Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Mol Ther ; 28(2): 677-689, 2020 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-31810863

RESUMEN

Mutations in hydroxymethylbilane synthase (HMBS) cause acute intermittent porphyria (AIP), an autosomal dominant disease where typically only one HMBS allele is mutated. In AIP, the accumulation of porphyrin precursors triggers life-threatening neurovisceral attacks and at long-term, entails an increased risk of hepatocellular carcinoma, kidney failure, and hypertension. Today, the only cure is liver transplantation, and a need for effective mechanism-based therapies, such as pharmacological chaperones, is prevailing. These are small molecules that specifically stabilize a target protein. They may be developed into an oral treatment, which could work curatively during acute attacks, but also prophylactically in asymptomatic HMBS mutant carriers. With the use of a 10,000 compound library, we identified four binders that further increased the initially very high thermal stability of wild-type HMBS and protected the enzyme from trypsin digestion. The best hit and a selected analog increased steady-state levels and total HMBS activity in human hepatoma cells overexpressing HMBS, and in an Hmbs-deficient mouse model with a low-expressed wild-type-like allele, compared to untreated controls. Moreover, the concentration of porphyrin precursors decreased in liver of mice treated with the best hit. Our findings demonstrate the great potential of these hits for the development of a pharmacological chaperone-based corrective treatment of AIP by enhancing wild-type HMBS function independently of the patients' specific mutation.


Asunto(s)
Biomarcadores , Descubrimiento de Drogas , Porfiria Intermitente Aguda/metabolismo , Animales , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Humanos , Ratones , Ratones Noqueados , Terapia Molecular Dirigida , Porfiria Intermitente Aguda/etiología , Porfiria Intermitente Aguda/terapia , Pliegue de Proteína , Proteínas/antagonistas & inhibidores , Proteínas/química , Proteínas/metabolismo , Reproducibilidad de los Resultados , Bibliotecas de Moléculas Pequeñas , Relación Estructura-Actividad
2.
Infect Immun ; 88(12)2020 11 16.
Artículo en Inglés | MEDLINE | ID: mdl-32989036

RESUMEN

Escherichia coli O25b:H4 sequence type 131 (ST131), which is resistant to fluoroquinolones and which is a producer of CTX-M-15, is globally one of the major extraintestinal pathogenic E. coli (ExPEC) lineages. Phylogenetic analyses showed that multidrug-resistant ST131 strains belong to clade C, which recently emerged from clade B by stepwise evolution. It has been hypothesized that features other than multidrug resistance could contribute to this dissemination since other major global ExPEC lineages (ST73 and ST95) are mostly antibiotic susceptible. To test this hypothesis, we compared early biofilm production, presence of ExPEC virulence factors (VFs), and in vivo virulence in a mouse sepsis model in 19 and 20 epidemiologically relevant strains of clades B and C, respectively. Clade B strains were significantly earlier biofilm producers (P < 0.001), carriers of more VFs (P = 4e-07), and faster killers of mice (P = 2e-10) than clade C strains. Gene inactivation experiments showed that the H30-fimB and ibeART genes were associated with in vivo virulence. Competition assays in sepsis, gut colonization, and urinary tract infection models between the most anciently diverged strain (B1 subclade), one C1 subclade strain, and a B4 subclade recombining strain harboring some clade C-specific genetic events showed that the B1 strain always outcompeted the C1 strain, whereas the B4 strain outcompeted the C1 strain, depending on the mouse niches. All these findings strongly suggest that clade C evolution includes a progressive loss of virulence involving multiple genes, possibly enhancing overall strain fitness by avoiding severe infections, even if it comes at the cost of a lower colonization ability.


Asunto(s)
Infecciones por Escherichia coli/microbiología , Proteínas de Escherichia coli/genética , Escherichia coli/genética , Escherichia coli Patógena Extraintestinal/genética , Sepsis/microbiología , Factores de Virulencia/genética , Virulencia/genética , Animales , Antibacterianos/farmacología , Biopelículas/efectos de los fármacos , Biopelículas/crecimiento & desarrollo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Modelos Animales de Enfermedad , Escherichia coli/efectos de los fármacos , Escherichia coli/crecimiento & desarrollo , Escherichia coli/patogenicidad , Proteínas de Escherichia coli/metabolismo , Evolución Molecular , Escherichia coli Patógena Extraintestinal/efectos de los fármacos , Escherichia coli Patógena Extraintestinal/patogenicidad , Genotipo , Integrasas/genética , Integrasas/metabolismo , Estimación de Kaplan-Meier , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Fenotipo , Infecciones Urinarias/microbiología , Secuenciación Completa del Genoma
3.
Gut ; 68(3): 522-532, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30301768

RESUMEN

OBJECTIVE: Sustained inflammation originating from macrophages is a driving force of fibrosis progression and resolution. Monoacylglycerol lipase (MAGL) is the rate-limiting enzyme in the degradation of monoacylglycerols. It is a proinflammatory enzyme that metabolises 2-arachidonoylglycerol, an endocannabinoid receptor ligand, into arachidonic acid. Here, we investigated the impact of MAGL on inflammation and fibrosis during chronic liver injury. DESIGN: C57BL/6J mice and mice with global invalidation of MAGL (MAGL -/- ), or myeloid-specific deletion of either MAGL (MAGLMye-/-), ATG5 (ATGMye-/-) or CB2 (CB2Mye-/-), were used. Fibrosis was induced by repeated carbon tetrachloride (CCl4) injections or bile duct ligation (BDL). Studies were performed on peritoneal or bone marrow-derived macrophages and Kupffer cells. RESULTS: MAGL -/- or MAGLMye-/- mice exposed to CCl4 or subjected to BDL were more resistant to inflammation and fibrosis than wild-type counterparts. Therapeutic intervention with MJN110, an MAGL inhibitor, reduced hepatic macrophage number and inflammatory gene expression and slowed down fibrosis progression. MAGL inhibitors also accelerated fibrosis regression and increased Ly-6Clow macrophage number. Antifibrogenic effects exclusively relied on MAGL inhibition in macrophages, since MJN110 treatment of MAGLMye-/- BDL mice did not further decrease liver fibrosis. Cultured macrophages exposed to MJN110 or from MAGLMye-/- mice displayed reduced cytokine secretion. These effects were independent of the cannabinoid receptor 2, as they were preserved in CB2Mye-/- mice. They relied on macrophage autophagy, since anti-inflammatory and antifibrogenic effects of MJN110 were lost in ATG5Mye-/- BDL mice, and were associated with increased autophagic flux and autophagosome biosynthesis in macrophages when MAGL was pharmacologically or genetically inhibited. CONCLUSION: MAGL is an immunometabolic target in the liver. MAGL inhibitors may show promising antifibrogenic effects during chronic liver injury.


Asunto(s)
Antiinflamatorios/uso terapéutico , Cirrosis Hepática Experimental/tratamiento farmacológico , Hígado/enzimología , Monoacilglicerol Lipasas/antagonistas & inhibidores , Animales , Antiinflamatorios/farmacología , Autofagia/efectos de los fármacos , Carbamatos/farmacología , Carbamatos/uso terapéutico , Tetracloruro de Carbono , Recuento de Células , Células Cultivadas , Citocinas/metabolismo , Progresión de la Enfermedad , Evaluación Preclínica de Medicamentos/métodos , Hidrolasas/metabolismo , Mediadores de Inflamación/antagonistas & inhibidores , Mediadores de Inflamación/metabolismo , Cirrosis Hepática Experimental/inducido químicamente , Cirrosis Hepática Experimental/enzimología , Cirrosis Hepática Experimental/patología , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Macrófagos/fisiología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Terapia Molecular Dirigida/métodos , Monoacilglicerol Lipasas/fisiología , Receptor Cannabinoide CB2/metabolismo , Succinimidas/farmacología , Succinimidas/uso terapéutico
4.
Gut ; 67(8): 1505-1516, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-28601846

RESUMEN

OBJECTIVE: Cirrhosis downregulates phagocyte oxidant production via their antibacterial superoxide-generating system, NADPH oxidase (NOX2) and increases patients' susceptibility to infection and mortality rate. To explore novel biochemical parameters that explain susceptibility to infections, we investigated the expression of NOX2 and partners in neutrophils of patients with severe alcoholic cirrhosis and have provided a novel approach to restore superoxide production capacity in patients' neutrophils and blood. DESIGN: Neutrophils were isolated from patients with decompensated alcoholic cirrhosis. NOX2 activity was assessed after stimulation of purified neutrophils or whole blood with the bacterial-derived peptide fMet-Leu-Phe. The expression of NOX2 and partners was studied by western blot analysis, flow cytometry and reverse transcription-PCR. RESULTS: The impaired superoxide production by patients' neutrophils was associated with a severe deficient expression of the NADPH oxidase catalytic core flavocytochrome-b558 (gp91 phox /NOX2 and p22 phox ), its cytosolic partner p47 phox but not p67 phox . NOX2 expression decreased rapidly by protein degradation involving elastase released during degranulation of healthy neutrophils stimulated with fMet-Leu-Phe, or highly present in patients' plasma. Interestingly, the deficient superoxide production was reversed by treatment of patients' neutrophils and whole blood with toll-like receptor 7/8 (TLR7/8) agonists. This treatment stimulated a rapid NOX2 transcription and translation through a process involving mammalian target of rapamycin (mTOR) whose expression was also deficient in patients' neutrophils. NOX2 expression was also increased by the TLR4 agonist lipopolysaccharide but with only a modest improvement of reactive oxygen species production. CONCLUSION: Impairment of neutrophil oxidants production in alcoholic cirrhosis is associated with NOX2 degradation and deficient mTOR-dependent translational machinery. The NOX2 depletion can be reversed via TRL7/8 activation and might be used to restore antimicrobial responses of immunocompromised patients.


Asunto(s)
Cirrosis Hepática Alcohólica/metabolismo , NADPH Oxidasas/metabolismo , Neutrófilos/fisiología , Especies Reactivas de Oxígeno/metabolismo , Receptor Toll-Like 7/metabolismo , Receptor Toll-Like 8/metabolismo , Estudios de Casos y Controles , Femenino , Humanos , Cirrosis Hepática Alcohólica/patología , Masculino , Persona de Mediana Edad
5.
Haematologica ; 102(2): 260-270, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-28143953

RESUMEN

Hemolysis occurring in hematologic diseases is often associated with an iron loading anemia. This iron overload is the result of a massive outflow of hemoglobin into the bloodstream, but the mechanism of hemoglobin handling has not been fully elucidated. Here, in a congenital erythropoietic porphyria mouse model, we evaluate the impact of hemolysis and regenerative anemia on hepcidin synthesis and iron metabolism. Hemolysis was confirmed by a complete drop in haptoglobin, hemopexin and increased plasma lactate dehydrogenase, an increased red blood cell distribution width and osmotic fragility, a reduced half-life of red blood cells, and increased expression of heme oxygenase 1. The erythropoiesis-induced Fam132b was increased, hepcidin mRNA repressed, and transepithelial iron transport in isolated duodenal loops increased. Iron was mostly accumulated in liver and spleen macrophages but transferrin saturation remained within the normal range. The expression levels of hemoglobin-haptoglobin receptor CD163 and hemopexin receptor CD91 were drastically reduced in both liver and spleen, resulting in heme- and hemoglobin-derived iron elimination in urine. In the kidney, the megalin/cubilin endocytic complex, heme oxygenase 1 and the iron exporter ferroportin were induced, which is reminiscent of significant renal handling of hemoglobin-derived iron. Our results highlight ironbound hemoglobin urinary clearance mechanism and strongly suggest that, in addition to the sequestration of iron in macrophages, kidney may play a major role in protecting hepatocytes from iron overload in chronic hemolysis.


Asunto(s)
Anemia Hemolítica/metabolismo , Hepatocitos/metabolismo , Hepcidinas/metabolismo , Hierro/metabolismo , Anemia Hemolítica/sangre , Anemia Hemolítica/complicaciones , Anemia Hemolítica/genética , Animales , Apoptosis , Transporte Biológico , Biomarcadores , Modelos Animales de Enfermedad , Eritrocitos/metabolismo , Eritropoyesis , Expresión Génica , Hemo/metabolismo , Hepcidinas/sangre , Hepcidinas/genética , Humanos , Hierro/orina , Sobrecarga de Hierro/etiología , Sobrecarga de Hierro/metabolismo , Macrófagos , Ratones , Ratones Noqueados , Ratones Transgénicos , Bazo/fisiología , Estrés Fisiológico
6.
J Am Soc Nephrol ; 27(3): 835-46, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26293821

RESUMEN

The iron-regulatory peptide hepcidin exhibits antimicrobial activity. Having previously shown hepcidin expression in the kidney, we addressed its role in urinary tract infection (UTI), which remains largely unknown. Experimental UTI was induced in wild-type (WT) and hepcidin-knockout (Hepc-/-) mice using the uropathogenic Escherichia coli CFT073 strain. Compared with infected WT mice, infected Hepc-/- mice showed a dramatic increase in renal bacterial load. Moreover, bacterial invasion was significantly dampened by the pretreatment of WT mice with hepcidin. Infected Hepc-/- mice exhibited decreased iron accumulation in the renal medulla and significant attenuation of the renal inflammatory response. Notably, we demonstrated in vitro bacteriostatic activity of hepcidin against CFT073. Furthermore, CFT073 repressed renal hepcidin, both in vivo and in cultured renal cells, and reduced phosphorylation of SMAD kinase in vivo, suggesting a bacterial strategy to escape the antimicrobial activities of hepcidin. In conclusion, we provide new mechanisms by which hepcidin contributes to renal host defense and suggest that targeting hepcidin offers a strategy to prevent bacterial invasion.


Asunto(s)
Antiinfecciosos/farmacología , Infecciones por Escherichia coli/metabolismo , Escherichia coli/efectos de los fármacos , Hepcidinas/metabolismo , Hepcidinas/farmacología , Infecciones Urinarias/metabolismo , Animales , Antiinfecciosos/metabolismo , Carga Bacteriana/genética , Células Cultivadas , Recuento de Colonia Microbiana , Citocinas/metabolismo , Infecciones por Escherichia coli/microbiología , Femenino , Hepcidinas/genética , Hierro/metabolismo , Médula Renal/citología , Médula Renal/metabolismo , Médula Renal/microbiología , Ratones , Ratones Endogámicos CBA , Ratones Noqueados , Nefritis/metabolismo , Nefritis/microbiología , Nefritis/patología , Neutrófilos , Fosforilación , ARN Mensajero/metabolismo , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Proteínas Smad/metabolismo , Infecciones Urinarias/microbiología
7.
Kidney Int ; 84(4): 756-66, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23615502

RESUMEN

Hepcidin, the key regulatory hormone of iron homeostasis, and iron carriers such as transferrin receptor1 (TFR1), divalent metal transporter1 (DMT1), and ferroportin (FPN) are expressed in kidney. Whether hepcidin plays an intrinsic role in the regulation of renal iron transport is unknown. Here, we analyzed the renal handling of iron in hemochromatosis Hepc(-/-) and Hjv(-/-) mouse models, as well as in phenylhydrazine (PHZ)-treated mice. We found a marked medullary iron deposition in the kidneys of Hepc(-/-) mice, and iron leak in the urine. The kidneys of Hepc(-/-) mice exhibited a concomitant decrease in TFR1 and increase in ferritin and FPN expression. Increased FPN abundance was restricted to the thick ascending limb (TAL). DMT1 protein remained unaffected despite a significant decrease of its mRNA level, suggesting that DMT1 protein is stabilized in the absence of hepcidin. Treatment of kidney sections from Hepc(-/-) mice with hepcidin decreased DMT1 protein, an effect confirmed in renal cell lines where hepcidin markedly decreased (55)Fe transport. In the kidneys of Hjv(-/-) mice exhibiting low hepcidin expression, the iron overload was similar to that in the kidneys of Hepc(-/-) mice. However, in PHZ mice, iron accumulation resulting from hemoglobin leak was detected in the proximal tubule. Thus, kidneys exhibit a tissue-specific handling of iron that depends on the extra iron source. Hepcidin may control the expression of iron transporters to prevent renal iron overload.


Asunto(s)
Hemocromatosis/metabolismo , Hepcidinas/metabolismo , Homeostasis/fisiología , Hierro/metabolismo , Túbulos Renales Distales/metabolismo , Animales , Proteínas de Transporte de Catión/metabolismo , Línea Celular , Células Cultivadas , Modelos Animales de Enfermedad , Proteínas Ligadas a GPI , Hemocromatosis/inducido químicamente , Hemocromatosis/genética , Proteína de la Hemocromatosis , Hepcidinas/deficiencia , Hepcidinas/genética , Técnicas In Vitro , Túbulos Renales Distales/patología , Asa de la Nefrona/metabolismo , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Noqueados , Zarigüeyas , Fenilhidrazinas/efectos adversos , Receptores de Transferrina/metabolismo
8.
J Hepatol ; 58(5): 936-48, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23321315

RESUMEN

BACKGROUND & AIMS: Lipopolysaccharide (LPS)-expressing bacteria cause severe inflammation in cirrhotic patients. The global gene response to LPS is unknown in cirrhotic immune cells. METHODS: Gene-expression profiling using Affymetrix Human Exon Array analyzed the expression of 14,851 genes in LPS-stimulated peripheral blood mononuclear cells (PBMCs) from 4 patients with cirrhosis and 4 healthy subjects. We performed validation studies using RT-qPCR in LPS-stimulated PBMCs from 52 patients and 9 healthy subjects and investigated the association of gene induction with mortality in 26 patients. RESULTS: Gene-expression profiling of LPS-stimulated cirrhotic cells showed 509 upregulated genes and 1588 downregulated genes. In LPS-stimulated "healthy" cells, 952 genes were upregulated and 838 genes downregulated. The 741 LPS-regulated genes shared by cirrhotic and "healthy" cells were involved in cytokine production/activity and induction of "immune paralysis". Comparison of functions associated with the 1356 genes, specifically regulated by LPS in cirrhotic cells, to functions of the 1049 genes, specifically regulated in "healthy" cells, allowed to define a cirrhosis-specific phenotype. Unlike in "healthy" cells, LPS failed to induce an interferon-mediated program in cirrhotic cells. In cirrhotic PBMCs, LPS specifically induced certain molecules involved in apoptosis and downregulated molecules involved in endocytic trafficking. RT-qPCR experiments showed that LPS-stimulated cirrhotic PBMCs had an enhanced induction of certain proinflammatory cytokines and chemokines. In the prognosis study, higher ex vivo LPS-induction of the inflammatory genes IL6 and CXCL5 was a significant predictor of mortality. CONCLUSIONS: Our results show that LPS-stimulated cirrhotic PBMCs exhibit an extensive and often unexpected transcriptional response.


Asunto(s)
Exones/genética , Perfilación de la Expresión Génica , Expresión Génica/efectos de los fármacos , Leucocitos Mononucleares/metabolismo , Lipopolisacáridos/farmacología , Cirrosis Hepática/metabolismo , Adulto , Anciano , Apoptosis/genética , Biomarcadores/metabolismo , Estudios de Casos y Controles , Quimiocina CXCL5/genética , Quimiocina CXCL5/metabolismo , Endocitosis/genética , Femenino , Humanos , Interleucina-6/genética , Interleucina-6/metabolismo , Leucocitos Mononucleares/efectos de los fármacos , Cirrosis Hepática/genética , Cirrosis Hepática/mortalidad , Masculino , Persona de Mediana Edad , Valor Predictivo de las Pruebas , Tasa de Supervivencia
9.
Gastroenterology ; 143(1): 166-76.e6, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22465620

RESUMEN

BACKGROUND & AIMS: Circulating membrane-shed microparticles (MPs) participate in regulation of vascular tone. We investigated the cellular origins of MPs in plasma from patients with cirrhosis and assessed the contribution of MPs to arterial vasodilation, a mechanism that contributes to portal hypertension. METHODS: We analyzed MPs from blood samples of 91 patients with cirrhosis and 30 healthy individuals (controls) using flow cytometry; their effects on the vascular response to vasoconstrictors were examined in vitro and in vivo. RESULTS: Circulating levels of leuko-endothelial (CD31(+)/41(-)), pan-leukocyte (CD11a(+)), lymphocyte (CD4(+)), and erythrocyte (CD235a(+)) MPs were higher in patients with cirrhosis than in controls. Plasma of patients with cirrhosis contained hepatocyte-derived MPs (cytokeratin-18(+)), whereas plasma from controls did not. The severity of cirrhosis and systemic inflammation were major determinants of the levels of leuko-endothelial and hepatocyte MPs. MPs from patients with advanced cirrhosis significantly impaired contraction of vessels in response to phenylephrine, whereas MPs from healthy controls or from patients of Child-Pugh class A did not. This effect depended on cyclooxygenase type 1 and required phosphatidylserine on the surface of MPs. Intravenous injection of MPs from patients with cirrhosis into BALB/C mice decreased mean arterial blood pressure. CONCLUSIONS: Cirrhosis is associated with increases in circulating subpopulations of MPs, likely resulting from systemic inflammation and liver cell damage. The overall pool of circulating MPs from patients with advanced cirrhosis impairs vasoconstrictor responses and decreases blood pressure, contributing to the arterial vasodilation associated with portal hypertension.


Asunto(s)
Micropartículas Derivadas de Células , Dilatación Patológica/fisiopatología , Hipertensión Portal/fisiopatología , Cirrosis Hepática/fisiopatología , Adulto , Femenino , Citometría de Flujo , Humanos , Cirrosis Hepática/sangre , Masculino , Persona de Mediana Edad , Vasoconstrictores/farmacología , Vasodilatación/efectos de los fármacos
10.
Mamm Genome ; 24(11-12): 427-38, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24121729

RESUMEN

Disorders of iron metabolism are among the most common acquired and constitutive diseases. Hemochromatosis has a solid genetic basis and in Northern European populations it is usually associated with homozygosity for the C282Y mutation in the HFE protein. However, the penetrance of this mutation is incomplete and the clinical presentation is highly variable. The rare and common variants identified so far as genetic modifiers of HFE-related hemochromatosis are unable to account for the phenotypic heterogeneity of this disorder. There are wide variations in the basal iron status of common inbred mouse strains, and this diversity may reflect the genetic background of the phenotypic diversity under pathological conditions. We therefore examined the genetic basis of iron homeostasis using quantitative trait loci mapping applied to the HcB-15 recombinant congenic strains for tissue and serum iron indices. Two highly significant QTL containing either the N374S Mon1a mutation or the Ferroportin locus were found to be major determinants in spleen and liver iron loading. Interestingly, when considering possible epistatic interactions, the effects of Mon1a on macrophage iron export are conditioned by the genotype at the Slc40a1 locus. Only mice that are C57BL/10ScSnA homozygous at both loci display a lower spleen iron burden. Furthermore, the liver-iron lowering effect of the N374S Mon1a mutation is observed only in mice that display a nonsense mutation in the Ceruloplasmin (Cp) gene. This study highlights the existence of genetic interactions between Cp, Mon1a, and the Slc40a1 locus in iron metabolism, suggesting that epistasis may be a crucial determinant of the variable biological and clinical presentations in iron disorders.


Asunto(s)
Proteínas Portadoras/genética , Proteínas de Transporte de Catión/genética , Ceruloplasmina/genética , Epistasis Genética , Hemocromatosis/veterinaria , Hierro/metabolismo , Ratones/genética , Enfermedades de los Roedores/genética , Animales , Femenino , Hemocromatosis/genética , Hemocromatosis/metabolismo , Hígado/metabolismo , Masculino , Ratones/metabolismo , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Sitios de Carácter Cuantitativo , Enfermedades de los Roedores/metabolismo , Bazo/metabolismo
11.
Gastroenterology ; 140(4): 1261-1271.e1, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21199652

RESUMEN

BACKGROUNDS & AIMS: The mechanism by which hepcidin regulates iron export from macrophages has been well established and is believed to involve degradation of ferroportin. However, in the small intestine, hepcidin's mechanisms of action are not known. We studied human polarized intestinal (Caco-2/TC7) cells and mouse duodenal segments, ex vivo, to investigate the molecular mechanisms by which hepcidin down-regulates intestinal transepithelial iron transport. METHODS: Iron transport was analyzed using 55FeNTA. Expression of Divalent Metal Transporter 1 (DMT1) and ferroportin was evaluated by reverse-transcription quantitative polymerase chain reaction and immunoblotting. Videomicroscopy analysis was performed on live cells that expressed either DMT1 or ferroportin fused to green fluorescent protein. RESULTS: In Caco-2/TC7 cells, physiologic doses of hepcidin (50-1000 nmol/L) inhibited transport of 55Fe in a dose-dependent manner; a half-maximum effect was observed at 75-100 nmol/L. However, 200 nmol/L hepcidin induced a significant decrease in DMT1 protein expression but no change in ferroportin protein levels, unlike macrophages. This result was confirmed ex vivo in isolated duodenal segments: 200 nmol/L hepcidin induced a significant reduction in iron transport and DMT1 protein levels but no change in ferroportin levels. In Caco-2/TC7 cells, the effect of hepcidin on the DMT1 protein level was completely abolished in the presence of a proteasome inhibitor (MG-132); DMT1 ubiquitination was induced by the addition of hepcidin. CONCLUSIONS: An acute increase in hepcidin concentration reduces intestinal iron absorption through ubiquitin-dependent proteasome degradation of DMT1.


Asunto(s)
Péptidos Catiónicos Antimicrobianos/metabolismo , Proteínas de Transporte de Catión/metabolismo , Duodeno/metabolismo , Hierro/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Animales , Células CACO-2 , Proteínas de Transporte de Catión/genética , Regulación hacia Abajo/fisiología , Expresión Génica/fisiología , Proteínas Fluorescentes Verdes/genética , Hepcidinas , Homeostasis/fisiología , Humanos , Mucosa Intestinal/metabolismo , Radioisótopos de Hierro/farmacocinética , Masculino , Ratones , Ratones Endogámicos C57BL , Ubiquitinación/fisiología
12.
Gastroenterology ; 141(4): 1509-19, 1519.e1-3, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21762662

RESUMEN

BACKGROUND & AIMS: Chronic, progressive hepatobiliary disease is the most severe complication of erythropoietic protoporphyria (EPP) and can require liver transplantation, although the mechanisms that lead to liver failure are unknown. We characterized protoporphyrin-IX (PPIX)-linked hepatobiliary disease in BALB/c and C57BL/6 (Fechm1Pas) mice with mutations in ferrochelatase as models for EPP. METHODS: Fechm1Pas and wild-type (control) mice were studied at 12-14 weeks of age. PPIX was quantified; its distribution in the liver, serum levels of lipoprotein-X, liver histology, contents of bile salt and cholesterol phospholipids, and expression of genes were compared in mice of the BALB/c and C57BL/6 backgrounds. The in vitro binding affinity of PPIX for bile components was determined. RESULTS: Compared with mice of the C57BL/6 background, BALB/c Fechm1Pas mice had a more severe pattern of cholestasis, fibrosis with portoportal bridging, bile acid regurgitation, sclerosing cholangitis, and hepatolithiasis. In C57BL/6 Fechm1Pas mice, PPIX was sequestrated mainly in the cytosol of hepatocytes and Kupffer cells, whereas, in BALB/c Fechm1Pas mice, PPIX was localized within enlarged bile canaliculi. Livers of C57BL/6 Fechm1Pas mice were protected through a combination of lower efflux of PPIX and reduced synthesis and export of bile acid. CONCLUSIONS: PPIX binds to bile components and disrupts the physiologic equilibrium of phospholipids, bile acids, and cholesterol in bile. This process might be involved in pathogenesis of sclerosing cholangitis from EPP; a better understanding might improve diagnosis and development of reagents to treat or prevent liver failure in patients with EPP.


Asunto(s)
Colangitis Esclerosante/prevención & control , Hepatocitos/metabolismo , Macrófagos del Hígado/metabolismo , Porfiria Eritropoyética/metabolismo , Protoporfirinas/metabolismo , Animales , Ácidos y Sales Biliares/metabolismo , Colangitis Esclerosante/genética , Colangitis Esclerosante/metabolismo , Colangitis Esclerosante/patología , Colesterol/metabolismo , Modelos Animales de Enfermedad , Ferroquelatasa/genética , Ferroquelatasa/metabolismo , Regulación de la Expresión Génica , Genotipo , Hepatocitos/patología , Macrófagos del Hígado/patología , Lipoproteína X/sangre , Cirrosis Hepática/metabolismo , Cirrosis Hepática/prevención & control , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Fenotipo , Fosfolípidos/metabolismo , Mutación Puntual , Porfiria Eritropoyética/complicaciones , Porfiria Eritropoyética/genética , Porfiria Eritropoyética/patología , Índice de Severidad de la Enfermedad
13.
Drug Metab Rev ; 44(1): 34-87, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21892896

RESUMEN

A frequent mechanism for drug-induced liver injury (DILI) is the formation of reactive metabolites that trigger hepatitis through direct toxicity or immune reactions. Both events cause mitochondrial membrane disruption. Genetic or acquired factors predispose to metabolite-mediated hepatitis by increasing the formation of the reactive metabolite, decreasing its detoxification, or by the presence of critical human leukocyte antigen molecule(s). In other instances, the parent drug itself triggers mitochondrial membrane disruption or inhibits mitochondrial function through different mechanisms. Drugs can sequester coenzyme A or can inhibit mitochondrial ß-oxidation enzymes, the transfer of electrons along the respiratory chain, or adenosine triphosphate (ATP) synthase. Drugs can also destroy mitochondrial DNA, inhibit its replication, decrease mitochondrial transcripts, or hamper mitochondrial protein synthesis. Quite often, a single drug has many different effects on mitochondrial function. A severe impairment of oxidative phosphorylation decreases hepatic ATP, leading to cell dysfunction or necrosis; it can also secondarily inhibit ß-oxidation, thus causing steatosis, and can also inhibit pyruvate catabolism, leading to lactic acidosis. A severe impairment of ß-oxidation can cause a fatty liver; further, decreased gluconeogenesis and increased utilization of glucose to compensate for the inability to oxidize fatty acids, together with the mitochondrial toxicity of accumulated free fatty acids and lipid peroxidation products, may impair energy production, possibly leading to coma and death. Susceptibility to parent drug-mediated mitochondrial dysfunction can be increased by factors impairing the removal of the toxic parent compound or by the presence of other medical condition(s) impairing mitochondrial function. New drug molecules should be screened for possible mitochondrial effects.


Asunto(s)
Enfermedad Hepática Inducida por Sustancias y Drogas/metabolismo , Hígado/metabolismo , Mitocondrias Hepáticas/metabolismo , Respiración de la Célula/efectos de los fármacos , Enfermedad Hepática Inducida por Sustancias y Drogas/patología , ADN Mitocondrial/biosíntesis , ADN Mitocondrial/efectos de los fármacos , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos , Hígado Graso/etiología , Hígado Graso/metabolismo , Hígado Graso/patología , Humanos , Sistema Inmunológico/metabolismo , Hígado/patología , Mitocondrias Hepáticas/efectos de los fármacos , Mitocondrias Hepáticas/patología , Mitocondrias Hepáticas/ultraestructura , Proteínas Mitocondriales/biosíntesis , Proteínas Mitocondriales/efectos de los fármacos , Oxidación-Reducción , Preparaciones Farmacéuticas/metabolismo , Especies Reactivas de Oxígeno/metabolismo
14.
Crit Care Med ; 40(7): 2141-8, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22564959

RESUMEN

OBJECTIVE: Anemia is common in critically ill patients, due to inflammation and blood loss. Anemia can be associated with iron deficiency and low serum hepcidin levels. However, iron administration in this setting remains controversial because of its potential toxicity, including oxidative stress induction and sepsis facilitation. The objective of this work was to determine the efficacy and toxicity of iron administration using a mouse model mimicking critical care anemia as well as a model of acute septicemia. DESIGN: Prospective, randomized, open label controlled animal study. SETTING: University-based research laboratory. SUBJECTS: C57BL/6 and OF1 mice. INTERVENTIONS: Intraperitoneal injection of zymosan inducing generalized inflammation in C57BL/6 mice, followed in our full model by repeated phlebotomies. A dose equivalent to 15 mg/kg of ferric carboxymaltose was injected intravenously on day 5. To assess the toxicity of iron in a septicemia model, OF1 mice were simultaneously injected with iron and different Escherichia coli strains. MEASUREMENTS AND MAIN RESULTS: To investigate the effect of iron on oxidative stress, we measured reactive oxygen species production in the blood using luminol-amplified chemiluminescence and superoxide dismutase 2 messenger RNA levels in the liver. These markers of oxidative stress were increased after iron administration in control mice but not in zymosan-treated mice. Liver catalase messenger RNA levels decreased in iron-treated control mice. Iron administration was not associated with increased mortality in the septicemia model or in the generalized inflammation model. Iron increased hemoglobin levels in mice fed with a low iron diet and subjected to phlebotomies and zymosan 2 wks after treatment administration. CONCLUSIONS: Adverse effects of intravenous iron supplementation by ferric carboxymaltose seem to be minimal in our animal models. Furthermore, iron appears to be effective in correcting anemia, despite inflammation. Studies of efficacy and safety of iron in critically ill patients are warranted.


Asunto(s)
Anemia/tratamiento farmacológico , Compuestos Férricos/administración & dosificación , Compuestos Férricos/toxicidad , Hematínicos/administración & dosificación , Hematínicos/toxicidad , Maltosa/análogos & derivados , Animales , Péptidos Catiónicos Antimicrobianos/genética , Péptidos Catiónicos Antimicrobianos/metabolismo , Catalasa/genética , Catalasa/metabolismo , Dieta , Modelos Animales de Enfermedad , Hemoglobinas , Hepcidinas , Inflamación/inducido químicamente , Inyecciones Intravenosas , Hierro/administración & dosificación , Hierro/análisis , Hígado/química , Hígado/metabolismo , Luminiscencia , Maltosa/administración & dosificación , Maltosa/toxicidad , Ratones , Ratones Endogámicos C57BL , Flebotomía , ARN Mensajero/metabolismo , Distribución Aleatoria , Especies Reactivas de Oxígeno/sangre , Sepsis/tratamiento farmacológico , Bazo/química , Superóxido Dismutasa/genética , Superóxido Dismutasa/metabolismo , Oligoelementos/administración & dosificación , Zimosan/farmacología
15.
Blood ; 116(26): 6072-81, 2010 Dec 23.
Artículo en Inglés | MEDLINE | ID: mdl-20844235

RESUMEN

Mouse bone marrow erythropoiesis is homeostatic, whereas after acute anemia, bone morphogenetic protein 4 (BMP4)-dependent stress erythropoiesis develops in the spleen. The aim of this work was to compare spleen stress erythropoiesis and bone marrow erythropoiesis in a mouse model of zymosan-induced generalized inflammation, which induces long-lasting anemia and to evaluate the ability of erythropoietin (Epo) injections to correct anemia in this setting. The effects of zymosan and/or Epo injections on erythroid precursor maturation and apoptosis, serum interferon-γ levels, hematologic parameters, and spleen BMP4 expression were analyzed, as well as the effect of zymosan on red blood cell half-life. We found that bone marrow erythropoiesis is suppressed by inflammation and does not respond to Epo administration, despite repression of erythroblast apoptosis. On the contrary, a robust erythropoietic response takes place in the spleen after Epo injections in both control and zymosan-induced generalized inflammation mice. This specific response implies Epo-mediated induction of BMP4 expression by F4/80(+) spleen macrophages, proliferation of stress burst-forming units-erythroid, and increased number of spleen erythroblasts. It allows only partial recovery of anemia, probably because of peripheral destruction of mature red cells. It is not clear whether similar BMP4-dependent stress erythropoiesis can occur in human bone marrow after Epo injections.


Asunto(s)
Anemia/prevención & control , Proteína Morfogenética Ósea 4/metabolismo , Modelos Animales de Enfermedad , Eritropoyesis/fisiología , Eritropoyetina/uso terapéutico , Inflamación/prevención & control , Anemia/inducido químicamente , Animales , Apoptosis , Western Blotting , Médula Ósea/metabolismo , Células Precursoras Eritroides/metabolismo , Humanos , Inflamación/etiología , Masculino , Ratones , Ratones Endogámicos C57BL , ARN Mensajero/genética , Receptores de Eritropoyetina/metabolismo , Proteínas Recombinantes , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Bazo/citología , Bazo/metabolismo , Zimosan/toxicidad
16.
FASEB J ; 25(9): 3271-8, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21670064

RESUMEN

Transient environmental influences, such as perinatal nutritional stress, may induce deleterious metabolic symptoms that last for the entire life of individuals, implying that epigenetic modifications play an important role in this process. We have investigated, in mice, the consequences of maternal undernutrition during gestation and lactation on DNA methylation and expression of the leptin gene, which plays a major regulatory role in coordinating nutritional state with many aspects of mammalian biology. We show that animals born to mothers fed a low-protein-diet (F1-LPD group) have a lower body weight/adiposity and exhibit a higher food intake than animals born to mothers fed a control diet (F1-CD group). These modifications persisted throughout life and were associated with lower levels of leptin mRNA and protein in starved F1-LPD mice, emphasizing that maternal protein-undernutrition affects the balance between food intake and energy expenditure in adults. Moreover, this nutritional stress resulted in the removal of methyls at CpGs located in the promoter of leptin, causing a permanent specific modification in the dynamics of the expression of leptin, which exhibits a stronger induction in the F1-LPD than in F1-CD mice in response to a meal. This study is an example of a molecular rationale linking transient environmental influences to permanent phenotypic consequences.


Asunto(s)
Dieta , Proteínas en la Dieta/farmacología , Leptina/metabolismo , Síndrome Metabólico/etiología , Fenómenos Fisiologicos de la Nutrición Prenatal , Animales , Secuencia de Bases , Composición Corporal , Islas de CpG , Femenino , Regulación de la Expresión Génica/fisiología , Humanos , Leptina/genética , Ratones , Ratones Endogámicos BALB C , Embarazo , Regiones Promotoras Genéticas , ARN Mensajero/genética , ARN Mensajero/metabolismo
17.
J Hepatol ; 55(4): 784-93, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21334395

RESUMEN

BACKGROUND & AIMS: In decompensated cirrhosis, the early innate immune response to the Toll-like receptor 4 (TLR4) agonist, lipopolysaccharides (LPS), is characterized by a hyper-production of pro-inflammatory cytokines and hypo-production of the anti-inflammatory cytokine IL-10. In LPS-stimulated non-cirrhotic immune cells, the constitutively active glycogen synthase kinase (GSK) 3 favors pro- vs. anti-inflammatory cytokines, by acting on gene induction. However, in these cells, TLR4 dampens its own pro-inflammatory response by inducing early (within minutes) AKT-mediated phosphorylation of GSK3ß (one of two GSK3 isoforms) on Ser9. Phosphorylation of GSK3ß (Ser9) inhibits its activity, decreases pro-inflammatory cytokines, and increases IL-10. Thus, we investigated the role of GSK3 in LPS-induced cytokine production by peripheral blood mononuclear cells (PBMCs) or monocytes from patients with advanced cirrhosis and normal subjects. METHODS: Cells were pre-incubated with or without GSK3 inhibitor (SB216763 or lithium chloride) for 1h and then stimulated with LPS. Cytokine production was assessed at mRNA and secreted proteins levels, by real-time RT-PCR at 1h and ELISA at 20 h, respectively. GSK3ß phosphorylation was assessed using Western blotting. RESULTS: In cirrhotic and normal PBMCs pretreated with GSK3 inhibitors, LPS-induced production of pro-inflammatory proteins TNF-α and IL-12p40 was significantly decreased while that of IL-10 was increased. LPS-induced, AKT-mediated phosphorylation of GSK3ß on Ser9 found in normal monocytes, was abolished in cirrhotic cells. CONCLUSIONS: GSK3 is involved in the early TLR4-mediated pro-inflammatory response in patients with decompensated cirrhosis. This was associated with a defect in AKT-mediated GSK3ß phosphorylation resulting in unrestricted 'pro-inflammatory' activity of the enzyme.


Asunto(s)
Glucógeno Sintasa Quinasa 3/inmunología , Glucógeno Sintasa Quinasa 3/metabolismo , Hepatitis/inmunología , Cirrosis Hepática/inmunología , Transducción de Señal/inmunología , Adyuvantes Inmunológicos/farmacología , Adulto , Anciano , Células Cultivadas , Femenino , Regulación Enzimológica de la Expresión Génica/inmunología , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Glucógeno Sintasa Quinasa 3/genética , Glucógeno Sintasa Quinasa 3 beta , Hepatitis/metabolismo , Humanos , Indoles/farmacología , Interleucina-10/inmunología , Interleucina-10/metabolismo , Interleucina-12/inmunología , Interleucina-12/metabolismo , Lipopolisacáridos/farmacología , Cloruro de Litio/farmacología , Cirrosis Hepática/metabolismo , Masculino , Maleimidas/farmacología , Persona de Mediana Edad , Monocitos/inmunología , Monocitos/metabolismo , Proteínas Proto-Oncogénicas c-akt/genética , Índice de Severidad de la Enfermedad , Receptor Toll-Like 4/inmunología , Receptor Toll-Like 4/metabolismo
18.
J Pharmacol Exp Ther ; 332(3): 886-97, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20016022

RESUMEN

Alcohol consumption increases reactive oxygen species (ROS) formation, which can damage mitochondrial DNA (mtDNA) and alter mitochondrial function. To test whether manganese superoxide dismutase (MnSOD) modulates acute alcohol-induced mitochondrial alterations, transgenic MnSOD-overexpressing (MnSOD(+++)) mice, heterozygous knockout (MnSOD(+/-)) mice, and wild-type (WT) littermates were sacrificed 2 or 24 h after intragastric ethanol administration (5 g/kg). Alcohol administration further increased MnSOD activity in MnSOD(+++) mice, but further decreased it in MnSOD(+/-) mice. In WT mice, alcohol administration transiently increased mitochondrial ROS formation, decreased mitochondrial glutathione, depleted and damaged mtDNA, and decreased complex I and V activities; alcohol durably increased inducible nitric-oxide synthase (NOS) expression, plasma nitrites/nitrates, and the nitration of tyrosine residues in complex V proteins. These effects were prevented in MnSOD(+++) mice and prolonged in MnSOD(+/-) mice. In alcoholized WT or MnSOD(+/-) mice, mtDNA depletion and the nitration of tyrosine residues in complex I and V proteins were prevented or attenuated by cotreatment with tempol (4-hydroxy-2,2,6,6-tetramethylpiperidine-N-oxyl), a superoxide scavenger; N(omega)-nitro-l-arginine methyl ester and N-[3-(aminomethyl)benzyl]acetamidine (1,400W), two NOS inhibitors; or uric acid, a peroxynitrite scavenger. In conclusion, MnSOD overexpression prevents, and MnSOD deficiency prolongs, mtDNA depletion after an acute alcohol binge in mice. The protective effects of MnSOD, tempol, NOS inhibitors, and uric acid point out a role of the superoxide anion reacting with NO to form mtDNA-damaging peroxynitrite.


Asunto(s)
ADN Mitocondrial/metabolismo , Etanol/envenenamiento , Hígado/metabolismo , Ácido Peroxinitroso/metabolismo , Superóxido Dismutasa/fisiología , Animales , Caspasa 3/metabolismo , Óxidos N-Cíclicos/farmacología , Proteínas de Unión al ADN/metabolismo , Complejo I de Transporte de Electrón/metabolismo , Depuradores de Radicales Libres/farmacología , Glutatión Peroxidasa/metabolismo , Proteínas del Grupo de Alta Movilidad/metabolismo , Hierro/metabolismo , Ratones , Ratones Noqueados , Mitocondrias Hepáticas/fisiología , ATPasas de Translocación de Protón Mitocondriales/metabolismo , NG-Nitroarginina Metil Éster/farmacología , Óxido Nítrico Sintasa de Tipo II/antagonistas & inhibidores , Óxido Nítrico Sintasa de Tipo II/biosíntesis , Especies Reactivas de Oxígeno/metabolismo , Marcadores de Spin , Superóxido Dismutasa/biosíntesis , Factores de Transcripción/metabolismo , Ácido Úrico/farmacología
19.
J Pharmacol Exp Ther ; 331(1): 23-34, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19587315

RESUMEN

Clinical studies suggest that moderate alcohol consumption can have beneficial effects, in particular regarding cardiovascular events, insulin resistance, and type 2 diabetes. In this study, lean and obese diabetic ob/ob mice were submitted or not to chronic ethanol intake via the drinking water for 6 months, which was associated with moderate levels of plasma ethanol. Plasma levels of alanine aminotransferase and aspartate aminotransferase were not increased by alcohol intake. Ethanol consumption progressively reduced the gain of body weight in ob/ob mice, but not in lean mice, and this was observed despite higher calorie intake. Increased plasma free fatty acids and glycerol in ethanol-treated ob/ob mice suggested peripheral lipolysis. Glycemia and insulinemia were significantly reduced, whereas adiponectinemia was increased in ethanol-treated ob/ob mice. Liver weight and triglycerides were significantly decreased in ethanol-treated ob/ob mice, and this was associated with less microvesicular steatosis. Hepatic levels of AMP-activated protein kinase and the phosphorylated form of acetyl-CoA carboxylase were higher in ethanol-treated ob/ob mice, suggesting better fatty acid oxidation. However, hepatic mRNA expression of several lipogenic genes was not reduced by ethanol consumption. Finally, mild oxidative stress was noticed in the liver of ethanol-treated mice, regardless of their genotype. Hence, our data are in keeping with clinical studies suggesting that moderate ethanol intake can have beneficial effects on type 2 diabetes and insulin sensitivity, at least in part through increased levels of plasma adiponectin. However, further studies are needed to determine whether long-term drinking of light-to-moderate amounts of ethanol is safe for the liver.


Asunto(s)
Consumo de Bebidas Alcohólicas/metabolismo , Diabetes Mellitus Experimental/metabolismo , Hígado/metabolismo , Triglicéridos/metabolismo , Aumento de Peso/fisiología , Animales , Peso Corporal/efectos de los fármacos , Peso Corporal/fisiología , Diabetes Mellitus Experimental/prevención & control , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/prevención & control , Etanol/administración & dosificación , Humanos , Hígado/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Triglicéridos/antagonistas & inhibidores , Aumento de Peso/efectos de los fármacos
20.
Toxicol Appl Pharmacol ; 234(3): 326-38, 2009 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-19063909

RESUMEN

Alcohol consumption increases reactive oxygen species formation and lipid peroxidation, whose products can damage mitochondrial DNA (mtDNA) and alter mitochondrial function. A possible role of manganese superoxide dismutase (MnSOD) on these effects has not been investigated. To test whether MnSOD overexpression modulates alcohol-induced mitochondrial alterations, we added ethanol to the drinking water of transgenic MnSOD-overexpressing (TgMnSOD) mice and their wild type (WT) littermates for 7 weeks. In TgMnSOD mice, alcohol administration further increased the activity of MnSOD, but decreased cytosolic glutathione as well as cytosolic glutathione peroxidase activity and peroxisomal catalase activity. Whereas ethanol increased cytochrome P-450 2E1 and mitochondrial ROS generation in both WT and TgMnSOD mice, hepatic iron, lipid peroxidation products and respiratory complex I protein carbonyls were only increased in ethanol-treated TgMnSOD mice but not in WT mice. In ethanol-fed TgMnSOD mice, but not ethanol-fed WT mice, mtDNA was depleted, and mtDNA lesions blocked the progress of polymerases. The iron chelator, DFO prevented hepatic iron accumulation, lipid peroxidation, protein carbonyl formation and mtDNA depletion in alcohol-treated TgMnSOD mice. Alcohol markedly decreased the activities of complexes I, IV and V of the respiratory chain in TgMnSOD, with absent or lesser effects in WT mice. There was no inflammation, apoptosis or necrosis, and steatosis was similar in ethanol-treated WT and TgMnSOD mice. In conclusion, prolonged alcohol administration selectively triggers iron accumulation, lipid peroxidation, respiratory complex I protein carbonylation, mtDNA lesions blocking the progress of polymerases, mtDNA depletion and respiratory complex dysfunction in TgMnSOD mice but not in WT mice.


Asunto(s)
Consumo de Bebidas Alcohólicas/efectos adversos , Daño del ADN , ADN Mitocondrial/metabolismo , Etanol/toxicidad , Hígado/efectos de los fármacos , Mitocondrias Hepáticas/efectos de los fármacos , Superóxido Dismutasa/metabolismo , Animales , Peso Corporal , Caspasa 3/metabolismo , Catalasa/metabolismo , Citocromo P-450 CYP2E1/metabolismo , Proteínas de Unión al ADN/metabolismo , Deferoxamina/farmacología , Regulación hacia Abajo , Complejo I de Transporte de Electrón/metabolismo , Etanol/sangre , Etanol/metabolismo , Glutatión/metabolismo , Glutatión Peroxidasa/metabolismo , Proteínas del Grupo de Alta Movilidad/metabolismo , Hierro/metabolismo , Quelantes del Hierro/farmacología , Peroxidación de Lípido/efectos de los fármacos , Hígado/enzimología , Hígado/ultraestructura , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mitocondrias Hepáticas/enzimología , Óxido Nítrico Sintasa de Tipo II/metabolismo , Estrés Oxidativo/efectos de los fármacos , Carbonilación Proteica/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Superóxido Dismutasa/genética , Sustancias Reactivas al Ácido Tiobarbitúrico/metabolismo , Factores de Transcripción/metabolismo , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA