Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
1.
J Cell Sci ; 129(16): 3153-66, 2016 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-27390173

RESUMO

Extracellular ligands control biological phenomena. Cells distinguish physiological stimuli from weak noise stimuli by establishing a ligand-concentration threshold. Hormonal control of the meiotic G2/M transition in oocytes is essential for reproduction. However, the mechanism for threshold establishment is unclear. In starfish oocytes, maturation-inducing hormones activate the PI3K-Akt pathway through the Gßγ complex of heterotrimeric G-proteins. Akt directly phosphorylates both Cdc25 phosphatase and Myt1 kinase, resulting in activation of cyclin-B-Cdk1, which then induces meiotic G2/M transition. Here, we show that cyclin-B-Cdk1 is partially activated after subthreshold hormonal stimuli, but this triggers negative feedback, resulting in dephosphorylation of Akt sites on Cdc25 and Myt1, thereby canceling the signal. We also identified phosphatase activity towards Akt substrates that exists independent of stimuli. In contrast to these negative regulatory activities, an atypical Gßγ-dependent pathway enhances PI3K-Akt-dependent phosphorylation. Based on these findings, we propose a model for threshold establishment in which hormonal dose-dependent competition between these new pathways establishes a threshold; the atypical Gßγ-pathway becomes predominant over Cdk-dependent negative feedback when the stimulus exceeds this threshold. Our findings provide a regulatory connection between cell cycle and signal transduction machineries.


Assuntos
Proteína Quinase CDC2/metabolismo , Ciclina B/metabolismo , Fase G2 , Meiose , Mitose , Estrelas-do-Mar/citologia , Estrelas-do-Mar/metabolismo , Adenina/análogos & derivados , Adenina/farmacologia , Animais , Ativação Enzimática/efeitos dos fármacos , Retroalimentação Fisiológica/efeitos dos fármacos , Fase G2/efeitos dos fármacos , Subunidades beta da Proteína de Ligação ao GTP/metabolismo , Humanos , Meiose/efeitos dos fármacos , Mitose/efeitos dos fármacos , Modelos Biológicos , Proteínas Mutantes/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação/efeitos dos fármacos , Fosfosserina/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Estrelas-do-Mar/efeitos dos fármacos , Especificidade por Substrato/efeitos dos fármacos , Fosfatases cdc25/metabolismo
2.
FASEB J ; 26(6): 2318-26, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22362895

RESUMO

Mallory-Denk bodies (MDBs) are hepatocyte inclusions that are associated with poor liver disease prognosis. The intermediate filament protein keratin 8 (K8) and its cross-linking by transglutaminase-2 (TG2) are essential for MDB formation. K8 hyperphosphorylation occurs in association with liver injury and MDB formation, but the link between keratin phosphorylation and MDB formation is unknown. We used a mutational approach to identify K8 Q70 as a residue that is important for K8 cross-linking to itself and other liver proteins. K8 cross-linking is markedly enhanced on treating cells with a phosphatase inhibitor and decreases dramatically on K8 S74A or Q70N mutation in the presence of phosphatase inhibition. K8 Q70 cross-linking, in the context of synthetic peptides or intact proteins transfected into cells, is promoted by phosphorylation at K8 S74 or by an S74D substitution and is inhibited by S74A mutation. Transgenic mice that express K8 S74A or a K8 G62C liver disease variant that inhibits K8 S74 phosphorylation have a markedly reduced ability to form MDBs. Our findings support a model in which the stress-triggered phosphorylation of K8 S74 induces K8 cross-linking by TG2, leading to MDB formation. These findings may extend to neuropathies and myopathies that are characterized by intermediate filament-containing inclusions.


Assuntos
Hepatócitos/ultraestrutura , Queratina-8/metabolismo , Hepatopatias/fisiopatologia , Corpos de Mallory/fisiologia , Sequência de Aminoácidos , Animais , Proteínas de Ligação ao GTP , Glutamina/metabolismo , Filamentos Intermediários/metabolismo , Queratina-8/genética , Camundongos , Camundongos Transgênicos , Fosforilação , Proteína 2 Glutamina gama-Glutamiltransferase , Transglutaminases
3.
Gastroenterology ; 138(4): 1607-17, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20064513

RESUMO

BACKGROUND & AIMS: Mallory-Denk bodies (MDBs) are keratin (K)-rich cytoplasmic hepatocyte inclusions commonly associated with alcoholic steatohepatitis. Given the significant gender differences in predisposition to human alcohol-related liver injury, and the strain difference in mouse MDB formation, we hypothesized that sex affects MDB formation. METHODS: MDBs were induced in male and female mice overexpressing K8, which are predisposed to MDB formation, and in nontransgenic mice by feeding the porphyrinogenic compound 3,5-diethoxycarbonyl-1,4-dihydrocollidine (DDC). MDB presence was determined by histologic, immunofluorescence, and biochemical analyses and correlated to liver injury using serologic and pathologic markers. Cytoskeletal and metabolic liver protein analysis, in vitro metabolism studies, and measurement of oxidative stress markers and protoporphyrin-IX were performed. RESULTS: Male mice formed significantly more MDBs, which was attenuated modestly by estradiol. MDB formation was accompanied by increased oxidative stress. Female mice had significantly fewer MDBs and oxidative stress-related changes, but had increased ductular reaction protoporphyrin-IX accumulation, and MDB-preventive K18 induction. Evaluation of the microsomal cytochrome-P450 (CYP) enzymes revealed significant gender differences in protein expression and activity in untreated and DDC-fed mice, and showed that DDC is metabolized by CYP3A. The changes in CYPs account for the gender differences in porphyria and DDC metabolism. DDC metabolite formation and oxidative injury accumulate on chronic DDC exposure in males, despite more efficient acute metabolism in females. CONCLUSIONS: Gender dimorphic formation of MDBs and porphyria associate with differences in CYPs, oxidative injury, and selective keratin induction. These findings may extend to human MDBs and other neuropathy- and myopathy-related inclusions.


Assuntos
Hepatócitos/metabolismo , Corpos de Inclusão/metabolismo , Estresse Oxidativo , Xenobióticos/metabolismo , Animais , Sistema Enzimático do Citocromo P-450/fisiologia , Feminino , Masculino , Camundongos , Microssomos Hepáticos/metabolismo , Porfirias/metabolismo , Protoporfirinas/metabolismo , Piridinas/farmacologia , Caracteres Sexuais
4.
Gastroenterology ; 139(3): 828-35, 835.e1-3, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20538000

RESUMO

BACKGROUND & AIMS: Keratins 8 and 18 (K8/K18) provide anti-apoptotic functions upon liver injury. The cytoprotective function of keratins explains the overrepresentation of K8/K18 variants in patients with cirrhosis. However, K8/K18 variant-associated susceptibility to acute liver injury, which is well-described in animal models, has not been studied in humans. METHODS: We analyzed the entire coding regions of KRT8 and KRT18 genes (15 total exons and their exon-intron boundaries) to determine the frequency of K8/K18 variants in 344 acute liver failure (ALF) patients (49% acetaminophen-related) and 2 control groups (African-American [n = 245] and previously analyzed white [n = 727] subjects). RESULTS: Forty-five ALF patients had significant amino-acid-altering K8/K18 variants, including 23 with K8 R341H and 11 with K8 G434S. K8 variants were significantly more common (total of 42 patients) than K18 variants (3 patients) (P < .001). We found increased frequency of variants in white ALF patients (9.1%) versus controls (3.7%) (P = .01). K8 R341H was more common in white (P = .01) and G434S was more common in African-American (P = .02) ALF patients versus controls. White patients with K8/K18 variants were less likely to survive ALF without transplantation (P = .02). K8 A333A and G434S variants associated exclusively with African Americans (23% combined frequency in African American but none in white controls; P < .0001), while overall, K18 variants were more common in non-white liver-disease subjects compared to whites (2.8% vs 0.6%, respectively; P = .008). CONCLUSIONS: KRT8 and KRT18 are important susceptibility genes for ALF development. Presence of K8/K18 variants predisposes to adverse ALF outcome, and some variants segregate with unique ethnic and race backgrounds.


Assuntos
Negro ou Afro-Americano/genética , Doença Hepática Induzida por Substâncias e Drogas/genética , Queratina-18/genética , Queratina-8/genética , Falência Hepática Aguda/genética , População Branca/genética , Acetaminofen/efeitos adversos , Adulto , Analgésicos não Narcóticos/efeitos adversos , Asiático/genética , Doença Hepática Induzida por Substâncias e Drogas/etnologia , Doença Hepática Induzida por Substâncias e Drogas/mortalidade , Distribuição de Qui-Quadrado , Feminino , Frequência do Gene , Predisposição Genética para Doença , Hispânico ou Latino/genética , Humanos , Indígenas Norte-Americanos/genética , Falência Hepática Aguda/induzido quimicamente , Falência Hepática Aguda/etnologia , Falência Hepática Aguda/mortalidade , Modelos Logísticos , Masculino , Pessoa de Meia-Idade , Razão de Chances , Fenótipo , Prognóstico , Estudos Prospectivos , Medição de Risco , Fatores de Risco , Estados Unidos/epidemiologia , Adulto Jovem
5.
Exp Cell Res ; 315(2): 119-26, 2009 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-19007772

RESUMO

Wilson disease is a genetic disorder characterized by the accumulation of copper in the body by defective biliary copper excretion. Wilson disease gene product (ATP7B) functions in copper incorporation to ceruloplasmin (Cp) and biliary copper excretion. However, copper metabolism in hepatocytes has been still unclear. Niemann-Pick disease type C (NPC) is a lipid storage disorder and the most commonly mutated gene is NPC1 and its gene product NPC1 is a late endosome protein and regulates intracellular vesicle traffic. In the present study, we induced NPC phenotype and examined the localization of ATP7B and secretion of holo-Cp, a copper-binding mature form of Cp. The vesicle traffic was modulated using U18666A, which induces NPC phenotype, and knock down of NPC1 by RNA interference. ATP7B colocalized with the late endosome markers, but not with the trans-Golgi network markers. U18666A and NPC1 knock down decreased holo-Cp secretion to culture medium, but did not affect the secretion of other secretory proteins. Copper accumulated in the cells after the treatment with U18666A. These findings suggest that ATP7B localizes in the late endosomes and that copper in the late endosomes is transported to the secretory compartment via NPC1-dependent pathway and incorporated into apo-Cp to form holo-Cp.


Assuntos
Adenosina Trifosfatases/metabolismo , Proteínas de Transporte/fisiologia , Proteínas de Transporte de Cátions/metabolismo , Cobre/metabolismo , Endossomos/metabolismo , Glicoproteínas de Membrana/fisiologia , Subunidades gama do Complexo de Proteínas Adaptadoras/metabolismo , Adenosina Trifosfatases/genética , Androstenos/farmacologia , Anticolesterolemiantes/farmacologia , Transporte Biológico/efeitos dos fármacos , Transporte Biológico/fisiologia , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patologia , Proteínas de Transporte/genética , Proteínas de Transporte de Cátions/genética , Linhagem Celular Tumoral , Ceruloplasmina/metabolismo , ATPases Transportadoras de Cobre , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patologia , Proteína 2 de Membrana Associada ao Lisossomo , Proteínas de Membrana Lisossomal/metabolismo , Glicoproteínas de Membrana/genética , Mutação , Proteína C1 de Niemann-Pick , Interferência de RNA , RNA Interferente Pequeno/genética , Proteínas rab de Ligação ao GTP/metabolismo , proteínas de unión al GTP Rab7
6.
Hepatology ; 48(3): 943-52, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18697208

RESUMO

UNLABELLED: Mallory-Denk bodies (MDBs) are hepatocyte inclusions found in several liver diseases and consist primarily of keratins 8 and 18 (K8/K18) and ubiquitin that are cross-linked by transglutaminase-2. We hypothesized that genetic variables contribute to the extent of MDB formation, because not all patients with an MDB-associated liver disease develop inclusions. We tested this hypothesis using five strains of mice (FVB/N, C3H/He, Balb/cAnN, C57BL/6, 129X1/Sv) fed for three months (eight mice per strain) the established MDB-inducing agent 3,5-diethoxycarbonyl-1,4-dihydrocollidine (DDC). MDB formation was compared using hematoxylin-and-eosin staining, or immunofluorescence staining with antibodies to K8/K18/ubiquitin, or biochemically by blotting with antibodies to transglutaminase-2/p62 proteins and to K8/K18/ubiquitin to detect keratin cross-linking. DDC feeding induced MDBs in all mouse strains, but there were dramatic strain differences that quantitatively varied 2.5-fold (P < 0.05). MDB formation correlated with hepatocyte ballooning, and most ballooned hepatocytes had MDBs. Immunofluorescence assessment was far more sensitive than hematoxylin-and-eosin staining in detecting small MDBs, which out-numbered (by approximately 30-fold to 90-fold) but did not parallel their large counterparts. MDB scores partially reflected the biochemical presence of cross-linked keratin-ubiquitin species but not the changes in liver size or injury in response to DDC. The extent of steatosis correlated with the total (large+small) number of MDBs, and there was a limited correlation between large MDBs and acidophil bodies. CONCLUSION: Mouse MDB formation has important genetic contributions that do not correlate with the extent of DDC-induced liver injury. If extrapolated to humans, the genetic contributions help explain why some patients develop MDBs whereas others are less likely to do so. Detection and classification of MDBs using MDB-marker-selective staining may offer unique links to specific histological features of DDC-induced liver injury.


Assuntos
Predisposição Genética para Doença/genética , Corpos de Inclusão/genética , Filamentos Intermediários/genética , Hepatopatias/genética , Camundongos Endogâmicos/genética , Animais , Dicarbetoxi-Di-Hidrocolidina/farmacologia , Hepatócitos/metabolismo , Hepatócitos/patologia , Hipertrofia , Corpos de Inclusão/metabolismo , Filamentos Intermediários/metabolismo , Queratina-18/metabolismo , Queratina-8/metabolismo , Fígado/efeitos dos fármacos , Fígado/metabolismo , Fígado/patologia , Hepatopatias/metabolismo , Hepatopatias/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Ubiquitina/metabolismo
7.
Hepatology ; 47(6): 2026-35, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18454506

RESUMO

UNLABELLED: The proteasomal and lysosomal/autophagy pathways in the liver and other tissues are involved in several biological processes including the degradation of misfolded proteins. Exposure of hepatocyte cell lines to proteasome inhibitors (PIs) results in the formation of inclusions that resemble Mallory-Denk bodies (MDBs). Keratins are essential for MDB formation and keratin 8 (K8)-overexpressing transgenic mice are predisposed to MDB formation. We tested the hypothesis that PIs induce MDBs in vivo and that autophagy participates in MDB turnover. The effect of the PI bortezomib (which is used to treat some malignancies) on MDB formation was tested in K8-overexpressing mice and in cultured cells. Inclusion formation was examined using immune and conventional electron microscopy (EM). Bortezomib induced MDB-like inclusions composed of keratins, ubiquitin, and p62 in cultured cells. Short-term exposure to bortezomib induced similar inclusions in K8-overexpressing but not in nontransgenic mice, without causing liver injury. In bortezomib-treated mice, autophagy was activated in hepatocytes as determined by EM and biochemical analysis. Further activation of autophagy by rapamycin (Rap) decreased the number of inclusions in bortezomib-treated K8 transgenic mice significantly. Rap also led to resorption of spontaneously formed MDBs in aging K8-overexpressing mice. Immune EM demonstrated K8-positive and ubiquitin-positive structures in autophagic vacuoles in the mouse liver. CONCLUSION: PIs alone are sufficient to induce MDBs in susceptible animals, while Rap-mediated activation of autophagy prevents MDB formation and causes MDB resorption. These findings suggest that some patients treated with PIs may become predisposed to MDB formation. Autophagy provides a potential cellular mechanism for the resorption of cytoplasmic inclusions.


Assuntos
Autofagia/efeitos dos fármacos , Hepatócitos/efeitos dos fármacos , Imunossupressores/farmacologia , Corpos de Inclusão/efeitos dos fármacos , Inibidores de Proteassoma , Sirolimo/farmacologia , Animais , Ácidos Borônicos/farmacologia , Bortezomib , Linhagem Celular Tumoral , Hepatócitos/metabolismo , Hepatócitos/patologia , Humanos , Corpos de Inclusão/metabolismo , Corpos de Inclusão/patologia , Queratina-8/genética , Queratina-8/metabolismo , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Modelos Animais , Inibidores de Proteases/farmacologia , Pirazinas/farmacologia , Transfecção
8.
Biochem Biophys Res Commun ; 367(1): 33-40, 2008 Feb 29.
Artigo em Inglês | MEDLINE | ID: mdl-18164256

RESUMO

Microtubules (MTs) and microfilaments (MFs) are known to modulate mitochondrial morphology, distribution and function. However, little is known evidence about the role of intermediate filaments (IFs) in modulating mitochondria except desmin. To investigate whether or not the IFs regulate mitochondrial morphology, distribution, and function, we manipulated the IFs of cultured epithelial cells to express a mutant keratin 18 (K18). In contrast to the filamentous expression of wild K18, mutant K18 induced aggregation of K8/18, showing no fine IF network in the cells. In mutant K18-transfected cells, the mitochondria were fragmented into small spheroids, although they were observed as mitochondrial fibers in un-transfected or wild K18-transfected cells. Fluorescence recovery after photobleaching of fluorescence-labeled mitochondria was markedly less in the mutant K18-transfected cells, although a significant recovery was confirmed in wild K18-transfected cells. These findings suggest that the IFs are important for the maintenance of normal mitochondrial structures.


Assuntos
Citoesqueleto de Actina/metabolismo , Células Epiteliais/patologia , Queratina-18/genética , Hepatopatias/patologia , Mitocôndrias Hepáticas/metabolismo , Citoesqueleto de Actina/genética , Citoesqueleto de Actina/patologia , Animais , Células Cultivadas , Células Epiteliais/metabolismo , Corantes Fluorescentes/química , Humanos , Hepatopatias/genética , Hepatopatias/metabolismo , Microscopia Confocal , Microscopia de Fluorescência , Mitocôndrias Hepáticas/genética , Mitocôndrias Hepáticas/patologia , Mutação , Transfecção
9.
Hepatol Res ; 37(10): 828-35, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17573944

RESUMO

AIM: Mallory bodies have been observed in various liver diseases, however, the precise mechanism and significance of these structures have yet to be determined. METHODS: Previously we reported on the redistribution of cytosolic proteins to keratin inclusions in mutant keratin 18-transfected cells. In this study, we treated green fluorescent protein-tagged wild-type keratin 18-transfected cells with several proteasome inhibitors and performed immunofluorescent analyses. RESULTS: Proteasome inhibitors induced intracellular keratin inclusions, and desmoplakin, zonula occludens-1 and beta-catenin were relocated to keratin inclusions, while theintegral membrane proteins were intact. The cytosolic proteins, 14-3-3 zeta protein and glucose-6-phosphate dehydrogenase were also relocated to inclusions. Moreover, E-cadherin, a basolateral membrane protein, was present on both the apical and basolateral domains in inclusion-containing cells. CONCLUSION: These data are identical to those in the mutant keratin 18 transfection study and suggest that keratin inclusions induced by different treatments affect localization of various cytosolic components, which may influence cellular functions performed by these proteins.

10.
Int J Mol Med ; 15(3): 431-6, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15702233

RESUMO

Intrahepatic cholestasis has been recognized as one of the characteristic features of the hepatic manifestations in graft-versus-host disease (GVHD). Tight junctions (TJs) play crucial roles in bile formation and alterations of TJs in hepatocytes and/or biliary epithelial cells (BECs) that cause intrahepatic cholestasis. To assess the changes of TJs in hepatocytes and BECs in a rat model of GVHD, we examined the localization of TJ-associated proteins, 7H6 and zonula occludens (ZO)-1. GVHD was induced by injecting spleen cells of parental strain rats (Brown Norway) into non-irradiated (Brown Norway x Lewis) F1 hybrid rats. Untreated F1 hybrid rats served as controls. Double-labeled immunofluorescent staining for 7H6 and ZO-1 was performed in liver sections. In control rats, immunostaining for 7H6 and ZO-1 colocalized to the apical site of BECs and was continuous along the bile canaliculi. In GVHD, 7H6 expression was decreased in BECs and was discontinuous along the bile canaliculi. On the other hand, the intensity of ZO-1 staining in hepatocytes increased and did not change in BECs compared with that of control rats. Changes in TJ-associated proteins of both hepatocytes and BECs may reflect the immunopathogenesis of GVHD-associated intrahepatic cholestasis.


Assuntos
Colestase Intra-Hepática/complicações , Colestase Intra-Hepática/metabolismo , Modelos Animais de Doenças , Doença Enxerto-Hospedeiro/complicações , Proteínas de Membrana/metabolismo , Fosfoproteínas/metabolismo , Junções Íntimas/metabolismo , Animais , Bile/metabolismo , Colestase Intra-Hepática/genética , Feminino , Doença Enxerto-Hospedeiro/genética , Doença Enxerto-Hospedeiro/metabolismo , Imuno-Histoquímica , Masculino , Ratos , Proteína da Zônula de Oclusão-1
11.
Int J Mol Med ; 10(5): 647-8, 2002 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-12373309

RESUMO

Lamivudine has previously been found to be effective not only in patients with compensated liver disease due to hepatitis B virus (HBV) but also in those with hepatic decompensation. However, long-term follow-up of patients with hepatic encephalopathy (HE) has not been previously reported. We describe a patient with recurrent HE associated with decompensated liver cirrhosis due to hepatitis B. After the initiation of treatment with lamivudine, manifestation of HE has not been observed for about 2 years and liver function has improved as well. This experience suggests that improved liver function using lamivudine may contribute to prevention from recurrence of HE.


Assuntos
Antivirais/uso terapêutico , Encefalopatia Hepática/prevenção & controle , Hepatite B/complicações , Hepatite B/tratamento farmacológico , Lamivudina/uso terapêutico , Cirrose Hepática/tratamento farmacológico , Cirrose Hepática/etiologia , Feminino , Humanos , Pessoa de Meia-Idade , Recidiva
12.
Int J Mol Med ; 9(6): 621-6, 2002 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12011979

RESUMO

Primary sclerosing cholangitis (PSC) is known to be frequently associated with inflammatory bowel diseases. In a rat with self-filling blind loop (SFBL), a proposed animal model for PSC, hepatobiliary inflammation has previously been demonstrated. In this study, we assessed the involvement of lipopolysaccharide (LPS), a bacterial endotoxin, in the pathogenesis of hepatobiliary inflammation of the SFBL model. The hepatic localization of LPS was examined by immunohistochemistry using an anti-lipid A antibody. The portal blood concentration of LPS was measured by an endotoxin-specific chromogenic Limulus test (Endospecy test). LPS was localized in the biliary epithelial cells (BECs) of rats with SFBL, and the portal blood concentration of LPS was significantly higher than that of sham-operated rats. Development of hepatobiliary inflammation, peribiliary fibrosis, and injury to the intestinal mucosa were histologically confirmed. Constriction in the biliary trees was radiologically demonstrated. These findings suggested that abnormal accumulation of LPS, which may be derived from portal blood, in BECs was involved in the pathogenesis of hepatobiliary inflammation with intestinal injury.


Assuntos
Ductos Biliares/anatomia & histologia , Ductos Biliares/patologia , Células Epiteliais/metabolismo , Lipopolissacarídeos/metabolismo , Animais , Colangiografia , Imuno-Histoquímica , Inflamação , Lipídeo A/imunologia , Lipopolissacarídeos/sangue , Masculino , Microscopia Eletrônica , Ligação Proteica , Ratos , Ratos Wistar , Fatores de Tempo
13.
Int J Mol Med ; 11(3): 293-8, 2003 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-12579329

RESUMO

Wilson disease is a genetic disorder characterized by the accumulation of copper in the body due to a defect of biliary copper excretion. The gene responsible for Wilson disease has been cloned, however, the precise localization of this gene product ATP7B, a copper-transporting ATPase, is still controversial. We examined the localization of ATP7B by expressing a chimeric protein, ATP7B-tagged with green fluorescent protein (GFP) (GFP-ATP7B), in HEK293, Hep3B and a highly polarized human hepatocyte line (OUMS29). Intracellular organelles were visualized by immunofluorescence microscopy. The effects of bathocuproine disulfonate, a copper chelator, and copper sulfate were examined. GFP-ATP7B colocalized with a late endosome marker, but not with endoplasmic reticulum, Golgi, or lysosome markers in a copper-depleting condition. Treatment with copper sulfate did not affect the localization of ATP7B. ATP7B is localized in the late endosomes in both copper-depleting and copper-loaded conditions. ATP7B seems to translocate copper from the cytosol into the late endosomes, and copper may be excreted to bile via lysosomes. We believe that the disturbed incorporation of copper into the late endosomes caused by mutated ATP7B is the main defect in Wilson disease.


Assuntos
Adenosina Trifosfatases/metabolismo , Proteínas de Transporte de Cátions/metabolismo , Endossomos/metabolismo , Hepatócitos/metabolismo , Adenosina Trifosfatases/efeitos dos fármacos , Adenosina Trifosfatases/genética , Proteínas de Transporte de Cátions/efeitos dos fármacos , Proteínas de Transporte de Cátions/genética , Linhagem Celular , Polaridade Celular , Quelantes/metabolismo , Quelantes/farmacologia , Cobre/metabolismo , Sulfato de Cobre/farmacologia , ATPases Transportadoras de Cobre , Endossomos/efeitos dos fármacos , Proteínas de Fluorescência Verde , Humanos , Proteínas Luminescentes/metabolismo , Mutação , Fenantrolinas/farmacologia , Proteínas Recombinantes/metabolismo , Transfecção
14.
Hepatol Res ; 26(2): 114-118, 2003 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12809938

RESUMO

A 77-year-old man with pneumonia associated with acute myeloid leukemia was introduced to the hepatology unit at our hospital for hyperbilirubinemia. He had been suffering from a high fever because of pneumonia. He was icteric and his serum concentrations of total and direct bilirubin were 13.1 and 7.9 mg/dl, respectively. However, the other standard biochemical examinations for hepatic function, such as serum concentrations of aspartate aminotransferase, alanine aminotransferase, gamma-glutamyl transpeptidase and alkaline phosphatase were normal except for lactate dehydrogenase. Lactate dehydrogenase isoenzyme analysis revealed that the high concentration was derived from leukemia cells. Ultrasonography of the abdomen revealed no abnormality in the liver or biliary tract. Administration of antibiotics for pneumonia decreased the serum bilirubin concentration, however, he died because of respiratory failure caused by the progression of pneumonia at 33 days after the admission. It was suggested that a disturbance in the bilirubin metabolism without hepatocyte necrosis or mechanical cholestasis might be involved in the pathogenesis of hyperbilirubinemia in patients with infectious diseases.

15.
J Histochem Cytochem ; 60(6): 475-83, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22473941

RESUMO

Mallory-Denk bodies (MDBs) are hepatocyte cytoplasmic inclusions found in several liver diseases and consist primarily of the cytoskeletal proteins, keratins 8 and 18 (K8/K18). Recent evidence indicates that the extent of stress-induced protein misfolding, a K8>K18 overexpression state, and transglutaminase-2 activation promote MDB formation. In addition, the genetic background and gender play an important role in mouse MDB formation, but the effect of aging on this process is unknown. Given that oxidative stress increases with aging, the authors hypothesized that aging predisposes to MDB formation. They used an established mouse MDB model-namely, feeding non-transgenic male FVB/N mice (1, 3, and 8 months old) with 3,5 diethoxycarbonyl-1,4-dihydrocollidine for 2 months. MDB formation was assessed using immunofluorescence staining and biochemically by demonstrating keratin and ubiquitin-containing crosslinks generated by transglutaminase-2. Immunofluorescence staining showed that old mice had a significant increase in MDB formation compared with young mice. MDB formation paralleled the generation of high molecular weight ubiquitinated keratin-containing complexes and induction of p62. Old mouse livers had increased oxidative stress. In addition, 20S proteasome activity and autophagy were decreased, and endoplasmic reticulum stress was increased in older livers. Therefore, aging predisposes to experimental MDB formation, possibly by decreased activity of protein degradation machinery.


Assuntos
Envelhecimento/patologia , Fígado/patologia , Corpos de Mallory/patologia , Estresse Oxidativo , Envelhecimento/metabolismo , Animais , Autofagia , Estresse do Retículo Endoplasmático , Imunofluorescência , Hipertrofia , Fígado/metabolismo , Masculino , Camundongos , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteólise
16.
Hepatol Res ; 42(6): 591-600, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22221855

RESUMO

AIM: Hepatitis C virus (HCV) core protein critically contributes to hepatocarcinogenesis, which is often observed in liver cirrhosis. Since the liver cirrhosis microenvironment is affected by hypoxia, we focused on the possible driving force of HCV core protein on signal relay from hypoxia-inducible factor (HIF)-1α to vascular endothelial growth factor (VEGF). METHODS: Human hepatocellular carcinoma cells stably overexpressing HCV core (Core cells) and NS5A (NS5A cells) were established; empty vector-transfected (EV) cells were used as controls. Hypoxia was induced by oxygen deprivation or by using cobalt chloride (CoCl(2) ). YC-1 was used to inhibit HIF-1α expression. Protein analyses for cultured cells and liver tissues obtained from CoCl(2) -treated HCV core-transgenic (Core-Tg) mice were performed by western blot and/or immunocytochemistry. Cellular mRNA levels were evaluated by quantitative real-time reverse transcription-polymerase chain reaction. RESULTS: Under hypoxia, the sustained expression of HIF-1α, but not HIF-2α, was profoundly observed in Core cells but, was faint in EV and NS5A cells. Immunocytochemistry revealed increased HIF-1α in the nucleus. HIF-1α mRNA levels were significantly higher in Core cells than in EV cells under both normoxia and hypoxia. The HIF-1α-targeted VEGF and Bcl-xL expressions were increased in Core cells under hypoxia and abolished by YC-1 treatment. Hypoxic liver samples of Core-Tg mice indicated significant increases in both HIF-1α and VEGF expression compared with the wild type. CONCLUSIONS: Hepatitis C virus core protein has the distinct potential to transcriptionally upregulate and sustain HIF-1α expression under hypoxia, thereby contributing to increased VEGF expression, a key regulator in the hypoxic milieu of liver cirrhosis.

17.
Mol Cell Biol ; 31(12): 2392-403, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21482667

RESUMO

Chronic exposure of the liver to hepatotoxic agents initiates an aberrant wound healing response marked by proinflammatory, as well as fibrotic, changes, leading to compromised organ structure and function. In a variety of pathological states, correlative links have been established between tissue fibrosis and the expression of transcription factors associated with the induction of epithelial-mesenchymal cell transition (EMT) programs similar to those engaged during development. However, the role played by endogenously derived, EMT-associated transcription factors in fibrotic states in vivo remains undefined. Using a mouse model of acute liver fibrosis, we demonstrate that hepatocytes upregulate the expression of the zinc finger transcriptional repressor, Snail1, during tissue remodeling. Hepatocyte-specific ablation of Snail1 demonstrates that this transcription factor plays a key role in liver fibrosis progression in vivo by triggering the proximal genetic programs that control multiple aspects of fibrogenesis, ranging from growth factor expression and extracellular matrix biosynthesis to the ensuing chronic inflammatory responses that characterize this class of pathological disorders.


Assuntos
Progressão da Doença , Hepatócitos/fisiologia , Cirrose Hepática/patologia , Cirrose Hepática/fisiopatologia , Fatores de Transcrição/metabolismo , Animais , Células Cultivadas , Transição Epitelial-Mesenquimal/fisiologia , Perfilação da Expressão Gênica , Hepatócitos/citologia , Masculino , Camundongos , Camundongos Transgênicos , Análise em Microsséries , Fatores de Transcrição da Família Snail , Fatores de Transcrição/genética
18.
J Cell Biol ; 195(2): 217-29, 2011 Oct 17.
Artigo em Inglês | MEDLINE | ID: mdl-22006949

RESUMO

Genetic factors impact liver injury susceptibility and disease progression. Prominent histological features of some chronic human liver diseases are hepatocyte ballooning and Mallory-Denk bodies. In mice, these features are induced by 3,5-diethoxycarbonyl-1,4-dihydrocollidine (DDC) in a strain-dependent manner, with the C57BL and C3H strains showing high and low susceptibility, respectively. To identify modifiers of DDC-induced liver injury, we compared C57BL and C3H mice using proteomic, biochemical, and cell biological tools. DDC elevated reactive oxygen species (ROS) and oxidative stress enzymes preferentially in C57BL livers and isolated hepatocytes. C57BL livers and hepatocytes also manifested significant down-regulation, aggregation, and nuclear translocation of glyceraldehyde 3-phosphate dehydrogenase (GAPDH). GAPDH knockdown depleted bioenergetic and antioxidant enzymes and elevated hepatocyte ROS, whereas GAPDH overexpression decreased hepatocyte ROS. On the other hand, C3H livers had higher expression and activity of the energy-generating nucleoside-diphosphate kinase (NDPK), and knockdown of hepatocyte NDPK augmented DDC-induced ROS formation. Consistent with these findings, cirrhotic, but not normal, human livers contained GAPDH aggregates and NDPK complexes. We propose that GAPDH and NDPK are genetic modifiers of murine DDC-induced liver injury and potentially human liver disease.


Assuntos
Gliceraldeído-3-Fosfato Desidrogenases/genética , Hepatócitos/patologia , Corpos de Inclusão/patologia , Hepatopatias/etiologia , Hepatopatias/patologia , Núcleosídeo-Difosfato Quinase/genética , Animais , Regulação Enzimológica da Expressão Gênica , Hepatócitos/enzimologia , Humanos , Hepatopatias/enzimologia , Hepatopatias/genética , Camundongos , Camundongos Endogâmicos , Estresse Oxidativo/genética , Piridinas , Espécies Reativas de Oxigênio
19.
Hepatol Res ; 41(5): 484-91, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21518405

RESUMO

AIM: Wilson disease is a genetic disorder of copper metabolism characterized by impaired biliary copper excretion. Wilson disease gene product (ATP7B) functions in copper incorporation to ceruloplasmin (Cp) and biliary copper excretion. Our previous study showed the late endosome localization of ATP7B and described the copper transport pathway from the late endosome to trans-Golgi network (TGN). However, the cellular localization of ATP7B and copper metabolism in hepatocytes remains controversial. The present study was performed to evaluate the role of Niemann-Pick type C (NPC) gene product NPC1 on intracellular copper transport in hepatocytes. METHODS: We induced the NPC phenotype using U18666A to modulate the vesicle traffic from the late endosome to TGN. Then, we examined the effect of NPC1 overexpression on the localization of ATP7B and secretion of holo-Cp, a copper-binding mature form of Cp. RESULTS: Overexpression of NPC1 increased holo-Cp secretion to culture medium of U18666A-treated cells, but did not affect the secretion of albumin. Manipulation of NPC1 function affected the localization of ATP7B and late endosome markers, but did not change the localization of a TGN marker. ATP7B co-localized with the late endosome markers, but not with the TGN marker. CONCLUSION: These findings suggest that ATP7B localizes in the late endosomes and that copper in the late endosomes is transported to the secretory compartment via an NPC1-dependent pathway and incorporated into Cp.

20.
J Clin Invest ; 119(7): 1794-805, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19587454

RESUMO

Simple epithelial keratins (SEKs) are found primarily in single-layered simple epithelia and include keratin 7 (K7), K8, K18-K20, and K23. Genetically engineered mice that lack SEKs or overexpress mutant SEKs have helped illuminate several keratin functions and served as important disease models. Insight into the contribution of SEKs to human disease has indicated that K8 and K18 are the major constituents of Mallory-Denk bodies, hepatic inclusions associated with several liver diseases, and are essential for inclusion formation. Furthermore, mutations in the genes encoding K8, K18, and K19 predispose individuals to a variety of liver diseases. Hence, as we discuss here, the SEK cytoskeleton is involved in the orchestration of several important cellular functions and contributes to the pathogenesis of human liver disease.


Assuntos
Queratinas/fisiologia , Animais , Biomarcadores Tumorais , Humanos , Queratinas/análise , Queratinas/química , Queratinas/genética , Hepatopatias/etiologia , Camundongos , Camundongos Transgênicos , Mutação , Metástase Neoplásica , Neoplasias Epiteliais e Glandulares/diagnóstico , Neoplasias Epiteliais e Glandulares/terapia , Proteínas/fisiologia
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa