Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Cell Oncol (Dordr) ; 45(3): 381-398, 2022 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-35426084

RESUMO

BACKGROUND: Solute carrier family 26 member (SLC26A9) is a Cl- uniporter with very high expression levels in the gastric mucosa. Here, we describe morphological and molecular alterations in gastric mucosa of slc26a9-/- mice and in selective parietal cell-deleted slc26a9fl/fl/Atp4b-Cre mice and correlate SLC26A9 expression levels with morphological and clinical parameters in a cohort of gastric cancer (GC) patients. METHODS: The expression patterns of genes related to transport and enzymatic function, proliferation, apoptosis, inflammation, barrier integrity, metaplasia and neoplasia development were studied by immunohistochemistry (IHC), quantitative RT-PCR, in situ hybridization and RNA microarray analysis. SLC26A9 expression and cellular/clinical phenotypes were studied in primary human GC tissues and GC cell lines. RESULTS: We found that both complete and parietal cell-selective Slc26a9 deletion in mice caused spontaneous development of gastric premalignant and malignant lesions. Dysregulated differentiation of gastric stem cells in an inflammatory environment, activated Wnt signaling, cellular hyperproliferation, apoptosis inhibition and metaplasia were observed. Analysis of human gastric precancerous and cancerous tissues revealed that SLC26A9 expression progressively decreased from atrophic gastritis to GC, and that downregulation of SLC26A9 was correlated with patient survival. Exogenous expression of SLC26A9 in GC cells induced upregulation of the Cl-/HCO3- exchanger AE2, G2/M cell cycle arrest and apoptosis and suppressed their proliferation, migration and invasion. CONCLUSIONS: Our data indicate that SLC26A9 deletion in parietal cells is sufficient to trigger gastric metaplasia and the development of neoplastic lesions. In addition, we found that SLC26A9 expression decreases during human gastric carcinogenesis, and that exogenous SLC26A9 expression in GC cells reduces their malignant behavior.


Assuntos
Antiporters , Lesões Pré-Cancerosas , Neoplasias Gástricas , Transportadores de Sulfato , Animais , Antiporters/genética , Antiporters/metabolismo , Mucosa Gástrica/metabolismo , Mucosa Gástrica/patologia , Humanos , Imuno-Histoquímica , Metaplasia/metabolismo , Metaplasia/patologia , Camundongos , Lesões Pré-Cancerosas/patologia , Neoplasias Gástricas/genética , Neoplasias Gástricas/patologia , Transportadores de Sulfato/genética , Transportadores de Sulfato/metabolismo
2.
Biochem Pharmacol ; 178: 114040, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32422138

RESUMO

BACKGROUND: The molecular basis for heat-stable Escherichia coli enterotoxin (STa) action and its synthetic analogue linaclotide is well understood at the enterocyte level. Pharmacologic strategies to prevent STa-induced intestinal fluid loss by inhibiting its effector molecules, however, have achieved insufficient inhibition in vivo. AIMS AND EXPERIMENTAL APPROACH: To investigate whether the currently discussed effector molecules and signaling mechanisms of STa/linaclotide-induced diarrhea have similar relevance in vivo than at the enterocyte level, we studied the effect of 10-7M of the STa analogue linaclotide on short circuit current (Isc) of chambered isolated jejunal mucosa, and on the in vivo action on fluid transport in a perfused segment of proximal jejunum of anesthetized mice. The selected mice were deficient of transport (NHE3, CFTR, Slc26a3/a6), adaptor (NHERF1-3), or signal transduction molecules [cGMP-dependent kinase II (GKII)] considered to be downstream effectors after STa/linaclotide binding to guanylate cyclase C (GCC). Selective NHE3 inhibition by tenapanor was also employed. KEY RESULTS, CONCLUSIONS AND IMPLICATIONS: The comparison allowed the separation of effectors for stimulation of electrogenic anion secretion and for inhibition of electrolyte/fluid absorption in response to STa/linaclotide. The cGKII-NHERF1-CFTR and cGKII-NHERF2-NHE3 interactions are indeed major effectors of small intestinal fluid loss downstream of GCC activation in vitro and in vivo, but 50% of the linaclotide-induced fluid loss in vivo, while dependent on CFTR activation and NHE3 inhibition, does not involve cGKII, and 30% does not depend on NHERF1 or NHERF2. A combined NHERF1 and NHERF2 inhibition appears nevertheless a good pharmacological strategy against STa-mediated fluid loss.


Assuntos
Diarreia/induzido quimicamente , Diarreia/metabolismo , Agonistas da Guanilil Ciclase C/farmacologia , Mucosa Intestinal/enzimologia , Jejuno/metabolismo , Proteínas Quinases/metabolismo , Animais , Células CACO-2 , Agonistas da Guanilil Ciclase C/efeitos adversos , Humanos , Mucosa Intestinal/efeitos dos fármacos , Transporte de Íons/efeitos dos fármacos , Transporte de Íons/fisiologia , Jejuno/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Peptídeos/efeitos adversos , Peptídeos/farmacologia , Fosfoproteínas/antagonistas & inibidores , Fosfoproteínas/metabolismo , Trocadores de Sódio-Hidrogênio/antagonistas & inibidores , Trocadores de Sódio-Hidrogênio/metabolismo
3.
Mol Reprod Dev ; 85(8-9): 682-695, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-30118583

RESUMO

Members of the solute carrier 26 (SLC26) family have emerged as important players in mediating anions fluxes across the plasma membrane of epithelial cells, in cooperation with the cystic fibrosis transmembrane conductance regulator (CFTR) chloride channel. Among them, SLC26A3 acts as a chloride/bicarbonate exchanger, highly expressed in the gastrointestinal, pancreatic and renal tissues. In humans, mutations in the SLC26A3 gene were shown to induce congenital chloride-losing diarrhea (CLD), a rare autosomal recessive disorder characterized by life-long secretory diarrhea. In view of some reports indicating subfertility in some male CLD patients together with SLC26-A3 and -A6 expression in the male genital tract and sperm cells, we analyzed the male reproductive parameters and functions of SLC26A3 deficient mice, which were previously reported to display CLD gastro-intestinal features. We show that in contrast to Slc26a6, deletion of Slc26a3 is associated with severe lesions and abnormal cytoarchitecture of the epididymis, together with sperm quantitative, morphological and functional defects, which altogether compromised male fertility. Overall, our work provides new insight into the pathophysiological mechanisms that may alter the reproductive functions and lead to male subfertility in CLD patients, with a phenotype reminiscent of that induced by CFTR deficiency in the male genital tract.


Assuntos
Antiporters/metabolismo , Epididimo/metabolismo , Epididimo/fisiopatologia , Fertilização , Infertilidade Masculina/metabolismo , Capacitação Espermática , Transportadores de Sulfato/metabolismo , Animais , Antiporters/genética , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Diarreia/congênito , Diarreia/etiologia , Masculino , Erros Inatos do Metabolismo/etiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mutação , Fenótipo , Contagem de Espermatozoides , Motilidade dos Espermatozoides , Espermatozoides/patologia , Transportadores de Sulfato/genética , Testículo/fisiopatologia
4.
Pflugers Arch ; 467(8): 1795-807, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25271043

RESUMO

A dysfunction of the Na(+)/H(+) exchanger isoform 3 (NHE3) significantly contributes to the reduced salt absorptive capacity of the inflamed intestine. We previously reported a strong decrease in the NHERF family member PDZK1 (NHERF3), which binds to NHE3 and regulates its function in a mouse model of colitis. The present study investigates whether a causal relationship exists between the decreased PDZK1 expression and the NHE3 dysfunction in human and murine intestinal inflammation. Biopsies from the colon of patients with ulcerative colitis, murine inflamed ileal and colonic mucosa, NHE3-transfected Caco-2BBe colonic cells with short hairpin RNA (shRNA) knockdown of PDZK1, and Pdzk1-gene-deleted mice were studied. PDZK1 mRNA and protein expression was strongly decreased in inflamed human and murine intestinal tissue as compared to inactive disease or control tissue, whereas that of NHE3 or NHERF1 was not. Inflamed human and murine intestinal tissues displayed correct brush border localization of NHE3 but reduced acid-activated NHE3 transport activity. A similar NHE3 transport defect was observed when PDZK1 protein content was decreased by shRNA knockdown in Caco-2BBe cells or when enterocyte PDZK1 protein content was decreased to similar levels as found in inflamed mucosa by heterozygote breeding of Pdzk1-gene-deleted and WT mice. We conclude that a decrease in PDZK1 expression, whether induced by inflammation, shRNA-mediated knockdown, or heterozygous breeding, is associated with a decreased NHE3 transport rate in human and murine enterocytes. We therefore hypothesize that inflammation-induced loss of PDZK1 expression may contribute to the NHE3 dysfunction observed in the inflamed intestine.


Assuntos
Proteínas de Transporte/metabolismo , Colite/metabolismo , Colo/metabolismo , Enterócitos/metabolismo , Ileíte/metabolismo , Íleo/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Trocadores de Sódio-Hidrogênio/metabolismo , Animais , Biópsia , Células CACO-2 , Proteínas de Transporte/genética , Colite/induzido quimicamente , Colite/genética , Colite/patologia , Colo/patologia , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Sulfato de Dextrana , Modelos Animais de Doenças , Regulação para Baixo , Enterócitos/patologia , Humanos , Ileíte/induzido quimicamente , Ileíte/genética , Ileíte/patologia , Íleo/patologia , Mediadores da Inflamação/metabolismo , Interleucina-10/genética , Interleucina-10/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/deficiência , Peptídeos e Proteínas de Sinalização Intracelular/genética , Proteínas de Membrana , Camundongos da Linhagem 129 , Camundongos Knockout , Microvilosidades/metabolismo , Interferência de RNA , RNA Mensageiro/metabolismo , Estudos Retrospectivos , Trocador 3 de Sódio-Hidrogênio , Trocadores de Sódio-Hidrogênio/genética , Transfecção , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo
5.
Pflugers Arch ; 467(6): 1261-75, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24965066

RESUMO

Slc26a9 is an anion transporter that is strongly expressed in the stomach and lung. Slc26a9 variants were recently found associated with a higher incidence of meconium ileus in cystic fibrosis (CF) infants, raising the question whether Slc26a9 is expressed in the intestine and what its functional role is. Slc26a9 messenger RNA (mRNA) was found highly expressed in the mucosae of the murine and human upper gastrointestinal tract, with an abrupt decrease in expression levels beyond the duodenum. Absence of SLC26a9 expression strongly increased the intestinally related mortality in cystic fibrosis transmembrane conductance regulator (CFTR)-deficient mice. Proximal duodenal JHCO3(-) and fluid secretion were reduced in the absence of Slc26a9 expression. In the proximal duodenum of young Slc26a9 KO mice, the glands and villi/crypts were elongated and proliferation was enhanced. This difference was lost with ageing, as were the alterations in fluid movement, whereas the reduction in JHCO3(-) remained. Laser dissection followed by qPCR suggested Slc26a9 expression to be crypt-predominant in the duodenum. In summary, deletion of Slc26a9 caused bicarbonate secretory and fluid absorptive changes in the proximal duodenal mucosa and increased the postweaning death rates in CFTR-deficient mice. Functional alterations in the duodenum were most prominent at young ages. We assume that the association of meconium ileus and Slc26a9 variants may be related to maldigestion and impaired downstream signaling caused by loss of upper GI tract digestive functions, aggravating the situation of lack of secretion and sticky mucus at the site of obstruction in CF intestine.


Assuntos
Antiporters/genética , Bicarbonatos/metabolismo , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Fibrose Cística/metabolismo , Duodeno/metabolismo , Absorção Intestinal , Animais , Antiporters/metabolismo , Proliferação de Células , Fibrose Cística/patologia , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Duodeno/crescimento & desenvolvimento , Duodeno/patologia , Humanos , Mucosa Intestinal/metabolismo , Transporte de Íons , Camundongos , Camundongos Endogâmicos C57BL , Transportadores de Sulfato
6.
Pflugers Arch ; 466(12): 2269-78, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24595473

RESUMO

Pseudomonas aeruginosa infections of the airway cells decrease apical expression of both wild-type (wt) and F508del CFTR through the inhibition of apical endocytic recycling. CFTR endocytic recycling is known to be regulated by its interaction with PDZ domain containing proteins. Recent work has shown that the PDZ domain scaffolding protein NHERF1 finely regulates both wt and F508delCFTR membrane recycling. Here, we investigated the effect of P. aeruginosa infection on NHERF1 post-translational modifications and how this affects CFTR expression in bronchial epithelial cells and in murine lung. Both in vitro in bronchial cells, and in vivo in mice, infection reduced CFTR expression and increased NHERF1 molecular weight through its hyper-phosphorylation and ubquitination as a consequence of both bacterial pilin- and flagellin-mediated host-cell interaction. The ability of P. aeruginosa to down-regulate mature CFTR expression was reduced both in vivo in NHERF1 knockout mice and in vitro after silencing NHERF1 expression or mutations blocking its phosphorylation at serines 279 and 301. These studies provide the first evidence that NHERF1 phosphorylation may negatively regulate its action and, therefore, the assembly and function of multiprotein NHERF1 complexes in response to infection. The identification of molecular mechanisms responsible for these effects could identify novel targets to block potential P. aeruginosa interference with the efficacy of potentiator and/or corrector compounds.


Assuntos
Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Fosfoproteínas/metabolismo , Processamento de Proteína Pós-Traducional , Infecções por Pseudomonas/metabolismo , Mucosa Respiratória/metabolismo , Trocadores de Sódio-Hidrogênio/metabolismo , Animais , Brônquios/citologia , Brônquios/metabolismo , Brônquios/microbiologia , Linhagem Celular , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Humanos , Pulmão/citologia , Pulmão/metabolismo , Pulmão/microbiologia , Camundongos , Mutação , Fosfoproteínas/genética , Fosforilação , Pseudomonas aeruginosa , Mucosa Respiratória/microbiologia , Trocadores de Sódio-Hidrogênio/genética , Ubiquitinação
7.
Crit Care Med ; 42(3): e177-88, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24368347

RESUMO

OBJECTIVES: A common potentially fatal disease of the pancreas is acute pancreatitis, for which there is no treatment. Most studies of this disorder focus on the damage to acinar cells since they are assumed to be the primary target of multiple stressors affecting the pancreas. However, increasing evidence suggests that the ducts may also have a crucial role in induction of the disease. To test this hypothesis, we sought to determine the specific role of the duct in the induction of acute pancreatitis using well-established disease models and mice with deletion of the Na/H exchanger regulatory factor-1 that have selectively impaired ductal function. DESIGN: Randomized animal study. SETTING: Animal research laboratory. SUBJECTS: Wild-type and Na/H exchanger regulatory factor-1 knockout mice. INTERVENTIONS: Acute necrotizing pancreatitis was induced by i.p. administration of cerulein or by intraductal administration of sodium taurocholate. The pancreatic expression of Na/H exchanger regulatory factor-1 and cystic fibrosis transmembrane conductance regulator (a key player in the control of ductal secretion) was analyzed by immunohistochemistry. In vivo pancreatic ductal secretion was studied in anesthetized mice. Functions of pancreatic acinar and ductal cells as well as inflammatory cells were analyzed in vitro. MEASUREMENTS AND MAIN RESULTS: Deletion of Na/H exchanger regulatory factor-1 resulted in gross mislocalization of cystic fibrosis transmembrane conductance regulator, causing marked reduction in pancreatic ductal fluid and bicarbonate secretion. Importantly, deletion of Na/H exchanger regulatory factor-1 had no deleterious effect on functions of acinar and inflammatory cells. Deletion of Na/H exchanger regulatory factor-1, which specifically impaired ductal function, increased the severity of acute pancreatitis in the two mouse models tested. CONCLUSIONS: Our findings provide the first direct evidence for the crucial role of ductal secretion in protecting the pancreas from acute pancreatitis and strongly suggest that improved ductal function should be an important modality in prevention and treatment of the disease.


Assuntos
Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Ductos Pancreáticos/metabolismo , Pancreatite Necrosante Aguda/metabolismo , Pancreatite Necrosante Aguda/patologia , Fosfoproteínas/metabolismo , Trocadores de Sódio-Hidrogênio/metabolismo , Sistemas de Transporte de Aminoácidos/metabolismo , Animais , Biomarcadores/metabolismo , Distribuição de Qui-Quadrado , Modelos Animais de Doenças , Imuno-Histoquímica , Camundongos , Camundongos Knockout , Pâncreas/metabolismo , Pâncreas/fisiologia , RNA Mensageiro/metabolismo , Distribuição Aleatória , Valores de Referência , Regeneração/fisiologia , Sensibilidade e Especificidade , Simportadores/metabolismo
8.
J Physiol ; 591(21): 5377-91, 2013 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-24018950

RESUMO

The duodenal villus brush border membrane expresses several ion transporters and/or channels, including the solute carrier 26 anion transporters Slc26a3 (DRA) and Slc26a6 (PAT-1), the Na(+)/H(+) exchanger isoform 3 (NHE3), as well as the anion channels cystic fibrosis transmembrane conductance regulator (CFTR) and Slc26a9. Using genetically engineered mouse models lacking Scl26a3, Slc26a6, Slc26a9 or Slc9a3 (NHE3), the study was carried out to assess the role of these transporters in mediating the protective duodenal bicarbonate secretory response (DBS-R) to luminal acid; and to compare it to their role in DBS-R elicited by the adenylyl cyclase agonist forskolin. While basal DBS was reduced in the absence of any of the three Slc26 isoforms, the DBS-R to forskolin was not altered. In contrast, the DBS-R to a 5 min exposure to luminal acid (pH 2.5) was strongly reduced in the absence of Slc26a3 or Slc26a9, but not Slc26a6. CFTR inhibitor [CFTR(Inh)-172] reduced the first phase of the acid-induced DBS-R, while NHE3 inhibition (or knockout) abolished the sustained phase of the DBS-R. Luminal acid exposure resulted in the activation of multiple intracellular signalling pathways, including SPAK, AKT and p38 phosphorylation. It induced a biphasic trafficking of NHE3, first rapidly into the brush border membrane, followed by endocytosis in the later stage. We conclude that the long-lasting DBS-R to luminal acid exposure activates multiple duodenocyte signalling pathways and involves changes in trafficking and/or activity of CFTR, Slc26 isoforms Slc26a3 and Slc26a9, and NHE3.


Assuntos
Antiporters/metabolismo , Bicarbonatos/metabolismo , Duodeno/metabolismo , Transporte de Íons , Trocadores de Sódio-Hidrogênio/metabolismo , Animais , Antiporters/genética , Benzoatos/farmacologia , Colforsina/farmacologia , Regulador de Condutância Transmembrana em Fibrose Cística/antagonistas & inibidores , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Endocitose , Concentração de Íons de Hidrogênio , Mucosa Intestinal/metabolismo , Sistema de Sinalização das MAP Quinases , Camundongos , Fosforilação , Trocador 3 de Sódio-Hidrogênio , Trocadores de Sódio-Hidrogênio/antagonistas & inibidores , Trocadores de Sódio-Hidrogênio/genética , Transportadores de Sulfato , Tiazolidinas/farmacologia
9.
J Clin Invest ; 122(10): 3629-34, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22945630

RESUMO

Asthma is a chronic condition with unknown pathogenesis, and recent evidence suggests that enhanced airway epithelial chloride (Cl-) secretion plays a role in the disease. However, the molecular mechanism underlying Cl- secretion and its relevance in asthma pathophysiology remain unknown. To determine the role of the solute carrier family 26, member 9 (SLC26A9) Cl- channel in asthma, we induced Th2-mediated inflammation via IL-13 treatment in wild-type and Slc26a9-deficient mice and compared the effects on airway ion transport, morphology, and mucus content. We found that IL-13 treatment increased Cl- secretion in the airways of wild-type but not Slc26a9-deficient mice. While IL-13-induced mucus overproduction was similar in both strains, treated Slc26a9-deficient mice exhibited airway mucus obstruction, which did not occur in wild-type controls. In a study involving healthy children and asthmatics, a polymorphism in the 3' UTR of SLC26A9 that reduced protein expression in vitro was associated with asthma. Our data demonstrate that the SLC26A9 Cl- channel is activated in airway inflammation and suggest that SLC26A9-mediated Cl- secretion is essential for preventing airway obstruction in allergic airway disease. These results indicate that SLC26A9 may serve as a therapeutic target for airway diseases associated with mucus plugging.


Assuntos
Obstrução das Vias Respiratórias/prevenção & controle , Antiporters/fisiologia , Asma/genética , Bronquite/fisiopatologia , Cloretos/metabolismo , Transporte de Íons/fisiologia , Muco/metabolismo , Traqueíte/fisiopatologia , Regiões 3' não Traduzidas , Obstrução das Vias Respiratórias/etiologia , Obstrução das Vias Respiratórias/fisiopatologia , Animais , Antiporters/deficiência , Antiporters/genética , Asma/fisiopatologia , Bronquite/induzido quimicamente , Bronquite/genética , Bronquite/imunologia , Cálcio/farmacologia , Criança , AMP Cíclico/farmacologia , Regulador de Condutância Transmembrana em Fibrose Cística/deficiência , Regulador de Condutância Transmembrana em Fibrose Cística/fisiologia , Modelos Animais de Doenças , Células Epiteliais/metabolismo , Predisposição Genética para Doença , Humanos , Interleucina-13/toxicidade , Pulmão/patologia , Camundongos , Camundongos Knockout , Transportadores de Sulfato , Células Th2/imunologia , Traqueíte/induzido quimicamente , Traqueíte/genética , Traqueíte/imunologia
10.
Am J Physiol Gastrointest Liver Physiol ; 303(12): G1312-21, 2012 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-23019197

RESUMO

The incidence of duodenal ulcer, especially Helicobacter pylori-negative duodenal ulcer, strongly increases with age. In humans, telomere length shortening is considered to be one critical factor in cellular senescence and organ survival. In this study, we compared basal and stimulated gastric acid and duodenal HCO(3)(-) secretory rates in aged late-generation (G(3)) telomerase-deficient (mTERC(-/-)) mice, which are characterized by severe telomere dysfunction due to the inability to elongate telomeres during cell division. We found that basal and forskolin-stimulated HCO(3)(-) secretion and short-circuit current (I(sc)) in isolated duodenal mucosa of G(3) mTERC(-/-) mice were markedly reduced compared with age-matched wild-type mice. In contrast, basal and forskolin-stimulated acid secretory rates in isolated G(3) mTERC(-/-) gastric mucosa were not significantly altered. Correspondingly, duodenal mucosa of G(3) mTERC(-/-) mice showed slimming and shortening of villi, whereas gastric mucosal histology was not significantly altered. However, the ratios of cystic fibrosis transmembrane conductance regulator (CFTR) and solute-linked carrier 26 gene family (Slc26a6) mRNA expression in relation to cytokeratin-18 were not altered in duodenal mucosa. The further knockout of p21, which is a downstream effector of telomere shortening-induced senescence, rescued villus atrophy of duodenal mucosa, and basal and forskolin-stimulated duodenal HCO(3)(-) secretion and I(sc) in mTERC(-/-) p21(-/-) double-knockout mice were not different from wild-type controls. In conclusion, genetic ablation of telomerase resulted in p21-dependent duodenal mucosal atrophy and reduced duodenal HCO(3)(-) secretory capacity, whereas gastric morphology and acid secretory function were preserved. This suggests that telomere shortening during aging may result in an imbalance between aggressive and protective secretions against duodenal mucosa and thus predispose to ulcer formation.


Assuntos
Envelhecimento/fisiologia , Bicarbonatos/metabolismo , Duodeno/fisiopatologia , Ácido Gástrico/metabolismo , Mucosa Intestinal/metabolismo , Encurtamento do Telômero/fisiologia , Animais , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
11.
J Physiol ; 590(21): 5317-34, 2012 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-22802588

RESUMO

This study investigated whether expression of the common cystic fibrosis transmembrane conductance regulator (CFTR) mutant F508del in the apical membrane of enterocytes confers increased bicarbonate secretory capacity on the intestinal epithelium of F508del mutant mice compared to that of CFTR knockout (KO) mice. CFTR KO mice, F508del mutant mice (F508del) and wild-type (WT) littermates were bred on the FVB/N background. F508del isolated brush border membrane (BBM) contained approximately 5-10% fully glycosylated band C protein compared to WT BBM. Similarly, the forskolin (FSK)-induced, CFTR-dependent short-circuit current (I(sc)) of F508del mucosa was approximately 5-10% of WT, whereas the HCO(3)(-) secretory response ( ) was almost half that of WT in both duodenum and mid-colon studied in vitro and in vivo. While WT intestine retained full FSK-induced in the absence of luminal Cl(-), the markedly higher than I(sc) in F508del intestine was dependent on the presence of luminal Cl(-), and was blocked by CFTR inhibitors. The Ste20-related proline-alanine-rich kinases (SPAK/OSR1), which are downstream of the with-no-lysine (K) protein kinases (WNK), were rapidly phosphorylated by FSK in WT and F508del, but significantly more slowly in CFTR KO intestine. In conclusion, the data demonstrate that low levels of F508del membrane expression in the intestine of F508del mice significantly increased FSK-induced HCO(3)(-) secretion mediated by Cl(-)/HCO(3)(-) exchange. However, in WT mucosa FSK elicited strong SPAK/OSR1 phosphorylation and Cl(-)-independent HCO(3)(-) efflux. This suggests that therapeutic strategies which deliver F508del to the apical membrane have the potential to significantly enhance epithelial HCO(3)(-) secretion.


Assuntos
Bicarbonatos/metabolismo , Cloretos/metabolismo , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Mucosa Intestinal/metabolismo , Animais , Membrana Celular/metabolismo , Colforsina/farmacologia , Colo/metabolismo , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Duodeno/metabolismo , Feminino , Masculino , Camundongos , Camundongos Endogâmicos CFTR , Microvilosidades/metabolismo , Mutação , Proteínas Serina-Treonina Quinases/metabolismo
12.
Inflamm Bowel Dis ; 18(1): 101-11, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21557395

RESUMO

BACKGROUND: Ileocolonic luminal pH has been reported to be abnormally low in inflammatory bowel disease (IBD) patients, and one of the causative factors may be reduced epithelial HCO(3)(-) secretory rate (J(HCO3)(-)). Disturbances in J(HCO3)(-) may occur due to inflammation-induced changes in the crypt and villous architecture, or due to the effect of proinflammatory cytokines on epithelial ion transporters. METHODS: To discriminate between these possibilities, the tumor necrosis factor alpha (TNF-α) overexpressing (TNF(+/ΔARE)) mouse model was chosen, which displays high proinflammatory cytokine levels in both ileum and colon, but develops only mild colonic histopathology and diarrhea. HCO(3)(-) secretion, mRNA expression, immunohistochemistry, and fluid absorptive capacity were measured in ileal and mid-colonic mucosa of TNF(+/ΔARE) and wildtype (WT) (TNF(+/+)) mice in Ussing chambers, and in anesthetized mice in vivo. RESULTS: The high basal J(HCO3)(-) observed in WT ileal and mid-colonic mucosa were luminal Cl(-) -dependent and strongly decreased in TNF(+/ΔARE) mice. Downregulated in adenoma (DRA) mRNA and protein expression was strongly decreased in TNF(+/ΔARE) ileocolon, whereas cystic fibrosis transmembrane conductance regulator (CFTR), Na(+) /H(+) exchanger 3 (NHE3), Na(+) /HCO(3)(-) cotransporter (NBC), and epithelial sodium channel (ENaC) expression was not significantly altered. This indicates that the severe defect in ileocolonic J(HCO3)(-) was due to DRA downregulation. Fluid absorption was severely depressed in the ileum but only mildly affected in the mid-distal colon, preventing the development of overt diarrhea. CONCLUSIONS: Even mild ileocolonic inflammation may result in a decrease of epithelial HCO(3)(-) secretion, which may contribute to alterations in surface pH, intestinal flora, and mucus barrier properties.


Assuntos
Antiporters/metabolismo , Bicarbonatos/metabolismo , Colite/patologia , Íleo/patologia , Inflamação/patologia , Fator de Necrose Tumoral alfa/fisiologia , Animais , Antiporters/genética , Western Blotting , Colite/metabolismo , Canais Epiteliais de Sódio/genética , Canais Epiteliais de Sódio/metabolismo , Feminino , Íleo/metabolismo , Inflamação/metabolismo , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patologia , Masculino , Camundongos , Camundongos Knockout , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Simportadores de Sódio-Bicarbonato/genética , Simportadores de Sódio-Bicarbonato/metabolismo , Trocador 3 de Sódio-Hidrogênio , Trocadores de Sódio-Hidrogênio/genética , Trocadores de Sódio-Hidrogênio/metabolismo , Transportadores de Sulfato
13.
J Biol Chem ; 286(16): 14120-8, 2011 Apr 22.
Artigo em Inglês | MEDLINE | ID: mdl-21367857

RESUMO

Kir4.1 channels were found to colocalize with the H(+)/K(+)-ATPase throughout the parietal cell (PC) acid secretory cycle. This study was undertaken to explore their functional role. Acid secretory rates, electrophysiological parameters, PC ultrastructure, and gene and protein expression were determined in gastric mucosae of 7-8-day-old Kir4.1-deficient mice and WT littermates. Kir4.1(-/-) mucosa secreted significantly more acid and initiated secretion significantly faster than WT mucosa. No change in PC number but a relative up-regulation of H(+)/K(+)-ATPase gene and protein expression (but not of other PC ion transporters) was observed. Electron microscopy revealed fully fused canalicular membranes and a lack of tubulovesicles in resting state Kir4.1(-/-) PCs, suggesting that Kir4.1 ablation may also interfere with tubulovesicle endocytosis. The role of this inward rectifier in the PC apical membrane may therefore be to balance between K(+) loss via KCNQ1/KCNE2 and K(+) reabsorption by the slow turnover of the H(+)/K(+)-ATPase, with consequences for K(+) reabsorption, inhibition of acid secretion, and membrane recycling. Our results demonstrate that Kir4.1 channels are involved in the control of acid secretion and suggest that they may also affect secretory membrane recycling.


Assuntos
Ácido Gástrico/metabolismo , Regulação da Expressão Gênica , Células Parietais Gástricas/citologia , Canais de Potássio Corretores do Fluxo de Internalização/fisiologia , Animais , Transporte Biológico , Eletrofisiologia/métodos , Endocitose , Mucosa Gástrica/metabolismo , ATPase Trocadora de Hidrogênio-Potássio/química , Camundongos , Camundongos Knockout , Microscopia Eletrônica de Transmissão/métodos , Microscopia de Fluorescência/métodos , Potássio/química , Canais de Potássio Corretores do Fluxo de Internalização/biossíntese
14.
Inflamm Bowel Dis ; 17(4): 884-98, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20722063

RESUMO

BACKGROUND: The absorption of water and ions (especially Na(+) and Cl(-)) is an important function of colonic epithelial cells in both physiological and pathophysiological conditions. Despite the comprehensive animal studies, there are only scarce available data on the ion transporter activities of the normal and inflamed human colon. METHODS: In this study, 128 healthy controls and 69 patients suffering from ulcerative colitis (UC) were involved. We investigated the expressional and functional characteristics of the Na(+)/H(+) exchangers (NHE) 1-3, the epithelial sodium channel (ENaC), and the SLC26A3 Cl(-)/HCO 3- exchanger downregulated in adenoma (DRA) in primary colonic crypts isolated from human biopsy and surgical samples using microfluorometry, patch clamp, and real-time reverse-transcription polymerase chain reaction (RT-PCR) techniques. RESULTS: Data collected from colonic crypts showed that the activities of electroneutral (via NHE3) and the electrogenic Na(+) absorption (via ENaC) are in inverse ratio to each other in the proximal and distal colon. We found no significant differences in the activity of NHE2 in different segments of the colon. Surface cell Cl(-)/HCO 3- exchange is more active in the distal part of the colon. Importantly, both sodium and chloride absorptions are damaged in UC, whereas NHE1, which has been shown to promote immune response, is upregulated by 6-fold. CONCLUSIONS: These results open up new therapeutic targets in UC.


Assuntos
Antiporters/metabolismo , Proteínas de Transporte de Cátions/metabolismo , Colite Ulcerativa/metabolismo , Colo/metabolismo , Canais Epiteliais de Sódio/metabolismo , Trocadores de Sódio-Hidrogênio/metabolismo , Antiporters/genética , Estudos de Casos e Controles , Proteínas de Transporte de Cátions/genética , Antiportadores de Cloreto-Bicarbonato , Cloretos/metabolismo , Colite Ulcerativa/genética , Colite Ulcerativa/cirurgia , Canais Epiteliais de Sódio/genética , Humanos , Transporte de Íons , Sódio/metabolismo , Trocador 1 de Sódio-Hidrogênio , Trocador 3 de Sódio-Hidrogênio , Trocadores de Sódio-Hidrogênio/genética , Transportadores de Sulfato
15.
J Physiol ; 588(Pt 24): 5049-63, 2010 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-20962002

RESUMO

Trafficking and regulation of the epithelial brush border membrane (BBM) Na+/H+ exchanger 3 (NHE3) in the intestine involves interaction with four different members of the NHERF family in a signal-dependent and possibly segment-specific fashion. The aim of this research was to study the role of NHERF2 (E3KARP) in intestinal NHE3 BBM localization and second messenger-mediated and receptor-mediated inhibition of NHE3. Immunolocalization of NHE3 in WT mice revealed predominant microvillar localization in jejunum and colon, a mixed distribution in the proximal ileum but localization near the terminal web in the distal ileum. The terminal web localization of NHE3 in the distal ileum correlated with reduced acid-activated NHE3 activity (fluorometrically assessed). NHERF2 ablation resulted in a shift of NHE3 to the microvilli and higher basal fluid absorption rates in the ileum, but no change in overall NHE3 protein or mRNA expression. Forskolin-induced NHE3 inhibition was preserved in the absence of NHERF2, whereas Ca2+ ionophore- or carbachol-mediated inhibition was abolished. Likewise, Escherichia coli heat stable enterotoxin peptide (STp) lost its inhibitory effect on intestinal NHE3. It is concluded that in native murine intestine, the NHE3 adaptor protein NHERF2 plays important roles in tethering NHE3 to a position near the terminal web and in second messenger inhibition of NHE3 in a signal- and segment-specific fashion, and is therefore an important regulator of intestinal fluid transport.


Assuntos
Cálcio/metabolismo , Membrana Celular/fisiologia , AMP Cíclico/metabolismo , GMP Cíclico/metabolismo , Fosfoproteínas/metabolismo , Trocadores de Sódio-Hidrogênio/metabolismo , Animais , Regulação da Expressão Gênica/fisiologia , Concentração de Íons de Hidrogênio , Mucosa Intestinal/metabolismo , Camundongos , Camundongos Knockout , Fosfoproteínas/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Trocador 3 de Sódio-Hidrogênio , Trocadores de Sódio-Hidrogênio/genética , Água/metabolismo
16.
J Physiol ; 588(Pt 10): 1763-77, 2010 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-20351045

RESUMO

Colonic epithelial K(+) secretion is a two-step transport process with initial K(+) uptake over the basolateral membrane followed by K(+) channel-dependent exit into the lumen. In this process the large-conductance, Ca(2+)-activated K(Ca)1.1 (BK) channel has been identified as the only apparent secretory K(+) channel in the apical membrane of the murine distal colon. The BK channel is responsible for both resting and Ca(2+)-activated colonic K(+) secretion and is up-regulated by aldosterone. Agonists (e.g. adrenaline) that elevate cAMP are potent activators of distal colonic K(+) secretion. However, the secretory K(+) channel responsible for cAMP-induced K(+) secretion remains to be defined. In this study we used the Ussing chamber to identify adrenaline-induced electrogenic K(+) secretion. We found that the adrenaline-induced electrogenic ion secretion is a compound effect dominated by anion secretion and a smaller electrically opposing K(+) secretion. Using tissue from (i) BK wildtype (BK(+/+)) and knockout (BK(/)) and (ii) cystic fibrosis transmembrane regulator (CFTR) wildtype (CFTR(+/+)) and knockout (CFTR(/)) mice we were able to isolate the adrenaline-induced K(+) secretion. We found that adrenaline-induced K(+) secretion: (1) is absent in colonic epithelia from BK(/) mice, (2) is greatly up-regulated in mice on a high K(+) diet and (3) is present as sustained positive current in colonic epithelia from CFTR(/) mice. We identified two known C-terminal BK alpha-subunit splice variants in colonic enterocytes (STREX and ZERO). Importantly, the ZERO variant known to be activated by cAMP is differentially up-regulated in enterocytes from animals on a high K(+) diet. In summary, these results strongly suggest that the adrenaline-induced distal colonic K(+) secretion is mediated by the BK channel and probably involves aldosterone-induced ZERO splice variant up-regulation.


Assuntos
Colo/metabolismo , Epinefrina/farmacologia , Subunidades alfa do Canal de Potássio Ativado por Cálcio de Condutância Alta/metabolismo , Bloqueadores dos Canais de Potássio , Potássio/metabolismo , Antagonistas Adrenérgicos beta/farmacologia , Aldosterona/sangue , Animais , Colo/efeitos dos fármacos , AMP Cíclico/fisiologia , Regulador de Condutância Transmembrana em Fibrose Cística/genética , DNA Complementar/biossíntese , DNA Complementar/isolamento & purificação , Cultura em Câmaras de Difusão , Eletrofisiologia , Enterócitos/efeitos dos fármacos , Enterócitos/metabolismo , Mucosa Intestinal/citologia , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/metabolismo , Subunidades alfa do Canal de Potássio Ativado por Cálcio de Condutância Alta/efeitos dos fármacos , Subunidades alfa do Canal de Potássio Ativado por Cálcio de Condutância Alta/genética , Camundongos , Camundongos Knockout , Potássio/farmacologia , Potássio na Dieta/farmacologia , Propranolol/farmacologia , RNA/biossíntese , RNA/isolamento & purificação , Reação em Cadeia da Polimerase Via Transcriptase Reversa
17.
Am J Physiol Cell Physiol ; 298(5): C1057-65, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20164375

RESUMO

CFTR has been recognized to function as both an anion channel and a key regulator of Slc26 anion transporters in heterologous expression systems. Whether this regulatory relationship between CFTR and Slc26 transporters is seen in native intestine, and whether this effect is coupled to CFTR transport function or other features of this protein, has not been studied. The duodena of anesthetized CFTR-, NHE3-, Slc26a6-, and Scl26a3-deficient mice and wild-type (WT) littermates were perfused, and duodenal bicarbonate (HCO(3)(-)) secretion (DBS) and fluid absorptive or secretory rates were measured. The selective NHE3 inhibitor S1611 or genetic ablation of NHE3 significantly reduced fluid absorptive rates and increased DBS. Slc26a6 (PAT1) or Slc26a3 (DRA) ablation reduced the S1611-induced DBS increase and reduced fluid absorptive rates, suggesting that the effect of S1611 or NHE3 ablation on HCO(3)(-) secretion may be an unmasking of Slc26a6- and Slc26a3-mediated Cl(-)/HCO(3)(-) exchange activity. In the absence of CFTR expression or after application of the CFTR(inh)-172, fluid absorptive rates were similar to those of WT, but S1611 induced virtually no increase in DBS, demonstrating that CFTR transport activity, and not just its presence, is required for Slc26-mediated duodenal HCO(3)(-) secretion. A functionally active CFTR is an absolute requirement for Slc26-mediated duodenal HCO(3)(-) secretion, but not for Slc26-mediated fluid absorption, in which these transporters operate in conjunction with the Na(+)/H(+) exchanger NHE3. This suggests that Slc26a6 and Slc26a3 need proton recycling via NHE3 to operate in the Cl(-) absorptive mode and Cl(-) exit via CFTR to operate in the HCO(3)(-) secretory mode.


Assuntos
Antiporters/metabolismo , Regulador de Condutância Transmembrana em Fibrose Cística/fisiologia , Animais , Benzoatos/farmacologia , Bicarbonatos/metabolismo , Duodeno/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/fisiologia , Mucosa Intestinal/metabolismo , Camundongos , Camundongos Endogâmicos CFTR , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Trocador 3 de Sódio-Hidrogênio , Trocadores de Sódio-Hidrogênio/genética , Trocadores de Sódio-Hidrogênio/metabolismo , Transportadores de Sulfato , Tiazolidinas/farmacologia
18.
Inflamm Bowel Dis ; 16(7): 1149-61, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20027604

RESUMO

BACKGROUND: A major causative factor of diarrhea in ulcerative colitis (UC) patients is the loss of Na(+) absorptive capacity of the inflamed colonic mucosa. Potential contributing mechanisms include reduced driving force for active transport, and impaired expression, mislocalization, or defective transport function of Na(+) absorptive proteins. We therefore studied the expression, brush border membrane (BBM) localization, and transport capacity of the major intestinal Na(+) absorptive protein, the Na(+)/H(+) exchanger isoform 3 (NHE3) in biopsies from UC patients. METHODS: In UC and control biopsies, inflammation was graded histologically, NHE3, tumor necrosis factor alpha (TNF-alpha), villin, as well as other housekeeping genes were analyzed by quantitative real-time polymerase chain reaction (PCR), BBM localization of NHE3 determined by immunohistochemistry, and confocal microscopy. Na(+) absorptive capacity was assessed by (22)Na(+) isotope fluxes and NHE3 transport activity measured microfluorometrically in BCECF-loaded surface colonocytes within isolated crypts. RESULTS: In mildly, moderately, and severely inflamed sigmoid colon of UC patients, neither NHE3 mRNA expression nor the abundance of NHE3 in the BBM was significantly altered compared to other structural components of the BBM. However, Na(+) absorption was strongly reduced by approximately 80% and acid-activated NHE3 transport activity was significantly decreased in the surface cells of sigmoid colonic crypts even in moderately inflamed mucosa. CONCLUSIONS: In the colonic mucosa of patients with active UC, NHE3 transport capacity was found significantly decreased despite correct NHE3 location and abundance in the brush border, independent of current treatment. These findings suggest functional NHE3 transport as a novel factor for inflammatory diarrhea in UC patients.


Assuntos
Colite Ulcerativa/metabolismo , Microvilosidades/metabolismo , Trocadores de Sódio-Hidrogênio/metabolismo , Sódio/metabolismo , Western Blotting , Colite Ulcerativa/genética , Colite Ulcerativa/patologia , Colo/metabolismo , Colo/patologia , Humanos , Técnicas Imunoenzimáticas , Absorção Intestinal , Transporte de Íons , Microvilosidades/genética , RNA Mensageiro/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Trocador 3 de Sódio-Hidrogênio , Trocadores de Sódio-Hidrogênio/genética , Distribuição Tecidual , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo
19.
Am J Physiol Gastrointest Liver Physiol ; 297(6): G1223-31, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19779011

RESUMO

A Na(+)/HCO(3)(-) cotransporter (NBC) is located in the basolateral membrane of the gastrointestinal epithelium, where it imports HCO(3)(-) during stimulated anion secretion. Having previously demonstrated secretagogue activation of NBC in murine colonic crypts, we now asked whether vesicle traffic and exocytosis are involved in this process. Electrogenic NBCe1-B was expressed at significantly higher levels than electroneutral NBCn1 in colonic crypts as determined by QRT-PCR. In cell surface biotinylation experiments, a time-dependent increase in biotinylated NBCe1 was observed, which occurred with a peak of +54.8% after 20 min with forskolin (P < 0.05) and more rapidly with a peak of +59.8% after 10 min with carbachol (P < 0.05) and which corresponded well with the time course of secretagogue-stimulated colonic bicarbonate secretion in Ussing chamber experiments. Accordingly, in isolated colonic crypts pretreated with forskolin and carbachol for 10 min, respectively, and subjected to immunohistochemistry, the NBCe1 signal showed a markedly stronger colocalization with the E-cadherin signal, which was used as a membrane marker, compared with the untreated control. Cytochalasin D did not change the observed increase in membrane abundance, whereas colchicine alone enhanced NBCe1 membrane expression without an additional increase after carbachol or forskolin, and LY294002 had a marked inhibitory effect. Taken together, our results demonstrate a secretagogue-induced increase of NBCe1 membrane expression. Vesicle traffic and exocytosis might thus represent a novel mechanism of intestinal NBC activation by secretagogues.


Assuntos
Bicarbonatos/metabolismo , Membrana Celular/efeitos dos fármacos , Colo/efeitos dos fármacos , Fármacos Gastrointestinais/farmacologia , Mucosa Intestinal/efeitos dos fármacos , Secreções Intestinais/efeitos dos fármacos , Simportadores de Sódio-Bicarbonato/metabolismo , Sódio/metabolismo , Adenilil Ciclases/metabolismo , Animais , Biotinilação , Caderinas/metabolismo , Carbacol/farmacologia , Membrana Celular/metabolismo , Agonistas Colinérgicos/farmacologia , Colchicina/farmacologia , Colforsina/farmacologia , Colo/metabolismo , AMP Cíclico/metabolismo , Citofotometria , Relação Dose-Resposta a Droga , Ativadores de Enzimas/farmacologia , Exocitose/efeitos dos fármacos , Concentração de Íons de Hidrogênio , Imuno-Histoquímica , Mucosa Intestinal/metabolismo , Potenciais da Membrana , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Reação em Cadeia da Polimerase , Vesículas Secretórias/efeitos dos fármacos , Vesículas Secretórias/metabolismo , Simportadores de Sódio-Bicarbonato/deficiência , Simportadores de Sódio-Bicarbonato/genética , Fatores de Tempo
20.
J Physiol ; 587(Pt 15): 3955-65, 2009 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-19491250

RESUMO

Parietal cell (PC) proton secretion via H(+)/K(+)-ATPase requires apical K(+) recycling. A variety of K(+) channels and transporters are expressed in the PC and the molecular nature of the apical K(+) recycling channel is under debate. This study was undertaken to delineate the exact function of KCNQ1 channels in gastric acid secretion. Acid secretory rates and electrophysiological parameters were determined in gastric mucosae of 7- to 8-day-old KCNQ1(+/+), (+/-) and (-/-) mice. Parietal cell ultrastructure, abundance and gene expression levels were quantified. Glandular structure and PC abundance, and housekeeping gene expression did not differ between the KCNQ1(-/-) and (+/+) mucosae. Microvillar secretory membranes were intact, but basal acid secretion was absent and forskolin-stimulated acid output reduced by approximately 90% in KCNQ1(-/-) gastric mucosa. Application of a high K(+) concentration to the luminal membrane restored normal acid secretory rates in the KCNQ1(-/-) mucosa. The study demonstrates that the KCNQ1 channel provides K(+) to the extracellular K(+) binding site of the H(+)/K(+)-ATPase during acid secretion, and no other gastric K(+) channel can substitute for this function.


Assuntos
Ácido Gástrico/metabolismo , Mucosa Gástrica/metabolismo , Canal de Potássio KCNQ1/metabolismo , Animais , Proteínas de Transporte de Ânions/metabolismo , Antiporters/metabolismo , Peso Corporal/fisiologia , Colforsina/farmacologia , Mucosa Gástrica/efeitos dos fármacos , Mucosa Gástrica/patologia , ATPase Trocadora de Hidrogênio-Potássio/metabolismo , Canal de Potássio KCNQ1/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Potássio/metabolismo , Canais de Potássio Corretores do Fluxo de Internalização/metabolismo , Proteínas SLC4A , Canal Kir5.1
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA