RESUMO
PURPOSE: Tamoxifen, a widely used drug for breast cancer treatment, is associated with adverse effects on the liver, including the development of fatty liver. This study aimed to investigate the potential protective effect of caffeine against tamoxifen-induced fatty liver in Wistar rats. METHODS: Rats were divided into normal control, tamoxifen + saline, and tamoxifen + caffeine. Plasma samples were assessed for biochemical markers related to oxidative stress, inflammation, liver function, and cell damage. Additionally, liver histopathology was examined to quantify the extent of fatty infiltration. RESULTS: In the tamoxifen + saline group, elevated levels of plasma malondialdehyde (MDA), tumor necrosis factor-alpha (TNF-α), alanine aminotransferase (ALT), cytokeratin 18, and soluble ST2 were observed compared to the normal control group, indicating increased oxidative stress, inflammation, and liver injury (p < 0.01). Moreover, histopathological examination revealed a significant increase in fatty infiltration (p < 0.001). However, in the tamoxifen + caffeine group, these markers were markedly reduced (p < 0.05, p < 0.01), and fatty infiltration was significantly mitigated (p < 0.001). CONCLUSIONS: The findings suggest that caffeine administration attenuates tamoxifen-induced fatty liver in rats by ameliorating oxidative stress, inflammation, liver injury, and cell damage. Histopathological evidence further supports the protective role of caffeine. This study highlights the potential of caffeine as a therapeutic intervention to counter tamoxifen-induced hepatic complications, contributing to the optimization of breast cancer treatment strategies.
Assuntos
Cafeína , Fígado Gorduroso , Malondialdeído , Estresse Oxidativo , Ratos Wistar , Tamoxifeno , Animais , Cafeína/farmacologia , Cafeína/uso terapêutico , Tamoxifeno/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Malondialdeído/análise , Fígado Gorduroso/induzido quimicamente , Fígado Gorduroso/prevenção & controle , Fígado Gorduroso/tratamento farmacológico , Feminino , Fígado/efeitos dos fármacos , Fígado/patologia , Alanina Transaminase/sangue , Ratos , Antineoplásicos Hormonais/farmacologia , Fator de Necrose Tumoral alfa/sangue , Fator de Necrose Tumoral alfa/análise , Biomarcadores/sangue , Biomarcadores/análise , Modelos Animais de DoençasRESUMO
Metabolic-associated fatty liver disease (MAFLD), also known as non-alcoholic fatty liver disease (NAFLD), is a worldwide liver disease without definitive or widely used therapeutic drugs in clinical practice. In this study, we confirm that 6-gingerol (6-G), an active ingredient of ginger (Zingiber officinale Roscoe) in traditional Chinese medicine (TCM), can alleviate fructose-induced hepatic steatosis. It was found that 6-G significantly decreased hyperlipidemia caused by high-fructose diets (HFD) in rats, and reversed the increase in hepatic de novo lipogenesis (DNL) and triglyceride (TG) levels induced by HFD, both in vivo and in vitro. Mechanistically, chemical proteomics and cellular thermal shift assay (CETSA)-proteomics approaches revealed that stearoyl-CoA desaturase (SCD) is a direct binding target of 6-G, which was confirmed by further CETSA assay and molecular docking. Meanwhile, it was found that 6-G could not alter SCD expression (in either mRNA or protein levels), but inhibited SCD activity (decreasing the desaturation levels of fatty acids) in HFD-fed rats. Furthermore, SCD deficiency mimicked the ability of 6-G to reduce lipid accumulation in HF-induced HepG2 cells, and impaired the improvement in hepatic steatosis brought about by 6-G treatment in HFD supplemented with oleic acid diet-induced SCD1 knockout mice. Taken together, our present study demonstrated that 6-G inhibits DNL by targeting SCD to alleviate fructose diet-induced hepatic steatosis.
Assuntos
Catecóis , Álcoois Graxos , Frutose , Lipogênese , Estearoil-CoA Dessaturase , Animais , Álcoois Graxos/farmacologia , Estearoil-CoA Dessaturase/metabolismo , Estearoil-CoA Dessaturase/genética , Lipogênese/efeitos dos fármacos , Frutose/metabolismo , Frutose/efeitos adversos , Ratos , Humanos , Masculino , Catecóis/farmacologia , Camundongos , Simulação de Acoplamento Molecular , Células Hep G2 , Hepatopatia Gordurosa não Alcoólica/metabolismo , Hepatopatia Gordurosa não Alcoólica/tratamento farmacológico , Hepatopatia Gordurosa não Alcoólica/induzido quimicamente , Hepatopatia Gordurosa não Alcoólica/etiologia , Ratos Sprague-Dawley , Triglicerídeos/metabolismo , Fígado Gorduroso/metabolismo , Fígado Gorduroso/tratamento farmacológico , Fígado Gorduroso/induzido quimicamente , Fígado Gorduroso/etiologia , Fígado/metabolismo , Fígado/efeitos dos fármacos , Fígado/patologia , Camundongos Endogâmicos C57BLRESUMO
Metabolic dysfunction-associated Steatohepatitis (MASH), is a prominent cause for liver cirrhosis. MASH-cirrhosis is responsible for liver complications and there is no specific treatment. To develop new therapeutic approaches, animal models are needed. The aim of this study was to develop a fast animal model of MASH-cirrhosis in rats reflecting the human disease. Carbon tetrachloride (CCl4) injections in combination with a high-fat Western diet (WD) were used to induce MASH-cirrhosis. To accelerate liver injury, animals received phenobarbital (PB) in their drinking water using two different regimens. Rats developed advanced MASH-cirrhosis characterized by portal hypertension, blood biochemistry, hepatic ballooning, steatosis, inflammation and fibrosis. Importantly, rats receiving low-dose PB for the long term (LT) showed ascites after 6 weeks, whereas rats with high-dose short-term (ST) PB developed ascites after 8 weeks. ST- and LT-treated rats showed increased portal pressure (PP) and decreased mean arterial pressure (MAP). Of note, hepatocyte ballooning was only observed in the LT group. The LT administration of low-dose PB with CCl4 intoxication and WD represents a fast and reproducible rat model mimicking decompensated MASH-cirrhosis in humans. Thus, CCl4 + WD with LT low-dose phenobarbital treatment might be the preferred rat animal model for drug development in MASH-cirrhosis.
Assuntos
Tetracloreto de Carbono , Modelos Animais de Doenças , Cirrose Hepática , Fenobarbital , Animais , Ratos , Tetracloreto de Carbono/toxicidade , Masculino , Cirrose Hepática/induzido quimicamente , Cirrose Hepática/patologia , Fígado Gorduroso/induzido quimicamente , Fígado Gorduroso/patologia , Fígado/efeitos dos fármacos , Fígado/patologia , Fígado/metabolismo , Dieta Hiperlipídica/efeitos adversosRESUMO
Fatty liver disease in laying hens, characterized by excessive lipid accumulation in hepatocytes, poses significant challenges to poultry health and production efficiency. In this study, we investigated the therapeutic potential of epigallocatechin gallate (EGCG), a bioactive compound found in green tea, in mitigating oleic acid (OA)-induced hepatic steatosis in primary chicken hepatocytes. Treatment with EGCG effectively attenuated lipid deposition by downregulating lipid synthesis-related genes. Moreover, EGCG mitigated oxidative stress, inflammation, DNA damage, and apoptosis induced by OA, thereby preserving hepatocyte viability. Mechanistically, EGCG exerted its protective effects by modulating the p38 MAPK signaling pathway. Our findings suggest that EGCG holds promise as a therapeutic agent for managing fatty liver disease in poultry, offering insights into novel strategies for improving poultry health and production outcomes.
Assuntos
Catequina , Galinhas , Fígado Gorduroso , Hepatócitos , Sistema de Sinalização das MAP Quinases , Ácido Oleico , Doenças das Aves Domésticas , Animais , Ácido Oleico/farmacologia , Catequina/análogos & derivados , Catequina/farmacologia , Catequina/administração & dosagem , Hepatócitos/efeitos dos fármacos , Fígado Gorduroso/veterinária , Fígado Gorduroso/induzido quimicamente , Fígado Gorduroso/tratamento farmacológico , Doenças das Aves Domésticas/induzido quimicamente , Doenças das Aves Domésticas/tratamento farmacológico , Feminino , Sistema de Sinalização das MAP Quinases/efeitos dos fármacosRESUMO
The rising prevalence of obesity-related illnesses, such as metabolic steatotic liver disease (MASLD), represents a significant global public health concern. This disease affects approximately 30 % of the adult population and is the result of metabolic abnormalities rather than alcohol consumption. Additionally, MASLD is associated with an increased risk of cardiovascular disease (CVD), chronic liver disease, and a variety of cancers, particularly gastrointestinal cancers. Clonal hematopoiesis (CH) is a biological state characterized by the expansion of a population of blood cells derived from a single mutated hematopoietic stem cell. The presence of CH in the absence of a diagnosed blood disorder or cytopenia is known as clonal hematopoiesis of indeterminate potential (CHIP), which itself increases the risk of hematological malignancies and CVD. Steatotic liver disease may also complicate the clinical course of cancer patients receiving antineoplastic agents, a condition referred to as chemotherapy induced steatohepatitis (CASH). This review will present an outline of the various aspects of MASLD, including complications. Furthermore, it will summarize the existing knowledge on the emerging association between CHIP and MASLD and present the available data on patient cases with concurrent MASLD and hematological neoplasms. Finally, it will provide a brief overview of the chemotherapeutic drugs associated with CASH, the underlying pathophysiologic mechanisms and their clinical implications.
Assuntos
Antineoplásicos , Fígado Gorduroso , Neoplasias Hematológicas , Humanos , Neoplasias Hematológicas/tratamento farmacológico , Neoplasias Hematológicas/complicações , Fígado Gorduroso/induzido quimicamente , Fígado Gorduroso/patologia , Antineoplásicos/efeitos adversos , Antineoplásicos/uso terapêutico , Hematopoiese ClonalRESUMO
BACKGROUND: Precision Health aims to revolutionize disease prevention by leveraging information across multiple omic datasets (multi-omics). However, existing methods generally do not consider personalized environmental risk factors (e.g., environmental pollutants). OBJECTIVE: To develop and apply a precision health framework which combines multiomic integration (including early, intermediate, and late integration, representing sequential stages at which omics layers are combined for modeling) with mediation approaches (including high-dimensional mediation to identify biomarkers, mediation with latent factors to identify pathways, and integrated/quasi-mediation to identify high-risk subpopulations) to identify novel biomarkers of prenatal mercury induced metabolic dysfunction-associated fatty liver disease (MAFLD), elucidate molecular pathways linking prenatal mercury with MAFLD in children, and identify high-risk children based on integrated exposure and multiomics data. METHODS: This prospective cohort study used data from 420 mother-child pairs from the Human Early Life Exposome (HELIX) project. Mercury concentrations were determined in maternal or cord blood from pregnancy. Cytokeratin 18 (CK-18; a MAFLD biomarker) and five omics layers (DNA Methylation, gene transcription, microRNA, proteins, and metabolites) were measured in blood in childhood (age 6-10 years). RESULTS: Each standard deviation increase in prenatal mercury was associated with a 0.11 [95% confidence interval: 0.02-0.21] standard deviation increase in CK-18. High dimensional mediation analysis identified 10 biomarkers linking prenatal mercury and CK-18, including six CpG sites and four transcripts. Mediation with latent factors identified molecular pathways linking mercury and MAFLD, including altered cytokine signaling and hepatic stellate cell activation. Integrated/quasi-mediation identified high risk subgroups of children based on unique combinations of exposure levels, omics profiles (driven by epigenetic markers), and MAFLD. CONCLUSIONS: Prenatal mercury exposure is associated with elevated liver enzymes in childhood, likely through alterations in DNA methylation and gene expression. Our analytic framework can be applied across many different fields and serve as a resource to help guide future precision health investigations.
Assuntos
Mercúrio , Efeitos Tardios da Exposição Pré-Natal , Humanos , Feminino , Gravidez , Mercúrio/sangue , Criança , Masculino , Estudos Prospectivos , Poluentes Ambientais/sangue , Fígado Gorduroso/induzido quimicamente , Biomarcadores/sangue , Medicina de Precisão , Adulto , Exposição Ambiental , Exposição Materna , MultiômicaRESUMO
Per- and polyfluoroalkyl substances (PFAS) are persistent organic pollutants previously associated with elevated liver enzymes in human cohorts and steatotic liver disease in animal models. We aimed to evaluate the associations between PFAS exposures, and liver enzymes and vibration controlled transient elastography (VCTE) biomarkers of metabolic dysfunction associated steatotic liver disease (MASLD) in adult National Health and Nutrition Examination Survey (NHANES) 2017 to 2018. VCTE was determined by FibroScan. Serum PFAS (n = 14), measured by mass spectrometry, were analyzed individually and by principal component (PC). Univariate and multivariable associations were determined between PFAS exposures and liver disease outcome variables: alanine aminotransferase (ALT), controlled attenuation parameter (CAP), liver stiffness measurement (LSM), FibroScan-based Score (FAST), using R. About 1,400 participants including 50% women with a mean age of 48 ± 19 years and a mean BMI of 29 ± 7 kg/m2 were analyzed. Four PFAS clustered to PC1, whereas 3 PFAS clustered to PC2. PC1 was significantly associated with ALT (ß = 0.028), CAP (ß = 0.041), LSM (ß = 0.025), and FAST (ß = 0.198) in univariate analysis. Individual PFAS exposures were oftentimes inversely associated with these measurements in multivariate analysis. In adult NHANES 2017-2018, PFAS may not be a significant burden for MASLD, because of the inconsistent associations between the environmental PFAS exposures and biomarkers of liver steatosis, inflammation, and fibrosis. More data are required to better understand the relationships between PFAS exposures and liver disease.
Assuntos
Poluentes Ambientais , Fluorocarbonos , Inquéritos Nutricionais , Humanos , Feminino , Masculino , Adulto , Pessoa de Meia-Idade , Fluorocarbonos/sangue , Fluorocarbonos/toxicidade , Poluentes Ambientais/sangue , Poluentes Ambientais/toxicidade , Fígado Gorduroso/sangue , Fígado Gorduroso/induzido quimicamente , Biomarcadores/sangue , Estados Unidos/epidemiologia , Exposição Ambiental/efeitos adversos , Técnicas de Imagem por Elasticidade , Alanina Transaminase/sangue , Fígado/efeitos dos fármacos , Fígado/patologia , Fígado/diagnóstico por imagemRESUMO
There is an increasing incidence and prevalence of fatty liver disease in the western world, with steatosis as the most prevalent variant. Known causes of steatosis include exposure to food-borne chemicals, and overconsumption of alcohol, carbohydrates and fat, and it is a well-known side effect of certain pharmaceuticals such as tetracycline, amiodarone and tamoxifen (drug-induced hepatic steatosis). Mechanistic knowledge on chemical-induced steatosis has greatly evolved and has been organized into adverse outcome pathways (AOPs) describing the chain of events from first molecular interaction of a substance with a biological system to the adverse outcome, intrahepatic lipid accumulation. In this study, three known steatosis-inducing pesticides (imazalil, clothianidin, and thiacloprid) were tested for their ability to induce hepatic triglyceride accumulation in the zebrafish (Danio rerio) embryo (ZFE) at 5 days post fertilization, both as single compounds and equipotent binary mixtures. The results indicate that the ZFE is very well applicable as a higher tier testing model to confirm effects in downstream key events in AOPs, that is, chemically-induced triglyceride accumulation in the whole organism and production of visible steatosis. Moreover, dose addition could be concluded for binary mixtures of substances with similar and with dissimilar modes of action.
Assuntos
Embrião não Mamífero , Fígado Gorduroso , Praguicidas , Triglicerídeos , Peixe-Zebra , Animais , Fígado Gorduroso/induzido quimicamente , Embrião não Mamífero/efeitos dos fármacos , Triglicerídeos/metabolismo , Praguicidas/toxicidade , Modelos Animais de Doenças , Relação Dose-Resposta a DrogaRESUMO
Valproic acid (VPA), a common antiepileptic drug, can cause liver steatosis after long-term therapy. However, an impact of ferroptosis on VPA-induced liver steatosis has not been investigated. In the study, treatment with VPA promoted ferroptosis in the livers of mice by elevating ferrous iron (Fe2+) levels derived from the increased absorption by transferrin receptor 1 (TFR1) and the decreased storage by ferritin (FTH1 and FTL), disrupting the redox balance via reduced levels of solute carrier family 7 member 11 (SLC7A11), glutathione (GSH), and glutathione peroxidase 4 (GPX4), and augmenting acyl-CoA synthetase long-chain family member 4 (ACSL4) -mediated lipid peroxide generation, accompanied by enhanced liver steatosis. All the changes were significantly reversed by co-treatment with an iron-chelating agent, deferoxamine mesylate (DFO) and a ferroptosis inhibitor, ferrostatin-1 (Fer-1). Similarly, the increases in Fe2+, TFR1, and ACSL4 levels, as well as the decreases in GSH, GPX4, and ferroportin (FPN) levels, were detected in VPA-treated HepG2 cells. These changes were also attenuated after co-treatment with Fer-1. It demonstrates that ferroptosis promotes VPA-induced liver steatosis through iron overload, inhibition of the GSH-GPX4 axis, and upregulation of ACSL4. It offers a potential therapy targeting ferroptosis for patients with liver steatosis following VPA treatment.
Assuntos
Coenzima A Ligases , Fígado Gorduroso , Ferroptose , Fosfolipídeo Hidroperóxido Glutationa Peroxidase , Receptores da Transferrina , Ácido Valproico , Ferroptose/efeitos dos fármacos , Animais , Ácido Valproico/toxicidade , Camundongos , Fosfolipídeo Hidroperóxido Glutationa Peroxidase/metabolismo , Coenzima A Ligases/metabolismo , Coenzima A Ligases/genética , Humanos , Fígado Gorduroso/induzido quimicamente , Fígado Gorduroso/metabolismo , Masculino , Receptores da Transferrina/metabolismo , Receptores da Transferrina/genética , Ferro/metabolismo , Camundongos Endogâmicos C57BL , Ferritinas/metabolismo , Glutationa/metabolismo , Células Hep G2 , Sistema y+ de Transporte de Aminoácidos/metabolismo , Sistema y+ de Transporte de Aminoácidos/genética , Fígado/efeitos dos fármacos , Fígado/metabolismoRESUMO
Metabolic dysfunction-associated steatotic liver disease (MASLD) is defined as morphofunctional changes in the liver. Studies have shown that Westernized eating patterns and environmental pollutants can directly induce the development of MASLD. This study evaluates the effect of co-exposure to interesterified palm oil (IPO) and 3,3',4,4',5-pentachlorobiphenyl (PCB-126) on the progression of MASLD in an animal model. C57BL/6 mice were fed IPO and co-exposed to PCB-126 for ten weeks. The co-exposure led to an imbalance in carbohydrate metabolism, increased systemic inflammation markers, and morphofunctional changes in the liver. These liver changes included the presence of inflammatory cells, fibrosis, alterations in aspartate transaminase (AST) and alanine transaminase (ALT) enzymes, and imbalance in gene expression related to fatty acid ß-oxidation, de novo lipogenesis, mitochondrial dynamics, and endoplasmic reticulum stress. Separate exposures to IPO and PCB-126 affected metabolism and MASLD progression. Nutritional and lifestyle factors may potentiate the onset and severity of MASLD.
Assuntos
Fígado , Camundongos Endogâmicos C57BL , Óleo de Palmeira , Bifenilos Policlorados , Animais , Bifenilos Policlorados/toxicidade , Camundongos , Fígado/efeitos dos fármacos , Fígado/metabolismo , Masculino , Fígado Gorduroso/induzido quimicamente , Fígado Gorduroso/metabolismo , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Poluentes Ambientais/toxicidadeRESUMO
Bioaccumulation of d-Limonene in environment due to the aggrandised usage of their natural sources like citrus food wastes and industrial day to day life products has raised concern to their biotoxicity to environment biotic health. Moreover, their after-usage discharge to aquatic system has enhanced the distress of posing threat and needs attention. This study entails mechanistic and molecular evaluation of in-vivo biotoxicity of d-Limonene in zebrafish embryo models. Experimental analysis excavated the controlled concentration-dependent morphological, physiological and cellular in-vivo impact of d-Limonene in zebrafish embryos through significant changes in oxidative stress, steatosis and apoptosis regulated via 6-fold and 5-fold mRNA expression change in p53 and Sod1 genes. Computational evaluation deduced the cellular mechanism of d-limonene biotoxicity as irregularities in oxidative stress, apoptosis and steatosis due of their intrinsic interaction with metabolic proteins like Zhe1a (-4.8 Kcal/mol), Sod1(-5.3 Kcal/mol), p53, caspase3 and apoa1 leading to influential change in structural and functional integrity of the metabolic proteins. The study unravelled the measured in-vivo biotoxicity of d-Limonene at cellular and molecular level to advocate the controlled usage of d-Limonene related natural and industrial product for a sustainable environmental health.
Assuntos
Apoptose , Limoneno , Estresse Oxidativo , Peixe-Zebra , Animais , Limoneno/toxicidade , Apoptose/efeitos dos fármacos , Poluentes Químicos da Água/toxicidade , Embrião não Mamífero/efeitos dos fármacos , Fígado Gorduroso/induzido quimicamenteRESUMO
Accumulating animal studies have demonstrated associations between ambient air pollution (AP) and metabolic dysfunction-associated fatty liver disease (MAFLD), but relevant epidemiological evidence is limited. We evaluated the association of long-term exposure to AP with the risk of incident MAFLD in Northwest China. The average AP concentration between baseline and follow-up was used to assess individual exposure levels. Cox proportional hazard models and restricted cubic spline functions (RCS) were used to estimate the association of PM2.5 and its constituents with the risk of MAFLD and the dose-response relationship. Quantile g-computation was used to assess the joint effects of mixed exposure to air pollutants on MAFLD and the weights of the various pollutants. We observed 1516 cases of new-onset MAFLD, with an incidence of 10.89%. Increased exposure to pollutants was significantly associated with increased odds of MAFLD, with hazard ratios (HRs) of 2.93 (95% CI: 1.22, 7.00), 2.86 (1.44, 5.66), 7.55 (3.39, 16.84), 4.83 (1.89, 12.38), 3.35 (1.35, 8.34), 1.89 (1.02, 1.62) for each interquartile range increase in PM2.5, SO42-, NO3-, NH4+, OM, and BC, respectively. Stratified analyses suggested that females, frequent exercisers and never-drinkers were more susceptible to MAFLD associated with ambient PM2.5 and its constituents. Mixed exposure to SO42-, NO3-, NH4+, OM and BC was associated with an increased risk of MAFLD, and the weight of BC had the strongest effect on MAFLD. Exposure to ambient PM2.5 and its constituents increased the risk of MAFLD.
Assuntos
Poluentes Atmosféricos , Material Particulado , Humanos , China/epidemiologia , Masculino , Feminino , Pessoa de Meia-Idade , Estudos de Coortes , Adulto , Exposição Ambiental/efeitos adversos , Fígado Gorduroso/induzido quimicamente , Fígado Gorduroso/epidemiologia , Modelos de Riscos Proporcionais , Incidência , Poluição do Ar/efeitos adversos , Doenças Metabólicas/epidemiologia , Doenças Metabólicas/induzido quimicamente , IdosoRESUMO
OBJECTIVE: This study evaluated the effects of long-term polystyrene microplastics (PS-MPs) exposure on hepatic lipid metabolism in vivo by lipidomics. RESULTS: H&E staining showed long-term PS-MPs exposure could trigger the hepatic inflammatory cell infiltration and hepatic steatosis in SD rats, indicating long-term PS-MPs exposure caused hepatoxicity. Lipidomics revealed that the concentrations of 8 lipid metabolites in the liver were altered after exposure to PS-MPs for both 6 and 12 months, namely LdMePE (16:0), LPC (18:1), LPC (18:2), LPC (20:4), PC (17:0_20:4), PC (18:2_22:6), PC (22:6_13:0) and SM (d18:1_24:0), which were all statistically different from the control groups detected at both time points after PS-MPs exposure, suggesting the mainly metabolic pathway was glycerolipid metabolism. CONCLUSION: This study showed chronic exposure to PS-MPs could cause hepatotoxicity and induce hepatic lipidomics alterations in vivo, which could provide an essential clue for the safety assessment of PS-MPs.
Assuntos
Metabolismo dos Lipídeos , Lipidômica , Fígado , Microplásticos , Poliestirenos , Ratos Sprague-Dawley , Animais , Poliestirenos/toxicidade , Fígado/efeitos dos fármacos , Fígado/metabolismo , Microplásticos/toxicidade , Metabolismo dos Lipídeos/efeitos dos fármacos , Masculino , Ratos , Lipídeos , Fatores de Tempo , Doença Hepática Induzida por Substâncias e Drogas/etiologia , Doença Hepática Induzida por Substâncias e Drogas/metabolismo , Doença Hepática Induzida por Substâncias e Drogas/patologia , Fígado Gorduroso/induzido quimicamente , Fígado Gorduroso/metabolismoRESUMO
Activation of pregnane X receptor (PXR) by xenobiotics has been associated with metabolic diseases. This study aimed to reveal the impact of PXR activation on hepatic metabolome and explore novel mechanisms underlying PXR-mediated lipid metabolism disorder in the liver. Wild-type and PXR-deficient male C57BL/6 mice were used as in vivo models, and hepatic steatosis was induced by pregnenolone-16α-carbonitrile, a typical rodent PXR agonist. Metabolomic analysis of liver tissues showed that PXR activation led to significant changes in metabolites involved in multiple metabolic pathways previously reported, including lipid metabolism, energy homeostasis, and amino acid metabolism. Moreover, the level of hepatic all-trans retinoic acid (ATRA), the main active metabolite of vitamin A, was significantly increased by PXR activation, and genes involved in ATRA metabolism exhibited differential expression following PXR activation or deficiency. Consistent with previous research, the expression of downstream target genes of peroxisome proliferator-activated receptor α (PPARα) was decreased. Analysis of fatty acids by Gas Chromatography-Mass Spectrometer further revealed changes in polyunsaturated fatty acid metabolism upon PXR activation, suggesting inhibition of PPARα activity. Taken together, our findings reveal a novel metabolomic signature of hepatic steatosis induced by PXR activation in mice.
Assuntos
Ácidos Graxos Insaturados , Fígado Gorduroso , Fígado , Metabolômica , Camundongos Endogâmicos C57BL , PPAR alfa , Receptor de Pregnano X , Tretinoína , Animais , Masculino , Receptor de Pregnano X/metabolismo , Receptor de Pregnano X/genética , Tretinoína/metabolismo , Fígado/metabolismo , Fígado/efeitos dos fármacos , Fígado Gorduroso/metabolismo , Fígado Gorduroso/induzido quimicamente , Ácidos Graxos Insaturados/metabolismo , PPAR alfa/metabolismo , PPAR alfa/genética , Metabolismo dos Lipídeos/efeitos dos fármacos , Camundongos , Camundongos Knockout , Carbonitrila de Pregnenolona/farmacologia , Modelos Animais de DoençasRESUMO
Antituberculosis drugs induce pharmacologic cholestatic liver injury with long-term administration. Liver injury resulting from rifampicin is potentially related to the bile acid nuclear receptor Farnesoid X Receptor (FXR). To investigate this, cholestasis was induced in both wild-type (C57BL/6N) mice and FXR knockout (FXR-null) mice through administration of rifampicin (200 mg/kg) via gavage for 7 consecutive days. Compared with C57BL/6N mice, FXR-null mice exhibited more severe liver injury after rifampicin administration, characterized by enlarged liver size, elevated transaminases, and increased inflammation. Moreover, under rifampicin treatment, FXR knockout impairs lipid secretion and exacerbates hepatic steatosis. Significantly, the expression of metabolism molecules BSEP increased, while NTCP and CYP7A1 decreased following rifampicin administration in C57BL/6N mice, whereas these changes were absent in FXR knockout mice. Furthermore, rifampicin treatment in both C57BL/6N and FXR-null mice was associated with elevated c-Jun N-terminal kinase phosphorylation (p-JNK) levels, with a more pronounced elevation in FXR-null mice. Our study suggests that rifampicin-induced liver injury, steatosis, and cholestasis are associated with FXR dysfunction and altered bile acid metabolism, and that the JNK signaling pathway is partially implicated in this injury. Based on these results, we propose that FXR might be a novel therapeutic target for addressing drug-induced liver injury.
Assuntos
Doença Hepática Induzida por Substâncias e Drogas , Fígado , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores Citoplasmáticos e Nucleares , Rifampina , Animais , Rifampina/efeitos adversos , Receptores Citoplasmáticos e Nucleares/metabolismo , Receptores Citoplasmáticos e Nucleares/genética , Doença Hepática Induzida por Substâncias e Drogas/tratamento farmacológico , Doença Hepática Induzida por Substâncias e Drogas/patologia , Fígado/patologia , Fígado/efeitos dos fármacos , Fígado/metabolismo , Masculino , Camundongos , Membro 11 da Subfamília B de Transportadores de Cassetes de Ligação de ATP/genética , Membro 11 da Subfamília B de Transportadores de Cassetes de Ligação de ATP/metabolismo , Colesterol 7-alfa-Hidroxilase/genética , Colesterol 7-alfa-Hidroxilase/metabolismo , Simportadores/genética , Simportadores/metabolismo , Ácidos e Sais Biliares/metabolismo , Transportadores de Ânions Orgânicos Dependentes de Sódio/genética , Transportadores de Ânions Orgânicos Dependentes de Sódio/metabolismo , Colestase/induzido quimicamente , Colestase/tratamento farmacológico , Colestase/metabolismo , Fígado Gorduroso/tratamento farmacológico , Fígado Gorduroso/induzido quimicamente , Fígado Gorduroso/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismoRESUMO
Dexamethasone (DXMS), a synthetic glucocorticoid, is known for its pharmacological effects on anti-inflammation, stress response enhancement and immune suppression, and has been widely used to treat potential premature delivery and related diseases. However, emerging evidence has shown that prenatal DXMS exposure leads to increased susceptibility to multiple diseases. In the present study, we used zebrafish as a model to study the effects of embryonic DXMS exposure on liver development and disease. We discovered that embryonic DXMS exposure upregulated the levels of total cholesterol and triglycerides in the liver, increased the glycolysis process and ultimately caused hepatic steatosis in zebrafish larvae. Furthermore, DXMS exposure exacerbated hepatic steatosis in a zebrafish model of fatty liver disease. In addition, we showed that embryonic DXMS exposure worsened liver injury induced by paracetamol (N-acetyl-p-aminophenol, APAP), increased the infiltration of macrophages and neutrophils, and promoted the expression of inflammatory factors, leading to impeded liver regeneration. Taken together, our results provide new evidence that embryonic DXMS exposure exacerbates hepatic steatosis by activating glycolytic pathway, aggravates APAP-induced liver damage and impeded regeneration under a persistent inflammation, calling attention to DXMS administration during pregnancy with probable clinical implications for offspring.
Assuntos
Acetaminofen , Doença Hepática Induzida por Substâncias e Drogas , Dexametasona , Fígado Gorduroso , Peixe-Zebra , Animais , Dexametasona/toxicidade , Acetaminofen/toxicidade , Fígado Gorduroso/induzido quimicamente , Fígado Gorduroso/patologia , Doença Hepática Induzida por Substâncias e Drogas/patologia , Fígado/efeitos dos fármacos , Fígado/patologia , Embrião não Mamífero/efeitos dos fármacos , Feminino , Modelos Animais de DoençasRESUMO
Hepatic steatosis is the first step in a series of events that drives hepatic disease and has been considerably associated with exposure to fine particulate matter (PM2.5). Although the chemical constituents of particles matter in the negative health effects, the specific components of PM2.5 that trigger hepatic steatosis remain unclear. New strategies prioritizing the identification of the key components with the highest potential to cause adverse effects among the numerous components of PM2.5 are needed. Herein, we established a high-resolution mass spectrometry (MS) data set comprising the hydrophobic organic components corresponding to 67 PM2.5 samples in total from Taiyuan and Guangzhou, two representative cities in North and South China, respectively. The lipid accumulation bioeffect profiles of the above samples were also obtained. Considerable hepatocyte lipid accumulation was observed in most PM2.5 extracts. Subsequently, 40 of 695 components were initially screened through machine learning-assisted data filtering based on an integrated bioassay with MS data. Next, nine compounds were further selected as candidates contributing to hepatocellular steatosis based on absorption, distribution, metabolism, and excretion evaluation and molecular dockingin silico. Finally, seven components were confirmed in vitro. This study provided a multilevel screening strategy for key active components in PM2.5 and provided insight into the hydrophobic PM2.5 components that induce hepatocellular steatosis.
Assuntos
Interações Hidrofóbicas e Hidrofílicas , Material Particulado , Fígado Gorduroso/induzido quimicamente , Humanos , China , Poluentes AtmosféricosRESUMO
This study explores the impact of environmental pollutants on nuclear receptors (CAR, PXR, PPARα, PPARγ, FXR, and LXR) and their heterodimerization partner, the Retinoid X Receptor (RXR). Such interaction may contribute to the onset of non-alcoholic fatty liver disease (NAFLD), which is initially characterized by steatosis and potentially progresses to steatohepatitis and fibrosis. Epidemiological studies have linked NAFLD occurrence to the exposure to environmental contaminants like PFAS. This study aims to assess the simultaneous activation of nuclear receptors via perfluorooctanoic acid (PFOA) and RXR coactivation via Tributyltin (TBT), examining their combined effects on steatogenic mechanisms. Mice were exposed to PFOA (10 mg/kg/day), TBT (5 mg/kg/day) or a combination of them for three days. Mechanisms underlying hepatic steatosis were explored by measuring nuclear receptor target gene and lipid metabolism key gene expressions, by quantifying plasma lipids and hepatic damage markers. This study elucidated the involvement of the Liver X Receptor (LXR) in the combined effect on steatosis and highlighted the permissive nature of the LXR/RXR heterodimer. Antagonistic effects of TBT on the PFOA-induced activation of the Pregnane X Receptor (PXR) and Peroxisome Proliferator-Activated Receptor Gamma (PPARγ) were also observed. Overall, this study revealed complex interactions between PFOA and TBT, shedding light on their combined impact on liver health.
Assuntos
Caprilatos , Fluorocarbonos , Compostos de Trialquitina , Animais , Compostos de Trialquitina/farmacologia , Caprilatos/farmacologia , Camundongos , Fluorocarbonos/toxicidade , Fluorocarbonos/farmacologia , Masculino , Camundongos Endogâmicos C57BL , Receptores X do Fígado/metabolismo , Fígado/metabolismo , Fígado/efeitos dos fármacos , Fígado/patologia , Receptores X de Retinoides/metabolismo , Fígado Gorduroso/metabolismo , Fígado Gorduroso/induzido quimicamente , Fígado Gorduroso/patologia , Receptores Citoplasmáticos e Nucleares/metabolismo , Metabolismo dos Lipídeos/efeitos dos fármacos , Hepatopatia Gordurosa não Alcoólica/metabolismo , Hepatopatia Gordurosa não Alcoólica/patologia , Hepatopatia Gordurosa não Alcoólica/induzido quimicamenteRESUMO
Chlormequat chloride (CCC), a widely used plant growth regulator, is a choline analogue that has been shown to have endocrine-disrupting effects. Previous studies have shown that maternal exposure to CCC could induce hyperlipidemia and growth disruption in rat offspring. This study aims to further investigate the effects of peripubertal exposure to CCC on pubertal development and lipid homeostasis, as well as the underlying mechanisms. In vivo, male weanling rats were exposed to CCC (0, 20, 75 and 200 mg/kg bw/day) from post-natal day 21-60 via daily oral gavage. The results in rats showed that 75 mg/kg CCC treatment induced hepatic steatosis, predominantly microvesicular steatosis with a small amount of macrovesicular steatosis, in rat livers and 200 mg/kg CCC treatment induced liver damage including inflammatory infiltration, hepatic sinusoidal dilation and necrosis. In vitro, HepG2 cells were treated with CCC (0, 30, 60, 120, 240 and 480 µg/mL) for 24 h. And the results showed that CCC above 120 µg/mL induced an increase in triglyceride and neutral lipid levels of HepG2 cells. Mechanism exploration revealed that CCC treatment promoted the activation of mTOR/SREBP1 signalling pathway and inhibited activation of AMPK in both in vivo rat livers and in vitro HepG2 cells. Treatment with AMPK activator Acadesine (AICAR) could alleviate the lipid accumulation in HepG2 cells induced by CCC. Collectively, the present results indicate that CCC might induce hepatic steatosis by promoting mTOR/SREBP1 mediated lipogenesis via AMPK inhibition.
Assuntos
Proteínas Quinases Ativadas por AMP , Clormequat , Fígado Gorduroso , Lipogênese , Proteína de Ligação a Elemento Regulador de Esterol 1 , Serina-Treonina Quinases TOR , Animais , Serina-Treonina Quinases TOR/metabolismo , Masculino , Fígado Gorduroso/induzido quimicamente , Fígado Gorduroso/metabolismo , Lipogênese/efeitos dos fármacos , Proteínas Quinases Ativadas por AMP/metabolismo , Humanos , Células Hep G2 , Proteína de Ligação a Elemento Regulador de Esterol 1/metabolismo , Proteína de Ligação a Elemento Regulador de Esterol 1/genética , Ratos , Clormequat/toxicidade , Ratos Sprague-Dawley , Fígado/efeitos dos fármacos , Fígado/metabolismoRESUMO
Exposure to fine particulate matter (PM2.5) is a significant risk factor for hepatic steatosis. The N6-methyladenosine (m6A) is implicated in metabolic disturbances triggered by exogenous environmental factors. However, the role of m6A in mediating PM2.5-induced hepatic steatosis remains unclear. Herein, male C57BL/6J mice were subjected to PM2.5 exposure throughout the entire heating season utilizing a real-ambient PM2.5 whole-body inhalation exposure system. Concurrently, HepG2 cell models exposed to PM2.5 were developed to delve the role of m6A methylation modification. Following PM2.5 exposure, significant hepatic lipid accumulation and elevated global m6A level were observed both in vitro and in vivo. The downregulation of YTHDC2, an m6A-binding protein, might contribute to this alteration. In vitro studies revealed that lipid-related genes CEPT1 and YWHAH might be targeted by m6A modification. YTHDC2 could bind to CDS region of them and increase their stability. Exposure to PM2.5 shortened mRNA lifespan and suppressed the expression of CEPT1 and YWHAH, which were reversed to baseline or higher level upon the enforced expression of YTHDC2. Consequently, our findings indicate that PM2.5 induces elevated m6A methylation modification of CEPT1 and YWHAH by downregulating YTHDC2, which in turn mediates the decrease in the mRNA stabilization and expression of these genes, ultimately resulting in hepatic steatosis.