Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
Add more filters

Country/Region as subject
Publication year range
1.
J Toxicol Environ Health B Crit Rev ; 26(8): 442-467, 2023 11 17.
Article in English | MEDLINE | ID: mdl-37786264

ABSTRACT

Drug-Induced Liver Injury (DILI) constitutes hepatic damage attributed to drug exposure. DILI may be categorized as hepatocellular, cholestatic or mixed and might also involve immune responses. When DILI occurs in dose-dependent manner, it is referred to as intrinsic, while if the injury occurs spontaneously, it is termed as idiosyncratic. This review predominately focused on idiosyncratic liver injury. The established molecular mechanisms for DILI include (1) mitochondria dysfunction, (2) increased reactive oxygen species levels, (3) presence of elevated apoptosis and necrosis, (4) and bile duct injuries associated with immune mediated pathways. However, it should be emphasized that the underlying mechanisms responsible for DILI are still unknown. Prevention strategies are critical as incidences occur frequently, and treatment options are limited once the injury has developed. The aim of this review was to utilize retrospective cohort studies from across the globe to gain insight into epidemiological patterns. This review considers (1) what is currently known regarding the mechanisms underlying DILI, (2) discusses potential risk factors and (3) implications of the coronavirus pandemic on DILI presentation and research. Future perspectives are also considered and discussed and include potential new biomarkers, causality assessment and reporting methods.


Subject(s)
Chemical and Drug Induced Liver Injury , Liver Diseases , Humans , Retrospective Studies , Chemical and Drug Induced Liver Injury/epidemiology , Chemical and Drug Induced Liver Injury/etiology , Liver Diseases/epidemiology , Risk Factors
2.
Arch Toxicol ; 96(1): 287-303, 2022 01.
Article in English | MEDLINE | ID: mdl-34668024

ABSTRACT

In an effort to replace, reduce and refine animal experimentation, there is an unmet need to advance current in vitro models that offer features with physiological relevance and enhanced predictivity of in vivo toxicological output. Hepatic toxicology is key following chemical, drug and nanomaterials (NMs) exposure, as the liver is vital in metabolic detoxification of chemicals as well as being a major site of xenobiotic accumulation (i.e., low solubility particulates). With the ever-increasing production of NMs, there is a necessity to evaluate the probability of consequential adverse effects, not only in health but also in clinically asymptomatic liver, as part of risk stratification strategies. In this study, two unique disease initiation and maintenance protocols were developed and utilised to mimic steatosis and pre-fibrotic NASH in scaffold-free 3D liver microtissues (MT) composed of primary human hepatocytes, hepatic stellate cells, Kupffer cells and sinusoidal endothelial cells. The characterized diseased MT were utilized for the toxicological assessment of a panel of xenobiotics. Highlights from the study included: 1. Clear experimental evidence for the pre-existing liver disease is important in the augmentation of xenobiotic-induced hepatotoxicity and 2. NMs are able to activate stellate cells. The data demonstrated that pre-existing disease is vital in the intensification of xenobiotic-induced liver damage. Therefore, it is imperative that all stages of the wide spectrum of liver disease are incorporated in risk assessment strategies. This is of significant consequence, as a substantial number of the general population suffer from sub-clinical liver injury without any apparent or diagnosed manifestations.


Subject(s)
Non-alcoholic Fatty Liver Disease , Animals , Endothelial Cells/metabolism , Hepatocytes , Humans , Kupffer Cells , Liver , Non-alcoholic Fatty Liver Disease/chemically induced , Non-alcoholic Fatty Liver Disease/metabolism
3.
Arch Toxicol ; 95(2): 573-589, 2021 02.
Article in English | MEDLINE | ID: mdl-33106934

ABSTRACT

The liver plays an important role in xenobiotic metabolism and represents a primary target for toxic substances. Many different in vitro cell models have been developed in the past decades. In this study, we used RNA-sequencing (RNA-Seq) to analyze the following human in vitro liver cell models in comparison to human liver tissue: cancer-derived cell lines (HepG2, HepaRG 3D), induced pluripotent stem cell-derived hepatocyte-like cells (iPSC-HLCs), cancerous human liver-derived assays (hPCLiS, human precision cut liver slices), non-cancerous human liver-derived assays (PHH, primary human hepatocytes) and 3D liver microtissues. First, using CellNet, we analyzed whether these liver in vitro cell models were indeed classified as liver, based on their baseline expression profile and gene regulatory networks (GRN). More comprehensive analyses using non-differentially expressed genes (non-DEGs) and differential transcript usage (DTU) were applied to assess the coverage for important liver pathways. Through different analyses, we noticed that 3D liver microtissues exhibited a high similarity with in vivo liver, in terms of CellNet (C/T score: 0.98), non-DEGs (10,363) and pathway coverage (highest for 19 out of 20 liver specific pathways shown) at the beginning of the incubation period (0 h) followed by a decrease during long-term incubation for 168 and 336 h. PHH also showed a high degree of similarity with human liver tissue and allowed stable conditions for a short-term cultivation period of 24 h. Using the same metrics, HepG2 cells illustrated the lowest similarity (C/T: 0.51, non-DEGs: 5623, and pathways coverage: least for 7 out of 20) with human liver tissue. The HepG2 are widely used in hepatotoxicity studies, however, due to their lower similarity, they should be used with caution. HepaRG models, iPSC-HLCs, and hPCLiS ranged clearly behind microtissues and PHH but showed higher similarity to human liver tissue than HepG2 cells. In conclusion, this study offers a resource of RNA-Seq data of several biological replicates of human liver cell models in vitro compared to human liver tissue.


