ijms-logo

Journal Browser

Journal Browser

Molecular Mechanisms of Hepatotoxicity

A special issue of International Journal of Molecular Sciences (ISSN 1422-0067). This special issue belongs to the section "Molecular Toxicology".

Deadline for manuscript submissions: closed (30 September 2022) | Viewed by 23377

Special Issue Editors


E-Mail Website
Guest Editor
UMR_S 1193, INSERM/Université Paris-Saclay, Fédération Hospitalo-Universitaire (FHU) Hépatinov, F-94807 Villejuif, France
Interests: organoids; human pluripotent stem cells; hepatic differentiation; biomaterials; biohybrid systems; bioartificial liver; bioengineering; bio-printing; imaging; light-sheet microscopy; biochemistry; pharmacology; organ-on-chip

E-Mail Website1 Website2
Guest Editor
UMR_S 1193, INSERM/Université Paris-Saclay, Fédération Hospitalo-Universitaire (FHU) Hépatinov, F-94807 Villejuif, France
Interests: end stage liver disease; rare liver disorders; liver regenerative medicine; liver transplantation; liver bioconstruction; bioengineering

Special Issue Information

Dear Colleagues,

Liver toxicity is the leading cause of the removal of approved drugs from the market as well as the constant increase in patients suffering from acute liver failure. Drug-induced liver injury (DILI) can be caused by numerous pharmaceutical agents, as well as dietary or herbal supplements. Moreover, with a few exceptions, the mechanisms by which these drugs and molecules cause hepatic injury are elusive. Since there is no real treatment available apart from orthotopic liver transplantation for the most severe cases, the characterization of the specific mechanisms that lead to hepatotoxicity, the identification of potential novel targets for the development of new therapeutic strategies, as well as the development of new devices for improving drug screening and pharmacological responses are major priorities in this field.

The aim of this Special Issue is to provide an overview of the main cellular and molecular mechanisms; to elucidate the causes and sources of hepatoxicity, which leads to the onset and progression of hepatic disorders, highlighting both the common and specific pathways involved in each one of them; and to propose new bio-technological approaches for studying toxicity.

Dr. Antonietta Messina
Prof. Dr. Jean-Charles Duclos-Vallée
Guest Editors

Manuscript Submission Information

Manuscripts should be submitted online at www.mdpi.com by registering and logging in to this website. Once you are registered, click here to go to the submission form. Manuscripts can be submitted until the deadline. All submissions that pass pre-check are peer-reviewed. Accepted papers will be published continuously in the journal (as soon as accepted) and will be listed together on the special issue website. Research articles, review articles as well as short communications are invited. For planned papers, a title and short abstract (about 100 words) can be sent to the Editorial Office for announcement on this website.

Submitted manuscripts should not have been published previously, nor be under consideration for publication elsewhere (except conference proceedings papers). All manuscripts are thoroughly refereed through a single-blind peer-review process. A guide for authors and other relevant information for submission of manuscripts is available on the Instructions for Authors page. International Journal of Molecular Sciences is an international peer-reviewed open access semimonthly journal published by MDPI.

Please visit the Instructions for Authors page before submitting a manuscript. There is an Article Processing Charge (APC) for publication in this open access journal. For details about the APC please see here. Submitted papers should be well formatted and use good English. Authors may use MDPI's English editing service prior to publication or during author revisions.

Keywords

  • hepatotoxicity
  • drug-induced liver injury (DILI)
  • epidemiology of liver toxicity
  • organoids
  • organ-on-chip
  • acute liver failure (ALF)
  • drugs
  • paracetamol
  • antibiotics
  • mitochondria
  • drug-screening
  • immuno-toxicity

Published Papers (9 papers)

Order results
Result details
Select all
Export citation of selected articles as:

Editorial

Jump to: Research, Review

4 pages, 205 KiB  
Editorial
Molecular Mechanisms of Hepatotoxicity
by Antonietta Messina and Jean-Charles Duclos-Vallée
Int. J. Mol. Sci. 2023, 24(4), 3791; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms24043791 - 14 Feb 2023
Viewed by 1608
Abstract
Drug-induced liver injury, also known as drug-induced hepatotoxicity (DILI), is a major cause of medicine withdrawal (prescription or over-the-counter) from the market [...] Full article
(This article belongs to the Special Issue Molecular Mechanisms of Hepatotoxicity)

Research

Jump to: Editorial, Review

14 pages, 2299 KiB  
Article
Plasma Sphingoid Base Profiles of Patients Diagnosed with Intrinsic or Idiosyncratic Drug-induced Liver Injury
by Zhibo Gai, Sophia L. Samodelov, Irina Alecu, Thorsten Hornemann, Jane I. Grove, Guruprasad P. Aithal, Michele Visentin and Gerd A. Kullak-Ublick
Int. J. Mol. Sci. 2023, 24(3), 3013; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms24033013 - 3 Feb 2023
Cited by 2 | Viewed by 2210
Abstract
Sphingolipids are exceptionally diverse, comprising hundreds of unique species. The bulk of circulating sphingolipids are synthesized in the liver, thereby plasma sphingolipid profiles represent reliable surrogates of hepatic sphingolipid metabolism and content. As changes in plasma sphingolipid content have been associated to exposure [...] Read more.
Sphingolipids are exceptionally diverse, comprising hundreds of unique species. The bulk of circulating sphingolipids are synthesized in the liver, thereby plasma sphingolipid profiles represent reliable surrogates of hepatic sphingolipid metabolism and content. As changes in plasma sphingolipid content have been associated to exposure to drugs inducing hepatotoxicity both in vitro and in rodents, in the present study the translatability of the preclinical data was assessed by analyzing the plasma of patients with suspected drug-induced liver injury (DILI) and control subjects. DILI patients, whether intrinsic or idiosyncratic cases, had no alterations in total sphingoid base levels and profile composition compared to controls, whereby cardiovascular disease (CVD) was a confounding factor. Upon exclusion of CVD individuals, elevation of 1-deoxysphingosine (1-deoxySO) in the DILI group emerged. Notably, 1-deoxySO values did not correlate with ALT values. While 1-deoxySO was elevated in all DILI cases, only intrinsic DILI cases concomitantly displayed reduction of select shorter chain sphingoid bases. Significant perturbation of the sphingolipid metabolism observed in this small exploratory clinical study is discussed and put into context, in the consideration that sphingolipids might contribute to the onset and progression of DILI, and that circulating sphingoid bases may function as mechanistic markers to study DILI pathophysiology. Full article
(This article belongs to the Special Issue Molecular Mechanisms of Hepatotoxicity)
Show Figures

Figure 1

17 pages, 2488 KiB  
Article
Methotrexate-Induced Liver Injury Is Associated with Oxidative Stress, Impaired Mitochondrial Respiration, and Endoplasmic Reticulum Stress In Vitro
by Saskia Schmidt, Catherine Jane Messner, Carine Gaiser, Carina Hämmerli and Laura Suter-Dick
Int. J. Mol. Sci. 2022, 23(23), 15116; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms232315116 - 1 Dec 2022
Cited by 8 | Viewed by 3495
Abstract
Low-dose methotrexate (MTX) is a standard therapy for rheumatoid arthritis due to its low cost and efficacy. Despite these benefits, MTX has been reported to cause chronic drug-induced liver injury, namely liver fibrosis. The hallmark of liver fibrosis is excessive scarring of liver [...] Read more.
Low-dose methotrexate (MTX) is a standard therapy for rheumatoid arthritis due to its low cost and efficacy. Despite these benefits, MTX has been reported to cause chronic drug-induced liver injury, namely liver fibrosis. The hallmark of liver fibrosis is excessive scarring of liver tissue, triggered by hepatocellular injury and subsequent activation of hepatic stellate cells (HSCs). However, little is known about the precise mechanisms through which MTX causes hepatocellular damage and activates HSCs. Here, we investigated the mechanisms leading to hepatocyte injury in HepaRG and used immortalized stellate cells (hTERT-HSC) to elucidate the mechanisms leading to HSC activation by exposing mono- and co-cultures of HepaRG and hTERT-HSC to MTX. The results showed that at least two mechanisms are involved in MTX-induced toxicity in HepaRG: (i) oxidative stress through depletion of glutathione (GSH) and (ii) impairment of cellular respiration in a GSH-independent manner. Furthermore, we measured increased levels of endoplasmic reticulum (ER) stress in activated HSC following MTX treatment. In conclusion, we established a human-relevant in vitro model to gain mechanistical insights into MTX-induced hepatotoxicity, linked oxidative stress in HepaRG to a GSH-dependent and -independent pathway, and hypothesize that not only oxidative stress in hepatocytes but also ER stress in HSCs contribute to MTX-induced activation of HSCs. Full article
(This article belongs to the Special Issue Molecular Mechanisms of Hepatotoxicity)
Show Figures

Figure 1

13 pages, 1407 KiB  
Article
Characterization of the RAS/RAF/ERK Signal Cascade as a Novel Regulating Factor in Alpha-Amanitin-Induced Cytotoxicity in Huh-7 Cells
by Doeun Kim, Min Seo Lee, Eunji Sung, Sangkyu Lee and Hye Suk Lee
Int. J. Mol. Sci. 2022, 23(20), 12294; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms232012294 - 14 Oct 2022
Cited by 4 | Viewed by 1668
Abstract
The well-known hepatotoxicity mechanism resulting from alpha-amanitin (α-AMA) exposure arises from RNA polymerase II (RNAP II) inhibition. RNAP Ⅱ inhibition occurs through the dysregulation of mRNA synthesis. However, the signaling pathways in hepatocytes that arise from α-AMA have not yet been fully elucidated. [...] Read more.
The well-known hepatotoxicity mechanism resulting from alpha-amanitin (α-AMA) exposure arises from RNA polymerase II (RNAP II) inhibition. RNAP Ⅱ inhibition occurs through the dysregulation of mRNA synthesis. However, the signaling pathways in hepatocytes that arise from α-AMA have not yet been fully elucidated. Here, we identified that the RAS/RAF/ERK signaling pathway was activated through quantitative phosphoproteomic and molecular biological analyses in Huh-7 cells. Bioinformatics analysis showed that α-AMA exposure increased protein phosphorylation in a time-dependent α-AMA exposure. In addition, phosphorylation increased not only the components of the ERK signaling pathway but also U2AF65 and SPF45, known splicing factors. Therefore, we propose a novel mechanism of α-AMA as follows. The RAS/RAF/ERK signaling pathway involved in aberrant splicing events is activated by α-AMA exposure followed by aberrant splicing events leading to cell death in Huh-7 cells. Full article
(This article belongs to the Special Issue Molecular Mechanisms of Hepatotoxicity)
Show Figures

Figure 1

20 pages, 2812 KiB  
Article
Possible Role of Extracellular Vesicles in Hepatotoxicity of Acetaminophen
by Martina Šrajer Gajdošik, Anamarija Kovač Peić, Marija Begić, Petra Grbčić, Kate E. Brilliant, Douglas C. Hixson and Djuro Josić
Int. J. Mol. Sci. 2022, 23(16), 8870; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms23168870 - 9 Aug 2022
Cited by 2 | Viewed by 2014
Abstract
We examined proteomic profiles of rat liver extracellular vesicles (EVs) shed following treatment with a sub-toxic dose (500 mg/kg) of the pain reliever drug, acetaminophen (APAP). EVs representing the entire complement of hepatic cells were isolated after perfusion of the intact liver and [...] Read more.
We examined proteomic profiles of rat liver extracellular vesicles (EVs) shed following treatment with a sub-toxic dose (500 mg/kg) of the pain reliever drug, acetaminophen (APAP). EVs representing the entire complement of hepatic cells were isolated after perfusion of the intact liver and analyzed with LC-MS/MS. The investigation was focused on revealing the function and cellular origin of identified EVs proteins shed by different parenchymal and non-parenchymal liver cells and their possible role in an early response of this organ to a toxic environment. Comparison of EV proteomic profiles from control and APAP-treated animals revealed significant differences. Alpha-1-macroglobulin and members of the cytochrome P450 superfamily were highly abundant proteins in EVs shed by the normal liver. In contrast, proteins like aminopeptidase N, metalloreductase STEAP4, different surface antigens like CD14 and CD45, and most members of the annexin family were detected only in EVs that were shed by livers of APAP-treated animals. In EVs from treated livers, there was almost a complete disappearance of members of the cytochrome P450 superfamily and a major decrease in other enzymes involved in the detoxification of xenobiotics. Additionally, there were proteins that predominated in non-parenchymal liver cells and in the extracellular matrix, like fibronectin, receptor-type tyrosine-protein phosphatase C, and endothelial type gp91. These differences indicate that even treatment with a sub-toxic concentration of APAP initiates dramatic perturbation in the function of this vital organ. Full article
(This article belongs to the Special Issue Molecular Mechanisms of Hepatotoxicity)
Show Figures

Figure 1

16 pages, 4146 KiB  
Article
Hepatocyte Specific gp130 Signalling Underlies APAP Induced Liver Injury
by Jinrui Dong, Wei-Wen Lim, Shamini G. Shekeran, Jessie Tan, Sze Yun Lim, Joyce Wei Ting Goh, Benjamin L. George, Sebastian Schafer, Stuart A. Cook and Anissa A. Widjaja
Int. J. Mol. Sci. 2022, 23(13), 7089; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms23137089 - 25 Jun 2022
Cited by 4 | Viewed by 2607
Abstract
N-acetyl-p-aminophenol (APAP)-induced liver damage is associated with upregulation of Interleukin-11 (IL11), which is thought to stimulate IL6ST (gp130)-mediated STAT3 activity in hepatocytes, as a compensatory response. However, recent studies have found IL11/IL11RA/gp130 signaling to be hepatotoxic. To investigate further the role of IL11 [...] Read more.
N-acetyl-p-aminophenol (APAP)-induced liver damage is associated with upregulation of Interleukin-11 (IL11), which is thought to stimulate IL6ST (gp130)-mediated STAT3 activity in hepatocytes, as a compensatory response. However, recent studies have found IL11/IL11RA/gp130 signaling to be hepatotoxic. To investigate further the role of IL11 and gp130 in APAP liver injury, we generated two new mouse strains with conditional knockout (CKO) of either Il11 (CKOIl11) or gp130 (CKOgp130) in adult hepatocytes. Following APAP, as compared to controls, CKOgp130 mice had lesser liver damage with lower serum Alanine Transaminase (ALT) and Aspartate Aminotransferase (AST), greatly reduced serum IL11 levels (90% lower), and lesser centrilobular necrosis. Livers from APAP-injured CKOgp130 mice had lesser ERK, JNK, NOX4 activation and increased markers of regeneration (PCNA, Cyclin D1, Ki67). Experiments were repeated in CKOIl11 mice that, as compared to wild-type mice, had lower APAP-induced ALT/AST, reduced centrilobular necrosis and undetectable IL11 in serum. As seen with CKOgp130 mice, APAP-treated CKOIl11 mice had lesser ERK/JNK/NOX4 activation and greater features of regeneration. Both CKOgp130 and CKOIl11 mice had normal APAP metabolism. After APAP, CKOgp130 and CKOIl11 mice had reduced Il6, Ccl2, Ccl5, Il1β, and Tnfα expression. These studies exclude IL11 upregulation as compensatory and establish autocrine, self-amplifying, gp130-dependent IL11 secretion from damaged hepatocytes as toxic and anti-regenerative. Full article
(This article belongs to the Special Issue Molecular Mechanisms of Hepatotoxicity)
Show Figures

Figure 1

14 pages, 7264 KiB  
Article
Silencing the Tlr4 Gene Alleviates Methamphetamine-Induced Hepatotoxicity by Inhibiting Lipopolysaccharide-Mediated Inflammation in Mice
by Li-Bin Wang, Li-Jian Chen, Qi Wang and Xiao-Li Xie
Int. J. Mol. Sci. 2022, 23(12), 6810; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms23126810 - 18 Jun 2022
Cited by 7 | Viewed by 2047
Abstract
Methamphetamine (METH) is a stimulant drug. METH abuse induces hepatotoxicity, although the mechanisms are not well understood. METH-induced hepatotoxicity was regulated by TLR4-mediated inflammation in BALB/c mice in our previous study. To further investigate the underlying mechanisms, the wild-type (C57BL/6) and Tlr4−/− [...] Read more.
Methamphetamine (METH) is a stimulant drug. METH abuse induces hepatotoxicity, although the mechanisms are not well understood. METH-induced hepatotoxicity was regulated by TLR4-mediated inflammation in BALB/c mice in our previous study. To further investigate the underlying mechanisms, the wild-type (C57BL/6) and Tlr4−/− mice were treated with METH. Transcriptomics of the mouse liver was performed via RNA-sequencing. Histopathological changes, serum levels of metabolic enzymes and lipopolysaccharide (LPS), and expression of TLR4-mediated proinflammatory cytokines were assessed. Compared to the control, METH treatment induced obvious histopathological changes and significantly increased the levels of metabolic enzymes in wild-type mice. Furthermore, inflammatory pathways were enriched in the liver of METH-treated mice, as demonstrated by expression analysis of RNA-sequencing data. Consistently, the expression of TLR4 pathway members was significantly increased by METH treatment. In addition, increased serum LPS levels in METH-treated mice indicated overproduction of LPS and gut microbiota dysbiosis. However, antibiotic pretreatment or silencing Tlr4 significantly decreased METH-induced hepatic injury, serum LPS levels, and inflammation. In addition, the dampening effects of silencing Tlr4 on inflammatory pathways were verified by the enrichment analysis of RNA-sequencing data in METH-treated Tlr4−/− mice compared to METH-treated wild-type mice. Taken together, these findings implied that Tlr4 silencing, comparable to antibiotic pretreatment, effectively alleviated METH-induced hepatotoxicity by inhibiting LPS-TLR4-mediated inflammation in the liver. Full article
(This article belongs to the Special Issue Molecular Mechanisms of Hepatotoxicity)
Show Figures

Figure 1

Review

Jump to: Editorial, Research

19 pages, 1511 KiB  
Review
The Farnesoid X Receptor as a Master Regulator of Hepatotoxicity
by Magdalena Rausch, Sophia L. Samodelov, Michele Visentin and Gerd A. Kullak-Ublick
Int. J. Mol. Sci. 2022, 23(22), 13967; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms232213967 - 12 Nov 2022
Cited by 7 | Viewed by 2657
Abstract
The nuclear receptor farnesoid X receptor (FXR, NR1H4) is a bile acid (BA) sensor that links the enterohepatic circuit that regulates BA metabolism and elimination to systemic lipid homeostasis. Furthermore, FXR represents a real guardian of the hepatic function, preserving, in a multifactorial [...] Read more.
The nuclear receptor farnesoid X receptor (FXR, NR1H4) is a bile acid (BA) sensor that links the enterohepatic circuit that regulates BA metabolism and elimination to systemic lipid homeostasis. Furthermore, FXR represents a real guardian of the hepatic function, preserving, in a multifactorial fashion, the integrity and function of hepatocytes from chronic and acute insults. This review summarizes how FXR modulates the expression of pathway-specific as well as polyspecific transporters and enzymes, thereby acting at the interface of BA, lipid and drug metabolism, and influencing the onset and progression of hepatotoxicity of varying etiopathogeneses. Furthermore, this review article provides an overview of the advances and the clinical development of FXR agonists in the treatment of liver diseases. Full article
(This article belongs to the Special Issue Molecular Mechanisms of Hepatotoxicity)
Show Figures

Graphical abstract

30 pages, 1401 KiB  
Review
In Vitro Models for Studying Chronic Drug-Induced Liver Injury
by M. Teresa Donato, Gloria Gallego-Ferrer and Laia Tolosa
Int. J. Mol. Sci. 2022, 23(19), 11428; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms231911428 - 28 Sep 2022
Cited by 15 | Viewed by 4241
Abstract
Drug-induced liver injury (DILI) is a major clinical problem in terms of patient morbidity and mortality, cost to healthcare systems and failure of the development of new drugs. The need for consistent safety strategies capable of identifying a potential toxicity risk early in [...] Read more.
Drug-induced liver injury (DILI) is a major clinical problem in terms of patient morbidity and mortality, cost to healthcare systems and failure of the development of new drugs. The need for consistent safety strategies capable of identifying a potential toxicity risk early in the drug discovery pipeline is key. Human DILI is poorly predicted in animals, probably due to the well-known interspecies differences in drug metabolism, pharmacokinetics, and toxicity targets. For this reason, distinct cellular models from primary human hepatocytes or hepatoma cell lines cultured as 2D monolayers to emerging 3D culture systems or the use of multi-cellular systems have been proposed for hepatotoxicity studies. In order to mimic long-term hepatotoxicity in vitro, cell models, which maintain hepatic phenotype for a suitably long period, should be used. On the other hand, repeated-dose administration is a more relevant scenario for therapeutics, providing information not only about toxicity, but also about cumulative effects and/or delayed responses. In this review, we evaluate the existing cell models for DILI prediction focusing on chronic hepatotoxicity, highlighting how better characterization and mechanistic studies could lead to advance DILI prediction. Full article
(This article belongs to the Special Issue Molecular Mechanisms of Hepatotoxicity)
Show Figures

Figure 1

Back to TopTop