Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 58
Filtrar
1.
Biotechnol Bioeng ; 118(11): 4331-4337, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34292591

RESUMO

Hepatic ammonia detoxification to urea is critical for the prevention of hyperammonemia and neurological damage. Hepatocyte mitochondrial aquaporin-8 (AQP8) channels have been involved in ammonia-derived ureagenesis. Herein, we studied whether the adenoviral gene transfer of human AQP8 (hAQP8) to hepatocyte mitochondria enhances ammonia conversion to urea. Using primary cultured rat hepatocytes, we first confirmed the mitochondrial expression of hAQP8 and then, using unlabeled or 15 N-labeled ammonia, we demonstrated that the urea synthesis was significantly enhanced in hAQP8-transduced hepatocytes. Studies using isolated hAQP8-expressing mitochondria also showed an increased ammonia metabolism. hAQP8 transduction was able to recover the impaired ammonia-derived ureagenesis in hepatotoxin-treated hepatocytes. Our data suggest that mitochondrially-expressed hAQP8 enhances and improves hepatocyte ammonia conversion to urea, a finding with potential therapeutic implications for liver disease with impaired ammonia detoxification.


Assuntos
Amônia/metabolismo , Aquaporinas/biossíntese , Hepatócitos/metabolismo , Transdução Genética , Ureia/metabolismo , Animais , Aquaporinas/genética , Humanos , Ratos
2.
J Pharmacol Exp Ther ; 371(3): 713-717, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31570498

RESUMO

The purpose of this minireview is to show that a new paradigm is developing regarding hepatic bile flow. The focus thus far has been on carrier-mediated transport of bile acids and other solutes, such as glutathione, which create an osmotic gradient for the transcellular and paracellular flow of water into canaliculi. In addition to the physicochemical properties of bile acids, which govern the osmotic gradient, data now exist showing that the tight junctions governing paracellular water flow and Aquaporin-8 water channels governing transcellular water flow are regulated independently. Thus, the rate of water flow into the canaliculus in response to bile acid transport is variable and determines canalicular bile acid concentration, which affects the production and solubilization of cholesterol-lecithin vesicles. These new considerations modify thinking regarding the occurrence of cholestasis and its progression and reorient the design of experimental studies that can distinguish the different determinants of bile flow. SIGNIFICANCE STATEMENT: The paradigm that water flow into the canaliculus is determined only by the rate of carrier-mediated transport has been challenged recently by the changes that occur in hepatic bile composition in the Claudin-2 knockout mouse and with the cholestatic effect of estradiol 17ß-d-glucuronide. Thus, a respective reduction in paracellular or transcellular canalicular water flow, probably via Aquaporin 8, has no significant effect on bile acid excretion.


Assuntos
Canalículos Biliares/metabolismo , Bile/fisiologia , Água Corporal/metabolismo , Animais , Aquaporinas/fisiologia , Ácidos e Sais Biliares/metabolismo , Transporte Biológico , Claudina-2/fisiologia , Estradiol/farmacologia , Humanos , Camundongos , Concentração Osmolar
3.
Biochim Biophys Acta Mol Basis Dis ; 1864(4 Pt A): 1072-1085, 2018 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-29355600

RESUMO

Impaired canalicular secretion due to increased endocytosis and intracellular retention of canalicular transporters such as BSEP and MRP2 is a main, common pathomechanism of cholestasis. Nevertheless, the mechanisms governing this process are unknown. We characterized this process in estradiol 17 ß-d-glucuronide (E17G)-induced cholestasis, an experimental model which partially mimics pregnancy-induced cholestasis. Inhibitors of clathrin-mediated endocytosis (CME) such as monodansylcadaverine (MDC) or K+ depletion, but not the caveolin-mediated endocytosis inhibitors filipin and genistein, prevented E17G-induced endocytosis of BSEP and MRP2, and the associated impairment of activity of these transporters in isolated rat hepatocyte couplets (IRHC). Immunofluorescence and confocal microscopy studies showed that, in E17G-treated IRHC, there was a significant increase in the colocalization of MRP2 with clathrin, AP2, and Rab5, three essential members of the CME machinery. Knockdown of AP2 by siRNA in sandwich-cultured rat hepatocytes completely prevented E17G-induced endocytosis of BSEP and MRP2. MDC significantly prevented this endocytosis, and the impairment of bile flow and biliary secretion of BSEP and MRP2 substrates, in isolated and perfused livers. BSEP and MRP2, which were mostly present in raft (caveolin-enriched) microdomains in control rats, were largely found in non-raft (clathrin-enriched) microdomains in livers from E17G-treated animals, from where they can be readily recruited for CME. In conclusion, our findings show that CME is the mechanism responsible for the internalization of the canalicular transporters BSEP and MRP2 in E17G-induced cholestasis. The shift of these transporters from raft to non-raft microdomains could be a prerequisite for the transporters to be endocytosed under cholestatic conditions.


Assuntos
Colestase/metabolismo , Endocitose , Hepatócitos/metabolismo , Fígado/metabolismo , Microdomínios da Membrana/metabolismo , Membro 11 da Subfamília B de Transportadores de Cassetes de Ligação de ATP/metabolismo , Transportadores de Cassetes de Ligação de ATP/metabolismo , Animais , Colestase/induzido quimicamente , Colestase/patologia , Modelos Animais de Doenças , Feminino , Hepatócitos/patologia , Fígado/patologia , Microdomínios da Membrana/patologia , Ratos , Ratos Wistar
4.
IUBMB Life ; 69(12): 978-984, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-29087027

RESUMO

Lipopolysaccharides (LPS) are known to cause cholestasis in sepsis. There is evidence that a defective expression of canalicular aquaporin water channels contributes to bile secretory failure in LPS-induced cholestasis. Thus, we studied whether the hepatic adenovirus-mediated transfer of human aquaporin-1 gene (haqp1) can improve the cholestasis induced by LPS. Adenoviral vector encoding hAQP1 (AdhAQP1) or control vector was administered to rats by retrograde intrabiliary infusion. Hepatocyte canalicular hAQP1 expression was assessed by liver immunostaining and immunoblotting in purified plasma membranes. LPS reduced bile flow and biliary bile acid excretion by 30% and 45%, respectively. AdhAQP1-treatment normalized both bile flow and biliary bile acid excretion in LPS-induced cholestasis. Moreover, markedly elevated serum bile acid levels in cholestatic rats, were also normalized with the AdhAQP1 hepatic transduction. Bile flow and serum or biliary bile acids in normal rats were not significantly altered by AdhAQP1. AdhAQP1 delivery unaffected the downregulated protein expression of canalicular bile salt export pump (BSEP/ABCB11) in cholestasis, but improved its transport activity restoring reduced canalicular cholesterol content. Our data suggest that the adenovirus-mediated hepatocyte hAQP1 expression improves LPS-induced cholestasis in rats by stimulating the BSEP/ABCB11-mediated biliary bile acid excretion; a finding that might contribute to the understanding and treatment of sepsis-associated cholestatic diseases. © 2017 IUBMB Life, 69(12):978-984, 2017.


Assuntos
Membro 11 da Subfamília B de Transportadores de Cassetes de Ligação de ATP/genética , Adenoviridae/genética , Aquaporina 1/genética , Ácidos e Sais Biliares/metabolismo , Colestase/terapia , Hepatócitos/metabolismo , Sepse/terapia , Membro 11 da Subfamília B de Transportadores de Cassetes de Ligação de ATP/metabolismo , Adenoviridae/metabolismo , Animais , Aquaporina 1/metabolismo , Transporte Biológico , Colestase/induzido quimicamente , Colestase/genética , Colestase/patologia , Colesterol/metabolismo , Expressão Gênica , Terapia Genética/métodos , Vetores Genéticos/química , Vetores Genéticos/metabolismo , Hepatócitos/patologia , Humanos , Lipopolissacarídeos , Fígado/metabolismo , Fígado/patologia , Masculino , Ratos , Ratos Wistar , Reologia , Sepse/induzido quimicamente , Sepse/genética , Sepse/patologia , Transgenes
5.
IUBMB Life ; 69(5): 341-346, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-28322010

RESUMO

Hepatocyte mitochondrial aquaporin-8 (mtAQP8) works as a multifunctional membrane channel protein that facilitates the uptake of ammonia for its detoxification to urea as well as the mitochondrial release of hydrogen peroxide. Since early oligonucleotide microarray studies in liver of cholesterol-fed mice showed an AQP8 downregulation, we tested whether alterations of cholesterol content per se modulate mtAQP8 expression in human hepatocyte-derived Huh-7 cells. Cholesterol loading with methyl-ß-cyclodextrin (mßCD):cholesterol complexes downregulated the proteolytic activation of cholesterol-responsive sterol regulatory element-binding protein (SREBP) transcriptions factors 1 and 2, and the expression of the target gene 3-hydroxy-3-methylglutaryl-CoA reductase (HMGCR). Under such conditions, mtAQP8 mRNA and protein expressions were significantly reduced. In contrast, cholesterol depletion using mßCD alone increased SREBP-1 and 2 activation and upregulated HMGCR and mtAQP8 mRNA and protein expressions. The results suggest that cholesterol can regulate transcriptionally human hepatocyte mtAQP8 expression likely via SREBPs. The functional implications of our findings are discussed. © 2017 IUBMB Life, 69(5):341-346, 2017.


Assuntos
Aquaporinas/metabolismo , Colesterol/metabolismo , Hepatócitos/metabolismo , Aquaporinas/genética , Linhagem Celular , Colesterol/farmacocinética , Hepatócitos/efeitos dos fármacos , Humanos , Hidroximetilglutaril-CoA Redutases/metabolismo , Mitocôndrias/metabolismo , Proteína de Ligação a Elemento Regulador de Esterol 1/metabolismo , Proteína de Ligação a Elemento Regulador de Esterol 2/metabolismo , beta-Ciclodextrinas/farmacocinética
6.
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
7.
Anal Biochem ; 495: 1-2, 2016 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-26656923

RESUMO

Lipid-based transfection reagents are widely used for delivery of small interfering RNA into cells. We examined whether the commonly used commercial transfection reagents DharmaFECT-4 and Lipofectamine 2000 can interfere with lipid metabolism by studying cholesterogenesis. Cholesterol de novo synthesis from [(14)C]acetate was assessed in human hepatocyte-derived Huh-7 cells. The results revealed that DharmaFECT, but not Lipofectamine, markedly inhibited cholesterol biosynthesis by approximately 70%. Cell viability was not significantly altered. These findings suggest that caution is required in the choice of certain lipid-based transfection reagents for gene silencing experiments, particularly when assessing cholesterol metabolism.


Assuntos
Colesterol/biossíntese , Lipídeos/química , Transfecção , Linhagem Celular , Humanos , Indicadores e Reagentes/farmacologia , L-Lactato Desidrogenase/metabolismo , Metabolismo dos Lipídeos/efeitos dos fármacos , Lipídeos/farmacologia , RNA Interferente Pequeno/metabolismo
8.
Bioorg Med Chem Lett ; 26(16): 3955-7, 2016 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-27422336

RESUMO

Levoglucosenone has been used as template for the synthesis of a wide variety of compounds with an impressive structural variability. However, scarce work has been done regarding the generation of new bioactive entities. Here we report the cytotoxic effect of levoglucosenone and some related derivatives against hepatocarcinoma cell lines. Compounds were obtained in only one synthetic step and one of them showed an activity within the range of IC50 values of cisplatin, a frequently administered chemotherapy drug.


Assuntos
Antineoplásicos/química , Compostos Bicíclicos Heterocíclicos com Pontes/química , Glucose/análogos & derivados , Antineoplásicos/síntese química , Antineoplásicos/toxicidade , Compostos Bicíclicos Heterocíclicos com Pontes/síntese química , Compostos Bicíclicos Heterocíclicos com Pontes/toxicidade , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Glucose/síntese química , Glucose/química , Glucose/toxicidade , Humanos , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patologia
9.
Int J Mol Sci ; 17(7)2016 Jul 09.
Artigo em Inglês | MEDLINE | ID: mdl-27409609

RESUMO

Aquaporins (AQPs) are proteinaceous channels widespread in nature where they allow facilitated permeation of water and uncharged through cellular membranes. AQPs play a number of important roles in both health and disease. This review focuses on the most recent advances and research trends regarding the expression and modulation, as well as physiological and pathophysiological functions of AQPs in hepatocytes and Sertoli cells (SCs). Besides their involvement in bile formation, hepatocyte AQPs are involved in maintaining energy balance acting in hepatic gluconeogenesis and lipid metabolism, and in critical processes such as ammonia detoxification and mitochondrial output of hydrogen peroxide. Roles are played in clinical disorders including fatty liver disease, diabetes, obesity, cholestasis, hepatic cirrhosis and hepatocarcinoma. In the seminiferous tubules, particularly in SCs, AQPs are also widely expressed and seem to be implicated in the various stages of spermatogenesis. Like in hepatocytes, AQPs may be involved in maintaining energy homeostasis in these cells and have a major role in the metabolic cooperation established in the testicular tissue. Altogether, this information represents the mainstay of current and future investigation in an expanding field.


Assuntos
Aquaporinas/metabolismo , Hepatócitos/metabolismo , Células de Sertoli/metabolismo , Metabolismo Energético , Hepatócitos/citologia , Humanos , Infertilidade Masculina/metabolismo , Infertilidade Masculina/patologia , Masculino , Mitocôndrias/metabolismo , Hepatopatia Gordurosa não Alcoólica/metabolismo , Hepatopatia Gordurosa não Alcoólica/patologia , Espécies Reativas de Oxigênio/metabolismo , Células de Sertoli/citologia
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.
Apoptosis ; 19(5): 851-9, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24415197

RESUMO

We previously found that mitochondrial aquaporin-8 (mtAQP8) channels facilitate mitochondrial H2O2 release in human hepatoma HepG2 cells and that their knockdown causes oxidant-induced mitochondrial dysfunction and loss of viability. Here, we studied whether apoptosis or necrosis is involved as the mode of cell death. We confirmed that siRNA-induced mtAQP8 knockdown significantly decreased HepG2 viability by MTT assay, LDH leakage, and trypan blue exclusion test. Analysis of mitochondrial proapoptotic Bax-to-antiapoptotic BclXL ratio, mitochondrial cytochrome c release and caspase-3 activation showed no alterations in mtAQP8-knockdown cells. This indicates a primary mechanism of cell death other than the intrinsic mitochondrial apoptotic pathway. Thus, nuclear staining with DAPI did not reveal any increase of apoptotic features, i.e. chromatin condensation or nuclear fragmentation. Flow cytometry studies after double cell staining with annexin V and propidium iodide confirmed lack of apoptosis and suggested necrosis as the primary mechanism of death in mtAQP8-knockdown HepG2 cells. Necrosis was further supported by the increased nuclear delocalization and extracellular release of the High Mobility Group Box 1 protein. The knockdown of mtAQP8 in another human hepatoma-derived cell line, i.e. HuH-7 cells, also induced necrotic but not apoptotic death. Our data suggest that mtAQP8 knockdown induces necrotic cell death in human neoplastic hepatic cells, a finding that might be relevant to therapeutic strategies against hepatoma cells.


Assuntos
Apoptose , Aquaporinas/metabolismo , Carcinoma Hepatocelular/patologia , Neoplasias Hepáticas/patologia , Mitocôndrias/metabolismo , Aquaporinas/genética , Carcinoma Hepatocelular/metabolismo , Linhagem Celular Tumoral , Técnicas de Silenciamento de Genes , Humanos , Neoplasias Hepáticas/metabolismo , Necrose
12.
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
13.
Hepatology ; 65(1): 387-388, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27459635
14.
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
15.
Biol Cell ; 104(6): 342-51, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22316404

RESUMO

BACKGROUND INFORMATION: Lipolytic glycerol, released from adipocytes, flows through the bloodstream to the liver, where its utilisation in supplying hepatocyte gluconeogenesis is rate-limited by the permeation step. An aquaglyceroporin expressed in hepatocytes, aquaporin-9 (AQP9), has been often linked to liver uptake of glycerol. However, the truthfulness of this postulation and the potential existence of additional pathways of glycerol import by hepatocytes have never been assessed directly. Here, we define the identity and extent of liver glycerol transport and evaluate the correlation between hepatic AQP9 expression and glycerol permeability (P(gly) ) in AQP9(+/+) wild-type mice in different nutritional states and circulating insulin levels. The liver P(gly) of AQP9 null mice is also assessed. RESULTS: By stopped-flow light scattering, facilitated diffusion of glycerol into hepatocytes was indicated by the low Arrhenius activation energy (3.5 kcal/mol) and strong inhibition by phloretin, an AQP9 blocker, that characterised the transport. Although fasting markedly increased hepatic AQP9, a straight parallelism was seen both in quantitative and time-space terms between P(gly) and AQP9 protein in AQP9(+/+) mice kept in fed or fasted/refed states. In line with these findings, the highest P(gly) (P(gly) ≈ 14.0 × 10(-6) cm/s at 20°C) at 18-h fasting coincided with the highest percent of phloretin inhibition (63%). Besides being markedly lower than that in AQP9(+/+) mice, the liver P(gly) of the AQP9 null mice did not increase during fasting. Reverse-transcription PCR analysis showed lack of compensation by AQP3 and AQP7, the other known murine glycerol facilitators, in AQP9 null mice. CONCLUSIONS: Overall, these results experimentally prove major functional significance for AQP9 in maximising liver glycerol import during states requiring increased glucose production. If any, alternative facilitated pathways would be of minor importance in transporting glucogenetic glycerol into hepatocytes during starvation. Refining the understanding of liver AQP9 in metabolic and energy homeostasis may reveal helpful for therapeutic purposes.


Assuntos
Aquaporinas/metabolismo , Glicerol/metabolismo , Fígado/metabolismo , Animais , Aquaporinas/análise , Aquaporinas/genética , Difusão Facilitada , Jejum , Deleção de Genes , Gluconeogênese , Hepatócitos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Permeabilidade
16.
Cells ; 12(11)2023 06 02.
Artigo em Inglês | MEDLINE | ID: mdl-37296655

RESUMO

We previously reported that, in cultured hepatocytes, mitochondrial aquaporin-8 (AQP8) channels facilitate the conversion of ammonia to urea and that the expression of human AQP8 (hAQP8) enhances ammonia-derived ureagenesis. In this study, we evaluated whether hepatic gene transfer of hAQP8 improves detoxification of ammonia to urea in normal mice as well as in mice with impaired hepatocyte ammonia metabolism. A recombinant adenoviral (Ad) vector encoding hAQP8, AdhAQP8, or a control Ad vector was administered via retrograde infusion into the bile duct of the mice. Hepatocyte mitochondrial expression of hAQP8 was confirmed using confocal immunofluorescence and immunoblotting. The normal hAQP8-transduced mice showed decreased plasma ammonia and increased liver urea. Enhanced ureagenesis was confirmed via the NMR studies assessing the synthesis of 15N-labeled urea from 15N-labeled ammonia. In separate experiments, we made use of the model hepatotoxic agent, thioacetamide, to induce defective hepatic metabolism of ammonia in mice. The adenovirus-mediated mitochondrial expression of hAQP8 was able to restore normal ammonemia and ureagenesis in the liver of the mice. Our data suggest that hAQP8 gene transfer to mouse liver improves detoxification of ammonia to urea. This finding could help better understand and treat disorders with defective hepatic ammonia metabolism.


Assuntos
Amônia , Aquaporinas , Humanos , Camundongos , Animais , Amônia/metabolismo , Ureia/metabolismo , Aquaporinas/genética , Aquaporinas/metabolismo , Fígado/metabolismo , Adenoviridae/genética , Adenoviridae/metabolismo
17.
J Cell Physiol ; 227(1): 160-71, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21374596

RESUMO

Hepatocytes are epithelial cells whose apical poles constitute the bile canaliculi. The establishment and maintenance of canalicular poles is a finely regulated process that dictates the efficiency of primary bile secretion. Protein kinase A (PKA) modulates this process at different levels. AKAP350 is an A-kinase anchoring protein that scaffolds protein complexes involved in modulating the dynamic structures of the Golgi apparatus and microtubule cytoskeleton, facilitating microtubule nucleation at this organelle. In this study, we evaluated whether AKAP350 is involved in the development of bile canaliculi-like structures in hepatocyte derived HepG2 cells. We found that AKAP350 recruits PKA to the centrosomes and Golgi apparatus in HepG2 cells. De-localization of AKAP350 from these organelles led to reduced apical cell polarization. A decrease in AKAP350 expression inhibited the formation of canalicular structures and impaired F-actin organization at canalicular poles. Furthermore, loss of AKAP350 expression led to diminished polarized expression of the p-glycoprotein (MDR1/ABCB1) at the apical "canalicular" membrane. AKAP350 knock down effects on canalicular structures formation and actin organization could be mimicked by inhibition of Golgi microtubule nucleation by depletion of CLIP associated proteins (CLASPs). Our data reveal that AKAP350 participates in mechanisms which determine the development of canalicular structures as well as accurate canalicular expression of distinct proteins and actin organization, and provide evidence on the involvement of Golgi microtubule nucleation in hepatocyte apical polarization.


Assuntos
Proteínas de Ancoragem à Quinase A/metabolismo , Canalículos Biliares/metabolismo , Canalículos Biliares/ultraestrutura , Polaridade Celular/fisiologia , Proteínas do Citoesqueleto/metabolismo , Hepatócitos/metabolismo , Hepatócitos/ultraestrutura , Citoesqueleto de Actina/metabolismo , Citoesqueleto de Actina/ultraestrutura , Centrossomo/metabolismo , Centrossomo/ultraestrutura , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Imunofluorescência , Complexo de Golgi/metabolismo , Complexo de Golgi/ultraestrutura , Células Hep G2 , Humanos , Immunoblotting , Microscopia Confocal , Microscopia de Fluorescência
18.
Am J Physiol Renal Physiol ; 303(3): F458-66, 2012 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-22622463

RESUMO

Mitochondrial ammonia synthesis in proximal tubules and its urinary excretion are key components of the renal response to maintain acid-base balance during metabolic acidosis. Since aquaporin-8 (AQP8) facilitates transport of ammonia and is localized in inner mitochondrial membrane (IMM) of renal proximal cells, we hypothesized that AQP8-facilitated mitochondrial ammonia transport in these cells plays a role in the response to acidosis. We evaluated whether mitochondrial AQP8 (mtAQP8) knockdown by RNA interference is able to impair ammonia excretion in the human renal proximal tubule cell line, HK-2. By RT-PCR and immunoblotting, we found that AQP8 is expressed in these cells and is localized in IMM. HK-2 cells were transfected with short-interfering RNA targeting human AQP8. After 48 h, the levels of mtAQP8 protein decreased by 53% (P < 0.05). mtAQP8 knockdown decreased the rate of ammonia released into culture medium in cells grown at pH 7.4 (-31%, P < 0.05) as well as in cells exposed to acid (-90%, P < 0.05). We also evaluated mtAQP8 protein expression in HK-2 cells exposed to acidic medium. After 48 h, upregulation of mtAQP8 (+74%, P < 0.05) was observed, together with higher ammonia excretion rate (+73%, P < 0.05). In vivo studies in NH(4)Cl-loaded rats showed that mtAQP8 protein expression was also upregulated after 7 days of acidosis in renal cortex (+51%, P < 0.05). These data suggest that mtAQP8 plays an important role in the adaptive response of proximal tubule to acidosis possibly facilitating mitochondrial ammonia transport.


Assuntos
Acidose/metabolismo , Aquaporinas/metabolismo , Túbulos Renais Proximais/metabolismo , Mitocôndrias/metabolismo , Animais , Animais Geneticamente Modificados , Aquaporinas/genética , Gasometria , Western Blotting , Linhagem Celular , Sobrevivência Celular , Humanos , Córtex Renal/metabolismo , Túbulos Renais Proximais/citologia , L-Lactato Desidrogenase/metabolismo , Masculino , Microscopia Confocal , Compostos de Amônio Quaternário/urina , Interferência de RNA , Ratos , Ratos Wistar , Reação em Cadeia da Polimerase em Tempo Real
19.
Biochimie ; 188: 16-19, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-33493534

RESUMO

We recently provided evidence suggesting that mitochondrial aquaporin-8 (mtAQP8), a channel protein able to conduct H2O2, is involved in the modulation of hepatocyte cholesterogenesis. To expand that study, we cultured human hepatocyte-derived Huh-7 cells in medium with lipoprotein-deficient serum (LPDS) to induce the de novo synthesis of cholesterol and fatty acids. We found that LPDS induced mtAQP8 expression and that AQP8 gene silencing significantly down-regulated the LPDS-induced synthesis of cholesterol and fatty acids as well as the expression of the corresponding key biosynthetic enzymes, 3-hydroxy-3-methylglutaryl-CoA reductase and fatty acid synthase. Our data further support a regulatory role of mtAQP8 in hepatocyte lipid homeostasis.


Assuntos
Aquaporinas/genética , Aquaporinas/metabolismo , Hepatócitos/metabolismo , Lipogênese/fisiologia , Mitocôndrias/metabolismo , Linhagem Celular Tumoral , Colesterol/biossíntese , Ácido Graxo Sintase Tipo I/metabolismo , Ácidos Graxos/biossíntese , Inativação Gênica , Homeostase , Humanos , Hidroximetilglutaril-CoA Redutases/metabolismo , Lipoproteínas/deficiência
20.
Biochimie ; 188: 12-15, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-33811938

RESUMO

Bile secretion by hepatocytes is an osmotic process. The output of bile salts and other organic anions (e.g. glutathione), through the bile salt transporter BSEP/ABCB11 and the organic anion transporter MRP2/ABCC2, respectively, are considered to be the major osmotic driving forces for water secretion into bile canaliculi mainly via aquaporin-8 (AQP8) channels. The down-regulated canalicular expression of these key solute transporters and AQP8 would be a primary event in the establishment of hepatocellular cholestasis. Recent studies in animal models of hepatocellular cholestasis show that the hepatic delivery of AdhAQP1, an adenovector encoding for the archetypical water channel human aquaporin-1 (hAQP1), improves bile secretion and restores to normal the elevated serum bile salt levels. AdhAQP1-transduced hepatocytes show that the canalicularly-expressed hAQP1 not only enhances osmotic membrane water permeability but also induces the transport activities of BSEP/ABCB11 and MRP2/ABCC2 by redistribution in canalicular cholesterol-rich microdomains likely through interactions with the cholesterol-binding protein caveolin-1. Thus, the hepatic gene transfer of hAQP1 improves the bile secretory failure in hepatocellular cholestasis by increasing both biliary output and choleretic efficiency of key osmotic solutes, such as, bile salts and glutathione. The study of hepatocyte aquaporins has provided new insights into the mechanisms of bile formation and cholestasis, and may lead to innovative treatments for cholestatic liver diseases.


Assuntos
Aquaporinas/genética , Colestase/genética , Colestase/terapia , Terapia Genética/métodos , Animais , Bile/metabolismo , Hepatócitos/metabolismo , Humanos , Proteína 2 Associada à Farmacorresistência Múltipla
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa