Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Nat Commun ; 13(1): 5212, 2022 09 05.
Artigo em Inglês | MEDLINE | ID: mdl-36064721

RESUMO

Life-threatening hyperammonemia occurs in both inherited and acquired liver diseases affecting ureagenesis, the main pathway for detoxification of neurotoxic ammonia in mammals. Protein O-GlcNAcylation is a reversible and nutrient-sensitive post-translational modification using as substrate UDP-GlcNAc, the end-product of hexosamine biosynthesis pathway. Here we show that increased liver UDP-GlcNAc during hyperammonemia increases protein O-GlcNAcylation and enhances ureagenesis. Mechanistically, O-GlcNAcylation on specific threonine residues increased the catalytic efficiency for ammonia of carbamoyl phosphate synthetase 1 (CPS1), the rate-limiting enzyme in ureagenesis. Pharmacological inhibition of O-GlcNAcase, the enzyme removing O-GlcNAc from proteins, resulted in clinically relevant reductions of systemic ammonia in both genetic (hypomorphic mouse model of propionic acidemia) and acquired (thioacetamide-induced acute liver failure) mouse models of liver diseases. In conclusion, by fine-tuned control of ammonia entry into ureagenesis, hepatic O-GlcNAcylation of CPS1 increases ammonia detoxification and is a novel target for therapy of hyperammonemia in both genetic and acquired diseases.


Assuntos
Amônia , Carbamoil-Fosfato Sintase (Amônia) , Hiperamonemia , Ureia , Difosfato de Uridina , Acetilglucosamina , Amônia/metabolismo , Animais , Biocatálise , Carbamoil-Fosfato Sintase (Amônia)/genética , Carbamoil-Fosfato Sintase (Amônia)/metabolismo , Modelos Animais de Doenças , Glicosilação , Humanos , Hiperamonemia/genética , Hiperamonemia/metabolismo , Mamíferos/metabolismo , Camundongos , N-Acetilglucosaminiltransferases/genética , N-Acetilglucosaminiltransferases/metabolismo , Acidemia Propiônica/genética , Acidemia Propiônica/metabolismo , Processamento de Proteína Pós-Traducional/genética , Ureia/metabolismo , Difosfato de Uridina/genética , Difosfato de Uridina/metabolismo
2.
EMBO Mol Med ; 13(2): e13158, 2021 02 05.
Artigo em Inglês | MEDLINE | ID: mdl-33369168

RESUMO

Urea cycle disorders (UCD) are inherited defects in clearance of waste nitrogen with high morbidity and mortality. Novel and more effective therapies for UCD are needed. Studies in mice with constitutive activation of autophagy unravelled Beclin-1 as druggable candidate for therapy of hyperammonemia. Next, we investigated efficacy of cell-penetrating autophagy-inducing Tat-Beclin-1 (TB-1) peptide for therapy of the two most common UCD, namely ornithine transcarbamylase (OTC) and argininosuccinate lyase (ASL) deficiencies. TB-1 reduced urinary orotic acid and improved survival under protein-rich diet in spf-ash mice, a model of OTC deficiency (proximal UCD). In AslNeo/Neo mice, a model of ASL deficiency (distal UCD), TB-1 increased ureagenesis, reduced argininosuccinate, and improved survival. Moreover, it alleviated hepatocellular injury and decreased both cytoplasmic and nuclear glycogen accumulation in AslNeo/Neo mice. In conclusion, Beclin-1-dependent activation of autophagy improved biochemical and clinical phenotypes of proximal and distal defects of the urea cycle.


Assuntos
Acidúria Argininossuccínica , Doença da Deficiência de Ornitina Carbomoiltransferase , Distúrbios Congênitos do Ciclo da Ureia , Animais , Autofagia , Proteína Beclina-1/genética , Camundongos
3.
Hum Mol Genet ; 28(R1): R42-R48, 2019 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-31227828

RESUMO

Urea cycle disorders (UCD) are inborn errors of metabolism caused by deficiency of enzymes required to transfer nitrogen from ammonia into urea. Current paradigms of treatment focus on dietary manipulations, ammonia scavenger drugs, and orthotopic liver transplantation. In the last years, there has been intense preclinical research aiming at developing more effective treatments for UCD, and as a result, several novel approaches based on new knowledge of the disease pathogenesis, cell and gene therapies are currently under clinical investigation. We provide an overview of the latest advances for the development of novel therapies for UCD.


Assuntos
Distúrbios Congênitos do Ciclo da Ureia/terapia , Animais , Terapia Baseada em Transplante de Células e Tecidos , Terapia Combinada , Gerenciamento Clínico , Suscetibilidade a Doenças , Terapia de Reposição de Enzimas , Terapia Genética , Humanos , Transplante de Fígado/métodos , Redes e Vias Metabólicas , Resultado do Tratamento , Ureia/metabolismo , Distúrbios Congênitos do Ciclo da Ureia/diagnóstico , Distúrbios Congênitos do Ciclo da Ureia/etiologia , Distúrbios Congênitos do Ciclo da Ureia/metabolismo
4.
J Inherit Metab Dis ; 42(6): 1128-1135, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-30724386

RESUMO

The urea cycle and glutamine synthetase (GS) are the two main pathways for waste nitrogen removal and their deficiency results in hyperammonemia. Here, we investigated the efficacy of liver-specific GS overexpression for therapy of hyperammonemia. To achieve hepatic GS overexpression, we generated a helper-dependent adenoviral (HDAd) vector expressing the murine GS under the control of a liver-specific expression cassette (HDAd-GS). Compared to mice injected with a control vector expressing an unrelated reporter gene (HDAd-alpha-fetoprotein), wild-type mice with increased hepatic GS showed reduced blood ammonia levels and a concomitant increase of blood glutamine after intraperitoneal injections of ammonium chloride, whereas blood urea was unaffected. Moreover, injection of HDAd-GS reduced blood ammonia levels at baseline and protected against acute hyperammonemia following ammonia challenge in a mouse model with conditional hepatic deficiency of carbamoyl phosphate synthetase 1 (Cps1), the initial and rate-limiting step of ureagenesis. In summary, we found that upregulation of hepatic GS reduced hyperammonemia in wild-type and Cps1-deficient mice, thus confirming a key role of GS in ammonia detoxification. These results suggest that hepatic GS augmentation therapy has potential for treatment of both primary and secondary forms of hyperammonemia.


Assuntos
Amônia/metabolismo , Terapia Genética/métodos , Glutamato-Amônia Ligase/genética , Hiperamonemia/genética , Hiperamonemia/terapia , Fígado/metabolismo , Amônia/toxicidade , Animais , Carbamoil-Fosfato Sintase (Amônia)/genética , Carbamoil-Fosfato Sintase (Amônia)/metabolismo , Doença da Deficiência da Carbamoil-Fosfato Sintase I/genética , Doença da Deficiência da Carbamoil-Fosfato Sintase I/metabolismo , Doença da Deficiência da Carbamoil-Fosfato Sintase I/terapia , Modelos Animais de Doenças , Feminino , Técnicas de Transferência de Genes , Glutamato-Amônia Ligase/metabolismo , Hiperamonemia/metabolismo , Hiperamonemia/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Especificidade de Órgãos/genética
5.
J Inherit Metab Dis ; 42(6): 1097-1104, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-30671986

RESUMO

(Macro)autophagy/autophagy is a highly regulated lysosomal degradative process by which cells recycle their own nutrients, such as amino acids and other metabolites, to be reused in different biosynthetic pathways. Ammonia is a diffusible compound generated daily from catabolism of nitrogen-containing molecules and from gastrointestinal microbiome. Ammonia homeostasis is tightly controlled in humans and ammonia is efficiently converted by the healthy liver into non-toxic urea (through ureagenesis) and glutamine (through glutamine synthetase). Impaired ammonia detoxification leads to systemic hyperammonemia, a life-threatening condition resulting in detrimental effects on central nervous system. Here, we review current understanding on the role of ammonia in modulation of autophagy and the potential implications in the pathogenesis and treatment of disorders with hyperammonemia.


Assuntos
Amônia/metabolismo , Autofagia/fisiologia , Hiperamonemia/etiologia , Animais , Glutamato-Amônia Ligase/metabolismo , Glutamina/metabolismo , Homeostase , Humanos , Hiperamonemia/metabolismo , Hiperamonemia/patologia , Fígado/metabolismo , Ureia/metabolismo , Distúrbios Congênitos do Ciclo da Ureia/complicações , Distúrbios Congênitos do Ciclo da Ureia/metabolismo , Distúrbios Congênitos do Ciclo da Ureia/patologia
6.
Autophagy ; 14(7): 1273-1275, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30035657

RESUMO

Ammonia is a highly neurotoxic metabolite that is efficiently converted into urea or glutamine. During liver failure due to hepatocellular dysfunction or in inherited deficiencies of urea cycle enzymes, ammonia clearance is impaired resulting in systemic hyperammonemia and hepatic encephalopathy that can rapidly progress into coma and death if left untreated. Because available therapeutic options are often unsatisfactory, the development of effective therapies for hyperammonemia is highly needed. Here, we review our recent findings on the role of hepatic macroautophagy/autophagy in ammonia detoxification. We found that during hyperammonemia, ammonia-induced depletion of liver alpha-ketoglutarate and its consequent inhibition of the mechanistic target of rapamycin kinase complex 1 results in autophagy induction. Metabolite recycling induced by enhanced hepatic autophagy increases the efficiency of ammonia detoxification by furnishing key urea cycle intermediates and ATP, and stimulating ureagenesis. Moreover, autophagy enhancement by liver-directed gene transfer of the master regulator of autophagy TFEB (transcription factor EB) or treatments with the autophagy enhancers rapamycin and Tat-beclin 1 improve ammonia detoxification during hyperammonemia occurring as a consequence of either acquired or inherited diseases.


Assuntos
Autofagia , Hiperamonemia , Amônia , Humanos , Fígado , Ureia
7.
Proc Natl Acad Sci U S A ; 115(2): 391-396, 2018 01 09.
Artigo em Inglês | MEDLINE | ID: mdl-29279371

RESUMO

Ammonia is a potent neurotoxin that is detoxified mainly by the urea cycle in the liver. Hyperammonemia is a common complication of a wide variety of both inherited and acquired liver diseases. If not treated early and thoroughly, it results in encephalopathy and death. Here, we found that hepatic autophagy is critically involved in systemic ammonia homeostasis by providing key urea-cycle intermediates and ATP. Hepatic autophagy is triggered in vivo by hyperammonemia through an α-ketoglutarate-dependent inhibition of the mammalian target of rapamycin complex 1, and deficiency of autophagy impairs ammonia detoxification. In contrast, autophagy enhancement by means of hepatic gene transfer of the master regulator of autophagy transcription factor EB or treatments with the autophagy enhancers rapamycin and Tat-Beclin-1 increased ureagenesis and protected against hyperammonemia in a variety of acute and chronic hyperammonemia animal models, including acute liver failure and ornithine transcarbamylase deficiency, the most frequent urea-cycle disorder. In conclusion, hepatic autophagy is an important mechanism for ammonia detoxification because of its support of urea synthesis, and its enhancement has potential for therapy of both primary and secondary causes of hyperammonemia.


Assuntos
Autofagia , Hiperamonemia/metabolismo , Fígado/metabolismo , Ureia/metabolismo , Amônia/metabolismo , Animais , Humanos , Masculino , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos
8.
Hepatology ; 66(1): 124-135, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28295475

RESUMO

α1 -Antitrypsin (AAT) deficiency is one of the most common genetic disorders and the liver disease due to the Z mutant of AAT (ATZ) is a prototype of conformational disorder due to protein misfolding with consequent aberrant intermolecular protein aggregation. In the present study, we found that livers of PiZ transgenic mice expressing human ATZ have altered expression of a network of hepatocyte transcriptional factors, including hepatocyte nuclear factor-4α, that is early down-regulated and induces a transcriptional repression of ATZ expression. Reduced hepatocyte nuclear factor-4α was associated with activation of ß-catenin, which regulates liver zonation. Livers of PiZ mice and human patients with AAT deficiency were both found to have a severe perturbation of liver zonation. Functionally, PiZ mice showed a severe defect of ureagenesis, as shown by increased baseline ammonia, and reduced urea production and survival after an ammonia challenge. Down-regulation of hepatocyte nuclear factor-4α expression and defective zonation in livers have not been recognized so far as features of the liver disease caused by ATZ and are likely involved in metabolic disturbances and in the increased risk of hepatocellular carcinoma in patients with AAT deficiency. CONCLUSION: The findings of this study are consistent with the concept that abnormal AAT protein conformation and intrahepatic accumulation have broad effects on metabolic liver functions. (Hepatology 2017;66:124-135).


Assuntos
Carcinoma Hepatocelular/genética , Regulação Neoplásica da Expressão Gênica , Fator 4 Nuclear de Hepatócito/genética , Neoplasias Hepáticas/patologia , Deficiência de alfa 1-Antitripsina/genética , Envelhecimento/genética , Análise de Variância , Animais , Carcinoma Hepatocelular/patologia , Modelos Animais de Doenças , Regulação para Baixo , Humanos , Neoplasias Hepáticas/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mutação , Distribuição Aleatória , Estatísticas não Paramétricas , Deficiência de alfa 1-Antitripsina/patologia
9.
Hepatology ; 64(2): 535-48, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-26999313

RESUMO

UNLABELLED: The adenoviral gene transfer of human aquaporin-1 (hAQP1) water channels to the liver of 17α-ethinylestradiol-induced cholestatic rats improves bile flow, in part by enhancing canalicular hAQP1-mediated osmotic water secretion. To gain insight into the mechanisms of 17α-ethinylestradiol cholestasis improvement, we studied the biliary output of bile salts (BS) and the functional expression of the canalicular BS export pump (BSEP; ABCB11). Adenovector encoding hAQP1 (AdhAQP1) or control vector was administered by retrograde intrabiliary infusion. AdhAQP1-transduced cholestatic rats increased the biliary output of major endogenous BS (50%-80%, P < 0.05) as well as that of taurocholate administered in choleretic or trace radiolabel amounts (around 60%, P < 0.05). Moreover, liver transduction with AdhAQP1 normalized serum BS levels, otherwise markedly elevated in cholestatic animals. AdhAQP1 treatment was unable to improve BSEP protein expression in cholestasis; however, its transport activity, assessed by adenosine triphosphate-dependent taurocholate transport in canalicular membrane vesicles, was induced by 90% (P < 0.05). AdhAQP1 administration in noncholestatic rats induced no significant changes in either biliary BS output or BSEP activity. Canalicular BSEP, mostly present in raft (high cholesterol) microdomains in control rats, was largely found in nonraft (low cholesterol) microdomains in cholestasis. Considering that BSEP activity directly depends on canalicular membrane cholesterol content, decreased BSEP presence in rafts may contribute to BSEP activity decline in 17α-ethinylestradiol cholestasis. In AdhAQP1-transduced cholestatic rats, BSEP showed a canalicular microdomain distribution similar to that of control rats, which provides an explanation for the improved BSEP activity. CONCLUSION: Hepatocyte canalicular expression of hAQP1 through adenoviral gene transfer promotes biliary BS output by modulating BSEP activity in estrogen-induced cholestasis, a novel finding that might help us to better understand and treat cholestatic disorders. (Hepatology 2016;64:535-548).


Assuntos
Transportadores de Cassetes de Ligação de ATP/metabolismo , Aquaporina 1/genética , Ácidos e Sais Biliares/metabolismo , Colestase/terapia , Terapia Genética , Membro 11 da Subfamília B de Transportadores de Cassetes de Ligação de ATP , Animais , Aquaporina 1/metabolismo , Ácidos e Sais Biliares/sangue , Canalículos Biliares/metabolismo , Colestase/induzido quimicamente , Etinilestradiol , Hepatócitos/metabolismo , Humanos , Masculino , Ratos Wistar
10.
Biochem Cell Biol ; 93(4): 417-20, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26194146

RESUMO

It has been proposed that, during metabolic acidosis, the liver downregulates mitochondrial ammonia detoxification via ureagenesis, a bicarbonate-consuming process. Since we previously demonstrated that hepatocyte mitochondrial aquaporin-8 channels (mtAQP8) facilitate the uptake of ammonia and its metabolism into urea, we studied whether mtAQP8 is involved in the liver adaptive response to acidosis. Primary cultured rat hepatocytes were adapted to acidosis by exposing them to culture medium at pH 7.0 for 40 h. Control cells were exposed to pH 7.4. Hepatocytes exposed to acid medium showed a decrease in mtAQP8 protein expression (-30%, p < 0.05). Ureagenesis from ammonia was assessed by incubating the cells with (15)N-labeled ammonia and measuring (15)N-labeled urea synthesis by nuclear magnetic resonance. Reduced ureagenesis was found in acidified hepatocytes (-31%, p < 0.05). In vivo studies in rats subjected to 7 days acidosis also showed decreased protein expression of hepatic mtAQP8 (-50%, p < 0.05) and reduced liver urea content (-35%; p < 0.05). In conclusion, our in vitro and in vivo data suggest that hepatic mtAQP8 expression is downregulated in acidosis, a mechanism that may contribute to decreased ureagenesis from ammonia in response to acidosis.


Assuntos
Acidose/metabolismo , Amônia/metabolismo , Aquaporinas/metabolismo , Regulação para Baixo , Mitocôndrias/metabolismo , Ureia/metabolismo , Animais , Células Cultivadas , Masculino , Ratos , Ratos Wistar
11.
FEBS Lett ; 588(9): 1686-91, 2014 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-24642373

RESUMO

We recently reported that hepatocyte mitochondrial aquaporin-8 (mtAQP8) channels facilitate the uptake of ammonia and its metabolism into urea. Here we studied the effect of bacterial lipopolysaccharides (LPS) on ammonia-derived ureagenesis. In LPS-treated rats, hepatic mtAQP8 protein expression and diffusional ammonia permeability (measured utilizing ammonia analogues) of liver inner mitochondrial membranes were downregulated. NMR studies using 15N-labeled ammonia indicated that basal and glucagon-induced ureagenesis from ammonia were significantly reduced in hepatocytes from LPS-treated rats. Our data suggest that hepatocyte mtAQP8-mediated ammonia removal via ureagenesis is impaired by LPS, a mechanism potentially relevant to the molecular pathogenesis of defective hepatic ammonia detoxification in sepsis.


Assuntos
Amônia/metabolismo , Aquaporinas/metabolismo , Hepatócitos/metabolismo , Lipopolissacarídeos/farmacologia , Mitocôndrias Hepáticas/metabolismo , Ureia/metabolismo , Animais , Transporte Biológico , Células Cultivadas , Glucagon/fisiologia , Hepatócitos/imunologia , Masculino , Metilaminas/metabolismo , Mitocôndrias Hepáticas/imunologia , Ratos , Ratos Wistar
12.
Clin Sci (Lond) ; 125(11): 521-32, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23642207

RESUMO

We have reported previously that centrally applied ET (endothelin)-1 and ET-3 induce either choleresis or cholestasis depending on the dose. In the present study, we sought to establish the role of these endothelins in the short-term peripheral regulation of bile secretion in the rat. Intravenously infused endothelins induced significant choleresis in a dose-dependent fashion, ET-1 being more potent than ET-3. Endothelins (with the exception of a higher dose of ET-1) did not affect BP (blood pressure), portal venous pressure or portal blood flow. ET-1 and ET-3 augmented the biliary excretion of bile salts, glutathione and electrolytes, suggesting enhanced bile acid-dependent and -independent bile flows. ET-induced choleresis was mediated by ET(B) receptors coupled to NO and inhibited by truncal vagotomy, atropine administration and capsaicin perivagal application, supporting the participation of vagovagal reflexes. RT (reverse transcription)-PCR and Western blot analysis revealed ETA and ET(B) receptor expression in the vagus nerve. Endothelins, through ET(B) receptors, augmented the hepatocyte plasma membrane expression of Ntcp (Na⁺/taurocholate co-transporting polypeptide; Slc10a1), Bsep (bile-salt export pump; Abcb11), Mrp2 (multidrug resistance protein-2; Abcc2) and Aqp8 (aquaporin 8). Endothelins also increased the mRNAs of these transporters. ET-1 and ET-3 induced choleresis mediated by ET(B) receptors coupled to NO release and vagovagal reflexes without involving haemodynamic changes. Endothelin-induced choleresis seems to be caused by increased plasma membrane translocation and transcriptional expression of key bile transporters. These findings indicate that endothelins are able to elicit haemodynamic-independent biological effects in the liver and suggest that these peptides may play a beneficial role in pathophysiological situations where bile secretion is impaired.


Assuntos
Colestase/induzido quimicamente , Endotelina-1/farmacologia , Endotelina-3/farmacologia , Óxido Nítrico/fisiologia , Receptor de Endotelina B/fisiologia , Nervo Vago/efeitos dos fármacos , Animais , Bile/metabolismo , Pressão Sanguínea/efeitos dos fármacos , Colagogos e Coleréticos/farmacologia , Colestase/metabolismo , Hemodinâmica/efeitos dos fármacos , Óxido Nítrico/metabolismo , Ratos , Ratos Sprague-Dawley , Receptor de Endotelina B/metabolismo , Reflexo/efeitos dos fármacos , Fluxo Sanguíneo Regional/efeitos dos fármacos , Vagotomia , Nervo Vago/metabolismo , Nervo Vago/fisiologia
13.
Hepatology ; 57(5): 2061-71, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23299935

RESUMO

UNLABELLED: Hepatocyte mitochondrial ammonia detoxification via ureagenesis is critical for the prevention of hyperammonemia and hepatic encephalopathy. Aquaporin-8 (AQP8) channels facilitate the membrane transport of ammonia. Because AQP8 is expressed in hepatocyte inner mitochondrial membranes (IMMs), we studied whether mitochondrial AQP8 (mtAQP8) plays a role in ureagenesis from ammonia. Primary cultured rat hepatocytes were transfected with small interfering RNAs (siRNAs) targeting two different regions of the rat AQP8 molecule or with scrambled control siRNA. After 48 hours, the levels of mtAQP8 protein decreased by approximately 80% (P < 0.05) without affecting cell viability. mtAQP8 knockdown cells in the presence of ammonium chloride showed a decrease in ureagenesis of approximately 30% (P < 0.05). Glucagon strongly stimulated ureagenesis in control hepatocytes (+120%, P < 0.05) but induced no significant stimulation in mtAQP8 knockdown cells. Contrarily, mtAQP8 silencing induced no significant change in basal and glucagon-induced ureagenesis when glutamine or alanine was used as a source of nitrogen. Nuclear magnetic resonance studies using 15N-labeled ammonia confirmed that glucagon-induced 15N-labeled urea synthesis was markedly reduced in mtAQP8 knockdown hepatocytes (-90%, P < 0.05). In vivo studies in rats showed that under glucagon-induced ureagenesis, hepatic mtAQP8 protein expression was markedly up-regulated (+160%, P < 0.05). Moreover, transport studies in liver IMM vesicles showed that glucagon increased the diffusional permeability to the ammonia analog [(14) C]methylamine (+80%, P < 0.05). CONCLUSION: Hepatocyte mtAQP8 channels facilitate the mitochondrial uptake of ammonia and its metabolism into urea, mainly under glucagon stimulation. This mechanism may be relevant to hepatic ammonia detoxification and in turn, avoid the deleterious effects of hyperammonemia.


Assuntos
Amônia/metabolismo , Aquaporinas/metabolismo , Hepatócitos/metabolismo , Inativação Metabólica/fisiologia , Mitocôndrias Hepáticas/metabolismo , Ureia/metabolismo , Animais , Aquaporinas/efeitos dos fármacos , Aquaporinas/genética , Células Cultivadas , Glucagon/farmacologia , Hepatócitos/patologia , Masculino , Membranas Mitocondriais/metabolismo , Modelos Animais , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/fisiologia , Ratos , Ratos Wistar , Transfecção , Regulação para Cima/efeitos dos fármacos
14.
Biochem Biophys Res Commun ; 393(2): 217-21, 2010 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-20132793

RESUMO

Aquaporin-8 (AQP8) is a membrane channel permeable to water and ammonia. As AQP8 is expressed in the inner mitochondrial membrane of several mammalian tissues, we studied the effect of the AQP8 expression on the mitochondrial transport of ammonia. Recombinant rat AQP8 was expressed in the yeast Saccharomyces cerevisiae. The presence of AQP8 in the inner membrane of yeast mitochondria was demonstrated by subcellular fractionation and immunoblotting analysis. The ammonia transport was determined in isolated mitochondria by stopped flow light scattering using formamide as ammonia analog. We found that the presence of AQP8 increased by threefold mitochondrial formamide transport. AQP8-facilitated mitochondrial formamide transport in rat native tissue was confirmed in liver (a mitochondrial AQP8-expressing tissue) vs. brain (a mitochondrial AQP8 non-expressing tissue). Comparative studies indicated that the AQP8-mediated mitochondrial movement of formamide was markedly higher than that of water. Together, our data suggest that ammonia diffusional transport is a major function for mitochondrial AQP8.


Assuntos
Amônia/metabolismo , Aquaporinas/metabolismo , Mitocôndrias/metabolismo , Animais , Aquaporinas/genética , Transporte Biológico , Encéfalo/metabolismo , Formamidas/metabolismo , Mitocôndrias Hepáticas/metabolismo , Ratos , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Água/metabolismo
15.
Am J Physiol Regul Integr Comp Physiol ; 296(4): R1274-81, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19193945

RESUMO

Glucagon stimulates the vesicle trafficking of aquaporin-8 (AQP8) water channels to the rat hepatocyte canalicular membranes, a process thought to be relevant to glucagon-induced bile secretion. In this study, we investigated whether glucagon is able to modulate the gene expression of hepatocyte AQP8. Glucagon was administered to rats at 0.2 mg/100 g body wt ip in 2, 3, or 6 equally spaced doses for 8, 16, and 36 h, respectively. Immunoblotting analysis showed that hepatic 34-kDa AQP8 was significantly increased by 79 and 107% at 16 and 36 h, respectively. Hepatic AQP9 protein expression remained unaltered. AQP8 mRNA expression, assessed by real-time PCR, was not modified over time, suggesting a posttranscriptional mechanism of AQP8 protein increase. Glucagon effects on AQP8 were directly studied in primary cultured rat hepatocytes. Immunoblotting and confocal immunofluorescence microscopy confirmed the specific glucagon-induced AQP8 upregulation. The RNA polymerase II inhibitor actinomycin D was unable to prevent glucagon effect, providing additional support to the nontranscriptional upregulation of AQP8. Cycloheximide also showed no effect, suggesting that glucagon-induced AQP8 expression does not depend on protein synthesis but rather on protein degradation. Inhibitory experiments suggest that a reduced calpain-mediated AQP8 proteolysis could be involved. The action of glucagon on hepatocyte AQP8 was mimicked by dibutyryl cAMP and suppressed by PKA or phosphatidylinositol-3-kinase (PI3K) inhibitors. In conclusion, our data suggest that glucagon induces the gene expression of rat hepatocyte AQP8 by reducing its degradation, a process that involves cAMP-PKA and PI3K signal pathways.


Assuntos
Aquaporinas/metabolismo , Glucagon/metabolismo , Hepatócitos/metabolismo , Animais , Aquaporinas/genética , Células Cultivadas , AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/antagonistas & inibidores , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Glucagon/administração & dosagem , Hepatócitos/efeitos dos fármacos , Hepatócitos/enzimologia , Injeções Intraperitoneais , Masculino , Inibidores da Síntese de Ácido Nucleico/farmacologia , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Inibidores de Proteases/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Processamento de Proteína Pós-Traducional , Inibidores da Síntese de Proteínas/farmacologia , RNA Mensageiro/metabolismo , Ratos , Ratos Wistar , Fatores de Tempo , Regulação para Cima
16.
Am J Physiol Gastrointest Liver Physiol ; 296(1): G93-100, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18948439

RESUMO

Aquaporin-8 (AQP8) water channels, which are expressed in rat hepatocyte bile canalicular membranes, are involved in water transport during bile formation. Nevertheless, there is no conclusive evidence that AQP8 mediates water secretion into the bile canaliculus. In this study, we directly evaluated whether AQP8 gene silencing by RNA interference inhibits canalicular water secretion in the human hepatocyte-derived cell line, HepG2. By RT-PCR and immunoblotting we found that HepG2 cells express AQP8 and by confocal immunofluorescence microscopy that it is localized intracellularly and on the canalicular membrane, as described in rat hepatocytes. We also verified the expression of AQP8 in normal human liver. Forty-eight hours after transfection of HepG2 cells with RNA duplexes targeting two different regions of human AQP8 molecule, the levels of AQP8 protein specifically decreased by 60-70%. We found that AQP8 knockdown cells showed a significant decline in the canalicular volume of approximately 70% (P < 0.01), suggesting an impairment in the basal (nonstimulated) canalicular water movement. We also found that the decreased AQP8 expression inhibited the canalicular water transport in response either to an inward osmotic gradient (-65%, P < 0.05) or to the bile secretory agonist dibutyryl cAMP (-80%, P < 0.05). Our data suggest that AQP8 plays a major role in water transport across canalicular membrane of HepG2 cells and support the notion that defective expression of AQP8 causes bile secretory dysfunction in human hepatocytes.


Assuntos
Aquaporinas/metabolismo , Canalículos Biliares/metabolismo , Técnicas de Silenciamento de Genes , Hepatócitos/metabolismo , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Água/metabolismo , Aquaporinas/genética , Canalículos Biliares/efeitos dos fármacos , Linhagem Celular Tumoral , CMP Cíclico/análogos & derivados , CMP Cíclico/farmacologia , Hepatócitos/efeitos dos fármacos , Humanos , Microscopia Confocal , Osmose , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Tempo , Transfecção
17.
World J Gastroenterol ; 14(46): 7059-67, 2008 Dec 14.
Artigo em Inglês | MEDLINE | ID: mdl-19084912

RESUMO

This review focuses on current knowledge on hepatocyte aquaporins (AQPs) and their significance in bile formation and cholestasis. Canalicular bile secretion results from a combined interaction of several solute transporters and AQP water channels that facilitate water flow in response to the osmotic gradients created. During choleresis, hepatocytes rapidly increase their canalicular membrane water permeability by modulating the abundance of AQP8. The question was raised as to whether the opposite process, i.e. a decreased canalicular AQP8 expression would contribute to the development of cholestasis. Studies in several experimental models of cholestasis, such as extrahepatic obstructive cholestasis, estrogen-induced cholestasis, and sepsis-induced cholestasis demonstrated that the protein expression of hepatocyte AQP8 was impaired. In addition, biophysical studies in canalicular plasma membranes revealed decreased water permeability associated with AQP8 protein downregulation. The combined alteration in hepatocyte solute transporters and AQP8 would hamper the efficient coupling of osmotic gradients and canalicular water flow. Thus cholestasis may result from a mutual occurrence of impaired solute transport and decreased water permeability.


Assuntos
Aquaporinas/fisiologia , Colestase Intra-Hepática/fisiopatologia , Aquaporinas/metabolismo , Bile/metabolismo , Colestase Intra-Hepática/patologia , Hepatócitos/metabolismo , Hepatócitos/patologia , Humanos , Osmose/fisiologia
18.
Am J Physiol Gastrointest Liver Physiol ; 294(2): G567-75, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18174273

RESUMO

Although bacterial lipopolysaccharides (LPS) are known to cause cholestasis in sepsis, the molecular mechanisms accounting for this effect are only partially known. Because aquaporin-8 (AQP8) seems to facilitate the canalicular osmotic water movement during hepatocyte bile formation, we studied its gene and functional expression in LPS-induced cholestasis. By subcellular fractionation and immunoblotting analysis, we found that 34-kDa AQP8 was significantly decreased by 70% in plasma (canalicular) and intracellular (vesicular) liver membranes. However, expression and subcellular localization of hepatocyte sinusoidal AQP9 were unaffected. Immunohistochemistry for liver AQPs confirmed these observations. Osmotic water permeability (P(f)) of canalicular membranes, measured by stopped-flow spectrophotometry, was significantly reduced (65 +/- 1 vs. 49 +/- 1 microm/s) by LPS, consistent with defective canalicular AQP8 functional expression. By Northern blot analysis, we found that 1.5-kb AQP8 mRNA expression was increased by 80%, suggesting a posttranscriptional mechanism of protein reduction. The tumor necrosis factor-alpha (TNF-alpha) receptor fusion protein TNFp75:Fc prevented the LPS-induced impairment of AQP8 expression and bile flow, suggesting the cytokine TNF-alpha as a major mediator of LPS effect. Accordingly, studies in hepatocyte primary cultures indicated that recombinant TNF-alpha downregulated AQP8. The effect of TNF-alpha was prevented by the lysosomal protease inhibitors leupeptin or chloroquine or by the proteasome inhibitors MG132 or lactacystin, suggesting a cytokine-induced AQP8 proteolysis. In conclusion, our data suggest that LPS induces the TNF-alpha-mediated posttranscriptional downregulation of AQP8 functional expression in hepatocytes, a mechanism potentially relevant to the molecular pathogenesis of sepsis-associated cholestasis.


Assuntos
Aquaporinas/fisiologia , Colestase/etiologia , Colestase/fisiopatologia , Lipopolissacarídeos/farmacologia , Fígado/metabolismo , Sepse/complicações , Sepse/fisiopatologia , Fator de Necrose Tumoral alfa/fisiologia , Animais , Northern Blotting , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Citocinas/sangue , Regulação para Baixo/efeitos dos fármacos , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Imuno-Histoquímica , Luz , Fígado/efeitos dos fármacos , Masculino , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Ratos , Ratos Wistar , Espalhamento de Radiação , Frações Subcelulares/efeitos dos fármacos , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Fator de Necrose Tumoral alfa/sangue
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...