Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
J Neurochem ; 157(6): 1930-1945, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33539571

RESUMO

Ketogenic diets (KDs) alter brain metabolism. Multiple mechanisms may account for their effects, and different brain regions may variably respond. Here, we considered how a KD affects brain neuron and astrocyte transcription. We placed male C57Bl6/N mice on either a 3-month KD or chow diet, generated enriched neuron and astrocyte fractions, and used RNA-Seq to assess transcription. Neurons from KD-treated mice generally showed transcriptional pathway activation while their astrocytes showed a mix of transcriptional pathway suppression and activation. The KD especially affected pathways implicated in mitochondrial and endoplasmic reticulum function, insulin signaling, and inflammation. An unbiased analysis of KD-associated expression changes strongly implicated transcriptional pathways altered in AD, which prompted us to explore in more detail the potential molecular relevance of a KD to AD. Our results indicate a KD differently affects neurons and astrocytes, and provide unbiased evidence that KD-induced brain effects are potentially relevant to neurodegenerative diseases such as AD.


Assuntos
Astrócitos/metabolismo , Encéfalo/metabolismo , Dieta Cetogênica/métodos , Corpos Cetônicos/metabolismo , Neurônios/metabolismo , Transcrição Gênica/fisiologia , Animais , Dieta Cetogênica/tendências , Corpos Cetônicos/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL
2.
Br J Cancer ; 124(1): 166-175, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33024269

RESUMO

BACKGROUND: Previously, we identified ITIH5 as a suppressor of pancreatic ductal adenocarcinoma (PDAC) metastasis in experimental models. Expression of ITIH5 correlated with decreased cell motility, invasion and metastasis without significant inhibition of primary tumour growth. Here, we tested whether secretion of ITIH5 is required to suppress liver metastasis and sought to understand the role of ITIH5 in human PDAC. METHODS: We expressed mutant ITIH5 with deletion of the N-terminal secretion sequence (ITIH5Δs) in highly metastatic human PDAC cell lines. We used a human tissue microarray (TMA) to compare ITIH5 levels in uninvolved pancreas, primary and metastatic PDAC. RESULTS: Secretion-deficient ITIH5Δs was sufficient to suppress liver metastasis. Similar to secreted ITIH5, expression of ITIH5Δs was associated with rounded cell morphology, reduced cell motility and reduction of liver metastasis. Expression of ITIH5 is low in both human primary PDAC and matched metastases. CONCLUSIONS: Metastasis suppression by ITIH5 may be mediated by an intracellular mechanism. In human PDAC, loss of ITIH5 may be an early event and ITIH5-low PDAC cells in primary tumours may be selected for liver metastasis. Further defining the ITIH5-mediated pathway in PDAC could establish future therapeutic exploitation of this biology and reduce morbidity and mortality associated with PDAC metastasis.


Assuntos
Carcinoma Ductal Pancreático/patologia , Neoplasias Hepáticas/secundário , Invasividade Neoplásica/patologia , Neoplasias Pancreáticas/patologia , Proteínas Secretadas Inibidoras de Proteinases/metabolismo , Animais , Carcinoma Ductal Pancreático/metabolismo , Linhagem Celular Tumoral , Xenoenxertos , Humanos , Camundongos , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas
3.
J Biol Chem ; 290(17): 10934-46, 2015 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-25752611

RESUMO

We previously demonstrated that pharmacological induction of autophagy protected against acetaminophen (APAP)-induced liver injury in mice by clearing damaged mitochondria. However, the mechanism for removal of mitochondria by autophagy is unknown. Parkin, an E3 ubiquitin ligase, has been shown to be required for mitophagy induction in cultured mammalian cells following mitochondrial depolarization, but its role in vivo is not clear. The purpose of this study was to investigate the role of Parkin-mediated mitophagy in protection against APAP-induced liver injury. We found that Parkin translocated to mitochondria in mouse livers after APAP treatment followed by mitochondrial protein ubiquitination and mitophagy induction. To our surprise, we found that mitophagy still occurred in Parkin knock-out (KO) mice after APAP treatment based on electron microscopy analysis and Western blot analysis for some mitochondrial proteins, and Parkin KO mice were protected against APAP-induced liver injury compared with wild type mice. Mechanistically, we found that Parkin KO mice had decreased activated c-Jun N-terminal kinase (JNK), increased induction of myeloid leukemia cell differentiation protein (Mcl-1) expression, and increased hepatocyte proliferation after APAP treatment in their livers compared with WT mice. In contrast to chronic deletion of Parkin, acute knockdown of Parkin in mouse livers using adenovirus shRNA reduced mitophagy and Mcl-1 expression but increased JNK activation after APAP administration, which exacerbated APAP-induced liver injury. Therefore, chronic deletion (KO) and acute knockdown of Parkin have differential responses to APAP-induced mitophagy and liver injury in mice.


