Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Antioxidants (Basel) ; 11(5)2022 Apr 30.
Article in English | MEDLINE | ID: mdl-35624761

ABSTRACT

Drug-induced liver injury (DILI) development is commonly associated with acetaminophen (APAP) overdose, where glutathione scavenging leads to mitochondrial dysfunction and hepatocyte death. DILI is a severe disorder without effective late-stage treatment, since N-acetyl cysteine must be administered 8 h after overdose to be efficient. Ammonia homeostasis is altered during liver diseases and, during DILI, it is accompanied by decreased glycine N-methyltransferase (GNMT) expression and S-adenosylmethionine (AdoMet) levels that suggest a reduced methionine cycle. Anti-miR-873-5p treatment prevents cell death in primary hepatocytes and the appearance of necrotic areas in liver from APAP-administered mice. In our study, we demonstrate a GNMT and methionine cycle activity restoration by the anti-miR-873-5p that reduces mitochondrial dysfunction and oxidative stress. The lack of hyperammoniemia caused by the therapy results in a decreased urea cycle, enhancing the synthesis of polyamines from ornithine and AdoMet and thus impacting the observed recovery of mitochondria and hepatocyte proliferation for regeneration. In summary, anti-miR-873-5p appears to be an effective therapy against APAP-induced liver injury, where the restoration of GNMT and the methionine cycle may prevent mitochondrial dysfunction while activating hepatocyte proliferative response.

2.
Sci Rep ; 11(1): 22119, 2021 11 11.
Article in English | MEDLINE | ID: mdl-34764412

ABSTRACT

REACH (Registration, Evaluation, Authorization and Restriction of Chemicals) is a global strategy and regulation policy of the EU that aims to improve the protection of human health and the environment through the better and earlier identification of the intrinsic properties of chemical substances. It entered into force on 1st June 2007 (EC 1907/2006). REACH and EU policies plead for the use of robust high-throughput "omic" techniques for the in vitro investigation of the toxicity of chemicals that can provide an estimation of their hazards as well as information regarding the underlying mechanisms of toxicity. In agreement with the 3R's principles, cultured cells are nowadays widely used for this purpose, where metabolomics can provide a real-time picture of the metabolic effects caused by exposure of cells to xenobiotics, enabling the estimations about their toxicological hazards. High quality and robust metabolomics data sets are essential for precise and accurate hazard predictions. Currently, the acquisition of consistent and representative metabolomic data is hampered by experimental drawbacks that hinder reproducibility and difficult robust hazard interpretation. Using the differentiated human liver HepG2 cells as model system, and incubating with hepatotoxic (acetaminophen and valproic acid) and non-hepatotoxic compounds (citric acid), we evaluated in-depth the impact of several key experimental factors (namely, cell passage, processing day and storage time, and compound treatment) and instrumental factors (batch effect) on the outcome of an UPLC-MS metabolomic analysis data set. Results showed that processing day and storage time had a significant impact on the retrieved cell's metabolome, while the effect of cell passage was minor. Meta-analysis of results from pathway analysis showed that batch effect corrections and quality control (QC) measures are critical to enable consistent and meaningful estimations of the effects caused by compounds on cells. The quantitative analysis of the changes in metabolic pathways upon bioactive compound treatment remained consistent despite the concurrent causes of metabolomic data variation. Thus, upon appropriate data retrieval and correction and by an innovative metabolic pathway analysis, the metabolic alteration predictions remained conclusive despite the acknowledged sources of variability.


Subject(s)
Chemical and Drug Induced Liver Injury/genetics , Liver/drug effects , Metabolomics/methods , Acetaminophen/toxicity , Cell Line, Tumor , Citric Acid/toxicity , Hep G2 Cells , Humans , Metabolic Networks and Pathways/drug effects , Metabolome/drug effects , Metabolome/genetics , Quality Control , Reproducibility of Results , Valproic Acid/toxicity , Xenobiotics/toxicity
3.
Anal Chim Acta ; 1187: 339173, 2021 Dec 01.
Article in English | MEDLINE | ID: mdl-34753565

ABSTRACT

Extraction of meaningful biological information from the vast array of data that metabolomics analyses generate is a major challenge in the field. A variety of computational and visual tools that help to identify changes in metabolic pathways have been proposed including functional analysis and pathway analysis. Meta-analysis of metabolomic data has emerged as a powerful source of information. In this work, the applicability of the Mantel's test for the correlation of functional results from metabolic pathway analysis is shown using experimental and simulated data sets as evaluation examples. The statistical significance of the correlation coefficient can be assessed by permutation testing requiring practically no computation time. The use of the Mantel's test can assist the critical comparison of different phenotypes, studies, methods, platforms, or data preprocessing strategies, as well as help to identify inconsistencies between metabolomic study outcomes, making this algorithm attractive for data interpretation and meta-analysis on a routine basis.


Subject(s)
Metabolic Networks and Pathways , Metabolomics , Research Design
4.
Anal Chem ; 92(21): 14542-14549, 2020 11 03.
Article in English | MEDLINE | ID: mdl-33084322

ABSTRACT

The estimation of steatosis in a liver graft is mandatory prior to liver transplantation, as the risk of graft failure increases with the level of infiltrated fat. However, the assessment of liver steatosis before transplantation is typically based on a qualitative or semiquantitative characterization by visual inspection and palpation and histological analysis. Thus, there is an unmet need for transplantation surgeons to have access to a diagnostic tool enabling an in situ fast classification of grafts prior to extraction. In this study, we have assessed an attenuated total reflection-Fourier transform infrared (ATR-FTIR) spectroscopic method compatible with the requirements of an operation room for the evaluation of the lipid contents in human livers. A set of 20 human liver biopsies obtained from organs intended for transplantation were analyzed by expert pathologists, ATR-FTIR spectroscopy, lipid biochemical analysis, and UPLC-ESI(+/-)TOFMS for lipidomic profiling. Comparative analysis of multisource data showed strong correlations between ATR-FTIR, clinical, and lipidomic information. Results show that ATR-FTIR captures a global picture of the lipid composition of the liver, along with information for the quantification of the triradylglycerol content in liver biopsies. Although the methodology performance needs to be further validated, results support the applicability of ATR-FTIR for the in situ determination of the grade of liver steatosis at the operation room as a fast, quantitative method, as an alternative to the qualitative and subjective pathological examination.


Subject(s)
Liver Transplantation , Operating Rooms , Spectrophotometry, Infrared/methods , Humans , Time Factors
5.
Biomolecules ; 10(3)2020 03 24.
Article in English | MEDLINE | ID: mdl-32213983

ABSTRACT

The vitamin D receptor (VDR) must be relevant to liver lipid metabolism because VDR deficient mice are protected from hepatosteatosis. Therefore, our objective was to define the role of VDR on the overall lipid metabolism in human hepatocytes. We developed an adenoviral vector for human VDR and performed transcriptomic and metabolomic analyses of cultured human hepatocytes upon VDR activation by vitamin D (VitD). Twenty percent of the VDR responsive genes were related to lipid metabolism, including MOGAT1, LPGAT1, AGPAT2, and DGAT1 (glycerolipid metabolism); CDS1, PCTP, and MAT1A (phospholipid metabolism); and FATP2, SLC6A12, and AQP3 (uptake of fatty acids, betaine, and glycerol, respectively). They were rapidly induced (4-6 h) upon VDR activation by 10 nM VitD or 100 µM lithocholic acid (LCA). Most of these genes were also upregulated by VDR/VitD in mouse livers in vivo. Ultra-performance liquid chromatography-tandem mass spectrometry (UPLC-MS) metabolomics demonstrated intracellular accumulation of triglycerides, with concomitant decreases in diglycerides and phosphatidates, at 8 and 24 h upon VDR activation. Significant alterations in phosphatidylcholines, increases in lyso-phosphatidylcholines and decreases in phosphatidylethanolamines and phosphatidylethanolamine plasmalogens were also observed. In conclusion, active VitD/VDR signaling in hepatocytes triggers an unanticipated coordinated gene response leading to triglyceride synthesis and to important perturbations in glycerolipids and phospholipids.


Subject(s)
Gene Expression Regulation , Hepatocytes/metabolism , Phospholipids/biosynthesis , Receptors, Calcitriol/metabolism , Triglycerides/biosynthesis , Animals , Hep G2 Cells , Humans , Mice , Mice, Knockout, ApoE , Phospholipids/genetics , Receptors, Calcitriol/genetics , Triglycerides/genetics
6.
Arch Toxicol ; 94(2): 589-607, 2020 02.
Article in English | MEDLINE | ID: mdl-31894354

ABSTRACT

Anabolic-androgenic steroids are testosterone derivatives, used by body-builders to increase muscle mass. Epistane (EPI) is an orally administered 17α-alkylated testosterone derivative with 2a-3a epithio ring. We identified four individuals who, after EPI consumption, developed long-lasting cholestasis. The bile acid (BA) profile of three patients was characterized, as well the molecular mechanisms involved in this pathology. The serum BA pool was increased from 14 to 61-fold, basically on account of primary conjugated BA (cholic acid (CA) conjugates), whereas secondary BA were very low. In in vitro experiments with cultured human hepatocytes, EPI caused the accumulation of glycoCA in the medium. Moreover, as low as 0.01 µM EPI upregulated the expression of key BA synthesis genes (CYP7A1, by 65% and CYP8B1, by 67%) and BA transporters (NTCP, OSTA and BSEP), and downregulated FGF19. EPI increased the uptake/accumulation of a fluorescent BA analogue in hepatocytes by 50-70%. Results also evidenced, that 40 µM EPI trans-activated the nuclear receptors LXR and PXR. More importantly, 0.01 µM EPI activated AR in hepatocytes, leading to an increase in the expression of CYP8B1. In samples from a human liver bank, we proved that the expression of AR was positively correlated with that of CYP8B1 in men. Taken together, we conclude that EPI could cause cholestasis by inducing BA synthesis and favouring BA accumulation in hepatocytes, at least in part by AR activation. We anticipate that the large phenotypic variability of BA synthesis enzymes and transport genes in man provide a putative explanation for the idiosyncratic nature of EPI-induced cholestasis.


Subject(s)
Bile Acids and Salts/blood , Cholestasis/chemically induced , Hepatocytes/drug effects , Hepatocytes/metabolism , Testosterone Congeners/toxicity , Adult , Bile Acids and Salts/biosynthesis , Bile Acids and Salts/metabolism , Chemical and Drug Induced Liver Injury/etiology , Chemical and Drug Induced Liver Injury/metabolism , Cholestasis/metabolism , Cholic Acid/metabolism , Female , Fibroblast Growth Factors/genetics , Gene Expression Regulation/drug effects , Hep G2 Cells , Humans , Liver-Specific Organic Anion Transporter 1/genetics , Male , Receptors, Androgen/genetics , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Solute Carrier Organic Anion Transporter Family Member 1B3/genetics , Steroid 12-alpha-Hydroxylase/genetics , Steroid 12-alpha-Hydroxylase/metabolism , Up-Regulation/drug effects , Young Adult
7.
Mol Nutr Food Res ; 63(20): e1900487, 2019 10.
Article in English | MEDLINE | ID: mdl-31322321

ABSTRACT

SCOPE: Gut microbiota contributes to non-alcoholic fatty liver disease (NAFLD) pathogenesis by multiple mechanisms not yet completely understood. Novel differential features between germ-free mice (GFm) transplanted with protective or non-protective cecal microbiota against NAFLD are investigated. METHODS AND RESULTS: Gut microbiota composition, plasma, and fecal bile acids (BAs) and liver mRNAs are quantified in GFm recipients from four donor mice differing in NAFLD severity (control diet, high-fat diet [HFD]-responder, HFD-non-responder, and quercetin-supplemented HFD). Transplanted GFm are on control or HFD for 16-weeks. Multivariate analysis shows that GFm colonized with microbiota from HFD-non-responder and quercetin supplemented-HFD donors (protected against NAFLD) clusters together, whereas GFm colonized with microbiota from control and HFD-responder mice (non-protected against NAFLD) establishes another cluster. Protected phenotype is associated with increased gut Desulfovibrio and Oscillospira, reduced gut Bacteroides and Oribacterium, lower primary and higher secondary BAs in plasma and feces, induction of hepatic BA transporters, and repression of hepatic lipogenic and BA synthesis genes. CONCLUSION: Protective gut microbiota associates with increased specific secondary BAs, which likely inhibit lipogenic pathways and enhance bile flow in the liver. This novel cross-talk between gut and liver, via plasma BAs, that promotes protection against NAFLD may have clinical and nutritional relevance.


Subject(s)
Bile Acids and Salts/blood , Gastrointestinal Microbiome , Liver/metabolism , Non-alcoholic Fatty Liver Disease/prevention & control , Animals , Diet, High-Fat , Ethanol/blood , Male , Mice , Mice, Inbred C57BL , Transcriptome
8.
Arch Toxicol ; 91(2): 865-883, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27161439

ABSTRACT

Understanding central mechanisms underlying drug-induced toxicity plays a crucial role in drug development and drug safety. However, a translation of cellular in vitro findings to an actual in vivo context remains challenging. Here, physiologically based pharmacokinetic (PBPK) modeling was used for in vivo contextualization of in vitro toxicity data (PICD) to quantitatively predict in vivo drug response over time by integrating multiple levels of biological organization. Explicitly, in vitro toxicity data at the cellular level were integrated into whole-body PBPK models at the organism level by coupling in vitro drug exposure with in vivo drug concentration-time profiles simulated in the extracellular environment within the organ. PICD was exemplarily applied on the hepatotoxicant azathioprine to quantitatively predict in vivo drug response of perturbed biological pathways and cellular processes in rats and humans. The predictive accuracy of PICD was assessed by comparing in vivo drug response predicted for rats with observed in vivo measurements. To demonstrate clinical applicability of PICD, in vivo drug responses of a critical toxicity-related pathway were predicted for eight patients following acute azathioprine overdoses. Moreover, acute liver failure after multiple dosing of azathioprine was investigated in a patient case study by use of own clinical data. Simulated pharmacokinetic profiles were therefore related to in vivo drug response predicted for genes associated with observed clinical symptoms and to clinical biomarkers measured in vivo. PICD provides a generic platform to investigate drug-induced toxicity at a patient level and thus may facilitate individualized risk assessment during drug development.


Subject(s)
Azathioprine/toxicity , Drug-Related Side Effects and Adverse Reactions , Models, Theoretical , Pharmacokinetics , Adult , Animals , Azathioprine/adverse effects , Chemical and Drug Induced Liver Injury/etiology , Drug Overdose/etiology , Humans , Male , Rats , Reproducibility of Results , Toxicity Tests/methods , Toxicity Tests, Acute/methods
9.
Anal Bioanal Chem ; 408(4): 1217-30, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26769129

ABSTRACT

MS-based metabolite profiling of adherent mammalian cells comprises several challenging steps such as metabolism quenching, cell detachment, cell disruption, metabolome extraction, and metabolite measurement. In LC-MS, the final metabolome coverage is strongly determined by the separation technique and the MS conditions used. Human liver-derived cell line HepG2 was chosen as adherent mammalian cell model to evaluate the performance of several commonly used procedures in both sample processing and LC-MS analysis. In a first phase, metabolite extraction and sample analysis were optimized in a combined manner. To this end, the extraction abilities of five different solvents (or combinations) were assessed by comparing the number and the levels of the metabolites comprised in each extract. Three different chromatographic methods were selected for metabolites separation. A HILIC-based method which was set to specifically separate polar metabolites and two RP-based methods focused on lipidome and wide-ranging metabolite detection, respectively. With regard to metabolite measurement, a Q-ToF instrument operating in both ESI (+) and ESI (-) was used for unbiased extract analysis. Once metabolite extraction and analysis conditions were set up, the influence of cell harvesting on metabolome coverage was also evaluated. Therefore, different protocols for cell detachment (trypsinization or scraping) and metabolism quenching were compared. This study confirmed the inconvenience of trypsinization as a harvesting technique, and the importance of using complementary extraction solvents to extend metabolome coverage, minimizing interferences and maximizing detection, thanks to the use of dedicated analytical conditions through the combination of HILIC and RP separations. The proposed workflow allowed the detection of over 300 identified metabolites from highly polar compounds to a wide range of lipids.


Subject(s)
Chromatography, Liquid/methods , Liver/metabolism , Metabolome , Metabolomics/methods , Animals , Cell Adhesion , Cells, Cultured , Chromatography, High Pressure Liquid/methods , Cytological Techniques , Hep G2 Cells/chemistry , Hep G2 Cells/metabolism , Humans , Liquid-Liquid Extraction/methods , Liver/cytology , Rats , Spectrometry, Mass, Electrospray Ionization/methods , Workflow
10.
World J Gastroenterol ; 22(2): 874-86, 2016 Jan 14.
Article in English | MEDLINE | ID: mdl-26811633

ABSTRACT

This review aims to share the lessons we learned over time during the setting of the hepatocyte transplantation (HT) program at the Hepatic Cell Therapy Unit at Hospital La Fe in Valencia. New sources of liver tissue for hepatocyte isolation have been explored. The hepatocyte isolation and cryopreservation procedures have been optimized and quality criteria for assessment of functionality of hepatocyte preparations and suitability for HT have been established. The results indicate that: (1) Only highly viable and functional hepatocytes allow to recover those functions lacking in the native liver; (2) Organs with steatosis (≥ 40%) and from elderly donors are declined since low hepatocyte yields, viability and cell survival after cryopreservation, are obtained; (3) Neonatal hepatocytes are cryopreserved without significant loss of viability or function representing high-quality cells to improve human HT; (4) Cryopreservation has the advantage of providing hepatocytes constantly available and of allowing the quality evaluation and suitability for transplantation; and (5) Our results from 5 adults with acute liver failure and 4 from children with inborn metabolic diseases, indicate that HT could be a very useful and safe cell therapy, as long as viable and metabolically functional human hepatocytes are used.


Subject(s)
Hepatocytes/transplantation , Liver Failure, Acute/surgery , Liver Transplantation/methods , Metabolism, Inborn Errors/surgery , Adolescent , Adult , Aged , Aged, 80 and over , Cell Separation/methods , Cell Survival , Cryopreservation/methods , Diffusion of Innovation , Donor Selection , Female , Forecasting , Graft Survival , Humans , Infant , Infant, Newborn , Liver Failure, Acute/diagnosis , Liver Failure, Acute/metabolism , Liver Transplantation/adverse effects , Liver Transplantation/trends , Male , Metabolism, Inborn Errors/diagnosis , Metabolism, Inborn Errors/metabolism , Middle Aged , Patient Selection , Risk Factors , Treatment Outcome
11.
Stem Cell Res Ther ; 6: 246, 2015 Dec 12.
Article in English | MEDLINE | ID: mdl-26652177

ABSTRACT

BACKGROUND: Hepatic cell therapy has become a viable alternative to liver transplantation for life-threatening liver diseases. However, the supply of human hepatocytes is limited due to the shortage of suitable donor organs required to isolate high-quality cells. Human pluripotent stem cells reflect a potential renewable source for generating functional hepatocytes. However, most differentiation protocols use undefined matrices or factors of animal origin; as such, the resulting hepatocytes are not Good Manufacturing Practice compliant. Moreover, the preclinical studies employed to assess safety and function of human embryonic stem cell (hESC)-derived hepatocytes are generally limited to immunodeficient mice. In the present study, we evaluate the generation of hepatocytes under defined conditions using a European hESC line (VAL9) which was derived under animal-free conditions. The function capacity of VAL9-derived hepatocytes was assessed by transplantation into mice with acetaminophen-induced acute liver failure, a clinically relevant model. METHODS: We developed a protocol that successfully differentiates hESCs into bipotent hepatic progenitors under defined conditions, without the use of chromatin modifiers such as dimethyl sulphoxide. These progenitors can be cryopreserved and are able to generate both committed precursors of cholangiocytes and neonate-like hepatocytes. RESULTS: Thirty days post-differentiation, hESCs expressed hepatocyte-specific markers such as asialoglycoprotein receptor and hepatic nuclear factors including HNF4α. The cells exhibited properties of mature hepatocytes such as urea secretion and UGT1A1 and cytochrome P450 activities. When transplanted into mice with acetaminophen-induced acute liver failure, a model of liver damage, the VAL9-derived hepatocytes efficiently engrafted and proliferated, repopulating up to 10 % of the liver. In these transplanted livers, we observed a significant decrease of liver transaminases and found no evidence of tumourigenicity. Thus, VAL9-derived hepatocytes were able to rescue hepatic function in acetaminophen-treated animals. CONCLUSIONS: Our study reveals an efficient protocol for differentiating VAL9 hESCs to neonatal hepatocytes which are then able to repopulate livers in vivo without tumour induction. The human hepatocytes are able to rescue liver function in mice with acetaminophen-induced acute toxicity. These results provide proof-of-concept that replacement therapies using hESC-derived hepatocytes are effective for treating liver diseases.


Subject(s)
Chemical and Drug Induced Liver Injury/therapy , Embryonic Stem Cells/cytology , Hepatocytes/cytology , Hepatocytes/transplantation , Acetaminophen/toxicity , Animals , Biliary Tract/cytology , Cell Differentiation , Cell Line , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/pathology , Hepatocytes/metabolism , Heterografts , Humans , Male , Mice , Mice, Inbred NOD , Mice, SCID
12.
Liver Transpl ; 21(6): 801-11, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25821167

ABSTRACT

Crigler-Najjar type 1 disease is a rare inherited metabolic disease characterized by high levels of unconjugated bilirubin due to the complete absence of hepatic uridine diphosphoglucuronate-glucuronosyltransferase activity. Hepatocyte transplantation (HT) has been proposed as an alternative treatment for Crigler-Najjar syndrome, but it is still limited by the quality and the low engraftment and repopulation ability of the cells used. Because of their attachment capability and expression of adhesion molecules as well as the higher proportion of hepatic progenitor cells, neonatal hepatocytes may have an advantage over adult cells. Adult or neonatal hepatocytes were transplanted into Gunn rats, a model for Crigler-Najjar disease. Engraftment and repopulation were studied and compared by immunofluorescence (IF). Additionally, the serum bilirubin levels, the presence of bilirubin conjugates in rat serum, and the expression of uridine diphosphate glucuronosyltransferase 1 family polypeptide A1 (UGT1A1) in rat liver samples were also analyzed. Here we show that neonatal HT results in long-term correction in Gunn rats. In comparison with adult cells, neonatal cells showed better engraftment and repopulation capability 3 days and 6 months after transplantation, respectively. Bilirubinemia decreased in the transplanted animals during the whole experimental follow-up (6 months). Bilirubin conjugates were also present in the serum of the transplanted animals. Western blots and IF confirmed the presence and expression of UGT1A1 in the liver. This work is the first to demonstrate the advantage of using neonatal hepatocytes for the treatment of Crigler-Najjar in vivo.


Subject(s)
Crigler-Najjar Syndrome/therapy , Hepatocytes/transplantation , Liver Regeneration , Aged , Aged, 80 and over , Animals , Bilirubin/blood , Cell Proliferation , Female , Glucuronosyltransferase/metabolism , Humans , Infant, Newborn , Liver/metabolism , Male , Middle Aged , Propranolol , Rats, Gunn
13.
Mol Pharmacol ; 87(4): 582-94, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25576488

ABSTRACT

The small heterodimer partner (SHP) (NR0B2) is an atypical nuclear receptor that lacks a DNA-binding domain. It interacts with and inhibits many transcription factors, affecting key metabolic processes, including bile acid, cholesterol, fatty acid, and drug metabolism. Our aim was to determine the influence of steatotic drugs and nonalcoholic fatty liver disease (NAFLD) on SHP expression and investigate the potential mechanisms. SHP was found to be repressed by steatotic drugs (valproate, doxycycline, tetracycline, and cyclosporin A) in cultured hepatic cells and the livers of different animal models of NAFLD: iatrogenic (tetracycline-treated rats), genetic (glycine N-methyltransferase-deficient mice), and nutritional (mice fed a methionine- and choline-deficient diet). Among the different transcription factors investigated, CCAAT-enhancer-binding protein α (C/EBPα) showed the strongest dominant-repressive effect on SHP expression in HepG2 and human hepatocytes. Reporter assays revealed that the inhibitory effect of C/EBPα and steatotic drugs colocalize between -340 and -509 base pair of the SHP promoter, and mutation of a predicted C/EBPα response element at -473 base pair abolished SHP repression by both C/EBPα and drugs. Moreover, inhibition of major stress signaling pathways demonstrated that the mitogen-activated protein kinase kinase 1/2 pathway activates, while the phosphatidylinositol 3 kinase pathway represses SHP in a C/EBP-dependent manner. We conclude that SHP is downregulated by several steatotic drugs and in advanced NAFLD. These conditions can activate signals that target C/EBPα and consequently repress SHP, thus favoring the progression and severity of NAFLD.


Subject(s)
Fatty Liver/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Animals , CCAAT-Enhancer-Binding Protein-alpha/genetics , CCAAT-Enhancer-Binding Protein-alpha/metabolism , Cells, Cultured , Cyclosporine/toxicity , Doxycycline/toxicity , Fatty Liver/chemically induced , Humans , Male , Mice , Mitogen-Activated Protein Kinase 1/metabolism , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/metabolism , Promoter Regions, Genetic , Receptors, Cytoplasmic and Nuclear/genetics , Signal Transduction , Tetracycline/toxicity , Thiazepines/toxicity , Transcription, Genetic , Valproic Acid/toxicity
14.
Arch Toxicol ; 88(4): 967-82, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24469900

ABSTRACT

It is estimated that only a few marketed drugs are able to directly induce liver steatosis. However, many other drugs may exacerbate or precipitate fatty liver in the presence of other risk factors or in patients prone to non-alcoholic fatty liver disease. On the other hand, current in vitro tests for drug-induced steatosis in preclinical research are scarce and not very sensitive or reproducible. In the present study, we have investigated the effect of well-characterized steatotic drugs on the expression profile of 47 transcription factors (TFs) in human hepatoma HepG2 cells and found that these drugs are able to up- and down-regulate a substantial number of these factors. Multivariate data analysis revealed a common TF signature for steatotic drugs, which consistently and significantly repressed FOXA1, HEX and SREBP1C in cultured cells. This signature was also observed in the livers of rats and in cultured human hepatocytes. Therefore, we selected these three TFs as predictive biomarkers for iatrogenic steatosis. With these biomarkers, a logistic regression analysis yielded a predictive model, which was able to correctly classify 92 % of drugs. The developed algorithm also predicted that ibuprofen, nifedipine and irinotecan are potential steatotic drugs, whereas troglitazone is not. In summary, this is a sensitive, specific and simple RT-PCR test that can be easily implemented in preclinical drug development to predict drug-induced steatosis. Our results also indicate that steatotic drugs affect expression of both common and specific subsets of TF and lipid metabolism genes, thus generating complex transcriptomic responses that cause or contribute to steatosis in hepatocytes.


Subject(s)
Chemical and Drug Induced Liver Injury/etiology , Chemical and Drug Induced Liver Injury/genetics , Gene Expression Profiling , Liver/drug effects , Non-alcoholic Fatty Liver Disease/chemically induced , Non-alcoholic Fatty Liver Disease/genetics , Toxicogenetics/methods , Transcription Factors/genetics , Aged , Algorithms , Animals , Disease Models, Animal , Gene Expression Regulation/drug effects , Genetic Markers , Hep G2 Cells , Humans , Lipid Metabolism/drug effects , Lipid Metabolism/genetics , Liver/metabolism , Logistic Models , Male , Middle Aged , Multivariate Analysis , Rats , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Risk Assessment , Transcription Factors/metabolism
15.
Biochim Biophys Acta ; 1831(4): 803-18, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23318274

ABSTRACT

Liver fatty acid binding protein (FABP1) prevents lipotoxicity of free fatty acids and regulates fatty acid trafficking and partition. Our objective is to investigate the transcription factors controlling the human FABP1 gene and their regulation in nonalcoholic fatty liver disease (NAFLD). Adenovirus-mediated expression of multiple transcription factors in HepG2 cells and cultured human hepatocytes demonstrated that FOXA1 and PPARα are among the most effective activators of human FABP1, whereas C/EBPα is a major dominant repressor. Moreover, FOXA1 and PPARα induced re-distribution of FABP1 protein and increased cytoplasmic expression. Reporter assays demonstrated that the major basal activity of the human FABP1 promoter locates between -96 and -229bp, where C/EBPα binds to a composite DR1-C/EBP element. Mutation of this element at -123bp diminished basal reporter activity, abolished repression by C/EBPα and reduced transactivation by HNF4α. Moreover, HNF4α gene silencing by shRNA in HepG2 cells caused a significant down-regulation of FABP1 mRNA expression. FOXA1 activated the FABP1 promoter through binding to a cluster of elements between -229 and -592bp, whereas PPARα operated through a conserved proximal element at -59bp. Finally, FABP1, FOXA1 and PPARα were concomitantly repressed in animal models of NAFLD and in human nonalcoholic fatty livers, whereas C/EBPα was induced or did not change. We conclude that human FABP1 has a complex mechanism of regulation where C/EBPα displaces HNF4α and hampers activation by FOXA1 and PPARα. Alteration of expression of these transcription factors in NAFLD leads to FABP1 gen repression and could exacerbate lipotoxicity and disease progression.


Subject(s)
CCAAT-Enhancer-Binding Protein-alpha/metabolism , Fatty Acid-Binding Proteins/metabolism , Fatty Liver/metabolism , Fatty Liver/therapy , Hepatocyte Nuclear Factor 3-alpha/metabolism , PPAR alpha/metabolism , Animals , CCAAT-Enhancer-Binding Protein-alpha/genetics , Cells, Cultured , Fatty Acid-Binding Proteins/genetics , Fatty Liver/genetics , Hep G2 Cells , Hepatocyte Nuclear Factor 3-alpha/genetics , Humans , Male , Mice , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease , PPAR alpha/genetics , Protein Binding
16.
PLoS One ; 7(1): e30014, 2012.
Article in English | MEDLINE | ID: mdl-22238690

ABSTRACT

Triglyceride accumulation in nonalcoholic fatty liver (NAFL) results from unbalanced lipid metabolism which, in the liver, is controlled by several transcription factors. The Foxa subfamily of winged helix/forkhead box (Fox) transcription factors comprises three members which play important roles in controlling both metabolism and homeostasis through the regulation of multiple target genes in the liver, pancreas and adipose tissue. In the mouse liver, Foxa2 is repressed by insulin and mediates fasting responses. Unlike Foxa2 however, the role of Foxa1 in the liver has not yet been investigated in detail. In this study, we evaluate the role of Foxa1 in two human liver cell models, primary cultured hepatocytes and HepG2 cells, by adenoviral infection. Moreover, human and rat livers were analyzed to determine Foxa1 regulation in NAFL. Results demonstrate that Foxa1 is a potent inhibitor of hepatic triglyceride synthesis, accumulation and secretion by repressing the expression of multiple target genes of these pathways (e.g., GPAM, DGAT2, MTP, APOB). Moreover, Foxa1 represses the fatty acid transporter protein FATP2 and lowers fatty acid uptake. Foxa1 also increases the breakdown of fatty acids by inducing peroxisomal fatty acid ß-oxidation and ketone body synthesis. Finally, Foxa1 is able to largely up-regulate UCP1, thereby dissipating energy and consistently decreasing the mitochondria membrane potential. We also report that human and rat NAFL have a reduced Foxa1 expression, possibly through a protein kinase C-dependent pathway. We conclude that Foxa1 is an antisteatotic factor that coordinately tunes several lipid metabolic pathways to block triglyceride accumulation in hepatocytes. However, Foxa1 is down-regulated in human and rat NAFL and, therefore, increasing Foxa1 levels could protect from steatosis. Altogether, we suggest that Foxa1 could be a novel therapeutic target for NAFL disease and insulin resistance.


Subject(s)
Fatty Liver/genetics , Fatty Liver/metabolism , Hepatocyte Nuclear Factor 3-alpha/physiology , Hepatocytes/metabolism , Lipid Metabolism/genetics , Adult , Aged , Animals , Cells, Cultured , Down-Regulation/genetics , Fatty Liver/pathology , Female , Hep G2 Cells , Hepatocyte Nuclear Factor 3-alpha/genetics , Hepatocyte Nuclear Factor 3-alpha/metabolism , Hepatocytes/pathology , Humans , Male , Middle Aged , Non-alcoholic Fatty Liver Disease , Primary Cell Culture , Rats , Young Adult
17.
Toxicol In Vitro ; 26(8): 1272-7, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22138474

ABSTRACT

In a number of adverse drug reactions leading to hepatotoxicity, drug metabolism is thought to be involved by the generation of reactive metabolites from non-toxic drugs. The use of hepatoma cell lines, such as HepG2 cell line, for the evaluation of drug-induced hepatotoxicity is hampered by their low cytochrome P450 expression which makes impossible the study of the toxicity produced by bioactivable compounds. Genetically manipulated cells constitute promising tools for hepatotoxicity applications. HepG2 cells were simultaneously transfected with recombinant adenoviruses encoding CYP1A2, CYP2C9 and CYP3A4 to confer them drug-metabolic competence. Upgraded cells (Adv-HepG2) were highly able to metabolize the toxin studied in contrast to the reduced metabolic capacity of HepG2 cells. Aflatoxin B1-induced hepatotoxicity was studied as a proof of concept in metabolically competent and non-competent HepG2 cells by using high content screening technology. Significant differences in mitochondrial membrane potential, intracellular calcium concentration, nuclear morphology and cell viability after treatment with aflatoxin B1 were observed in Adv-HepG2 when compared to HepG2 cells. Rotenone (non bioactivable) and citrate (non hepatotoxic) were analysed as negative controls. This cell model showed to be a suitable hepatic model to test hepatotoxicity of bioactivable drugs and constitutes a valuable alternative for hepatotoxicity testing.


Subject(s)
Adenoviridae/genetics , Aflatoxin B1/toxicity , Chemical and Drug Induced Liver Injury/etiology , Models, Biological , Aflatoxin B1/metabolism , Aryl Hydrocarbon Hydroxylases/genetics , Calcium/metabolism , Cell Survival/drug effects , Chemical and Drug Induced Liver Injury/pathology , Citric Acid/administration & dosage , Citric Acid/metabolism , Cytochrome P-450 CYP1A2/genetics , Cytochrome P-450 CYP2C9 , Cytochrome P-450 CYP3A/genetics , Genetic Vectors , Hep G2 Cells , Humans , Membrane Potential, Mitochondrial/drug effects , Rotenone/administration & dosage , Rotenone/metabolism , Transfection
18.
Gastroenterol Hepatol ; 31(5): 299-309, 2008 May.
Article in Spanish | MEDLINE | ID: mdl-18448061

ABSTRACT

Currently, the only effective treatment for end-stage liver disease is liver transplantation. The number of patients on the waiting list increases considerably each year, giving rise to a wide imbalance between supply and demand for healthy livers. Knowledge of stem cells and their possible use have awakened great interest in the field of hepatology, these cells being one of the most promising short-term alternatives. Hepatic stem cell therapy consists of the implantation of healthy cells capable of performing the functions that damaged cells are unable to carry out. Recent observations indicate that several stem cells can differentiate into distinct cell lineages. Hepatic differentiation of adult stem cells from several origins has yielded highly promising results. Adipose tissue in adults contains a reservoir of stem cells that can be induced and differentiated into different types of cells, showing a high degree of plasticity. Because of its abundance and easy access, adipose tissue is a promising source of adult stem cells for hepatic stem cell therapy. The present article reviews the progress made in the differentiation of adult stem cells from adipose tissue into cells with hepatic phenotype. We also discuss the potential application of this technique as a therapy for temporary metabolic support in patients with end-stage liver failure awaiting whole organ transplantation, as a method to support liver function and facilitate regeneration of the native liver in cases of fulminant hepatic failure, and as a treatment in patients with genetic metabolic defects in vital liver functions.


Subject(s)
Adipose Tissue/cytology , Liver Failure/surgery , Stem Cell Transplantation , Cell Differentiation , Forecasting , Hepatocytes , Humans , Mesenchymal Stem Cells , Phenotype
19.
Gastroenterol. hepatol. (Ed. impr.) ; 31(5): 299-309, mayo 2008. tab, ilus
Article in Spanish | IBECS | ID: ibc-84648

ABSTRACT

Actualmente el único tratamiento efectivo para las enfermedadeshepáticas en estadio terminal es el trasplante de hígado.El número de pacientes en lista de espera aumentaconsiderablemente cada año, dando lugar a una mayor desproporciónentre la oferta y la demanda de un hígado sano.El conocimiento y el posible uso de las células madre ha despertadoun gran interés en el campo de la hepatología, haciendode ellas una de las alternativas más prometedoras acorto plazo. La terapia celular hepática permitiría suplir alhígado de células sanas capaces de llevar a cabo las funcionesque las células dañadas no son capaces de desarrollar.Observaciones recientes han puesto de manifiesto la capacidadde las células madre de diferenciarse hacia diferenteslinajes celulares. La diferenciación hepática de células madreadultas de diversos orígenes ha dado resultados muyprometedores. El tejido adiposo contiene en el individuoadulto un reservorio de células madre capaces de ser inducidasy diferenciadas hacia diferentes estirpes celulares, presentandoun elevado grado de plasticidad celular. La abundanciade este tejido y su fácil accesibilidad hacen de él unaprometedora fuente de células madre adultas para su uso enterapia celular hepática. Se presenta una revisión de losavances obtenidos en la diferenciación de células madre procedentesdel tejido adiposo hacia células de fenotipo hepáticoy sus posible aplicaciones como un método terapéuticocon la finalidad de mantener la función hepática del pacientedurante el período de espera hasta recibir el trasplante, opara facilitar la regeneración hepática en casos de fallo hepáticofulminante, y para el tratamiento de pacientes conmetabolopatías congénitas (AU)


Currently, the only effective treatment for end-stage liverdisease is liver transplantation. The number of patients onthe waiting list increases considerably each year, giving riseto a wide imbalance between supply and demand for healthylivers. Knowledge of stem cells and their possible use haveawakened great interest in the field of hepatology, these cellsbeing one of the most promising short-term alternatives. Hepaticstem cell therapy consists of the implantation of healthycells capable of performing the functions that damagedcells are unable to carry out. Recent observationsindicate that several stem cells can differentiate into distinctcell lineages. Hepatic differentiation of adult stem cells fromseveral origins has yielded highly promising results. Adiposetissue in adults contains a reservoir of stem cells that can beinduced and differentiated into different types of cells, showinga high degree of plasticity. Because of its abundanceand easy access, adipose tissue is a promising source of adultstem cells for hepatic stem cell therapy. The present articlereviews the progress made in the differentiation of adultstem cells from adipose tissue into cells with hepatic phenotype.We also discuss the potential application of thistechnique as a therapy for temporary metabolic support inpatients with end-stage liver failure awaiting whole organtransplantation, as a method to support liver function andfacilitate regeneration of the native liver in cases of fulminanthepatic failure, and as a treatment in patients with geneticmetabolic defects in vital liver functions (AU)


Subject(s)
Humans , Stem Cell Transplantation , Cell- and Tissue-Based Therapy/methods , Liver Failure, Acute/surgery , Adipose Tissue , Liver Transplantation
20.
Toxicol Lett ; 168(3): 286-91, 2007 Feb 05.
Article in English | MEDLINE | ID: mdl-17184936

ABSTRACT

Chromaffin cells in culture show high neuropathy target esterase (NTE) activity. It is well known that inhibition and specific modification of NTE by some organophosphorus (OPs) compounds induces a neurodegenerative neuropathy. It has been suggested that NTE is responsible for phosphatidylcholine homeostasis, although its role in neuropathy induction remains unclear. The cDNA of human NTE (4.4kbp) was inserted into an adenoviral vector. Bovine chromaffin cells cultured at 50,000 cells/well were incubated with the vector for 2h and after removing the volume of infection, cells were maintained in the incubator. After 24h, NTE activity was 6.8+/-0.5mU/10(6) cells in untreated cells and 14.8+/-1.5mU/10(6) cells, 19.3+/-2.9mU/10(6) cells, 24.8+/-0.9mU/10(6) cells and 30.9+/-1.0mU/10(6) cells in cells incubated with 2, 4, 8 and 16microl of vector, respectively. After 60min of inhibition with mipafox increased concentrations, the calculated I(50) (60min) values were 5.5, 6.2 and 6.6microM for cells infected with 0, 2 and 10microl of vector preparation. We confirm that the adenoviral vector containing the human NTE gene is active in bovine chromaffin cells in culture and that the NTE activity expressed by the vector shows the same inhibition pattern by the neuropathic OP mipafox as the NTE activity of bovine chromaffin cells and cells remained viable after the high NTE activity expression.


Subject(s)
Adenoviridae/genetics , Carboxylic Ester Hydrolases/metabolism , Chromaffin Cells/enzymology , Animals , Carboxylic Ester Hydrolases/antagonists & inhibitors , Cattle , Cells, Cultured , Genetic Vectors , Humans , Isoflurophate/analogs & derivatives , Isoflurophate/toxicity , Paraoxon/toxicity , Transfection , Valerates/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL