ijms-logo

Journal Browser

Journal Browser

Molecular Mechanisms of Specific Target Organ Toxicity

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 (31 October 2022) | Viewed by 25083

Special Issue Editor

Special Issue Information

Dear Colleagues, 

The field of toxicology is moving away from classical strategies that use animals to generate apical toxic outcomes assumed to be extrapolatable to humans toward more predictive human-based approaches devoid of animals. One of the historical milestones in this regard was the publication of the report “Toxicity testing in the 21st century: a vision and a strategy” by the US National Academy of Sciences in 2007, which fully embraces these novel approaches and that highlighted the importance of mechanistic toxicology. In this respect, human-based in vitro (cell culture) and in silico (computational) methodologies are strongly preferred over complex animal models to elucidate and predict mechanisms of toxicological action. This research field is still gaining momentum and has now evolved to become a multidisciplinary science feeding from various areas.

This Special Issue of the International Journal of Molecular Sciences focuses on the use of in vitro and in silico tools for the study and prediction of specific target organ toxicity at the mechanistic level. Submission of manuscripts, both original research papers and review papers, related to basic and applied animal-free and human-based toxicology involving a broad chemical space, such as pharmaceuticals, food ingredients, cosmetics, biocides, and industrial chemicals, is encouraged. The target audience includes undergraduates to full professionals in academic, industrial, and regulatory settings.

Prof. Dr. Mathieu Vinken
Guest Editor

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

  • toxicity
  • mechanism
  • adverse outcome pathway
  • cell and tissue culture
  • computational modeling

Published Papers (8 papers)

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

Research

Jump to: Review

13 pages, 2248 KiB  
Article
Transcriptomic Analysis in Human 3D Skin Model Injected with Resorbable Hyaluronic Acid Fillers Reveals Foreign Body Response
by Danyel G. J. Jennen, Marcel van Herwijnen, Marlon Jetten, Rob J. Vandebriel, Peter Keizers, Robert E. Geertsma, Wim H. de Jong and Jos C. S. Kleinjans
Int. J. Mol. Sci. 2022, 23(21), 13046; https://doi.org/10.3390/ijms232113046 - 27 Oct 2022
Cited by 2 | Viewed by 1764
Abstract
Usage of injectable dermal fillers applied for aesthetic purposes has extensively increased over the years. As such, the number of related adverse reactions has increased, including patients showing severe complications such as product migration, topical swelling and inflammatory reactions of the skin. In [...] Read more.
Usage of injectable dermal fillers applied for aesthetic purposes has extensively increased over the years. As such, the number of related adverse reactions has increased, including patients showing severe complications such as product migration, topical swelling and inflammatory reactions of the skin. In order to understand the underlying molecular events of these adverse reactions we performed a genome-wide gene expression study on the multi-cell type human Phenion® Full-Thickness Skin Model exposed to five experimental hyaluronic acid (HA) preparations with increasing cross-linking degree, four commercial fillers from Perfectha®, and non-resorbable filler Bio-Alcamid®. In addition, we evaluated whether cross-linking degree or particle size of the HA-based fillers could be associated with the occurrence of adverse effects. In all cases, exposure to different HA fillers resulted in a clearly elevated gene expression of cytokines and chemokines related to acute inflammation as part of the foreign body response. Furthermore, for one experimental filler genes of OXPHOS complexes I-V were significantly down-regulated (adjusted p-value < 0.05), resulting in mitochondrial dysfunction which can be linked to over-expression of pro-inflammatory cytokines TNFα and IL-1β and chemokine CCL2. Our hypothesis that cross-linking degree or particle size of the HA-based fillers is related to the biological responses induced by these fillers could only partially be confirmed for particle size. In conclusion, our innovative approach resulted in gene expression changes from a human 3D skin model exposed to dermal fillers that mechanistically substantiate aforementioned adverse reactions, and thereby adds to the weight of evidence that these fillers may induce inflammatory and fibrotic responses. Full article
(This article belongs to the Special Issue Molecular Mechanisms of Specific Target Organ Toxicity)
Show Figures

Figure 1

26 pages, 5340 KiB  
Article
The Joint Influence of Tl+ and Thiol-Modifying Agents on Rat Liver Mitochondrial Parameters In Vitro
by Sergey M. Korotkov and Artemy V. Novozhilov
Int. J. Mol. Sci. 2022, 23(16), 8964; https://doi.org/10.3390/ijms23168964 - 11 Aug 2022
Cited by 3 | Viewed by 1140
Abstract
Recent data have shown that the mitochondrial permeability transition pore (MPTP) is the complex of the Ca2+-modified adenine nucleotide translocase (ANT) and the Ca2+-modified ATP synthase. We found in a previous study that ANT conformational changes may be involved [...] Read more.
Recent data have shown that the mitochondrial permeability transition pore (MPTP) is the complex of the Ca2+-modified adenine nucleotide translocase (ANT) and the Ca2+-modified ATP synthase. We found in a previous study that ANT conformational changes may be involved in Tl+-induced MPTP opening in the inner membrane of Ca2+-loaded rat liver mitochondria. In this study, the effects of thiol-modifying agents (eosin-5-maleimide (EMA), fluorescein isothiocyanate (FITC), Cu(o-phenanthroline)2 (Cu(OP)2), and embelin (Emb)), and MPTP inhibitors (ADP, cyclosporine A (CsA), n-ethylmaleimide (NEM), and trifluoperazine (TFP)) on MPTP opening were tested simultaneously with increases in swelling, membrane potential (ΔΨmito) decline, decreases in state 3, 4, and 3UDNP (2,4-dinitrophenol-uncoupled) respiration, and changes in the inner membrane free thiol group content. The effects of these thiol-modifying agents on the studied mitochondrial characteristics were multidirectional and showed a clear dependence on their concentration. This research suggests that Tl+-induced MPTP opening in the inner membrane of calcium-loaded mitochondria may be caused by the interaction of used reagents (EMA, FITC, Emb, Cu(OP)2) with active groups of ANT, the mitochondrial phosphate carrier (PiC) and the mitochondrial respiratory chain complexes. This study provides further insight into the causes of thallium toxicity and may be useful in the development of new treatments for thallium poisoning. Full article
(This article belongs to the Special Issue Molecular Mechanisms of Specific Target Organ Toxicity)
Show Figures

Figure 1

21 pages, 4110 KiB  
Article
Effects of Drugs Formerly Suggested for COVID-19 Repurposing on Pannexin1 Channels
by Anne Caufriez, Andrés Tabernilla, Raf Van Campenhout, Axelle Cooreman, Kaat Leroy, Julen Sanz Serrano, Prashant Kadam, Bruna dos Santos Rodrigues, Arthur Lamouroux, Steven Ballet and Mathieu Vinken
Int. J. Mol. Sci. 2022, 23(10), 5664; https://doi.org/10.3390/ijms23105664 - 18 May 2022
Cited by 1 | Viewed by 1959
Abstract
Although many efforts have been made to elucidate the pathogenesis of COVID-19, the underlying mechanisms are yet to be fully uncovered. However, it is known that a dysfunctional immune response and the accompanying uncontrollable inflammation lead to troublesome outcomes in COVID-19 patients. Pannexin1 [...] Read more.
Although many efforts have been made to elucidate the pathogenesis of COVID-19, the underlying mechanisms are yet to be fully uncovered. However, it is known that a dysfunctional immune response and the accompanying uncontrollable inflammation lead to troublesome outcomes in COVID-19 patients. Pannexin1 channels are put forward as interesting drug targets for the treatment of COVID-19 due to their key role in inflammation and their link to other viral infections. In the present study, we selected a panel of drugs previously tested in clinical trials as potential candidates for the treatment of COVID-19 early on in the pandemic, including hydroxychloroquine, chloroquine, azithromycin, dexamethasone, ribavirin, remdesivir, favipiravir, lopinavir, and ritonavir. The effect of the drugs on pannexin1 channels was assessed at a functional level by means of measurement of extracellular ATP release. Immunoblot analysis and real-time quantitative reversetranscription polymerase chain reaction analysis were used to study the potential of the drugs to alter pannexin1 protein and mRNA expression levels, respectively. Favipiravir, hydroxychloroquine, lopinavir, and the combination of lopinavir with ritonavir were found to inhibit pannexin1 channel activity without affecting pannexin1 protein or mRNA levels. Thusthree new inhibitors of pannexin1 channels were identified that, though currently not being used anymore for the treatment of COVID-19 patients, could be potential drug candidates for other pannexin1-related diseases. Full article
(This article belongs to the Special Issue Molecular Mechanisms of Specific Target Organ Toxicity)
Show Figures

Figure 1

18 pages, 1808 KiB  
Article
Prediction of the Neurotoxic Potential of Chemicals Based on Modelling of Molecular Initiating Events Upstream of the Adverse Outcome Pathways of (Developmental) Neurotoxicity
by Domenico Gadaleta, Nicoleta Spînu, Alessandra Roncaglioni, Mark T. D. Cronin and Emilio Benfenati
Int. J. Mol. Sci. 2022, 23(6), 3053; https://doi.org/10.3390/ijms23063053 - 11 Mar 2022
Cited by 11 | Viewed by 3493
Abstract
Developmental and adult/ageing neurotoxicity is an area needing alternative methods for chemical risk assessment. The formulation of a strategy to screen large numbers of chemicals is highly relevant due to potential exposure to compounds that may have long-term adverse health consequences on the [...] Read more.
Developmental and adult/ageing neurotoxicity is an area needing alternative methods for chemical risk assessment. The formulation of a strategy to screen large numbers of chemicals is highly relevant due to potential exposure to compounds that may have long-term adverse health consequences on the nervous system, leading to neurodegeneration. Adverse Outcome Pathways (AOPs) provide information on relevant molecular initiating events (MIEs) and key events (KEs) that could inform the development of computational alternatives for these complex effects. We propose a screening method integrating multiple Quantitative Structure–Activity Relationship (QSAR) models. The MIEs of existing AOP networks of developmental and adult/ageing neurotoxicity were modelled to predict neurotoxicity. Random Forests were used to model each MIE. Predictions returned by single models were integrated and evaluated for their capability to predict neurotoxicity. Specifically, MIE predictions were used within various types of classifiers and compared with other reference standards (chemical descriptors and structural fingerprints) to benchmark their predictive capability. Overall, classifiers based on MIE predictions returned predictive performances comparable to those based on chemical descriptors and structural fingerprints. The integrated computational approach described here will be beneficial for large-scale screening and prioritisation of chemicals as a function of their potential to cause long-term neurotoxic effects. Full article
(This article belongs to the Special Issue Molecular Mechanisms of Specific Target Organ Toxicity)
Show Figures

Figure 1

24 pages, 3035 KiB  
Article
A Transcriptomic Approach to Elucidate the Mechanisms of Gefitinib-Induced Toxicity in Healthy Human Intestinal Organoids
by Daniela Rodrigues, Bram Herpers, Sofia Ferreira, Heeseung Jo, Ciarán Fisher, Luke Coyle, Seung-Wook Chung, Jos C. S. Kleinjans, Danyel G. J. Jennen and Theo M. de Kok
Int. J. Mol. Sci. 2022, 23(4), 2213; https://doi.org/10.3390/ijms23042213 - 17 Feb 2022
Cited by 6 | Viewed by 2698
Abstract
Gefitinib is a tyrosine kinase inhibitor (TKI) that selectively inhibits the epidermal growth factor receptor (EGFR), hampering cell growth and proliferation. Due to its action, gefitinib has been used in the treatment of cancers that present abnormally increased expression of EGFR. However, side [...] Read more.
Gefitinib is a tyrosine kinase inhibitor (TKI) that selectively inhibits the epidermal growth factor receptor (EGFR), hampering cell growth and proliferation. Due to its action, gefitinib has been used in the treatment of cancers that present abnormally increased expression of EGFR. However, side effects from gefitinib therapy may occur, among which diarrhoea is most common, that can lead to interruption of the planned therapy in the more severe cases. The mechanisms underlying intestinal toxicity induced by gefitinib are not well understood. Therefore, this study aims at providing insight into these mechanisms based on transcriptomic responses induced in vitro. A 3D culture of healthy human colon and small intestine (SI) organoids was exposed to 0.1, 1, 10 and 30 µM of gefitinib, for a maximum of three days. These drug concentrations were selected using physiologically-based pharmacokinetic simulation considering patient dosing regimens. Samples were used for the analysis of viability and caspase 3/7 activation, image-based analysis of structural changes, as well as RNA isolation and sequencing via high-throughput techniques. Differential gene expression analysis showed that gefitinib perturbed signal transduction pathways, apoptosis, cell cycle, FOXO-mediated transcription, p53 signalling pathway, and metabolic pathways. Remarkably, opposite expression patterns of genes associated with metabolism of lipids and cholesterol biosynthesis were observed in colon versus SI organoids in response to gefitinib. These differences in the organoids’ responses could be linked to increased activated protein kinase (AMPK) activity in colon, which can influence the sensitivity of the colon to the drug. Therefore, this study sheds light on how gefitinib induces toxicity in intestinal organoids and provides an avenue towards the development of a potential tool for drug screening and development. Full article
(This article belongs to the Special Issue Molecular Mechanisms of Specific Target Organ Toxicity)
Show Figures

Figure 1

28 pages, 5049 KiB  
Article
Unravelling Mechanisms of Doxorubicin-Induced Toxicity in 3D Human Intestinal Organoids
by Daniela Rodrigues, Luke Coyle, Barbara Füzi, Sofia Ferreira, Heeseung Jo, Bram Herpers, Seung-Wook Chung, Ciarán Fisher, Jos C. S. Kleinjans, Danyel Jennen and Theo M. de Kok
Int. J. Mol. Sci. 2022, 23(3), 1286; https://doi.org/10.3390/ijms23031286 - 24 Jan 2022
Cited by 12 | Viewed by 3781
Abstract
Doxorubicin is widely used in the treatment of different cancers, and its side effects can be severe in many tissues, including the intestines. Symptoms such as diarrhoea and abdominal pain caused by intestinal inflammation lead to the interruption of chemotherapy. Nevertheless, the molecular [...] Read more.
Doxorubicin is widely used in the treatment of different cancers, and its side effects can be severe in many tissues, including the intestines. Symptoms such as diarrhoea and abdominal pain caused by intestinal inflammation lead to the interruption of chemotherapy. Nevertheless, the molecular mechanisms associated with doxorubicin intestinal toxicity have been poorly explored. This study aims to investigate such mechanisms by exposing 3D small intestine and colon organoids to doxorubicin and to evaluate transcriptomic responses in relation to viability and apoptosis as physiological endpoints. The in vitro concentrations and dosing regimens of doxorubicin were selected based on physiologically based pharmacokinetic model simulations of treatment regimens recommended for cancer patients. Cytotoxicity and cell morphology were evaluated as well as gene expression and biological pathways affected by doxorubicin. In both types of organoids, cell cycle, the p53 signalling pathway, and oxidative stress were the most affected pathways. However, significant differences between colon and SI organoids were evident, particularly in essential metabolic pathways. Short time-series expression miner was used to further explore temporal changes in gene profiles, which identified distinct tissue responses. Finally, in silico proteomics revealed important proteins involved in doxorubicin metabolism and cellular processes that were in line with the transcriptomic responses, including cell cycle and senescence, transport of molecules, and mitochondria impairment. This study provides new insight into doxorubicin-induced effects on the gene expression levels in the intestines. Currently, we are exploring the potential use of these data in establishing quantitative systems toxicology models for the prediction of drug-induced gastrointestinal toxicity. Full article
(This article belongs to the Special Issue Molecular Mechanisms of Specific Target Organ Toxicity)
Show Figures

Figure 1

Review

Jump to: Research

24 pages, 2255 KiB  
Review
Review of Knowledge of Uranium-Induced Kidney Toxicity for the Development of an Adverse Outcome Pathway to Renal Impairment
by Yann Guéguen and Marie Frerejacques
Int. J. Mol. Sci. 2022, 23(8), 4397; https://doi.org/10.3390/ijms23084397 - 15 Apr 2022
Cited by 15 | Viewed by 3071
Abstract
An adverse outcome pathway (AOP) is a conceptual construct of causally and sequentially linked events, which occur during exposure to stressors, with an adverse outcome relevant to risk assessment. The development of an AOP is a means of identifying knowledge gaps in order [...] Read more.
An adverse outcome pathway (AOP) is a conceptual construct of causally and sequentially linked events, which occur during exposure to stressors, with an adverse outcome relevant to risk assessment. The development of an AOP is a means of identifying knowledge gaps in order to prioritize research assessing the health risks associated with exposure to physical or chemical stressors. In this paper, a review of knowledge was proposed, examining experimental and epidemiological data, in order to identify relevant key events and potential key event relationships in an AOP for renal impairment, relevant to stressors such as uranium (U). Other stressors may promote similar pathways, and this review is a necessary step to compare and combine knowledge reported for nephrotoxicants. U metal ions are filtered through the glomerular membrane of the kidneys, then concentrate in the cortical and juxtaglomerular areas, and bind to the brush border membrane of the proximal convoluted tubules. U uptake by epithelial cells occurs through endocytosis and the sodium-dependent phosphate co-transporter (NaPi-IIa). The identified key events start with the inhibition of the mitochondria electron transfer chain and the collapse of mitochondrial membrane potential, due to cytochrome b5/cytochrome c disruption. In the nucleus, U directly interacts with negatively charged DNA phosphate, thereby inducing an adduct formation, and possibly DNA strand breaks or cross-links. U also compromises DNA repair by inhibiting zing finger proteins. Thereafter, U triggers the Nrf2, NF-κB, or endoplasmic reticulum stress pathways. The resulting cellular key events include oxidative stress, DNA strand breaks and chromosomal aberrations, apoptosis, and pro-inflammatory effects. Finally, the main adverse outcome is tubular damage of the S2 and S3 segments of the kidneys, leading to tubular cell death, and then kidney failure. The attribution of renal carcinogenesis due to U is controversial, and specific experimental or epidemiological studies must be conducted. A tentative construction of an AOP for uranium-induced kidney toxicity and failure was proposed. Full article
(This article belongs to the Special Issue Molecular Mechanisms of Specific Target Organ Toxicity)
Show Figures

Figure 1

37 pages, 1661 KiB  
Review
Mitochondria as the Target of Hepatotoxicity and Drug-Induced Liver Injury: Molecular Mechanisms and Detection Methods
by Milos Mihajlovic and Mathieu Vinken
Int. J. Mol. Sci. 2022, 23(6), 3315; https://doi.org/10.3390/ijms23063315 - 18 Mar 2022
Cited by 31 | Viewed by 6122
Abstract
One of the major mechanisms of drug-induced liver injury includes mitochondrial perturbation and dysfunction. This is not a surprise, given that mitochondria are essential organelles in most cells, which are responsible for energy homeostasis and the regulation of cellular metabolism. Drug-induced mitochondrial dysfunction [...] Read more.
One of the major mechanisms of drug-induced liver injury includes mitochondrial perturbation and dysfunction. This is not a surprise, given that mitochondria are essential organelles in most cells, which are responsible for energy homeostasis and the regulation of cellular metabolism. Drug-induced mitochondrial dysfunction can be influenced by various factors and conditions, such as genetic predisposition, the presence of metabolic disorders and obesity, viral infections, as well as drugs. Despite the fact that many methods have been developed for studying mitochondrial function, there is still a need for advanced and integrative models and approaches more closely resembling liver physiology, which would take into account predisposing factors. This could reduce the costs of drug development by the early prediction of potential mitochondrial toxicity during pre-clinical tests and, especially, prevent serious complications observed in clinical settings. Full article
(This article belongs to the Special Issue Molecular Mechanisms of Specific Target Organ Toxicity)
Show Figures

Figure 1

Back to TopTop