Assuntos
Acetaminofen/efeitos adversos , Analgésicos não Narcóticos/efeitos adversos , Doença Hepática Induzida por Substâncias e Drogas , Deleção de Genes , Mitofagia , Ubiquitina-Proteína Ligases/deficiência , Acetaminofen/farmacologia , Analgésicos não Narcóticos/farmacologia , Animais , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/genética , Doença Hepática Induzida por Substâncias e Drogas/genética , Doença Hepática Induzida por Substâncias e Drogas/metabolismo , Doença Hepática Induzida por Substâncias e Drogas/patologia , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/genética , Técnicas de Silenciamento de Genes , Hepatócitos/metabolismo , Hepatócitos/patologia , Fígado/metabolismo , Fígado/patologia , Camundongos , Camundongos Knockout , Proteínas Mitocondriais/genética , Proteínas Mitocondriais/metabolismo , Mitofagia/efeitos dos fármacos , Mitofagia/genética , Proteína de Sequência 1 de Leucemia de Células Mieloides/biossíntese , Proteína de Sequência 1 de Leucemia de Células Mieloides/genética , Ubiquitinação/efeitos dos fármacos , Ubiquitinação/genética
4.
J Biol Chem ; 287(50): 42379-88, 2012 Dec 07.
Artigo em Inglês | MEDLINE | ID: mdl-23095748

RESUMO

Mitochondrial homeostasis via mitochondrial dynamics and quality control is crucial to normal cellular functions. Mitophagy (mitochondria removed by autophagy) stimulated by a mitochondrial uncoupler, carbonyl cyanide m-chlorophenylhydrazone (CCCP), requires Parkin, but it is not clear why Parkin is crucial to this process. We found that in the absence of Parkin, carbonyl cyanide m-chlorophenylhydrazone induced the formation of mitochondrial spheroids. Mitochondrial spheroid formation is also induced in vivo in the liver by acetaminophen overdose, a condition causing severe oxidative mitochondrial damages and liver injury. Mitochondrial spheroids could undergo a maturation process by interactions with acidic compartments. The formation of this new structure required reactive oxygen species and mitofusins. Parkin suppressed these mitochondrial dynamics by promoting mitofusin degradation. Consistently, genetic deletion of mitofusins without concomitant expression of Parkin was sufficient to prevent mitochondrial spheroid formation and resumed mitophagy. Mitochondrial spheroid formation and mitophagy could represent different strategies of mitochondrial homeostatic response to oxidative stress and are reciprocally regulated by mitofusins and Parkin.


Assuntos
GTP Fosfo-Hidrolases/metabolismo , Mitocôndrias Hepáticas/metabolismo , Dinâmica Mitocondrial/fisiologia , Mitofagia/fisiologia , Ubiquitina-Proteína Ligases/metabolismo , Acetaminofen/efeitos adversos , Acetaminofen/farmacologia , Analgésicos não Narcóticos/efeitos adversos , Analgésicos não Narcóticos/farmacologia , Animais , Carbonil Cianeto m-Clorofenil Hidrazona/farmacologia , Células Cultivadas , Doença Hepática Induzida por Substâncias e Drogas/metabolismo , Doença Hepática Induzida por Substâncias e Drogas/patologia , GTP Fosfo-Hidrolases/genética , Fígado/metabolismo , Fígado/ultraestrutura , Camundongos , Camundongos Knockout , Mitocôndrias Hepáticas/ultraestrutura , Dinâmica Mitocondrial/efeitos dos fármacos , Mitofagia/efeitos dos fármacos , Proteólise/efeitos dos fármacos , Ionóforos de Próton/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Ubiquitina-Proteína Ligases/genética
5.
Cell Rep ; 42(10): 113291, 2023 10 31.
Artigo em Inglês | MEDLINE | ID: mdl-37862166

RESUMO

Dysfunctional mitochondria are removed via multiple pathways, such as mitophagy, a selective autophagy process. Here, we identify an intracellular hybrid mitochondria-lysosome organelle (termed the mitochondria-lysosome-related organelle [MLRO]), which regulates mitochondrial homeostasis independent of canonical mitophagy during hepatocyte dedifferentiation. The MLRO is an electron-dense organelle that has either a single or double membrane with both mitochondria and lysosome markers. Mechanistically, the MLRO is likely formed from the fusion of mitochondria-derived vesicles (MDVs) with lysosomes through a PARKIN-, ATG5-, and DRP1-independent process, which is negatively regulated by transcription factor EB (TFEB) and associated with mitochondrial protein degradation and hepatocyte dedifferentiation. The MLRO, which is galectin-3 positive, is reminiscent of damaged lysosome and could be cleared by overexpression of TFEB, resulting in attenuation of hepatocyte dedifferentiation. Together, results from this study suggest that the MLRO may act as an alternative mechanism for mitochondrial quality control independent of canonical autophagy/mitophagy involved in cell dedifferentiation.


Assuntos
Mitocôndrias , Organelas , Mitocôndrias/metabolismo , Organelas/metabolismo , Lisossomos/metabolismo , Autofagia/fisiologia , Mitofagia/fisiologia
6.
J Alzheimers Dis ; 85(1): 381-394, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-34806611

RESUMO

BACKGROUND: Amyloid-ß (Aß), which derives from the amyloid-ß protein precursor (AßPP), forms plaques and serves as a fluid biomarker in Alzheimer's disease (AD). How Aß forms from AßPP is known, but questions relating to AßPP and Aß biology remain unanswered. AD patients show mitochondrial dysfunction, and an Aß/AßPP mitochondria relationship exists. OBJECTIVE: We considered how mitochondrial biology may impact AßPP and Aß biology. METHODS: SH-SY5Y cells were transfected with AßPP constructs. After treatment with FCCP (uncoupler), Oligomycin (ATP synthase inhibitor), or starvation Aß levels were measured. ß-secretase (BACE1) expression was measured. Mitochondrial localized full-length AßPP was also measured. All parameters listed were measured in ρ0 cells on an SH-SY5Y background. iPSC derived neurons were also used to verify key results. RESULTS: We showed that mitochondrial depolarization routes AßPP to, while hyperpolarization routes AßPP away from, the organelle. Mitochondrial AßPP and cell Aß secretion inversely correlate, as cells with more mitochondrial AßPP secrete less Aß, and cells with less mitochondrial AßPP secrete more Aß. An inverse relationship between secreted/extracellular Aß and intracellular Aß was observed. CONCLUSION: Our findings indicate mitochondrial function alters AßPP localization and suggest enhanced mitochondrial activity promotes Aß secretion while depressed mitochondrial activity minimizes Aß secretion. Our data complement other studies that indicate a mitochondrial, AßPP, and Aß nexus, and could help explain why cerebrospinal fluid Aß is lower in those with AD. Our data further suggest Aß secretion could serve as a biomarker of cell or tissue mitochondrial function.


Assuntos
Doença de Alzheimer/genética , Secretases da Proteína Precursora do Amiloide/metabolismo , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Potencial da Membrana Mitocondrial , Doença de Alzheimer/patologia , Ácido Aspártico Endopeptidases/metabolismo , Encéfalo/patologia , Linhagem Celular Tumoral , Humanos , Mitocôndrias/metabolismo , Neuroblastoma/patologia , Neurônios/metabolismo
7.
Cell Death Dis ; 13(2): 114, 2022 02 04.
Artigo em Inglês | MEDLINE | ID: mdl-35121743

RESUMO

Obesity creates a localized inflammatory reaction in the adipose, altering secretion of adipocyte-derived factors that contribute to pathologies including cancer. We have previously shown that adiponectin inhibits pancreatic cancer by antagonizing leptin-induced STAT3 activation. Yet, the effects of adiponectin on pancreatic cancer cell metabolism have not been addressed. In these studies, we have uncovered a novel metabolic function for the synthetic adiponectin-receptor agonist, AdipoRon. Treatment of PDAC cells with AdipoRon led to mitochondrial uncoupling and loss of ATP production. Concomitantly, AdipoRon-treated cells increased glucose uptake and utilization. This metabolic switch further correlated with AMPK mediated inhibition of the prolipogenic factor acetyl coenzyme A carboxylase 1 (ACC1), which is known to initiate fatty acid catabolism. Yet, measurements of fatty acid oxidation failed to detect any alteration in response to AdipoRon treatment, suggesting a deficiency for compensation. Additional disruption of glycolytic dependence, using either a glycolysis inhibitor or low-glucose conditions, demonstrated an impairment of growth and survival of all pancreatic cancer cell lines tested. Collectively, these studies provide evidence that pancreatic cancer cells utilize metabolic plasticity to upregulate glycolysis in order to adapt to suppression of oxidative phosphorylation in the presence of AdipoRon.


Assuntos
Neoplasias Pancreáticas , Receptores Artificiais , Adiponectina/metabolismo , Adiponectina/farmacologia , Ácidos Graxos , Glicólise , Humanos , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/patologia , Piperidinas , Receptores de Adiponectina/metabolismo , Receptores Artificiais/metabolismo , Neoplasias Pancreáticas
8.
J Pharmacol Exp Ther ; 337(2): 471-8, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21303922

RESUMO

Bile duct epithelial cells (BDECs) contribute to liver fibrosis by expressing αVß6 integrin, a critical activator of latent transforming growth factor ß (TGF-ß). ß6 integrin (Itgß6) mRNA induction and αVß6 integrin expression in BDECs are partially TGF-ß-dependent. However, the signaling pathways required for TGF-ß-dependent Itgß6 mRNA induction in BDECs are not known. We tested the hypothesis that the p38 mitogen-activated protein kinase (MAPK) signaling pathway contributes to TGF-ß1 induction of Itgß6 mRNA by activating SMAD and activator protein 1 (AP-1) transcription factors. Pretreatment of transformed human BDECs (MMNK-1 cells) with two different p38 MAPK inhibitors, but not a control compound, inhibited TGF-ß1 induction of Itgß6 mRNA. Inhibition of p38 also reduced TGF-ß1 activation of a SMAD-dependent reporter construct. Expression of a dominant-negative SMAD3 (SMAD3ΔC) significantly reduced TGF-ß1-induced Itgß6 mRNA expression. Expression of JunB mRNA, but not other AP-1 proteins, increased in TGF-ß1-treated MMNK-1 cells, and induction of JunB expression was p38-dependent. Consistent with a requirement for de novo induction of JunB protein, cycloheximide pretreatment inhibited TGF-ß1 induction of Itgß6 mRNA. Expression of a dominant-negative AP-1 mutant (TAM67) also inhibited TGF-ß1 induction of Itgß6 mRNA. Overall, the results suggest that p38 contributes to TGF-ß1-induced Itgß6 mRNA expression in MMNK-1 cells by regulating activation of both SMAD and AP-1 transcription factors.


Assuntos
Ductos Biliares/metabolismo , Células Epiteliais/metabolismo , Cadeias beta de Integrinas/biossíntese , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Ductos Biliares/citologia , Western Blotting , Linhagem Celular , Citosol/metabolismo , DNA Complementar/biossíntese , DNA Complementar/genética , Densitometria , Humanos , Imunoprecipitação , Luciferases/metabolismo , RNA/biossíntese , RNA/genética , RNA/isolamento & purificação , RNA Interferente Pequeno , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/fisiologia , Proteína Smad3/metabolismo , Fator de Transcrição AP-1/metabolismo , Fator de Crescimento Transformador beta1/fisiologia , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores
9.
J Pharmacol Exp Ther ; 339(2): 487-98, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21856859

RESUMO

Fatty acid-induced lipotoxicity plays a critical role in the pathogenesis of nonalcoholic liver disease. Saturated fatty acids and unsaturated fatty acids have differential effects on cell death and steatosis, but the mechanisms responsible for these differences are not known. Using cultured HepG2 cells and primary mouse hepatocytes, we found that unsaturated and saturated fatty acids differentially regulate autophagy and apoptosis. The unsaturated fatty acid, oleic acid, promoted the formation of triglyceride-enriched lipid droplets and induced autophagy but had a minimal effect on apoptosis. In contrast, the saturated fatty acid, palmitic acid, was poorly converted into triglyceride-enriched lipid droplets, suppressed autophagy, and significantly induced apoptosis. Subsequent studies revealed that palmitic acid-induced apoptosis suppressed autophagy by inducing caspase-dependent Beclin 1 cleavage, indicating cross-talk between apoptosis and autophagy. Moreover, our data suggest that the formation of triglyceride-enriched lipid droplets and induction of autophagy are protective mechanisms against fatty acid-induced lipotoxicity. In line with our in vitro findings, we found that high-fat diet-induced hepatic steatosis was associated with autophagy in the mouse liver. Potential modulation of autophagy may be a novel approach that has therapeutic benefits for obesity-induced steatosis and liver injury.


Assuntos
Apoptose/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Ácidos Graxos Insaturados/farmacologia , Fígado Gorduroso/fisiopatologia , Hepatócitos/fisiologia , Ácido Oleico/farmacologia , Ácido Palmítico/farmacologia , Adenoviridae/fisiologia , Animais , Proteínas Reguladoras de Apoptose/metabolismo , Proteína Beclina-1 , Caspase 3/metabolismo , Dieta Hiperlipídica , Ácidos Graxos Insaturados/fisiologia , Fígado Gorduroso/patologia , Células Hep G2 , Humanos , Masculino , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Espécies Reativas de Oxigênio/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Triglicerídeos/análise
10.
J Mol Med (Berl) ; 95(9): 951-963, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28597070

RESUMO

The shift by cancer cells toward aerobic glycolysis (Warburg effect) confers selective advantages by utilizing nutrients (e.g., lipids, amino acids, and nucleotides) to build biomass. Lipogenesis is generally enhanced, and its inhibition diminishes proliferation and survival. Re-expression of the metastasis suppressor KISS1 in human melanoma cells results in greater mitochondrial biogenesis, inhibition of glycolysis, utilization of beta-oxidation to provide energy, elevated oxidation of exogenous fatty acids, and increased expression of early-phase lipogenesis genes at both mRNA and protein levels. Correspondingly, the energy sensor AMPKß is phosphorylated, resulting in inhibitory phosphorylation of acetyl-CoA carboxylase (ACC), which is linked to enhanced beta-oxidation. Furthermore, PGC1α is required for KISS1-mediated phosphorylation of ACC and metastasis suppression. Collectively, these data further support the linkages between macromolecular metabolism and metastasis. KEY MESSAGES: • KISS1 alters fatty acid metabolism. • There may be connections between metastasis and metabolism. • PGC1alpha appears to be downstream mediator of KISS1 metastasis suppression.


Assuntos
Glicólise , Kisspeptinas/genética , Mitocôndrias/genética , Mitocôndrias/metabolismo , Oxirredução , Linhagem Celular Tumoral , Regulação da Expressão Gênica , Glucose/metabolismo , Humanos , Kisspeptinas/metabolismo , Metabolismo dos Lipídeos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Modelos Biológicos , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/metabolismo , Fosforilação , Estresse Fisiológico , Transcrição Gênica
12.
Acta Pharm Sin B ; 5(2): 158-67, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26579442

RESUMO

Alcoholic liver disease (ALD) is one of the major causes of liver morbidity and mortality worldwide. Chronic alcohol consumption leads to development of liver pathogenesis encompassing steatosis, inflammation, fibrosis, cirrhosis, and in extreme cases, hepatocellular carcinoma. Moreover, ALD may also associate with cholestasis. Emerging evidence now suggests that farnesoid X receptor (FXR) and bile acids also play important roles in ALD. In this review, we discuss the effects of alcohol consumption on FXR, bile acids and gut microbiome as well as their impacts on ALD. Moreover, we summarize the findings on FXR, FoxO3a (forkhead box-containing protein class O3a) and PPARα (peroxisome proliferator-activated receptor alpha) in regulation of autophagy-related gene transcription program and liver injury in response to alcohol exposure.

13.
World J Gastroenterol ; 20(36): 12908-33, 2014 Sep 28.
Artigo em Inglês | MEDLINE | ID: mdl-25278688

RESUMO

Alcoholic liver disease is a major health problem in the United States and worldwide. Chronic alcohol consumption can cause steatosis, inflammation, fibrosis, cirrhosis and even liver cancer. Significant progress has been made to understand key events and molecular players for the onset and progression of alcoholic liver disease from both experimental and clinical alcohol studies. No successful treatments are currently available for treating alcoholic liver disease; therefore, development of novel pathophysiological-targeted therapies is urgently needed. This review summarizes the recent progress on animal models used to study alcoholic liver disease and the detrimental factors that contribute to alcoholic liver disease pathogenesis including miRNAs, S-adenosylmethionine, Zinc deficiency, cytosolic lipin-1ß, IRF3-mediated apoptosis, RIP3-mediated necrosis and hepcidin. In addition, we summarize emerging adaptive protective effects induced by alcohol to attenuate alcohol-induced liver pathogenesis including FoxO3, IL-22, autophagy and nuclear lipin-1α.


Assuntos
Desenho de Fármacos , Hepatopatias Alcoólicas/tratamento farmacológico , Fígado/efeitos dos fármacos , Terapia de Alvo Molecular , Animais , Modelos Animais de Doenças , Predisposição Genética para Doença , Humanos , Fígado/metabolismo , Fígado/patologia , Hepatopatias Alcoólicas/diagnóstico , Hepatopatias Alcoólicas/genética , Hepatopatias Alcoólicas/metabolismo , Fenótipo , Fatores de Proteção , Fatores de Risco , Transdução de Sinais/efeitos dos fármacos , Resultado do Tratamento
14.
Toxicol Sci ; 137(2): 478-90, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24189133

RESUMO

Retention of bile acids (BAs) in the liver during cholestasis plays an important role in the development of cholestatic liver injury. Several studies have reported that high concentrations of certain BAs induce cell death and inflammatory response in the liver, and BAs may promote liver tumorigenesis. Macroautophagy (hereafter referred to as autophagy) is a lysosomal degradation process that regulates organelle and protein homeostasis and serves as a cell survival mechanism under a variety of stress conditions. However, it is not known if BAs modulate autophagy in hepatocytes. In the present study, we determined autophagic flux in livers of farnesoid X receptor (FXR) knockout (KO) mice that have increased concentrations of hepatic BAs and in primary cultured mouse hepatocytes treated with BAs. The results showed that autophagic flux was impaired in livers of FXR KO mice and in BA-treated primary mouse hepatocytes. Mechanistically, BAs did not affect the activities of cathepsin or the proteasome, but impaired autophagosomal-lysosomal fusion likely due to reduction of Rab7 protein expression and targeting to autophagosomes. In conclusion, BAs suppress autophagic flux in hepatocytes by impairing autophagosomal-lysosomal fusion, which may be implicated in bile acid-induced liver tumor promotion observed in FXR KO mice.


Assuntos
Autofagia/efeitos dos fármacos , Ácidos e Sais Biliares/metabolismo , Hepatócitos/efeitos dos fármacos , Fígado/efeitos dos fármacos , Animais , Ácidos e Sais Biliares/farmacologia , Catepsina B/metabolismo , Técnicas de Cultura de Células , Células Cultivadas , Hepatócitos/metabolismo , Hepatócitos/ultraestrutura , Fígado/metabolismo , Fígado/patologia , Camundongos , Camundongos Knockout , Microscopia Confocal , Microscopia Eletrônica , Microscopia de Fluorescência , Proteínas Associadas aos Microtúbulos/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Receptores Citoplasmáticos e Nucleares/genética , Fator de Transcrição TFIIH , Fatores de Transcrição/metabolismo
15.
PLoS One ; 9(12): e115849, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25536043

RESUMO

Chronic alcohol causes liver hypoxia and steatosis, which eventually develops into alcoholic liver disease (ALD). While it has been known that alcohol consumption activates hepatic hypoxia inducing factor-1α (HIF-1α), conflicting results regarding the role of HIF-1α in alcohol-induced liver injury and steatosis in mice have been reported. In the present study, we aimed to use hepatocyte-specific HIF-1ß knockout mice to eliminate the possible compensatory effects of the single knockout of the 1α subunit of HIF to study the role of HIFs in ALD. C57BL/6 wild type mice were treated with acute ethanol to mimic human binge drinking. Matched wild-type and hepatocyte specific HIF-1ß knockout mice were also subjected to a recently established Gao-binge alcohol model to mimic chronic plus binge conditions, which is quite common in human alcoholics. We found that acute alcohol treatment increased BNIP3 and BNIP3L/NIX expression in primary cultured hepatocytes and in mouse livers, suggesting that HIF may be activated in these models. We further found that hepatocyte-specific HIF-1ß knockout mice developed less steatosis and liver injury following the Gao-binge model or acute ethanol treatment compared with their matched wild type mice. Mechanistically, protection against Gao-binge treatment-induced steatosis and liver injury was likely associated with increased FoxO3a activation and subsequent induction of autophagy in hepatocyte-specific HIF-1ß knockout mice.


Assuntos
Translocador Nuclear Receptor Aril Hidrocarboneto/metabolismo , Autofagia/efeitos dos fármacos , Fígado Gorduroso Alcoólico/metabolismo , Fígado Gorduroso Alcoólico/patologia , Fígado/patologia , Animais , Translocador Nuclear Receptor Aril Hidrocarboneto/genética , Células Cultivadas , Etanol , Fígado Gorduroso Alcoólico/genética , Regulação da Expressão Gênica , Hepatócitos/metabolismo , Hepatócitos/patologia , Fígado/metabolismo , Masculino , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Mitocondriais/genética , Proteínas Mitocondriais/metabolismo
16.
Redox Biol ; 2: 991-1002, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25460735

RESUMO

Alcoholic liver disease encompasses a wide spectrum of pathogenesis including steatosis, fibrosis, cirrhosis, and alcoholic steatohepatitis. Autophagy is a lysosomal degradation process that degrades cellular proteins and damaged/excess organelles, and serves as a protective mechanism in response to various stresses. Acute alcohol treatment induces autophagy via FoxO3a-mediated autophagy gene expression and protects against alcohol-induced steatosis and liver injury in mice. Farnesoid X Receptor (FXR) is a nuclear receptor that regulates cellular bile acid homeostasis. In the present study, wild type and FXR knockout (KO) mice were treated with acute ethanol for 16h. We found that ethanol treated-FXR KO mice had exacerbated hepatotoxicity and steatosis compared to wild type mice. Furthermore, we found that ethanol treatment had decreased expression of various essential autophagy genes and several other FoxO3 target genes in FXR KO mice compared with wild type mice. Mechanistically, we did not find a direct interaction between FXR and FoxO3. Ethanol-treated FXR KO mice had increased Akt activation, increased phosphorylation of FoxO3 resulting in decreased FoxO3a nuclear retention and DNA binding. Furthermore, ethanol treatment induced hepatic mitochondrial spheroid formation in FXR KO mice but not in wild type mice, which may serve as a compensatory alternative pathway to remove ethanol-induced damaged mitochondria in FXR KO mice. These results suggest that lack of FXR impaired FoxO3a-mediated autophagy and in turn exacerbated alcohol-induced liver injury.


Assuntos
Autofagia/efeitos dos fármacos , Etanol/toxicidade , Proteína Forkhead Box O3/metabolismo , Hepatopatias Alcoólicas/metabolismo , Fígado/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , Animais , Proteína Forkhead Box O3/genética , Fígado/patologia , Hepatopatias Alcoólicas/patologia , Camundongos , Camundongos Knockout , Receptores Citoplasmáticos e Nucleares/genética
17.
Exp Biol Med (Maywood) ; 238(5): 525-38, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23856904

RESUMO

p62/sequestosome-1/A170/ZIP (hereafter referred to as p62) is a scaffold protein that has multiple functions, such as signal transduction, cell proliferation, cell survival, cell death, inflammation, tumourigenesis and oxidative stress response. While p62 is an autophagy substrate and is degraded by autophagy, p62 serves as an autophagy receptor for selective autophagic clearance of protein aggregates and organelles. Moreover, p62 functions as a signalling hub for various signalling pathways, including NF-κB, Nrf2 and mTOR. In this review, we discuss the pathophysiological role of p62 in the liver, including formation of hepatic inclusion bodies, cholestasis, obesity, insulin resistance, liver cell death and tumourigenesis.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Transformação Celular Neoplásica/metabolismo , Neoplasias Hepáticas , Fígado , Proteínas de Neoplasias/metabolismo , Animais , Autofagia , Proliferação de Células , Transformação Celular Neoplásica/patologia , Humanos , Corpos de Inclusão/metabolismo , Corpos de Inclusão/patologia , Inflamação/metabolismo , Inflamação/patologia , Inflamação/fisiopatologia , Resistência à Insulina , Fígado/metabolismo , Fígado/patologia , Fígado/fisiopatologia , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patologia , Neoplasias Hepáticas/fisiopatologia , Fator 2 Relacionado a NF-E2/metabolismo , NF-kappa B/metabolismo , Estresse Oxidativo , Proteína Sequestossoma-1 , Transdução de Sinais , Serina-Treonina Quinases TOR/metabolismo
18.
Exp Biol Med (Maywood) ; 236(5): 546-56, 2011 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-21478210

RESUMO

Autophagy is a highly conserved intracellular catabolic pathway that degrades cellular long-lived proteins and organelles. Autophagy is normally activated in response to nutrient deprivation and other stresses as a cell survival mechanism. Accumulating evidence indicates that autophagy plays a critical role in liver pathophysiology, in addition to maintaining hepatic energy and nutrient balance. Alcohol consumption causes hepatic metabolic changes, oxidative stress, accumulation of lipid droplets and damaged mitochondria; all of these can be regulated by autophagy. This review summarizes the recent findings about the role and mechanisms of autophagy in alcoholic liver disease (ALD), and the possible intervention for treating ALD by modulating autophagy.


Assuntos
Autofagia , Hepatopatias Alcoólicas/patologia , Animais , Etanol/efeitos adversos , Humanos , Hepatopatias Alcoólicas/metabolismo , Mamíferos/metabolismo , Modelos Biológicos , Transdução de Sinais
19.
Am J Physiol Gastrointest Liver Physiol ; 296(2): G388-98, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19074641

RESUMO

Human esophageal epithelial cells play a key role in esophageal inflammation in response to acidic pH during gastroesophageal reflux disease (GERD), increasing secretion of IL-6 and IL-8. The mechanisms underlying IL-6 and IL-8 expression and secretion in esophageal epithelial cells after acid stimulation are not well characterized. We investigated the role of PKC, MAPK, and NF-kappaB signaling pathways and transcriptional regulation of IL-6 and IL-8 expression in HET-1A cells exposed to acid. Exposure of HET-1A cells to pH 4.5 induced NF-kappaB activity and enhanced IL-6 and IL-8 secretion and mRNA and protein expression. Acid stimulation of HET-1A cells also resulted in activation of MAPKs and PKC (alpha and epsilon). Curcumin, as well as inhibitors of NF-kappaB (SN-50), PKC (chelerythrine), and p44/42 MAPK (PD-098059) abolished the acid-induced expression of IL-6 and IL-8. The JNK inhibitor SP-600125 blocked expression/secretion of IL-6 but only partially attenuated IL-8 expression. The p38 MAPK inhibitor SB-203580 did not inhibit IL-6 expression but exerted a stronger inhibitory effect on IL-8 expression. Together, these data demonstrate that 1) acid is a potent inducer of IL-6 and IL-8 production in HET-1A cells; 2) MAPK and PKC signaling play a key regulatory role in acid-mediated IL-6 and IL-8 expression via NF-kappaB activation; and 3) the anti-inflammatory plant compound curcumin inhibits esophageal activation in response to acid. Thus IL-6 and IL-8 expression by acid may contribute to the pathobiology of mucosal injury in GERD, and inhibition of the NF-kappaB/proinflammatory cytokine pathways may emerge as important therapeutic targets for treatment of esophageal inflammation.


Assuntos
Anti-Inflamatórios/farmacologia , Curcumina/farmacologia , Células Epiteliais/efeitos dos fármacos , Esôfago/efeitos dos fármacos , Interleucina-6/metabolismo , Interleucina-8/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , NF-kappa B/metabolismo , Proteína Quinase C/metabolismo , Antracenos/farmacologia , Benzofenantridinas/farmacologia , Linhagem Celular , Ativação Enzimática , Células Epiteliais/enzimologia , Células Epiteliais/imunologia , Esôfago/enzimologia , Esôfago/imunologia , Flavonoides/farmacologia , Humanos , Concentração de Íons de Hidrogênio , Imidazóis/farmacologia , Interleucina-6/genética , Interleucina-8/genética , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Mucosa/efeitos dos fármacos , Mucosa/enzimologia , Mucosa/imunologia , NF-kappa B/antagonistas & inibidores , Peptídeos/farmacologia , Proteína Quinase C/antagonistas & inibidores , Inibidores de Proteínas Quinases/farmacologia , Piridinas/farmacologia , Transdução de Sinais/efeitos dos fármacos , Telomerase/metabolismo , Fatores de Tempo , Transcrição Gênica/efeitos dos fármacos , Regulação para Cima
20.
Am J Physiol Gastrointest Liver Physiol ; 295(3): G581-90, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18635600

RESUMO

Tissue remodeling and mesenchymal cell accumulation accompanies chronic inflammatory disorders involving joints, lung, vasculature, and bowel. Chronic inflammation may alter DNA-mismatch repair (MMR) systems in mesenchymal cells, but is not defined in Crohn's disease (CD) and its associated intestinal remodeling and stricture formation. We determined whether DNA-MMR alteration plays a role in the pathogenesis of CD tissue remodeling. Control and CD bowel tissues were used to generate primary cultures of muscularis mucosa myofibroblasts, which were assessed directly or following stimulation with TNF-alpha/LPS or H2O2. MutS homolog (MSH)2, MSH3, and MSH6 expression in tissues and myofibroblasts was determined. Immunohistochemical staining revealed an increased expression of MSH2 in CD muscularis mucosa and submucosal tissues compared with controls or uninvolved CD tissue, and MSH2 expression was increased in CD myofibroblasts compared with control cells. TNF-alpha/LPS and H2O2 further enhanced MSH2 expression in both control and CD cells, which were decreased by simvastatin. There were no significant changes in MSH3 and MSH6 expression. Proliferating cell nuclear antigen and Ki67 staining of CD tissue revealed increased proliferation in the muscularis mucosa and submucosa of chronically inflamed tissues, and enhanced proliferation was seen in CD myofibroblasts compared with controls. Simvastatin reversed the effects of inflammatory stress on the DNA-MMR and inhibited proliferation of control and CD myofibroblasts. Gene silencing with MSH2 siRNA selectively decreased CD myofibroblast proliferation. These data demonstrate a potential role for MSH2 in the pathogenesis of nonneoplastic mesenchymal cell accumulation and intestinal remodeling in CD chronic inflammation.


Assuntos
Proliferação de Células , Doença de Crohn/enzimologia , Fibroblastos/enzimologia , Obstrução Intestinal/etiologia , Intestinos/enzimologia , Proteína 2 Homóloga a MutS/metabolismo , Adulto , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Doença de Crohn/complicações , Doença de Crohn/genética , Doença de Crohn/patologia , Proteínas de Ligação a DNA/metabolismo , Feminino , Fibroblastos/efeitos dos fármacos , Fibroblastos/patologia , Imunofluorescência , Humanos , Peróxido de Hidrogênio/farmacologia , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Imuno-Histoquímica , Obstrução Intestinal/enzimologia , Obstrução Intestinal/genética , Obstrução Intestinal/patologia , Intestinos/efeitos dos fármacos , Intestinos/patologia , Lipopolissacarídeos/farmacologia , Masculino , Instabilidade de Microssatélites , Pessoa de Meia-Idade , Proteína 2 Homóloga a MutS/genética , Proteína 3 Homóloga a MutS , Antígeno Nuclear de Célula em Proliferação/metabolismo , Interferência de RNA , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/metabolismo , Sinvastatina/farmacologia , Timidina/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Regulação para Cima
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA