Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
2.
FEBS Lett ; 2024 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-38884367

RESUMEN

Inherited metabolic disorders (IMDs) are a growing group of genetic diseases caused by defects in enzymes that mediate cellular metabolism, often resulting in the accumulation of toxic substrates. The liver is a highly metabolically active organ that hosts several thousands of chemical reactions. As such, it is an organ frequently affected in IMDs. In this article, we review current approaches for liver-directed gene-based therapy aimed at metabolite detoxification in a variety of IMDs. Moreover, we discuss current unresolved challenges in gene-based therapies for IMDs.

3.
Nat Metab ; 6(7): 1294-1309, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38858597

RESUMEN

Downregulation of the urea cycle enzyme argininosuccinate synthase (ASS1) in multiple tumors is associated with a poor prognosis partly because of the metabolic diversion of cytosolic aspartate for pyrimidine synthesis, supporting proliferation and mutagenesis owing to nucleotide imbalance. Here, we find that prolonged loss of ASS1 promotes DNA damage in colon cancer cells and fibroblasts from subjects with citrullinemia type I. Following acute induction of DNA damage with doxorubicin, ASS1 expression is elevated in the cytosol and the nucleus with at least a partial dependency on p53; ASS1 metabolically restrains cell cycle progression in the cytosol by restricting nucleotide synthesis. In the nucleus, ASS1 and ASL generate fumarate for the succination of SMARCC1, destabilizing the chromatin-remodeling complex SMARCC1-SNF5 to decrease gene transcription, specifically in a subset of the p53-regulated cell cycle genes. Thus, following DNA damage, ASS1 is part of the p53 network that pauses cell cycle progression, enabling genome maintenance and survival. Loss of ASS1 contributes to DNA damage and promotes cell cycle progression, likely contributing to cancer mutagenesis and, hence, adaptability potential.


Asunto(s)
Argininosuccinato Sintasa , Núcleo Celular , Citosol , Daño del ADN , Proteína p53 Supresora de Tumor , Humanos , Proteína p53 Supresora de Tumor/metabolismo , Proteína p53 Supresora de Tumor/genética , Citosol/metabolismo , Argininosuccinato Sintasa/metabolismo , Argininosuccinato Sintasa/genética , Núcleo Celular/metabolismo , Ciclo Celular/genética
4.
Nat Commun ; 13(1): 5212, 2022 09 05.
Artículo en Inglés | MEDLINE | ID: mdl-36064721

RESUMEN

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.


Asunto(s)
Amoníaco , Carbamoil-Fosfato Sintasa (Amoniaco) , Hiperamonemia , Urea , Uridina Difosfato , Acetilglucosamina , Amoníaco/metabolismo , Animales , Biocatálisis , Carbamoil-Fosfato Sintasa (Amoniaco)/genética , Carbamoil-Fosfato Sintasa (Amoniaco)/metabolismo , Modelos Animales de Enfermedad , Glicosilación , Humanos , Hiperamonemia/genética , Hiperamonemia/metabolismo , Mamíferos/metabolismo , Ratones , N-Acetilglucosaminiltransferasas/genética , N-Acetilglucosaminiltransferasas/metabolismo , Acidemia Propiónica/genética , Acidemia Propiónica/metabolismo , Procesamiento Proteico-Postraduccional/genética , Urea/metabolismo , Uridina Difosfato/genética , Uridina Difosfato/metabolismo
5.
EMBO Mol Med ; 13(2): e13158, 2021 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-33369168

RESUMEN

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.


Asunto(s)
Aciduria Argininosuccínica , Enfermedad por Deficiencia de Ornitina Carbamoiltransferasa , Trastornos Innatos del Ciclo de la Urea , Animales , Autofagia , Beclina-1/genética , Ratones
6.
Hum Mol Genet ; 28(R1): R42-R48, 2019 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-31227828

RESUMEN

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.


Asunto(s)
Trastornos Innatos del Ciclo de la Urea/terapia , Animales , Tratamiento Basado en Trasplante de Células y Tejidos , Terapia Combinada , Manejo de la Enfermedad , Susceptibilidad a Enfermedades , Terapia de Reemplazo Enzimático , Terapia Genética , Humanos , Trasplante de Hígado/métodos , Redes y Vías Metabólicas , Resultado del Tratamiento , Urea/metabolismo , Trastornos Innatos del Ciclo de la Urea/diagnóstico , Trastornos Innatos del Ciclo de la Urea/etiología , Trastornos Innatos del Ciclo de la Urea/metabolismo
7.
J Inherit Metab Dis ; 42(6): 1128-1135, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-30724386

RESUMEN

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.


Asunto(s)
Amoníaco/metabolismo , Terapia Genética/métodos , Glutamato-Amoníaco Ligasa/genética , Hiperamonemia/genética , Hiperamonemia/terapia , Hígado/metabolismo , Amoníaco/toxicidad , Animales , Carbamoil-Fosfato Sintasa (Amoniaco)/genética , Carbamoil-Fosfato Sintasa (Amoniaco)/metabolismo , Enfermedad por Deficiencia de Carbamoil-Fosfato Sintasa I/genética , Enfermedad por Deficiencia de Carbamoil-Fosfato Sintasa I/metabolismo , Enfermedad por Deficiencia de Carbamoil-Fosfato Sintasa I/terapia , Modelos Animales de Enfermedad , Femenino , Técnicas de Transferencia de Gen , Glutamato-Amoníaco Ligasa/metabolismo , Hiperamonemia/metabolismo , Hiperamonemia/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Especificidad de Órganos/genética
8.
J Inherit Metab Dis ; 42(6): 1097-1104, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-30671986

RESUMEN

(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.


Asunto(s)
Amoníaco/metabolismo , Autofagia/fisiología , Hiperamonemia/etiología , Animales , Glutamato-Amoníaco Ligasa/metabolismo , Glutamina/metabolismo , Homeostasis , Humanos , Hiperamonemia/metabolismo , Hiperamonemia/patología , Hígado/metabolismo , Urea/metabolismo , Trastornos Innatos del Ciclo de la Urea/complicaciones , Trastornos Innatos del Ciclo de la Urea/metabolismo , Trastornos Innatos del Ciclo de la Urea/patología
9.
Autophagy ; 14(7): 1273-1275, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30035657

RESUMEN

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.


Asunto(s)
Autofagia , Hiperamonemia , Amoníaco , Humanos , Hígado , Urea
10.
Proc Natl Acad Sci U S A ; 115(2): 391-396, 2018 01 09.
Artículo en Inglés | MEDLINE | ID: mdl-29279371

RESUMEN

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.


Asunto(s)
Autofagia , Hiperamonemia/metabolismo , Hígado/metabolismo , Urea/metabolismo , Amoníaco/metabolismo , Animales , Humanos , Masculino , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos
11.
Hepatology ; 66(1): 124-135, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28295475

RESUMEN

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).


Asunto(s)
Carcinoma Hepatocelular/genética , Regulación Neoplásica de la Expresión Génica , Factor Nuclear 4 del Hepatocito/genética , Neoplasias Hepáticas/patología , Deficiencia de alfa 1-Antitripsina/genética , Envejecimiento/genética , Análisis de Varianza , Animales , Carcinoma Hepatocelular/patología , Modelos Animales de Enfermedad , Regulación hacia Abajo , Humanos , Neoplasias Hepáticas/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutación , Distribución Aleatoria , Estadísticas no Paramétricas , Deficiencia de alfa 1-Antitripsina/patología
12.
Hepatology ; 64(2): 535-48, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-26999313

RESUMEN

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).


Asunto(s)
Transportadoras de Casetes de Unión a ATP/metabolismo , Acuaporina 1/genética , Ácidos y Sales Biliares/metabolismo , Colestasis/terapia , Terapia Genética , Miembro 11 de la Subfamilia B de Transportador de Casetes de Unión al ATP , Animales , Acuaporina 1/metabolismo , Ácidos y Sales Biliares/sangre , Canalículos Biliares/metabolismo , Colestasis/inducido químicamente , Etinilestradiol , Hepatocitos/metabolismo , Humanos , Masculino , Ratas Wistar
13.
Biochem Cell Biol ; 93(4): 417-20, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26194146

RESUMEN

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.


Asunto(s)
Acidosis/metabolismo , Amoníaco/metabolismo , Acuaporinas/metabolismo , Regulación hacia Abajo , Mitocondrias/metabolismo , Urea/metabolismo , Animales , Células Cultivadas , Masculino , Ratas , Ratas Wistar
14.
FEBS Lett ; 588(9): 1686-91, 2014 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-24642373

RESUMEN

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.


Asunto(s)
Amoníaco/metabolismo , Acuaporinas/metabolismo , Hepatocitos/metabolismo , Lipopolisacáridos/farmacología , Mitocondrias Hepáticas/metabolismo , Urea/metabolismo , Animales , Transporte Biológico , Células Cultivadas , Glucagón/fisiología , Hepatocitos/inmunología , Masculino , Metilaminas/metabolismo , Mitocondrias Hepáticas/inmunología , Ratas , Ratas Wistar
15.
Clin Sci (Lond) ; 125(11): 521-32, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23642207

RESUMEN

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.


Asunto(s)
Colestasis/inducido químicamente , Endotelina-1/farmacología , Endotelina-3/farmacología , Óxido Nítrico/fisiología , Receptor de Endotelina B/fisiología , Nervio Vago/efectos de los fármacos , Animales , Bilis/metabolismo , Presión Sanguínea/efectos de los fármacos , Colagogos y Coleréticos/farmacología , Colestasis/metabolismo , Hemodinámica/efectos de los fármacos , Óxido Nítrico/metabolismo , Ratas , Ratas Sprague-Dawley , Receptor de Endotelina B/metabolismo , Reflejo/efectos de los fármacos , Flujo Sanguíneo Regional/efectos de los fármacos , Vagotomía , Nervio Vago/metabolismo , Nervio Vago/fisiología
16.
Hepatology ; 57(5): 2061-71, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23299935

RESUMEN

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.


Asunto(s)
Amoníaco/metabolismo , Acuaporinas/metabolismo , Hepatocitos/metabolismo , Inactivación Metabólica/fisiología , Mitocondrias Hepáticas/metabolismo , Urea/metabolismo , Animales , Acuaporinas/efectos de los fármacos , Acuaporinas/genética , Células Cultivadas , Glucagón/farmacología , Hepatocitos/patología , Masculino , Membranas Mitocondriales/metabolismo , Modelos Animales , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/fisiología , Ratas , Ratas Wistar , Transfección , Regulación hacia Arriba/efectos de los fármacos
17.
Biochem Biophys Res Commun ; 393(2): 217-21, 2010 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-20132793

RESUMEN

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.


Asunto(s)
Amoníaco/metabolismo , Acuaporinas/metabolismo , Mitocondrias/metabolismo , Animales , Acuaporinas/genética , Transporte Biológico , Encéfalo/metabolismo , Formamidas/metabolismo , Mitocondrias Hepáticas/metabolismo , Ratas , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Agua/metabolismo
18.
Am J Physiol Regul Integr Comp Physiol ; 296(4): R1274-81, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19193945

RESUMEN

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.


Asunto(s)
Acuaporinas/metabolismo , Glucagón/metabolismo , Hepatocitos/metabolismo , Animales , Acuaporinas/genética , Células Cultivadas , AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/antagonistas & inhibidores , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Glucagón/administración & dosificación , Hepatocitos/efectos de los fármacos , Hepatocitos/enzimología , Inyecciones Intraperitoneales , Masculino , Inhibidores de la Síntesis del Ácido Nucleico/farmacología , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Inhibidores de Proteasas/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Procesamiento Proteico-Postraduccional , Inhibidores de la Síntesis de la Proteína/farmacología , ARN Mensajero/metabolismo , Ratas , Ratas Wistar , Factores de Tiempo , Regulación hacia Arriba
19.
Am J Physiol Gastrointest Liver Physiol ; 296(1): G93-100, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18948439

RESUMEN

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.


Asunto(s)
Acuaporinas/metabolismo , Canalículos Biliares/metabolismo , Técnicas de Silenciamiento del Gen , Hepatocitos/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Agua/metabolismo , Acuaporinas/genética , Canalículos Biliares/efectos de los fármacos , Línea Celular Tumoral , CMP Cíclico/análogos & derivados , CMP Cíclico/farmacología , Hepatocitos/efectos de los fármacos , Humanos , Microscopía Confocal , Ósmosis , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Tiempo , Transfección
20.
World J Gastroenterol ; 14(46): 7059-67, 2008 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-19084912

RESUMEN

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.


Asunto(s)
Acuaporinas/fisiología , Colestasis Intrahepática/fisiopatología , Acuaporinas/metabolismo , Bilis/metabolismo , Colestasis Intrahepática/patología , Hepatocitos/metabolismo , Hepatocitos/patología , Humanos , Ósmosis/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA