholipase A2 genes, Pla2g4c and Pla2g5 exhibited increased expression in CF mice at P24. Prostaglandin degradative genes, Hpgd and Ptgr1, showed lower expression in CF as compared with WT at P16 and P24, respectively. PGE2 levels were significantly greater in CF mice at most ages from P7 through adulthood. CONCLUSIONS: The results clearly demonstrate that lack of CFTR itself does not cause smooth muscle dysfunction, because the circular muscle from P5 CF mice had normal activity and dysfunction developed between P7 and P14.


Assuntos
Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Fibrose Cística/fisiopatologia , Trânsito Gastrointestinal/fisiologia , Enteropatias/fisiopatologia , Intestino Delgado/fisiopatologia , Contração Muscular , Músculo Liso/fisiopatologia , Animais , Bactérias/crescimento & desenvolvimento , Colinérgicos , Fibrose Cística/genética , Fibrose Cística/metabolismo , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Dinoprostona/metabolismo , Trânsito Gastrointestinal/genética , Expressão Gênica , Enteropatias/metabolismo , Enteropatias/microbiologia , Intestino Delgado/metabolismo , Intestino Delgado/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CFTR , Camundongos Knockout , Contração Muscular/genética , Fosfolipases A2/genética , Fosfolipases A2/metabolismo
13.
Pediatr Res ; 71(1): 4-12, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22289844

RESUMO

INTRODUCTION: Improving fat absorption remains a challenge in cystic fibrosis (CF). Antibiotics (AB) treatment has been shown to improve body weight in CF mice. The mechanism may include improvement in fat absorption. We aimed to determine the effect of AB on fat absorption in two CF mouse models. RESULTS: AB did not improve total fat absorption. Interestingly, AB accelerated the absorption of isotope-labeled fats, in both Δ/Δ and WT mice. The changes observed were not related to the solubilization capacity of bile or to changes in the bacteria in the small intestine. AB reduced the fecal excretion of cholate by ~50% (P < 0.05) in both CF mouse models, indicating improved intestinal bile salt absorption. DISCUSSION: In conclusion, AB treatment does not improve total fat absorption in CF mice but does decrease fecal loss of bile salts and accelerate long-chain fatty acid (LCFA) absorption. METHODS: For 3 weeks, we administered oral AB (ciprofloxacin/metronidazole) or control treatment to homozygous ΔF508 (Δ/Δ), cystic fibrosis transmembrane conductance regulator (CFTR) knockout (-/-), and wild-type (WT) mice and quantified fat absorption using a 72-h fat balance test. In Δ/Δ mice, we assessed fat absorption kinetics by administering tri-1-(13)C-palmitin and 1-(13)C-stearate intragastrically and determining the appearance of stable isotope-labeled fats in plasma. We quantified biliary and fecal bile salts (gas chromatography) and small intestinal bacteria (quantitative-PCR).


Assuntos
Antibacterianos/farmacologia , Fibrose Cística/fisiopatologia , Gorduras na Dieta/metabolismo , Absorção Intestinal/efeitos dos fármacos , Animais , Ácidos e Sais Biliares/análise , Peso Corporal/efeitos dos fármacos , Ciprofloxacina/farmacologia , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Modelos Animais de Doenças , Fezes/química , Feminino , Humanos , Metronidazol/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
14.
J Pediatr Gastroenterol Nutr ; 53(4): 371-9, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21970994

RESUMO

OBJECTIVES: The intestinal mucosal barrier protects the body from the large numbers of microbes that inhabit the intestines and the molecules they release. Intestinal barrier function is impaired in humans with cystic fibrosis (CF), including reduced activity of the lipopolysaccharide-detoxifying enzyme intestinal alkaline phosphatase (IAP) and increased permeability. The objective of this study was to determine the suitability of using the CF mouse to investigate intestinal barrier function, and whether interventions that are beneficial for the CF mouse intestinal phenotype (antibiotics or laxative), would improve barrier function. Also tested were the effects of exogenous IAP administration. MATERIALS AND METHODS: The Cftr(tm1UNC) mouse was used. IAP expression (encoded by the murine Akp3 gene) was measured by quantitative reverse transcription-polymerase chain reaction and enzyme activity. Intestinal permeability was assessed by measuring rhodamine-dextran plasma levels following gavage. RESULTS: CF mice had 40% Akp3 mRNA expression and 30% IAP enzyme activity, as compared with wild-type mice. Oral antibiotics and laxative treatments normalized Akp3 expression and IAP enzyme activity in the CF intestine. CF mice had a 5-fold greater transfer of rhodamine-dextran from gut lumen to blood. Antibiotic and laxative treatments reduced intestinal permeability in CF mice. Administration of exogenous purified IAP to CF mice reduced intestinal permeability to wild-type levels and reduced small intestinal bacterial overgrowth by >80%. CONCLUSIONS: The CF mouse intestine has impaired mucosal barrier function, similar to human CF. Interventions that improve other aspects of the CF intestinal phenotype (antibiotics and laxative) also increase IAP activity and decrease intestinal permeability in CF mice. Exogenous IAP improve permeability and strongly reduce bacterial overgrowth in CF mice, suggesting this may be a useful therapy for CF.


Assuntos
Fibrose Cística/tratamento farmacológico , Mucosa Intestinal/metabolismo , Intestino Delgado/efeitos dos fármacos , Intestino Delgado/metabolismo , Administração Oral , Fosfatase Alcalina/genética , Fosfatase Alcalina/metabolismo , Animais , Antibacterianos/farmacologia , Infecções Bacterianas/tratamento farmacológico , Carga Bacteriana/efeitos dos fármacos , Fibrose Cística/metabolismo , Fibrose Cística/microbiologia , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Modelos Animais de Doenças , Laxantes/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fenótipo , RNA Mensageiro
15.
BMC Gastroenterol ; 10: 107, 2010 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-20843337

RESUMO

BACKGROUND: Cystic fibrosis (CF) is caused by mutations in the CFTR gene that impair the function of CFTR, a cAMP-regulated anion channel. In the small intestine loss of CFTR function creates a dehydrated, acidic luminal environment which is believed to cause an accumulation of mucus, a phenotype characteristic of CF. CF mice have small intestinal bacterial overgrowth, an altered innate immune response, and impaired intestinal transit. We investigated whether lubiprostone, which can activate the CLC2 Cl- channel, would improve the intestinal phenotype in CF mice. METHODS: Cftr(tm1UNC) (CF) and wildtype (WT) littermate mice on the C57BL/6J background were used. Lubiprostone (10 µg/kg-day) was administered by gavage for two weeks. Mucus accumulation was estimated from crypt lumen widths in periodic acid-Schiff base, Alcian blue stained sections. Luminal bacterial load was measured by qPCR for the bacterial 16S gene. Gastric emptying and small intestinal transit in fasted mice were assessed using gavaged rhodamine dextran. Gene expression was evaluated by Affymetrix Mouse430 2.0 microarray and qRT-PCR. RESULTS: Crypt width in control CF mice was 700% that of WT mice (P < 0.001). Lubiprostone did not affect WT crypt width but, unexpectedly, increased CF crypt width 22% (P = 0.001). Lubiprostone increased bacterial load in WT mice to 490% of WT control levels (P = 0.008). Conversely, lubiprostone decreased bacterial overgrowth in CF mice by 60% (P = 0.005). Lubiprostone increased gastric emptying at 20 min postgavage in both WT (P < 0.001) and CF mice (P < 0.001). Lubiprostone enhanced small intestinal transit in WT mice (P = 0.024) but not in CF mice (P = 0.377). Among other innate immune markers, expression of mast cell genes was elevated 4-to 40-fold in the CF intestine as compared to WT, and lubiprostone treatment of CF mice decreased expression to WT control levels. CONCLUSIONS: These results indicate that lubiprostone has some benefits for the CF intestinal phenotype, especially on bacterial overgrowth and the innate immune response. The unexpected observation of increased mucus accumulation in the crypts of lubiprostone-treated CF mice suggests the possibility that lubiprostone increases mucus secretion.


Assuntos
Alprostadil/análogos & derivados , Fibrose Cística/tratamento farmacológico , Imunidade Inata , Alprostadil/uso terapêutico , Animais , Bactérias/crescimento & desenvolvimento , Carga Bacteriana , Canais de Cloreto , Fibrose Cística/genética , Fibrose Cística/patologia , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Modelos Animais de Doenças , Intestino Delgado/imunologia , Intestino Delgado/microbiologia , Intestino Delgado/patologia , Lubiprostona , Camundongos , Camundongos Endogâmicos C57BL , Fenótipo
16.
J Clin Invest ; 119(9): 2535-7, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19726878

RESUMO

Accumulation of thick, sticky mucus is a hallmark of the genetic disease cystic fibrosis (CF) and has a central role in CF pathophysiology. Mutations in the CF transmembrane regulator (CFTR) ion channel are known to result in abnormally thick and sticky mucus; however, why mucus accumulates in CF is still not completely understood. In this issue of the JCI, Garcia and colleagues show that mucin--the heavily glycosylated protein contained within mucus--requires CFTR and bicarbonate in order to be released from mouse intestine (see the related article beginning on page 2613). The authors propose a model whereby CFTR-mediated bicarbonate secretion must be concurrent with mucin exocytosis for proper mucin release.


Assuntos
Bicarbonatos/metabolismo , Mucinas/metabolismo , Animais , Fibrose Cística/etiologia , Fibrose Cística/genética , Fibrose Cística/fisiopatologia , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Regulador de Condutância Transmembrana em Fibrose Cística/fisiologia , Exocitose , Humanos , Camundongos , Camundongos Endogâmicos CFTR , Modelos Biológicos
17.
PLoS One ; 4(1): e4283, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19172182

RESUMO

BACKGROUND: Cystic fibrosis (CF) has many effects on the gastrointestinal tract and a common problem in this disease is poor nutrition. In the CF mouse there is an innate immune response with a large influx of mast cells into the muscularis externa of the small intestine and gastrointestinal dysmotility. The aim of this study was to evaluate the potential role of mast cells in gastrointestinal dysmotility using the CF mouse (Cftr(tm1UNC), Cftr knockout). METHODOLOGY: Wild type (WT) and CF mice were treated for 3 weeks with mast cell stabilizing drugs (ketotifen, cromolyn, doxantrazole) or were treated acutely with a mast cell activator (compound 48/80). Gastrointestinal transit was measured using gavage of a fluorescent tracer. RESULTS: In CF mice gastric emptying at 20 min post-gavage did not differ from WT, but was significantly less than in WT at 90 min post-gavage. Gastric emptying was significantly increased in WT mice by doxantrazole, but none of the mast cell stabilizers had any significant effect on gastric emptying in CF mice. Mast cell activation significantly enhanced gastric emptying in WT mice but not in CF mice. Small intestinal transit was significantly less in CF mice as compared to WT. Of the mast cell stabilizers, only doxantrazole significantly affected small intestinal transit in WT mice and none had any effect in CF mice. Mast cell activation resulted in a small but significant increase in small intestinal transit in CF mice but not WT mice. CONCLUSIONS: The results indicate that mast cells are not involved in gastrointestinal dysmotility but their activation can stimulate small intestinal transit in cystic fibrosis.


Assuntos
Fibrose Cística/patologia , Trânsito Gastrointestinal/fisiologia , Mastócitos/metabolismo , Animais , Fibrose Cística/metabolismo , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Modelos Animais de Doenças , Esvaziamento Gástrico/efeitos dos fármacos , Mucosa Gástrica/metabolismo , Trato Gastrointestinal/patologia , Imunidade Inata , Intestino Delgado/patologia , Mastócitos/citologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Tioxantenos/farmacologia , Xantonas/farmacologia
18.
J Pediatr Gastroenterol Nutr ; 47(4): 406-16, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18852632

RESUMO

OBJECTIVES: Imbalances in essential fatty acid levels have been reported in cystic fibrosis (CF), which may relate to elevated proinflammatory eicosanoid generation. The aim of this work was to better define eicosanoid metabolism in the CF intestine. MATERIALS AND METHODS: We used the small intestine of the cystic fibrosis transmembrane conductance regulator knockout mouse (CF mouse) to measure eicosanoid metabolic gene expression by quantitative reverse transcription polymerase chain reaction and Western blot, and eicosanoid levels by enzyme immunoassay, as compared with wild-type (WT) littermates. RESULTS: In the CF small intestine, expression of the secretory phospholipase A2 Pla2g5 mRNA was upregulated to 980% of WT levels. The following were downregulated: leukotriene C4 synthase Ltc4s (mRNA 55% of WT); omega-hydroxylase cytochrome P450s Cyp2c40 (mRNA 54% of WT), and Cyp4a10 (mRNA 4% of WT); and the major prostaglandin degradative enzymes prostaglandin dehydrogenase Hpgd (mRNA 27% of WT) and leukotriene B4 12-hydroxydehydrogenase/15-oxo-prostaglandin 13-reductase Ltb4dh (mRNA 64% and protein 30% of WT). The prostaglandins PGE2 and PGF2alpha were increased to 400% to 600% of WT levels in the CF mouse intestine, and the hydroxyeicosatetraenoic acids (HETEs) 12-, 15-, and 20-HETE were decreased to 3% to 20% of WT levels. CONCLUSIONS: There are changes in eicosanoid metabolic gene expression that are accompanied by significant changes in specific eicosanoid levels. These changes are expected to play important roles in the pathophysiology of CF in the intestine.


Assuntos
Regulador de Condutância Transmembrana em Fibrose Cística/deficiência , Fibrose Cística/metabolismo , Eicosanoides/metabolismo , Regulação Enzimológica da Expressão Gênica , Intestino Delgado/metabolismo , Animais , Western Blotting , Fibrose Cística/enzimologia , Fibrose Cística/fisiopatologia , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Feminino , Imuno-Histoquímica , Intestino Delgado/enzimologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos
19.
Am J Physiol Gastrointest Liver Physiol ; 294(3): G717-27, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18202109

RESUMO

The Dmbt1 gene encodes alternatively spliced glycoproteins that are either membrane-associated or secreted epithelial products. Functions proposed for Dmbt1 include it being a tumor suppressor, having roles in innate immune defense and inflammation, and being a Golgi-sorting receptor in the exocrine pancreas. The heavily sulfated membrane glycoprotein mucin-like glycoprotein (Muclin) is a Dmbt1 product that is strongly expressed in organs of the gastrointestinal (GI) system. To explore Muclin's functions in the GI system, the Dmbt1 gene was targeted to produce Muclin-deficient mice. Muclin-deficient mice have normal body weight gain and are fertile. The Muclin-deficient mice did not develop GI tumors, even when crossed with mice lacking the known tumor suppressor p53. When colitis was induced by dextran sulfate sodium, there was no significant difference in disease severity in Muclin-deficient mice. Also, when acute pancreatitis was induced with supraphysiological caerulein, there was no difference in disease severity in the Muclin-deficient mice. Exocrine pancreatic function was impaired, as measured by attenuated neurohormonal-stimulated amylase release from Muclin-deficient acinar cells. Also, by [(35)S]Met/Cys pulse-chase analysis, traffic of newly synthesized protein to the stimulus-releasable pool was significantly retarded in Muclin-deficient cells compared with wild type. Thus Muclin deficiency impairs trafficking of regulated proteins to a stimulus-releasable pool in the exocrine pancreas.


Assuntos
Trato Gastrointestinal/fisiologia , Mucinas/deficiência , Amilases/metabolismo , Animais , Western Blotting , Proteínas de Ligação ao Cálcio , Ceruletídeo , Colite/induzido quimicamente , Colite/genética , Colite/patologia , Proteínas de Ligação a DNA , Sulfato de Dextrana , Eletroforese em Gel de Poliacrilamida , Neoplasias Gastrointestinais/epidemiologia , Neoplasias Gastrointestinais/genética , Neoplasias Gastrointestinais/patologia , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mucinas/genética , Pâncreas/citologia , Pâncreas/efeitos dos fármacos , Pâncreas/metabolismo , Pancreatite/induzido quimicamente , Pancreatite/patologia , Proteína Supressora de Tumor p53/deficiência , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/fisiologia , Proteínas Supressoras de Tumor
20.
Am J Physiol Gastrointest Liver Physiol ; 293(3): G577-84, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17615175

RESUMO

The accumulation of mucus in affected organs is characteristic of cystic fibrosis (CF). The CF mouse small intestine has dramatic mucus accumulation and exhibits slower interdigestive intestinal transit. These factors are proposed to play cooperative roles that foster small intestinal bacterial overgrowth (SIBO) and contribute to the innate immune response of the CF intestine. It was hypothesized that decreasing the mucus accumulation would reduce SIBO and might improve other aspects of the CF intestinal phenotype. To test this, solid chow-fed CF mice were treated with an osmotic laxative to improve gut hydration or liquid-fed mice were treated orally with N-acetylcysteine (NAC) to break mucin disulfide bonds. Treatment with laxative or NAC reduced mucus accumulation by 43% and 50%, respectively, as measured histologically as dilation of the intestinal crypts. Laxative and NAC also reduced bacterial overgrowth in the CF intestine by 92% and 63%, respectively. Treatment with laxative normalized small intestinal transit in CF mice, whereas NAC did not. The expression of innate immune response-related genes was significantly reduced in laxative-treated CF mice, whereas there was no significant effect in NAC-treated CF mice. In summary, laxative and NAC treatments of CF mice reduced mucus accumulation to a similar extent, but laxative was more effective than NAC at reducing bacterial load. Eradication of bacterial overgrowth by laxative treatment was associated with normalized intestinal transit and a reduction in the innate immune response. These results suggest that both mucus accumulation and slowed interdigestive small intestinal transit contribute to SIBO in the CF intestine.


Assuntos
Acetilcisteína/farmacologia , Bactérias/efeitos dos fármacos , Catárticos/farmacologia , Fibrose Cística/tratamento farmacológico , Expectorantes/farmacologia , Trânsito Gastrointestinal/efeitos dos fármacos , Inflamação/tratamento farmacológico , Intestino Delgado/efeitos dos fármacos , Muco/metabolismo , Polietilenoglicóis/farmacologia , Acetilcisteína/uso terapêutico , Animais , Bactérias/genética , Bactérias/crescimento & desenvolvimento , Peso Corporal/efeitos dos fármacos , Catárticos/uso terapêutico , Fibrose Cística/metabolismo , Fibrose Cística/microbiologia , Fibrose Cística/fisiopatologia , Modelos Animais de Doenças , Expectorantes/uso terapêutico , Esvaziamento Gástrico/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Imunidade Inata/efeitos dos fármacos , Imunidade Inata/genética , Inflamação/metabolismo , Inflamação/microbiologia , Inflamação/fisiopatologia , Intestino Delgado/metabolismo , Intestino Delgado/microbiologia , Intestino Delgado/fisiopatologia , Camundongos , Camundongos Endogâmicos CFTR , Polietilenoglicóis/uso terapêutico , RNA Bacteriano/metabolismo , RNA Ribossômico 16S/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...