Subject(s)
Computational Biology/methods , Hepatocytes/metabolism , Liver Neoplasms/metabolism , Liver/metabolism , Transcriptome , Cell Differentiation , Cell Line, Tumor , Cells, Cultured , Gene Expression Profiling , Gene Expression Regulation , Gene Regulatory Networks , Hep G2 Cells , High-Throughput Nucleotide Sequencing , Humans , In Vitro Techniques , Induced Pluripotent Stem Cells/metabolism , Models, Biological , RNA-Seq
4.
Chem Res Toxicol ; 33(1): 258-270, 2020 01 21.
Article in English | MEDLINE | ID: mdl-31820940

ABSTRACT

The importance of adsorption, distribution, metabolism, excretion, and toxicity (ADMET) analysis is expected to grow substantially due to recent failures in detecting severe toxicity issues of new chemical entities during preclinical/clinical development. Traditionally, safety risk assessment studies for humans have been conducted in animals during advanced preclinical or clinical phase of drug development. However, potential drug toxicity in humans now needs to be detected in the drug discovery process as soon as possible without reliance on animal studies. The "omics", such as genomics, proteomics, and metabolomics, have recently entered pharmaceutical research in both drug discovery and drug development, but to the best of our knowledge, no applications in high-throughput safety risk assessment have been attempted so far. This paper reports an innovative method to anticipate adverse drug effects in an early discovery phase based on lipid fingerprints using human three-dimensional microtissues. The risk of clinical hepatotoxicity potential was evaluated for a data set of 22 drugs belonging to five different therapeutic chemical classes and with various drug-induced liver injury effect. The treatment of microtissues with repeated doses of each drug allowed collecting lipid fingerprints for five time points (2, 4, 7, 9, and 11 days), and multivariate statistical analysis was applied to search for correlations with the hepatotoxic effect. The method allowed clustering of the drugs based on their hepatotoxic effect, and the observed lipid impairments for a number of drugs was confirmed by literature sources. Compared to traditional screening methods, here multiple interconnected variables (lipids) are measured simultaneously, providing a snapshot of the cellular status from the lipid perspective at a molecular level. Applied here to hepatotoxicity, the proposed workflow can be applied to several tissues, being tridimensional microtissues from various origins.


Subject(s)
Chemical and Drug Induced Liver Injury , Lipidomics , Humans , Liver , Models, Statistical , Risk Assessment/methods , Spheroids, Cellular , Workflow
5.
Arch Toxicol ; 94(7): 2435-2461, 2020 07.
Article in English | MEDLINE | ID: mdl-32632539

ABSTRACT

Hazard assessment, based on new approach methods (NAM), requires the use of batteries of assays, where individual tests may be contributed by different laboratories. A unified strategy for such collaborative testing is presented. It details all procedures required to allow test information to be usable for integrated hazard assessment, strategic project decisions and/or for regulatory purposes. The EU-ToxRisk project developed a strategy to provide regulatorily valid data, and exemplified this using a panel of > 20 assays (with > 50 individual endpoints), each exposed to 19 well-known test compounds (e.g. rotenone, colchicine, mercury, paracetamol, rifampicine, paraquat, taxol). Examples of strategy implementation are provided for all aspects required to ensure data validity: (i) documentation of test methods in a publicly accessible database; (ii) deposition of standard operating procedures (SOP) at the European Union DB-ALM repository; (iii) test readiness scoring accoding to defined criteria; (iv) disclosure of the pipeline for data processing; (v) link of uncertainty measures and metadata to the data; (vi) definition of test chemicals, their handling and their behavior in test media; (vii) specification of the test purpose and overall evaluation plans. Moreover, data generation was exemplified by providing results from 25 reporter assays. A complete evaluation of the entire test battery will be described elsewhere. A major learning from the retrospective analysis of this large testing project was the need for thorough definitions of the above strategy aspects, ideally in form of a study pre-registration, to allow adequate interpretation of the data and to ensure overall scientific/toxicological validity.


Subject(s)
Documentation , Electronic Data Processing/legislation & jurisprudence , Government Regulation , Toxicity Tests , Toxicology/legislation & jurisprudence , Animals , Cells, Cultured , Europe , Humans , Policy Making , Reproducibility of Results , Retrospective Studies , Risk Assessment , Terminology as Topic , Zebrafish/embryology
6.
Part Fibre Toxicol ; 16(1): 42, 2019 11 19.
Article in English | MEDLINE | ID: mdl-31739797

ABSTRACT

BACKGROUND: With ever-increasing exposure to engineered nanomaterials (NMs), there is an urgent need to evaluate the probability of consequential adverse effects. The potential for NM translocation to distal organs is a realistic prospect, with the liver being one of the most important target organs. Traditional in vitro or ex vivo hepatic toxicology models are often limiting (i.e. short life-span, reduced metabolic activity, lacking important cell populations, etc.). In this study, we scrutinize a 3D human liver microtissue (MT) model (composed of primary hepatocytes and non-parenchymal cells). This unique experiment benefits from long-term (3 weeks) repeated very low exposure concentrations, as well as incorporation of recovery periods (up to 2 weeks), in an attempt to account for the liver's recovery capacity in vivo. As a means of assessing the toxicological potential of NMs, cell cytotoxicity (cell membrane integrity and aspartate aminotransferase (AST) activity), pro/anti-inflammatory response and hepatic function were investigated. RESULTS: The data showed that 2 weeks of cell culture might be close to limits before subtle ageing effects start to overshadow low sub-lethal NM-induced cellular responses in this test system (adenylate kinase (AK) cytotoxicity assay). We showed that in vitro AST measurement are not suitable in a nanotoxicological context. Moreover, the cytokine analysis (IL6, IL8, IL10 and TNF-α) proved useful in highlighting recovery periods as being sufficient for allowing a reduction in the pro-inflammatory response. Next, low soluble NM-treated MT showed a concentration-dependent penetration of materials deep into the tissue. CONCLUSION: In this study the advantages and pitfalls of the multi-cellular primary liver MT are discussed. Furthermore, we explore a number of important considerations for allowing more meaningful in vitro vs. in vivo comparisons in the field of hepatic nanotoxicology.


Subject(s)
Chemical and Drug Induced Liver Injury/etiology , Hepatocytes/drug effects , Kupffer Cells/drug effects , Liver/drug effects , Nanostructures/toxicity , Tissue Culture Techniques/methods , Albumins/metabolism , Cell Survival/drug effects , Coculture Techniques , Cytokines/metabolism , Hepatocytes/metabolism , Hepatocytes/pathology , Humans , Kupffer Cells/metabolism , Kupffer Cells/pathology , Liver/metabolism , Liver/pathology , Liver Function Tests
7.
Arch Toxicol ; 93(6): 1609-1637, 2019 06.
Article in English | MEDLINE | ID: mdl-31250071

ABSTRACT

Drug-induced liver injury (DILI) cannot be accurately predicted by animal models. In addition, currently available in vitro methods do not allow for the estimation of hepatotoxic doses or the determination of an acceptable daily intake (ADI). To overcome this limitation, an in vitro/in silico method was established that predicts the risk of human DILI in relation to oral doses and blood concentrations. This method can be used to estimate DILI risk if the maximal blood concentration (Cmax) of the test compound is known. Moreover, an ADI can be estimated even for compounds without information on blood concentrations. To systematically optimize the in vitro system, two novel test performance metrics were introduced, the toxicity separation index (TSI) which quantifies how well a test differentiates between hepatotoxic and non-hepatotoxic compounds, and the toxicity estimation index (TEI) which measures how well hepatotoxic blood concentrations in vivo can be estimated. In vitro test performance was optimized for a training set of 28 compounds, based on TSI and TEI, demonstrating that (1) concentrations where cytotoxicity first becomes evident in vitro (EC10) yielded better metrics than higher toxicity thresholds (EC50); (2) compound incubation for 48 h was better than 24 h, with no further improvement of TSI after 7 days incubation; (3) metrics were moderately improved by adding gene expression to the test battery; (4) evaluation of pharmacokinetic parameters demonstrated that total blood compound concentrations and the 95%-population-based percentile of Cmax were best suited to estimate human toxicity. With a support vector machine-based classifier, using EC10 and Cmax as variables, the cross-validated sensitivity, specificity and accuracy for hepatotoxicity prediction were 100, 88 and 93%, respectively. Concentrations in the culture medium allowed extrapolation to blood concentrations in vivo that are associated with a specific probability of hepatotoxicity and the corresponding oral doses were obtained by reverse modeling. Application of this in vitro/in silico method to the rat hepatotoxicant pulegone resulted in an ADI that was similar to values previously established based on animal experiments. In conclusion, the proposed method links oral doses and blood concentrations of test compounds to the probability of hepatotoxicity.


Subject(s)
Chemical and Drug Induced Liver Injury/diagnosis , Drug-Related Side Effects and Adverse Reactions/diagnosis , Administration, Oral , Algorithms , Animals , Cell Line , Cell Survival/drug effects , Computer Simulation , Gene Expression/drug effects , Hepatocytes/drug effects , Humans , In Vitro Techniques , Maximum Tolerated Dose , Pharmaceutical Preparations/administration & dosage , Pharmaceutical Preparations/blood , Pharmacokinetics , Reproducibility of Results , Sensitivity and Specificity , Support Vector Machine
8.
Arch Toxicol ; 92(12): 3505-3515, 2018 12.
Article in English | MEDLINE | ID: mdl-30317417

ABSTRACT

Primary human hepatocytes (PHHs) remain the gold standard for in vitro testing in the field of pharmacology and toxicology. One crucial parameter influencing the results of in vitro tests is the incubation period with test compounds. It has been suggested that longer incubation periods may be critical for the prediction of repeated dose toxicity. However, a study that systematically analyzes the relationship between incubation period and cytotoxicity in PHHs is not available. To close this gap, 30 compounds were tested in a concentration-dependent manner for cytotoxicity in cultivated cryopreserved PHHs (three donors per compound) for 1, 2 and 7 days. The median of the EC50 values of all compounds decreased 1.78-fold on day 2 compared to day 1, and 1.89-fold on day 7 compared to day 1. Median values of EC50 ratios of all compounds at day 2 and day 7 were close to one but for individual compounds the ratio increased up to almost six. Strong correlations were obtained for EC50 on day 1 and day 7 (R = 0.985; 95% CI 0.960-0.994), day 1 and day 2 (R = 0.964; 95% CI 0.910-0.986), as well as day 2 and day 7 (R = 0.981; 95% CI 0.955-0.992). However, compound specific differences also occurred. Whereas, for example, busulfan showed a relatively strong increase on day 7 compared to day 1, cytotoxicity of acetaminophen did not increase during longer incubation periods. To validate the observed correlations, a publicly available data set, containing data on the cytotoxicity of human hepatocytes cultivated as spheroids for incubation periods of 5 and 14 days, was analyzed. A high correlation coefficient of EC50 values at day 5 and day 14 was obtained (R = 0.894; 95% CI 0.798-0.945). In conclusion, the median cytotoxicity of the test compounds increased between 1 and 2 days of incubation, with no or only a minimal further increase until day 7. It remains to be studied whether the different results obtained for some individual compounds after longer exposure periods would correspond better to human-repeated dose toxicity.


Subject(s)
Chemical and Drug Induced Liver Injury/etiology , Drug-Related Side Effects and Adverse Reactions/diagnosis , Hepatocytes/drug effects , Toxicity Tests/methods , Acetaminophen/toxicity , Cell Culture Techniques/methods , Cells, Cultured , Cryopreservation , Dose-Response Relationship, Drug , Humans , Time Factors
9.
Pflugers Arch ; 468(4): 739-50, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26676951

ABSTRACT

The epithelial cells lining the proximal tubules of the kidney mediate complex transport processes and are particularly vulnerable to drug toxicity. Drug toxicity studies are classically based on two-dimensional cultures of immortalized proximal tubular cells. Such immortalized cells are dedifferentiated, and lose transport properties (including saturable endocytic uptake) encountered in vivo. Generating differentiated, organotypic human microtissues would potentially alleviate these limitations and facilitate drug toxicity studies. Here, we describe the generation and characterization of kidney microtissues from immortalized (HK-2) and primary (HRPTEpiC) human renal proximal tubular epithelial cells under well-defined conditions. Microtissue cultures were done in hanging drop GravityPLUS™ culture plates and were characterized for morphology, proliferation and differentiation markers, and by monitoring the endocytic uptake of albumin. Kidney microtissues were successfully obtained by co-culturing HK-2 or HRPTEpiC cells with fibroblasts. The HK-2 microtissues formed highly proliferative, but dedifferentiated microtissues within 10 days of culture, while co-culture with fibroblasts yielded spherical structures already after 2 days. Low passage HRPTEpiC microtissues (mono- and co-culture) were less proliferative and expressed tissue-specific differentiation markers. Electron microscopy evidenced epithelial differentiation markers including microvilli, tight junctions, endosomes, and lysosomes in the co-cultured HRPTEpiC microtissues. The co-cultured HRPTEpiC microtissues showed specific uptake of albumin that could be inhibited by cadmium and gentamycin. In conclusion, we established a reliable hanging drop protocol to obtain functional kidney microtissues with proximal tubular epithelial cell lines. These microtissues could be used for high-throughput drug and toxicology screenings, with endocytosis as a functional readout.


Subject(s)
Epithelial Cells/cytology , Kidney Tubules, Proximal/cytology , Primary Cell Culture/methods , Cell Differentiation , Cell Line , Cell Proliferation , Cells, Cultured , Coculture Techniques/methods , Culture Media, Conditioned/pharmacology , Epithelial Cells/drug effects , Epithelial Cells/physiology , Fibroblasts/metabolism , Humans , Tissue Culture Techniques/methods
10.
Proc Natl Acad Sci U S A ; 108(7): 2945-50, 2011 Feb 15.
Article in English | MEDLINE | ID: mdl-21282654

ABSTRACT

The function of the liver is well-preserved during the aging process, although some evidence suggests that liver regeneration might be impaired with advanced age. We observed a decreased ability of the liver to restore normal volume after partial hepatectomy in elderly mice, and we identified a pathway that rescued regeneration and was triggered by serotonin. 2,5-dimethoxy-4-iodoamphetamine (DOI), a serotonin receptor agonist, reversed the age-related pseudocapillarization of old liver and improved hepatosinusoidal blood flow. After hepatectomy, the open fenestrae were associated with a restored attachment of platelets to endothelium and the initiation of a normal regenerative response, including the up-regulation of essential growth mediators and serotonin receptors. In turn, hepatocyte proliferation recovered along with regain of liver volume and animal survival. DOI operates through the release of VEGF, and its effects could be blocked with anti-VEGF antibodies both in vitro and in vivo. These results suggest that pseudocapillarization in the aged acts as a barrier to liver regeneration. DOI breaks this restraint through an endothelium-dependent mechanism driven by VEGF. This pathway highlights a target for reversing the age-associated decline in the capacity of the liver to regenerate.


Subject(s)
Amphetamines/pharmacology , Liver Regeneration/physiology , Liver/blood supply , Liver/physiology , Serotonin Receptor Agonists/pharmacology , Age Factors , Animals , Cell Proliferation/drug effects , Hepatectomy , Hepatocytes/drug effects , Hepatocytes/physiology , Hepatocytes/ultrastructure , Immunohistochemistry , Liver/surgery , Liver Regeneration/drug effects , Male , Mice , Mice, Inbred C57BL , Microcirculation/physiology , Microscopy, Electron, Scanning , Microscopy, Electron, Transmission , Polymerase Chain Reaction , Regional Blood Flow/drug effects , Statistics, Nonparametric , Vascular Endothelial Growth Factor A/metabolism
11.
Chemosphere ; 364: 143032, 2024 Aug 05.
Article in English | MEDLINE | ID: mdl-39111678

ABSTRACT

Nano and microplastics are defined as particles smaller than 100 nm and 5 mm respectively. The widespread production and use of plastics in everyday life has resulted in significant accumulation of plastic debris in the environment. Over the last two decades there are increased concerns regarding the potential entry and accumulation of plastics in the human body with ingestion being one of the most important routes of exposure. However, the magnitude and nature of potential toxic effects of plastic exposure to human health is not yet fully understood. The liver is the body's principal detoxification organ and critically to this study recognized as the main accumulation site for particulates. In this study as the first of its kind the health impacts of long term low repeated polystyrene microplastics (1 and 5 µm) exposure was investigated in a functionally active 3D liver microtissue model, composed of primary human hepatocytes, Kupffer cells, sinusoidal endothelial cells and hepatic stellate cells. The highlight from the data includes microplastic-induced dose (3.125-25 µg/ml) and time dependent (up to 504 h) increase in cell death and inflammation manifested by enhanced release of IL6, IL8 and TNF-α. The exposure to repeated dosing of the plastics also resulted in notable pathology manifested as aberrant tissue architecture, such as dilated bile canaliculi and large lipid droplets inside the hepatic cells. This toxicity matched extremely well to the accumulation of the materials with the cells of microtissue predominately in the organ macrophages. This study highlights the real issue and danger of microplastic exposure with potential for long-term accumulation and adverse effects of non-biodegradable plastics within the liver.

12.
Hepatology ; 56(1): 209-18, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22290718

ABSTRACT

UNLABELLED: Obstructive cholestasis induces liver injury, postoperative complications, and mortality after surgery. Adaptive control of cholestasis, including bile salt homeostasis, is necessary for recovery and survival. Peripheral serotonin is a cytoprotective neurotransmitter also associated with liver regeneration. The effect of serotonin on cholestatic liver injury is not known. Therefore, we tested whether serotonin affects the severity of cholestatic liver injury. We induced cholestasis by ligation of the bile duct (BDL) in either wild-type (WT) mice or mice lacking peripheral serotonin (Tph1(-/-) and immune thrombocytopenic [ITP] mice). Liver injury was assessed by the levels of plasma aspartate aminotransferase (AST), alanine aminotransferase (ALT) and tissue necrosis. Bile salt-regulating genes were measured by quantitative polymerase chain reaction and confirmed by western blotting and immunohistochemistry. Tph1(-/-) mice displayed higher levels of plasma AST, ALT, bile salts, and hepatic necrosis after 3 days of BDL than WT mice. Likewise, liver injury was disproportional in ITP mice. Moreover, severe cholestatic complications and mortality after prolonged BDL were increased in Tph1(-/-) mice. Despite the elevation in toxic bile salts, expression of genes involved in bile salt homeostasis and detoxification were not affected in Tph1(-/-) livers. In contrast, the bile salt reabsorption transporters Ostα and Ostß were up-regulated in the kidneys of Tph1(-/-) mice, along with a decrease in urinary bile salt excretion. Serotonin reloading of Tph1(-/-) mice reversed this phenotype, resulting in a reduction of circulating bile salts and liver injury. CONCLUSION: We propose a physiological function of serotonin is to ameliorate liver injury and stabilize the bile salt pool through adaptation of renal transporters in cholestasis.


Subject(s)
Bile Acids and Salts/metabolism , Liver Diseases/prevention & control , Liver/immunology , Serotonin/metabolism , Animals , Bile Ducts/surgery , Cells, Cultured , Cholestasis/metabolism , Cholestasis/pathology , Disease Models, Animal , Hepatocytes/cytology , Hepatocytes/metabolism , Ligation/methods , Liver/pathology , Liver Diseases/blood , Liver Function Tests , Male , Mass Spectrometry , Mice , Mice, Inbred C57BL , Mice, Knockout , Random Allocation , Sensitivity and Specificity , Serotonin/pharmacology
13.
Phys Chem Chem Phys ; 15(26): 11054-60, 2013 Jul 14.
Article in English | MEDLINE | ID: mdl-23714784

ABSTRACT

The adsorption geometry of 1,3,5-tris(4-mercaptophenyl)benzene (TMB) on Cu(111) is determined with high precision using two independent methods, experimentally by quantitative low energy electron diffraction (LEED-I(V)) and theoretically by dispersion corrected density functional theory (DFT-vdW). Structural refinement using both methods consistently results in similar adsorption sites and geometries. Thereby a level of confidence is reached that allows deduction of subtle structural details such as molecular deformations or relaxations of copper substrate atoms.

14.
Gut ; 61(1): 117-27, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21997548

ABSTRACT

OBJECTIVE: The purpose of this study was to assess non-invasive imaging modalities including MRI and CT and compare the quantitative amount of fat with data provided by the pathologist and a chemical lipid assay in leptin-deficient mouse livers. METHODS: A liver/fat phantom was first used to assess the accuracy of small-animal MRI and human MRI and CT, followed by correlation analysis with ob/ob mouse liver fat quantified by an accurate chemical lipid assay. Similarly, the authors compared the pathologist's quantification and the automated software quantification of fat with the lipid assay. The authors then investigated whether hepatic steatosis assessed by MRI correlates with the degree of liver injury in a model of ischaemia/reperfusion in leptin-deficient mice as well as with serious postoperative complications in patients undergoing major liver resection (NCT01234714). RESULTS: The authors designed lipid/liver mixtures at various ratios to mimic a wide range of fat liver contents. Small-animal and human MRI detected this fat with a high correlation to the actual fat contents. Mouse livers assessed by human MRI correlated best with total intrahepatic fat by chemical lipid analysis (r=0.975). Human CT, the pathologist's assessment and the automated software were less reliable (r=-0.873, 0.512 and 0.873, respectively). There was a significant correlation of the MRI fat quantification with several parameters of liver injury, and MRI data could predict mouse survival after ischaemia/reperfusion injury. In patients undergoing major liver resection, higher liver fat content was associated with more serious postoperative complications, such as liver or multiorgan failure and sepsis, necessitating admission to the intensive care unit. CONCLUSIONS: With the use of a well-defined set of biological standards, MRI can predict intrahepatic fat with high accuracy. In contrast to biopsies, this method is non-invasive, giving a representative assessment of the whole liver.


Subject(s)
Fatty Liver/diagnosis , Lipids/analysis , Magnetic Resonance Imaging , Adult , Aged , Animals , Cattle , Fatty Liver/diagnostic imaging , Fatty Liver/surgery , Female , Hepatectomy , Humans , Liver/chemistry , Liver/pathology , Magnetic Resonance Imaging/standards , Male , Mice , Mice, Inbred C57BL , Middle Aged , Postoperative Complications , Reference Standards , Reperfusion Injury/etiology , Reperfusion Injury/mortality , Retrospective Studies , Tomography, X-Ray Computed
15.
Hepatology ; 53(1): 253-62, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21254175

ABSTRACT

UNLABELLED: The implantation of grafts below 30% of the normal liver volume is associated with a high risk of failure known as small-for-size (SFS) syndrome. Strategies to rescue small grafts may have a dramatic impact on organ shortage. Serotonin is a potent growth factor for the liver. The goal of this study was to determine whether enhanced serotonin signaling could prevent the deleterious effects of SFS syndrome. We performed 30% normal liver volume transplantations in wild-type C57/BL6 and interleukin-6 (IL-6)(-/-) mice. Some animals received α-methyl-5-HT (DOI), an agonist of serotonin receptor-2 (5-HT2B). Endpoints included long-term survival, serum and hepatic markers of liver injury and regeneration, assessment of hepatic microcirculation by intravital fluorescence microscopy and scanning electron microscopy, and transcript levels of a variety of serotonin receptors, tumor necrosis factor α, and IL-6. All recipients of small grafts (controls) died within 2-4 days of transplantation, whereas half of those receiving DOI survived permanently. Control animals disclosed major liver injury, including diffuse microvesicular steatosis in hepatocytes, impairment of microcirculation, and a failure of regeneration, whereas these parameters were dramatically improved in animals subjected to DOI. Blockage of 5-HT2B blunted the protective effects of DOI. Whereas IL-6 levels were higher in DOI-treated animals, IL-6(-/-) mice were still protected by DOI, suggesting a protective pathway independent of IL-6. CONCLUSION: Serotonin through its action on receptor-2B protects SFS liver grafts from injury and prevents microcirculation and regeneration. The mechanism of hepato-protection is independent of IL-6.


Subject(s)
Graft Rejection/drug therapy , Liver Regeneration/drug effects , Liver Transplantation/methods , Receptor, Serotonin, 5-HT2B/metabolism , Serotonin 5-HT2 Receptor Agonists/pharmacology , Animals , Interleukin-6/metabolism , Liver/blood supply , Male , Mice , Mice, Inbred C57BL , Microcirculation/drug effects , Serotonin/analogs & derivatives , Serotonin/pharmacology , Tumor Necrosis Factor-alpha/metabolism
16.
SLAS Technol ; 27(4): 267-275, 2022 08.
Article in English | MEDLINE | ID: mdl-35584761

ABSTRACT

Cell dissemination during tumor development is a characteristic of cancer metastasis. Dissemination from three-dimensional spheroid models on extracellular matrices designed to mimic tissue-specific physiological microenvironments may allow us to better elucidate the mechanism behind cancer metastasis and the response to therapeutic agents. The orientation of fibrillar collagen plays a key role in cellular processes and mediates metastasis through contact-guidance. Understanding how cells migrate on aligned collagen fibrils requires in vitro assays with reproducible and standardized orientation of collagen fibrils on the macro-to-nanoscale. Herein, we implement a spheroid-based migration assay, integrated with a fibrillar type I collagen matrix, in a manner compatible with high throughput image acquisition and quantitative analysis. The migration of highly proliferating U2OS osteosarcoma cell spheroids onto an aligned fibrillar type I collagen matrix was quantified. Cell dissemination from the spheroid was polarized with increased invasion in the direction of fibril alignment. The resulting area of cell dissemination had an aspect ratio of 1.2 ± 0.1 and an angle of maximum invasion distance of 5° ± 44° relative to the direction of collagen fibril alignment. The assay described here can be applied to a fully automated imaging and analysis pipeline for the assessment of tumor cell migration with high throughput screening.


Subject(s)
Collagen Type I , Neoplasms , Biomimetics , Cell Line, Tumor , Collagen Type I/metabolism , Extracellular Matrix , Fibrillar Collagens/metabolism
17.
Toxicol In Vitro ; 85: 105473, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36108805

ABSTRACT

To reduce, replace, and refine in vivo testing, there is increasing emphasis on the development of more physiologically relevant in vitro test systems to improve the reliability of non-animal-based methods for hazard assessment. When developing new approach methodologies, it is important to standardize the protocols and demonstrate the methods can be reproduced by multiple laboratories. The aim of this study was to assess the transferability and reproducibility of two advanced in vitro liver models, the Primary Human multicellular microtissue liver model (PHH) and the 3D HepG2 Spheroid Model, for nanomaterial (NM) and chemical hazard assessment purposes. The PHH model inter-laboratory trial showed strong consistency across the testing sites. All laboratories evaluated cytokine release and cytotoxicity following exposure to titanium dioxide (TiO2) and zinc oxide (ZnO) nanoparticles. No significant difference was observed in cytotoxicity or IL-8 release for the test materials. The data were reproducible with all three laboratories with control readouts within a similar range. The PHH model ZnO induced the greatest cytotoxicity response at 50.0 µg/mL and a dose-dependent increase in IL-8 release. For the 3D HepG2 spheroid model, all test sites were able to construct the model and demonstrated good concordance in IL-8 cytokine release and genotoxicity data. This trial demonstrates the successful transfer of new approach methodologies across multiple laboratories, with good reproducibility for several hazard endpoints.


Subject(s)
Zinc Oxide , Humans , Zinc Oxide/toxicity , Reproducibility of Results , Interleukin-8 , Liver , Cell Line , Spheroids, Cellular
18.
J Hepatol ; 55(6): 1291-9, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21703192

ABSTRACT

BACKGROUND & AIMS: Chemical composition of hepatic lipids is an evolving player in steatotic liver ischemia/reperfusion (I/R) injury. Thromboxane A(2) (TXA(2)) is a vasoactive pro-inflammatory lipid mediator derived from arachidonic acid (AA), an omega-6 fatty acid (Ω-6 FA). Reduced tolerance of the macrosteatotic liver to I/R may be related to increased TXA(2) synthesis due to the predominance of Ω-6 FAs. METHODS: TXA(2) levels elicited by I/R in ob/ob and wild type mice were assessed by ELISA. Ob/ob mice were fed Ω-3 FAs enriched diet to reduce hepatic synthesis of AA and TXA(2) or treated with selective TXA(2) receptor blocker before I/R. RESULTS: I/R triggered significantly higher hepatic TXA(2) production in ob/ob than wild type animals. Compared with ob/ob mice on regular diet, Ω-3 FAs supplementation markedly reduced hepatic AA levels before ischemia and consistently blunted hepatic TXA(2) synthesis after reperfusion. Sinusoidal perfusion and hepatocellular damage were significantly ameliorated despite downregulation of heme oxygenase-1. Hepatic transcript and protein levels of IL-1ß and neutrophil recruitment were significantly diminished after reperfusion. Moreover, TXA(2) receptor blockage conferred similar protection without modification of the histological pattern of steatosis. A stronger protection was achieved in the steatotic compared with lean animals. CONCLUSIONS: Enhanced I/R injury in the macrosteatotic liver is explained, at least partially, by TXA(2) mediated microcirculatory failure rather than size-related mechanical compression of the sinusoids by lipid droplets. TXA(2) blockage may be a simple strategy to include steatotic organs and overcome the shortage of donor organs for liver transplantation.


Subject(s)
Fatty Liver/metabolism , Lipids/chemistry , Liver/injuries , Liver/metabolism , Reperfusion Injury/etiology , Reperfusion Injury/metabolism , Thromboxane A2/metabolism , 6-Ketoprostaglandin F1 alpha/biosynthesis , Animals , Arachidonic Acid/metabolism , Fatty Acids, Omega-3/administration & dosage , Fatty Liver/complications , Fatty Liver/pathology , Lipid Metabolism , Liver/blood supply , Liver/drug effects , Macrophage Activation/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Microcirculation/drug effects , Neutrophil Activation/drug effects , Oxidative Stress/drug effects , Receptors, Thromboxane A2, Prostaglandin H2/antagonists & inhibitors , Reperfusion Injury/pathology
19.
Hepatology ; 51(4): 1244-54, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20099302

ABSTRACT

UNLABELLED: In addition to its function as a neurotransmitter and vascular active molecule, serotonin is also a mitogen for hepatocytes and promotes liver regeneration. A possible role in hepatocellular cancer has not yet been investigated. Human hepatocellular cancer cell lines Huh7 and HepG2 were used to assess the function of serotonin in these cell lines. Characteristics of autophagy were detected with transmission electron microscopy, immunoblots of microtubule-associated protein light chain 3(LC3) and p62 (sequestosome 1). Immunoblots of the mammalian target of rapamycin (mTOR) and its downstream targets p70S6K and 4E-BP1 were used to investigate signaling pathways of serotonin. Two different animal models served as principle of proof of in vitro findings. Clinical relevance of the experimental findings was evaluated with a tissue microarray from 168 patients with hepatocellular carcinoma. Serotonin promotes tumor growth and survival in starved hepatocellular carcinoma cells. During starvation hepatocellular carcinoma cells exhibited characteristics of autophagy, which disappeared in serotonin-treated cells. Rapamycin, an inhibitor of mTOR, is known to induce autophagy. Serotonin could override rapamycin by an mTOR-independent pathway and activate common downstream signals such as p70S6K and 4E-BP1. In two tumor models of the mouse, inhibition of serotonin signaling consistently impaired tumor growth. Human biopsies revealed expression of the serotonin receptor HTR2B, correlating with downstream signals, e.g., phosphorylated p70S6K and proliferation. CONCLUSION: This study provides evidence that serotonin is involved in tumor growth of hepatocellular cancer by activating downstream targets of mTOR, and therefore serotonin-related pathways might represent a new treatment strategy.


Subject(s)
Carcinoma, Hepatocellular/pathology , Liver Neoplasms/pathology , Serotonin/pharmacology , Animals , Autophagy/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cytoprotection , Disease Models, Animal , Female , Humans , Intracellular Signaling Peptides and Proteins/physiology , Mice , Protein Serine-Threonine Kinases/physiology , Receptor, Serotonin, 5-HT2B/physiology , Signal Transduction , TOR Serine-Threonine Kinases
20.
Liver Int ; 31(3): 313-21, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21281431

ABSTRACT

BACKGROUND: New chemotherapy regimens are increasingly used in metastatic colorectal cancer to the liver before surgery. Some clinical observations have suggested that chemotherapy may affect liver regeneration. AIMS: The aim of this study was to evaluate liver damage and liver regeneration after chemotherapy treatment in a model of partial hepatectomy. METHODS: C57BL/6 mice were repeatedly treated with intraperitoneal injections of either saline or different chemotherapy regimens including the drugs 5-fluorouracyl (5-FU), irinotecan, oxaliplatin, gemcitabine and combined treatments with 5-FU/irinotecan, 5-FU/oxaliplatin. A 70% partial hepatectomy was performed 1 week after the last injection. Ki-67 and PCNA immunohistochemistry were performed to assess liver regeneration, serum liver enzymes and histology analysis to evaluate injury. RESULTS: A variety of chemotherapeutic agents used at maximum tolerated doses compatible with survival affected body weight and blood cell levels. However, these regimens did not affect liver injury before and after hepatectomy nor did they impair liver regeneration. Liver histology showed no steatosis, fibrosis or inflammation before hepatectomy. We therefore tested whether chemotherapy in presence of diet-induced steatosis may trigger injury. Even under these conditions, we did not observe histological signs of inflammation or sinusoidal injury. CONCLUSIONS: Liver injury and liver regeneration are not impaired after neoadjuvant chemotherapy with 5-FU, irinotecan, oxaliplatin and gemcitabine in non-tumoural liver parenchyma. In addition, combined treatments disclose no adverse effects on liver regeneration. Chemotherapy alone induces no histological alterations even in the presence of steatosis.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/toxicity , Chemical and Drug Induced Liver Injury/etiology , Disease Models, Animal , Hepatectomy , Liver Regeneration/drug effects , Liver/drug effects , Alanine Transaminase/blood , Animals , Aspartate Aminotransferases/blood , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/pathology , Fatty Liver/complications , Fatty Liver/pathology , Ki-67 Antigen/metabolism , Liver/metabolism , Liver/pathology , Liver Regeneration/physiology , Mice , Mice, Inbred C57BL , Proliferating Cell Nuclear Antigen/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL