Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Am J Physiol Gastrointest Liver Physiol ; 311(3): G423-30, 2016 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-27390324

RESUMO

Divalent metal-ion transporter-1 (DMT1), the principal mechanism by which nonheme iron is taken up at the intestinal brush border, is energized by the H(+)-electrochemical potential gradient. The provenance of the H(+) gradient in vivo is unknown, so we have explored a role for brush-border Na(+)/H(+) exchanger (NHE) isoforms by examining iron homeostasis and intestinal iron handling in mice lacking NHE2 or NHE3. We observed modestly depleted liver iron stores in NHE2-null (NHE2(-/-)) mice stressed on a low-iron diet but no change in hematological or blood iron variables or the expression of genes associated with iron metabolism compared with wild-type mice. Ablation of NHE3 strongly depleted liver iron stores, regardless of diet. We observed decreases in blood iron variables but no overt anemia in NHE3-null (NHE3(-/-)) mice on a low-iron diet. Intestinal expression of DMT1, the apical surface ferrireductase cytochrome b reductase-1, and the basolateral iron exporter ferroportin was upregulated in NHE3(-/-) mice, and expression of liver Hamp1 (hepcidin) was suppressed compared with wild-type mice. Absorption of (59)Fe from an oral dose was substantially impaired in NHE3(-/-) compared with wild-type mice. Our data point to an important role for NHE3 in generating the H(+) gradient that drives DMT1-mediated iron uptake at the intestinal brush border.


Assuntos
Regulação da Expressão Gênica/fisiologia , Ferro/metabolismo , Microvilosidades/fisiologia , Trocadores de Sódio-Hidrogênio/metabolismo , Animais , Transporte Biológico , Humanos , Mucosa Intestinal/metabolismo , Camundongos , Camundongos Knockout , Oócitos , Sódio/metabolismo , Trocador 3 de Sódio-Hidrogênio , Trocadores de Sódio-Hidrogênio/genética , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Desequilíbrio Hidroeletrolítico , Xenopus
2.
Am J Physiol Gastrointest Liver Physiol ; 309(8): G635-47, 2015 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-26294671

RESUMO

Divalent metal-ion transporter-1 (DMT1) is a widely expressed iron-preferring membrane-transport protein that serves a critical role in erythroid iron utilization. We have investigated its role in intestinal metal absorption by studying a mouse model lacking intestinal DMT1 (i.e., DMT1(int/int)). DMT1(int/int) mice exhibited a profound hypochromic-microcytic anemia, splenomegaly, and cardiomegaly. That the anemia was due to iron deficiency was demonstrated by the following observations in DMT1(int/int) mice: 1) blood iron and tissue nonheme-iron stores were depleted; 2) mRNA expression of liver hepcidin (Hamp1) was depressed; and 3) intraperitoneal iron injection corrected the anemia, and reversed the changes in blood iron, nonheme-iron stores, and hepcidin expression levels. We observed decreased total iron content in multiple tissues from DMT1(int/int) mice compared with DMT1(+/+) mice but no meaningful change in copper, manganese, or zinc. DMT1(int/int) mice absorbed (64)Cu and (54)Mn from an intragastric dose to the same extent as did DMT1(+/+) mice but the absorption of (59)Fe was virtually abolished in DMT1(int/int) mice. This study reveals a critical function for DMT1 in intestinal nonheme-iron absorption for normal growth and development. Further, this work demonstrates that intestinal DMT1 is not required for the intestinal transport of copper, manganese, or zinc.


Assuntos
Proteínas de Transporte de Cátions/metabolismo , Cobre/metabolismo , Absorção Intestinal/fisiologia , Ferro/metabolismo , Manganês/metabolismo , Anemia Hipocrômica/genética , Anemia Hipocrômica/patologia , Animais , Proteínas de Transporte de Cátions/genética , Transportador de Cobre 1 , Regulação da Expressão Gênica/fisiologia , Homeostase/fisiologia , Camundongos , Camundongos Knockout , Zinco/metabolismo
3.
J Physiol ; 593(8): 1809-27, 2015 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-25605613

RESUMO

KEY POINTS: An in vitro approach to study gastric development is primary mouse-derived epithelium cultured as three-dimensional spheroids known as organoids. We have devised two unique gastric fundic-derived organoid cultures: model 1 for the expansion of gastric fundic stem cells, and model 2 for the maintenance of mature cell lineages. Organoids maintained in co-culture with immortalized stomach mesenchymal cells express robust numbers of surface pit, mucous neck, chief, endocrine and parietal cells. Histamine induced a significant decrease in intraluminal pH that was reversed by omeprazole in fundic organoids and indicated functional activity and regulation of parietal cells. Localized photodamage resulted in rapid cell exfoliation coincident with migration of neighbouring cells to the damaged area, sustaining epithelial continuity. We report the use of these models for studies of epithelial cell biology and cell damage and repair. ABSTRACT: Studies of gastric function and disease have been limited by the lack of extended primary cultures of the epithelium. An in vitro approach to study gastric development is primary mouse-derived antral epithelium cultured as three-dimensional spheroids known as organoids. There have been no reports on the use of organoids for gastric function. We have devised two unique gastric fundic-derived organoid cultures: model 1 for the expansion of gastric fundic stem cells, and model 2 for the maintenance of mature cell lineages. Both models were generated from single glands dissociated from whole fundic tissue and grown in basement membrane matrix (Matrigel) and organoid growth medium. Model 1 enriches for a stem cell-like niche via simple passage of the organoids. Maintained in Matrigel and growth medium, proliferating organoids expressed high levels of stem cell markers CD44 and Lgr5. Model 2 is a system of gastric organoids co-cultured with immortalized stomach mesenchymal cells (ISMCs). Organoids maintained in co-culture with ISMCs express robust numbers of surface pit, mucous neck, chief, endocrine and parietal cells. Histamine induced a significant decrease in intraluminal pH that was reversed by omeprazole in fundic organoids and indicated functional activity and regulation of parietal cells. Localized photodamage resulted in rapid cell exfoliation coincident with migration of neighbouring cells to the damaged area, sustaining epithelial continuity. Thus, we report the use of these models for studies of epithelial cell biology and cell damage and repair.


Assuntos
Técnicas de Cocultura/métodos , Células Epiteliais/citologia , Mucosa Gástrica/citologia , Organoides/citologia , Células-Tronco/citologia , Animais , Diferenciação Celular , Camundongos
4.
Am J Physiol Gastrointest Liver Physiol ; 308(6): G497-509, 2015 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-25552580

RESUMO

Clostridium difficile infection (CDI) is principally responsible for hospital acquired, antibiotic-induced diarrhea and colitis and represents a significant financial burden on our healthcare system. Little is known about C. difficile proliferation requirements, and a better understanding of these parameters is critical for development of new therapeutic targets. In cell lines, C. difficile toxin B has been shown to inhibit Na(+)/H(+) exchanger 3 (NHE3) and loss of NHE3 in mice results in an altered intestinal environment coupled with a transformed gut microbiota composition. However, this has yet to be established in vivo in humans. We hypothesize that C. difficile toxin inhibits NHE3, resulting in alteration of the intestinal environment and gut microbiota. Our results demonstrate that CDI patient biopsy specimens have decreased NHE3 expression and CDI stool has elevated Na(+) and is more alkaline compared with stool from healthy individuals. CDI stool microbiota have increased Bacteroidetes and Proteobacteria and decreased Firmicutes phyla compared with healthy subjects. In vitro, C. difficile grows optimally in the presence of elevated Na(+) and alkaline pH, conditions that correlate to changes observed in CDI patients. To confirm that inhibition of NHE3 was specific to C. difficile, human intestinal organoids (HIOs) were injected with C. difficile or healthy and CDI stool supernatant. Injection of C. difficile and CDI stool decreased NHE3 mRNA and protein expression compared with healthy stool and control HIOs. Together these data demonstrate that C. difficile inhibits NHE3 in vivo, which creates an altered environment favored by C. difficile.


Assuntos
Clostridioides difficile/crescimento & desenvolvimento , Colo/metabolismo , Colo/microbiologia , Enterocolite Pseudomembranosa/metabolismo , Enterocolite Pseudomembranosa/microbiologia , Microbiota , Trocadores de Sódio-Hidrogênio/metabolismo , Adulto , Idoso , Proteínas de Bactérias/metabolismo , Toxinas Bacterianas/metabolismo , Estudos de Casos e Controles , Células Cultivadas , Clostridioides difficile/isolamento & purificação , Clostridioides difficile/metabolismo , Regulação para Baixo , Fezes/microbiologia , Feminino , Interações Hospedeiro-Patógeno , Humanos , Concentração de Íons de Hidrogênio , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiologia , Masculino , Pessoa de Meia-Idade , Organoides , Células-Tronco Pluripotentes/metabolismo , Células-Tronco Pluripotentes/microbiologia , RNA Mensageiro/metabolismo , Sódio/metabolismo , Trocador 3 de Sódio-Hidrogênio , Trocadores de Sódio-Hidrogênio/genética
5.
Dig Dis Sci ; 59(3): 569-82, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24202649

RESUMO

BACKGROUND: Bone marrow-derived mesenchymal stem cells (BM-MSCs) promote gastric cancer in response to gastritis. In culture, BM-MSCs are prone to mutation with continued passage but it is unknown whether a similar process occurs in vivo in response to gastritis. AIM: The purpose of this study was to identify the role of chronic gastritis in the transformation of BM-MSCs leading to an activated cancer-promoting phenotype. METHODS: Age matched C57BL/6 (BL/6) and gastrin deficient (GKO) mice were used for isolation of stomach, serum and mesenchymal stem cells (MSCs) at 3 and 6 months of age. MSC activation was assessed by growth curve analysis, fluorescence-activated cell sorting and xenograft assays. To allow for the isolation of bone marrow-derived stromal cells and assay in response to chronic gastritis, IRG/Vav-1(Cre) mice that expressed both enhanced green fluorescent protein-expressing hematopoietic cells and red fluorescent protein-expressing stromal cells were generated. In a parabiosis experiment, IRG/Vav-1(Cre) mice were paired to either an uninfected Vav-1(Cre) littermate or a BL/6 mouse inoculated with Helicobacter pylori. RESULTS: GKO mice displayed severe atrophic gastritis accompanied by elevated gastric tissue and circulating transforming growth factor beta (TGFß) by 3 months of age. Compared to BM-MSCs isolated from uninflamed BL/6 mice, BM-MSCs isolated from GKO mice displayed an increased proliferative rate and elevated phosphorylated-Smad3 suggesting active TGFß signaling. In xenograft assays, mice injected with BM-MSCs from 6-month-old GKO animals displayed tumor growth. RFP+ stromal cells were rapidly recruited to the gastric mucosa of H. pylori parabionts and exhibited changes in gene expression. CONCLUSIONS: Gastritis promotes the in vivo activation of BM-MSCs to a phenotype reminiscent of a cancer-promoting cell.


Assuntos
Transformação Celular Neoplásica , Mucosa Gástrica/patologia , Gastrite Atrófica/patologia , Células-Tronco Mesenquimais/patologia , Fenótipo , Animais , Biomarcadores/metabolismo , Proliferação de Células , Mucosa Gástrica/metabolismo , Mucosa Gástrica/microbiologia , Gastrinas/deficiência , Gastrite Atrófica/metabolismo , Gastrite Atrófica/microbiologia , Proteínas Hedgehog/metabolismo , Infecções por Helicobacter/patologia , Helicobacter pylori , Immunoblotting , Células-Tronco Mesenquimais/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Parabiose , Reação em Cadeia da Polimerase em Tempo Real , Proteína Smad3/metabolismo , Fator de Crescimento Transformador beta/metabolismo
6.
Am J Physiol Gastrointest Liver Physiol ; 294(2): G429-40, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18032481

RESUMO

Although colonic lumen NH(4)(+) levels are high, 15-44 mM normal range in humans, relatively few studies have addressed the transport mechanisms for NH(4)(+). More extensive studies have elucidated the transport of NH(4)(+) in the kidney collecting duct, which involves a number of transporter processes also present in the distal colon. Similar to NH(4)(+) secretion in the renal collecting duct, we show that the distal colon secretory model, T84 cell line, has the capacity to secrete NH(4)(+) and maintain an apical-to-basolateral NH(4)(+) gradient. NH(4)(+) transport in the secretory direction was supported by basolateral NH(4)(+) loading on NKCC1, Na(+)-K(+)-ATPase, and the NH(4)(+) transporter, RhBG. NH(4)(+) was transported on NKCC1 in T84 cells nearly as well as K(+) as determined by bumetanide-sensitive (86)Rb-uptake. (86)Rb-uptake and ouabain-sensitive current measurement indicated that NH(4)(+) is transported by Na(+)-K(+)-ATPase in these cells to an equal extent as K(+). T84 cells expressed mRNA for the basolateral NH(4)(+) transporter RhBG and the apical NH(4)(+) transporter RhCG. Net NH(4)(+) transport in the secretory direction determined by (14)C-methylammonium (MA) uptake and flux occurred in T84 cells suggesting functional RhG protein activity. The occurrence of NH(4)(+) transport in the secretory direction within a colonic crypt cell model likely serves to minimize net absorption of NH(4)(+) because of surface cell NH(4)(+) absorption. These findings suggest that we rethink the present limited understanding of NH(4)(+) handling by the distal colon as being due solely to passive absorption.


Assuntos
Proteínas Sanguíneas/fisiologia , Colo/citologia , Colo/metabolismo , Glicoproteínas de Membrana/fisiologia , Compostos de Amônio Quaternário/metabolismo , Simportadores de Cloreto de Sódio-Potássio/fisiologia , Adenosina Trifosfatases/metabolismo , Transporte Biológico Ativo/fisiologia , Linhagem Celular , AMP Cíclico/farmacologia , DNA Complementar/genética , Humanos , Concentração de Íons de Hidrogênio , Metilaminas/metabolismo , Plasmídeos/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Radioisótopos de Rubídio , ATPase Trocadora de Sódio-Potássio/metabolismo , Membro 2 da Família 12 de Carreador de Soluto
7.
Am J Physiol Gastrointest Liver Physiol ; 292(1): G447-55, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16901991

RESUMO

The role of Slc26a6 (PAT1) on apical Cl-/HCO3- exchange and bicarbonate secretion in pancreatic duct cells was investigated using Slc26a6 null and wild-type (WT) mice. Apical Cl-/HCO3- exchange activity was measured with the pH-sensitive dye BCECF in microperfused interlobular ducts. The HCO3(-)-influx mode of apical [Cl-]i/[HCO3-]o exchange (where brackets denote concentration and subscripts i and o denote intra- and extracellular, respectively) was dramatically upregulated in Slc26a6 null mice (P < 0.01 vs. WT), whereas the HCO3(-)-efflux mode of apical [Cl-]o/[HCO3-]i exchange was decreased in Slc26a6 null mice (P < 0.05 vs. WT), suggesting the unidirectionality of the Slc26a6-mediated HCO3- transport. Fluid secretory rate in interlobular ducts were comparable in WT and Slc26a6 null mice (P > 0.05). In addition, when pancreatic juice was collected from whole animal in basal and secretin-stimulated conditions, neither juice volume nor its pH showed differences between WT and Slc26a6 null mice. Semiquantitative RT-PCR demonstrated more than fivefold upregulation in Slc26a3 (DRA) expression in Slc26a6 knockout pancreas. In conclusion, these results point to the role of Slc26a6 in HCO3- efflux at the apical membrane and also suggest the presence of a robust Slc26a3 compensatory upregulation, which can replace the function of Slc26a6 in pancreatic ducts.


Assuntos
Antiporters/deficiência , Antiporters/genética , Antiporters/fisiologia , Antiportadores de Cloreto-Bicarbonato/fisiologia , AMP Cíclico/farmacologia , Ductos Pancreáticos/fisiologia , Animais , Bicarbonatos/metabolismo , Camundongos , Camundongos Knockout , Ductos Pancreáticos/efeitos dos fármacos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Secretina/farmacologia , Transportadores de Sulfato
8.
J Biol Chem ; 281(49): 37962-71, 2006 Dec 08.
Artigo em Inglês | MEDLINE | ID: mdl-17001077

RESUMO

Mutations in the SLC26A3 (DRA (down-regulated in adenoma)) gene constitute the molecular etiology of congenital chloride-losing diarrhea in humans. To ascertain its role in intestinal physiology, gene targeting was used to prepare mice lacking slc26a3. slc26a3-deficient animals displayed postpartum lethality at low penetrance. Surviving dra-deficient mice exhibited high chloride content diarrhea, volume depletion, and growth retardation. In addition, the large intestinal loops were distended, with colonic mucosa exhibiting an aberrant growth pattern and the colonic crypt proliferative zone being greatly expanded in slc26a3-null mice. Apical membrane chloride/base exchange activity was sharply reduced, and luminal content was more acidic in slc26a3-null mouse colon. The epithelial cells in the colon displayed unique adaptive regulation of ion transporters; NHE3 expression was enhanced in the proximal and distal colon, whereas colonic H,K-ATPase and the epithelial sodium channel showed massive up-regulation in the distal colon. Plasma aldosterone was increased in slc26a3-null mice. We conclude that slc26a3 is the major apical chloride/base exchanger and is essential for the absorption of chloride in the colon. In addition, slc26a3 regulates colonic crypt proliferation. Deletion of slc26a3 results in chloride-rich diarrhea and is associated with compensatory adaptive up-regulation of ion-absorbing transporters.


Assuntos
Antiporters/deficiência , Cloretos/metabolismo , Colo/metabolismo , Colo/patologia , Animais , Antiporters/genética , Antiporters/fisiologia , Sequência de Bases , Proliferação de Células , Primers do DNA/genética , Feminino , ATPase Trocadora de Hidrogênio-Potássio/metabolismo , Transporte de Íons , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Biológicos , Trocador 3 de Sódio-Hidrogênio , Trocadores de Sódio-Hidrogênio/genética , Trocadores de Sódio-Hidrogênio/metabolismo , Transportadores de Sulfato , Regulação para Cima
9.
Am J Physiol Gastrointest Liver Physiol ; 289(4): G768-78, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16002564

RESUMO

Normal human colonic luminal (NH(4)(+)) concentration ([NH(4)(+)]) ranges from approximately 10 to 100 mM. However, the nature of the effects of NH(4)(+) on transport, as well as NH(4)(+) transport itself, in colonic epithelium is poorly understood. We elucidate here the effects of apical NH(4)(+) on cAMP-stimulated Cl(-) secretion in colonic T84 cells. In HEPES-buffered solutions, 10 mM apical NH(4)(+) had no significant effect on cAMP-stimulated current. In contrast, 10 mM apical NH(4)(+) reduced current within 5 min to 61 +/- 4% in the presence of 25 mM HCO(3)(-). Current inhibition was not simply due to an increase in extracellular K(+)-like cations, in that the current magnitude was 95 +/- 5% with 10 mM apical K(+) and 46 +/- 3% with 10 mM apical NH(4)(+) relative to that with 5 mM apical K(+). We previously demonstrated that inhibition of Cl(-) secretion by basolateral NH(4)(+) occurs in HCO(3)(-)-free conditions and exhibits anomalous mole fraction behavior. In contrast, apical NH(4)(+) inhibition of current in HCO(3)(-) buffer did not show anomalous mole fraction behavior and followed the absolute [NH(4)(+)] in K(+)-NH(4)(+) mixtures, where K(+) concentration + [NH(4)(+)] = 10 mM. The apical NH(4)(+) inhibitory effect was not prevented by 100 microM methazolamide, suggesting no role for apical carbonic anhydrase. However, apical NH(4)(+) inhibition of current was prevented by 10 min of pretreatment of the apical surface with 500 microM DIDS, 100 microM 4,4'-dinitrostilbene-2,2'-disulfonic acid (DNDS), or 25 microM niflumic acid, suggesting a role for NH(4)(+) action through an apical anion exchanger. mRNA and protein for the apical anion exchangers SLC26A3 [downregulated in adenoma (DRA)] and SLC26A6 [putative anion transporter (PAT1)] were detected in T84 cells by RT-PCR and Northern and Western blots. DRA and PAT1 appear to associate with CFTR in the apical membrane. We conclude that the HCO(3)(-) dependence of apical NH(4)(+) inhibition of secretion is due to the action of NH(4)(+) on an apical anion exchanger.


Assuntos
Bicarbonatos/metabolismo , Colo/metabolismo , AMP Cíclico/antagonistas & inibidores , AMP Cíclico/farmacologia , Compostos de Amônio Quaternário/farmacologia , Ácido 4,4'-Di-Isotiocianoestilbeno-2,2'-Dissulfônico/farmacologia , Animais , Antiporters/efeitos dos fármacos , Antiporters/metabolismo , Northern Blotting , Western Blotting , Carbacol/farmacologia , Inibidores da Anidrase Carbônica/farmacologia , Linhagem Celular , Canais de Cloreto/antagonistas & inibidores , Colo/efeitos dos fármacos , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Humanos , Imuno-Histoquímica , Proteínas de Membrana Transportadoras/metabolismo , Agonistas Muscarínicos/farmacologia , Ratos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Estilbenos/farmacologia , Transportadores de Sulfato
10.
Am J Physiol Gastrointest Liver Physiol ; 288(1): G135-42, 2005 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15358594

RESUMO

Ischemia is the central pathogenic factor underlying a spectrum of intestinal disorders. The study of the cellular signaling responses to ischemic stress in nonepithelial cells has progressed substantially in the previous several years, but little is known about the response in epithelial cells. Unique features of the epithelial response to ischemic stress suggest differential regulation with regards to signaling. The PKC family of proteins has been implicated in ischemic stress in nonepithelial systems. The role of PKC isoforms in chemical ischemia in intestinal epithelial cells is evaluated in this study. Additionally, the phosphorylation of the F-actin cross-linking protein myristoylated alanine-rich C kinase substrate (MARCKS) is also studied. Chemical ischemia resulted in the transient activation of only the isoform PKC-epsilon as detected by translocation employing the subcellular fractionation technique. The pharmacological agonists phorbol 12-myristate 13-acetate and carbachol also led to the translocation of PKC-epsilon. By immunofluoresence, MARCKS is noted to be located at the lateral membrane under control conditions. In response to carbachol, MARCKS translocates to the cytosol, indicating its phosphorylation, which is additionally confirmed biochemically. Consistent with this observation, carbachol induces the translocation of PKC-epsilon to proximity with MARCKS at the lateral membrane. In response to chemical ischemia, MARCKS fails to translocate and phosphorylation does not increase. Additionally, the translocation of PKC-epsilon is not to the lateral membrane but rather basally. The data suggest that the differential translocation of PKC-epsilon in response to pharmacological agonists versus ischemic stress may lead to different effects on downstream targets.


Assuntos
Intestinos/citologia , Intestinos/fisiologia , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas de Membrana/metabolismo , Proteína Quinase C/farmacologia , Carbacol/farmacologia , Agonistas Colinérgicos/farmacologia , Células Epiteliais/fisiologia , Humanos , Hipóxia , Substrato Quinase C Rico em Alanina Miristoilada , Fosforilação , Proteína Quinase C/metabolismo , Proteína Quinase C-épsilon , Acetato de Tetradecanoilforbol/farmacologia
11.
Am J Physiol Gastrointest Liver Physiol ; 286(1): G14-22, 2004 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-12946942

RESUMO

A significant amount of ammonium (NH4+) is absorbed by the colon. The nature of NH4+ effects on transport and NH4+ transport itself in colonic epithelium is poorly understood. The goal of this study was to elucidate the effects of NH4+ on cAMP-stimulated Cl- secretion in the colonic cell line T84. In HEPES-buffered solutions, application of basolateral NH4+ resulted in a reduced level of Cl- secretory current. The effect of NH4+ appears to occur by at least three mechanisms: 1) basolateral membrane depolarization, 2) a competitive effect with K+, and 3) a long-term (>20 min) increase in transepithelial resistance (TER). The competitive effect with K+ exhibits anomalous mole fraction behavior. Transepithelial current relative to that in 10 mM basolateral K+ was inhibited 15% by 10 mM NH4+ alone and by 30% with a mixture of 2 mM K+ and 8 mM NH4+. A mole fraction mix of 2 mM K+:8 mM NH4+ produced a greater inhibition of basolateral membrane K+ current than pure K+ or NH4+ alone. Similar anomalous behavior was also observed for inhibition of bumetanide-sensitive 36Cl- uptake, e.g., Na+-K+-2Cl- -cotransporter (NKCC-1). No anomalous effect was observed on Na+-K+-ATPase current. Both NKCC-1 and Na+-K+-ATPase activity were elevated in 10 mM NH4+ with respect to 10 mM K+. The effect on TER did not exhibit anomalous mole fraction behavior. The overall effect of basolateral NH4+ on cAMP-stimulated transport is dependent on the [K+]o /[NH4+]o ratio at the basolateral membrane, where o is outside of the cell.


Assuntos
Cloretos/metabolismo , Colo/metabolismo , Compostos de Amônio Quaternário/farmacologia , Linhagem Celular , Cloro/metabolismo , Colforsina/farmacologia , Colo/efeitos dos fármacos , Dinoprostona/farmacologia , Epitélio/metabolismo , Humanos , Concentração de Íons de Hidrogênio , Potássio/metabolismo , Canais de Potássio/efeitos dos fármacos , Canais de Potássio/metabolismo , Radioisótopos , Simportadores de Cloreto de Sódio-Potássio/metabolismo , ATPase Trocadora de Sódio-Potássio/metabolismo , Estimulação Química
12.
Adv Exp Med Biol ; 559: 131-9, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-18727234

RESUMO

Basolateral ammonium produces an inhibition of Cl- secretion the magnitude of which is dependent on the NH4+ to K+ concentration ratio. Inhibition is maximal at a mole fraction ratio of 0.25 K+ to NH4+. This anomalous mole fraction effect is due to effects on the basolateral K+ channel as well as Na(+)-K(+)-2Cl- cotransporter. However, only Cl- loading, not K+ loading, appears affected in an anomalous mole fraction manner. Transepithelial current is only slightly inhibited relative to equilmolar K+ by NH4+. As in other systems, both Na(+)-K(+)-ATPase and Na(+)-K(+)-2Cl- can act in Na(+)-NH4(+)-ATPase and Na(+)-NH4(+)-2Cl- transport modes. NH4+ conducts through most K+ channels and thus likely through the apical K+ channel present in native crypt cells. This suggests that, similar to the kidney, colonic secretory cells have the capacity to secrete NH4+ when in a K(+)-secreting mode with elevated basolateral NH4+ levels.


Assuntos
Colo/citologia , Colo/metabolismo , Canais Iônicos/metabolismo , Proteínas de Membrana Transportadoras/metabolismo , Compostos de Amônio Quaternário/metabolismo , Animais , Humanos , Concentração de Íons de Hidrogênio , ATPase Trocadora de Sódio-Potássio/metabolismo
13.
Am J Physiol Cell Physiol ; 285(2): C300-9, 2003 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12660149

RESUMO

Protein kinase C (PKC) is known to regulate epithelial barrier function. However, the effect of specific PKC isozymes, and their mechanism of action, are largely unknown. We determined that the nonphorbol ester PKC agonist bryostatin-1 increased transepithelial electrical resistance (TER), a marker of barrier function, in confluent T84 epithelia. Bryostatin-1, which has been shown to selectively activate PKC-alpha, -epsilon, and -delta (34), was associated with a shift in the subcellular distribution of the tight junction proteins claudin-1 and ZO-2 from a detergent-soluble fraction into a detergent-insoluble fraction. Bryostatin-1 also led to the appearance of a higher-molecular-weight form of occludin previously shown to correspond to protein phosphorylation. These changes were attenuated by the conventional and novel PKC inhibitor Gö-6850 but not the conventional PKC inhibitor Gö-6976 or the PKC-delta inhibitor röttlerin, implicating a novel isozyme, likely PKC-epsilon. The results suggest that enhanced epithelial barrier function induced by bryostatin-1 involves a PKC-epsilon-dependent signaling pathway leading to recruitment of claudin-1 and ZO-2, and phosphorylation of occludin, into the tight junctional complex.


Assuntos
Células Epiteliais/enzimologia , Lactonas/farmacologia , Proteínas de Membrana/metabolismo , Proteína Quinase C/metabolismo , Junções Íntimas/enzimologia , Regulação para Cima/fisiologia , Briostatinas , Linhagem Celular , Permeabilidade da Membrana Celular/efeitos dos fármacos , Permeabilidade da Membrana Celular/fisiologia , Claudina-1 , Impedância Elétrica , Inibidores Enzimáticos/farmacologia , Células Epiteliais/efeitos dos fármacos , Humanos , Macrolídeos , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Proteínas de Membrana/efeitos dos fármacos , Ocludina , Proteína Quinase C/efeitos dos fármacos , Proteína Quinase C-épsilon , Transporte Proteico/efeitos dos fármacos , Transporte Proteico/fisiologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Junções Íntimas/efeitos dos fármacos , Regulação para Cima/efeitos dos fármacos , Proteína da Zônula de Oclusão-2
14.
Am J Physiol Gastrointest Liver Physiol ; 284(4): G703-12, 2003 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-12505880

RESUMO

Tumor necrosis factor (TNF) increases epithelial permeability in many model systems. Protein kinase C (PKC) isozymes regulate epithelial barrier function and alter ligand-receptor interactions. We sought to define the impact of PKC on TNF-induced barrier dysfunction in T84 intestinal epithelia. TNF induced a dose- and time-dependent fall in transepithelial electrical resistance (TER) and an increase in [(3)H]mannitol flux. The TNF-induced fall in TER was not PKC mediated but was prevented by pretreatment with bryostatin-1, a PKC agonist. As demonstrated by a pattern of sensitivity to pharmacological inhibitors of PKC, this epithelial barrier preservation was mediated by novel PKC isozymes. Bryostatin-1 reduced TNF receptor (TNF-R1) surface availability, as demonstrated by radiolabeled TNF binding and cell surface biotinylation assays, and increased TNF-R1 receptor shedding. The pattern of sensitivity to isozyme-selective PKC inhibitors suggested that these effects were mediated by activation of PKC-epsilon. In addition, after bryostatin-1 treatment, PKC-delta and TNF-R1 became associated, as determined by mutual coimmunoprecipitation assay, which has been shown to lead to receptor desensitization in neutrophils. TNF-induced barrier dysfunction occurs independently of PKC, but selective modulation of novel PKC isozymes may regulate TNF-R1 signaling.


Assuntos
Antineoplásicos/farmacologia , Mucosa Intestinal/metabolismo , Lactonas/farmacologia , Proteína Quinase C/metabolismo , Fator de Necrose Tumoral alfa/farmacologia , Antígenos CD/metabolismo , Transporte Biológico/efeitos dos fármacos , Transporte Biológico/fisiologia , Briostatinas , Células Cultivadas , Relação Dose-Resposta a Droga , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Humanos , Mucosa Intestinal/citologia , Radioisótopos do Iodo , Macrolídeos , Manitol/farmacocinética , Proteína Quinase C-delta , Proteína Quinase C-épsilon , Receptores do Fator de Necrose Tumoral/metabolismo , Receptores Tipo I de Fatores de Necrose Tumoral , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Trítio , Fator de Necrose Tumoral alfa/metabolismo
15.
Pancreas ; 25(3): 290-5, 2002 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-12370541

RESUMO

INTRODUCTION: Calcium-activated chloride conductance has been identified in normal pancreatic duct cells. Recent in vitro evidence suggests that angiotensin II (AngII) stimulates pancreatic secretion in both cystic fibrosis (CFPAC) and transformed pancreatic cells. AIMS: To investigate calcium-mediated stimulatory effects of AngII in both nontransformed dog pancreatic duct epithelial (DPDE) and CFPAC cells. METHODS: Western blots were performed in both cells seeking AngII receptors. In additional studies, DPDE and CFPAC cells were grown on vitrogen-coated glass cover slips and loaded with Indo-1-AM dye. Cells were placed in a confocal microscope's perfusion chamber and perfused with 100 microM AngII or ATP (control). Cells were excited with UV light, and intracellular calcium ([Ca+2]i) was read using fluorescence emission at 405 and 530 nm. Finally, single channels in the DPDE cells were examined using cell-attached patch clamps. Current amplitude histograms provided estimates of the conductance and open probability of channels. RESULTS: Western blots demonstrated presence of both AT and AT AngII receptors in DPDE and CFPAC cells; the density of AT receptors appeared lower than that of AT receptors. Basal intracellular calcium concentrations did not differ between DPDE (109 +/- 11 nM) and CFPAC (103 +/- 8 nM) cells. AngII significantly increased measured intracellular calcium concentrations in both DPDE (909 +/- 98 nM) and CFPAC (879 +/- 207 nM) cells, as did ATP (DPDE = 1722 +/- 228 nM; CFPAC = 1522 +/- 245 nM). In the patch clamp studies, a variety of different channels were observed; they appeared to be an 11pS nonselective cation (NSC) channel, a 4.6pS Na+ channel, a 3pS anion channel, and an 8pS chloride channel. The latter channel had characteristics similar to cystic fibrosis transmembrane conductance regulator (CFTR). Apical or basolateral application of AngII activated both the 11pS NSC and the 3pS channels. CONCLUSION: In nontransformed DPDE and CFPAC cells, specific AngII receptors mediate increases in [Ca ]. The latter effect of AngII may elicit activation of calcium-mediated chloride channels, suggesting a role for AngII as an alternative mediator of pancreatic ductal secretion.


Assuntos
Angiotensina II/farmacologia , Sinalização do Cálcio , Ductos Pancreáticos/fisiologia , Animais , Western Blotting , Linhagem Celular , Células Cultivadas , Canais de Cloreto/fisiologia , Fibrose Cística/fisiopatologia , Cães , Condutividade Elétrica , Células Epiteliais/química , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/fisiologia , Ductos Pancreáticos/citologia , Técnicas de Patch-Clamp , Receptores de Angiotensina/análise
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA