1
|
Borgne-Sanchez A, Fromenty B. Mitochondrial dysfunction in drug-induced hepatic steatosis: Recent findings and current concept. Clin Res Hepatol Gastroenterol 2025; 49:102529. [PMID: 39798918 DOI: 10.1016/j.clinre.2025.102529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 12/19/2024] [Accepted: 01/08/2025] [Indexed: 01/15/2025]
Abstract
Mitochondrial activity is necessary for the maintenance of many liver functions. In particular, mitochondrial fatty acid oxidation (FAO) is required for energy production and lipid homeostasis. This key metabolic pathway is finely tuned by the mitochondrial respiratory chain (MRC) activity and different transcription factors such as peroxisome proliferator-activated receptor α (PPARα). Many drugs have been shown to cause mitochondrial dysfunction, which can lead to acute and chronic liver lesions. While severe inhibition of mitochondrial FAO would eventually cause microvesicular steatosis, hypoglycemia, and liver failure, moderate impairment of this metabolic pathway can induce macrovacuolar steatosis, which can progress in the long term to steatohepatitis and cirrhosis. Drugs can impair mitochondrial FAO through several mechanisms including direct inhibition of FAO enzymes, sequestration of coenzyme A and l-carnitine, impairment of the activity of one or several MRC complexes and reduced PPARα expression. In drug-induced macrovacuolar steatosis, non-mitochondrial mechanisms can also be involved in lipid accumulation including increased de novo lipogenesis and reduced very-low-density lipoprotein secretion. Nonetheless, mitochondrial dysfunction and subsequent oxidative stress appear to be key events in the progression of steatosis to steatohepatitis. Patients suffering from metabolic dysfunction-associated steatotic liver disease (MASLD) and treated with mitochondriotoxic drugs should be closely monitored to reduce the risk of acute liver injury or a faster transition of steatosis to steatohepatitis. Therapies based on the mitochondrial cofactor l-carnitine, the antioxidant N-acetylcysteine, or thyromimetics might be useful to prevent or treat drug-induced mitochondrial dysfunction, steatosis, and steatohepatitis.
Collapse
Affiliation(s)
| | - Bernard Fromenty
- INSERM, INRAE, Univ Rennes, Institut NUMECAN, UMR_S1317, 35000 Rennes, France.
| |
Collapse
|
2
|
Kim D, Shin Y, Baek YW, Kang H, Lim J, Bae ON. The effect of biocide chloromethylisothiazolinone/methylisothiazolinone (CMIT/MIT) mixture on C2C12 muscle cell damage attributed to mitochondrial reactive oxygen species overproduction and autophagy activation. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART A 2025; 88:137-151. [PMID: 39446036 DOI: 10.1080/15287394.2024.2420083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2024]
Abstract
The mixture of 5-chloro-2-methyl-4-isothiazolin-3-one and 2-methyl-4-isothiazolin-3-one (CMIT/MIT) is a biocide widely used as a preservative in various commercial products. This biocide has also been used as an active ingredient in humidifier disinfectants in South Korea, resulting in serious health effects among users. Recent evidence suggests that the underlying mechanism of CMIT/MIT-initiated toxicity might be associated with defects in mitochondrial functions. The aim of this study was to utilize the C2C12 skeletal muscle model to investigate the effects of CMIT/MIT on mitochondrial function and relevant molecular pathways associated with skeletal muscle dysfunction. Data demonstrated that exposure to CMIT/MIT during myogenic differentiation induced significant mitochondrial excess production of reactive oxygen species (ROS) and a decrease in intracellular ATP levels. Notably, CMIT/MIT significantly inhibited mitochondrial oxidative phosphorylation (Oxphos) and reduced mitochondrial mass at a lower concentration than the biocide amount, which diminished the viability of myotubes. CMIT/MIT induced activation of autophagy flux and decreased protein expression levels of myosin heavy chain (MHC). Taken together, CMIT/MIT exposure produced damage in C2C12 myotubes by impairing mitochondrial bioenergetics and activating autophagy. Our findings contribute to an increased understanding of the underlying mechanisms associated with CMIT/MIT-induced adverse skeletal muscle health effects.
Collapse
Affiliation(s)
- Donghyun Kim
- College of Pharmacy Institute of Pharmaceutical Science and Technology, Hanyang University ERICA Campus, Ansan, South Korea
| | - Yusun Shin
- College of Pharmacy Institute of Pharmaceutical Science and Technology, Hanyang University ERICA Campus, Ansan, South Korea
| | - Yong-Wook Baek
- Humidifier Disinfectant Health Center, Environmental Health Research, National Institute of Environmental Research, Incheon, Republic of Korea
| | - HanGoo Kang
- Humidifier Disinfectant Health Center, Environmental Health Research, National Institute of Environmental Research, Incheon, Republic of Korea
| | - Jungyun Lim
- Humidifier Disinfectant Health Center, Environmental Health Research, National Institute of Environmental Research, Incheon, Republic of Korea
| | - Ok-Nam Bae
- College of Pharmacy Institute of Pharmaceutical Science and Technology, Hanyang University ERICA Campus, Ansan, South Korea
| |
Collapse
|
3
|
Mitchell RJ, Havrylyuk D, Hachey AC, Heidary DK, Glazer EC. Photodynamic therapy photosensitizers and photoactivated chemotherapeutics exhibit distinct bioenergetic profiles to impact ATP metabolism. Chem Sci 2025; 16:721-734. [PMID: 39629492 PMCID: PMC11609979 DOI: 10.1039/d4sc05393a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Accepted: 11/18/2024] [Indexed: 12/07/2024] Open
Abstract
Energy is essential for all life, and mammalian cells generate and store energy in the form of ATP by mitochondrial (oxidative phosphorylation) and non-mitochondrial (glycolysis) metabolism. These processes can now be evaluated by extracellular flux analysis (EFA), which has proven to be an indispensable tool in cell biology, providing previously inaccessible information regarding the bioenergetic landscape of cell lines, complex tissues, and in vivo models. Recently, EFA demonstrated its utility as a screening tool in drug development, both by providing insights into small molecule-organelle interactions, and by revealing the peripheral and potentially undesired off-target effects small molecules have within cells. Surprisingly, technologies to quantify cellular bioenergetics have not been systematically applied in phototherapy development, leaving open several questions about how the mechanism of action of a compound can impact essential cellular functions. Here, we utilized the Seahorse analyzer to address this question for photosensitizers (PSs) for photodynamic therapy (PDT) and contrast these systems to molecules that photo-release a ligand and thus act as photocages or photoactivated chemotherapeutics (PACT), intending to understand the influence these two classes of compounds have on cellular bioenergetics. EFA results show that acute treatment of A549 lung adenocarcinoma cells with PDT agents induces a quiescent bioenergetic response as a result of mitochondrial respiration shutdown. The loss of oxidative phosphorylation is followed by disruption of glycolysis, which occurs after an initial increase in glycolytic respiration is unable to compensate for the interruption of the electron transport chain (ETC). In contrast, the PACT agents tested had little impact on cellular respiration, and the minor inhibition of these metabolic processes was not related to the mechanism of action, as reflected by a lack of correlation with photoejection efficiency. Notably, a system capable of both generating 1O2 and photo-releasing a ligand exhibited the dominant profile of a PDT agent and induced the quiescent bioenergetic state, indicating potential implications on cellular bioenergetics for so-called dual-action agents. These findings are presented with the aim to provide the necessary groundwork for expanding the application and utility of EFA to phototherapeutics and to highlight the role of metabolic alterations in PDT.
Collapse
|
4
|
Nandre RM, Newman AH, Terse PS. In vitro safety evaluation of dopamine D3R antagonist, R-VK4-116, as a potential medication for the treatment of opioid use disorder. PLoS One 2024; 19:e0315569. [PMID: 39680602 DOI: 10.1371/journal.pone.0315569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 11/20/2024] [Indexed: 12/18/2024] Open
Abstract
R-VK4-116 is currently being developed as a medication to treat opioid use disorder (OUD). To characterize in vitro safety properties of R-VK4-116, metabolic stability in hepatocytes or liver microsomes, metabolite identification, metabolism/transporter-mediated drug interactions, lysosomal perturbation, mitochondrial toxicity, off-target enzyme effects, cellular and nuclear receptor functional assays, electrophysiological assays, CiPA, KINOMEscanTM, plasma protein binding, phospholipidosis and steatosis assays were performed. Overall, R-VK4-116 was metabolically stable in hepatocytes and microsomes. Four major metabolites were detected: two mono-oxidation products, one di-oxidation product, and one demethylated plus di-oxidation product. CYP2D6 and CYP3A4 were major contributors in R-VK4-116 metabolism. Further, R-VK4-116 did not induce/inhibit CYP enzymes. However, R-VK4-116 inhibited BCRP/P-gp, and showed antagonist effects on α1A(h), H1(h) and agonist effect on hGABAA∞1β2γ2 at 10 μM. R-VK4-116 inhibited hERG and Kir2.1 at a high concentration of 100 μM. KINOMEscanTM provided 5 hits (CHEK2, HPK1, MARK3, SRPK2 and TNK1) with Kds of >10 μM. Further, R-VK4-116 was bound to human, rat and dog plasma proteins (~83-93%). R-VK4-116 did not induce lysosome perturbation, mitochondrial toxicity, phospholipidosis, or steatosis at ≤10 μM. These results demonstrated that R-VK4-116 possesses favorable in vitro safety properties and supports further development as a potential medication for OUD.
Collapse
Affiliation(s)
- Rahul M Nandre
- Therapeutic Development Branch, Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institute of Health, Rockville, Maryland, United States of America
| | - Amy Hauck Newman
- Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse-Intramural Research Program, National Institute of Health, Baltimore, Maryland, United States of America
| | - Pramod S Terse
- Therapeutic Development Branch, Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institute of Health, Rockville, Maryland, United States of America
| |
Collapse
|
5
|
Nakamura H, Matsui T, Shinozawa T. Triclocarban induces lipid droplet accumulation and oxidative stress responses by inhibiting mitochondrial fatty acid oxidation in HepaRG cells. Toxicol Lett 2024; 396:11-18. [PMID: 38631510 DOI: 10.1016/j.toxlet.2024.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 03/05/2024] [Accepted: 04/12/2024] [Indexed: 04/19/2024]
Abstract
Mitochondrial fatty acid oxidation (mtFAO) plays an important role in hepatic energy metabolism. Severe mtFAO injury leads to nonalcoholic fatty liver disease (NAFLD) and liver failure. Several drugs have been withdrawn owing to safety issues, such as induction of fatty liver disease through mtFAO disruption. For instance, the antimicrobial triclocarban (TCC), an environmental contaminant that was removed from the market due to its unknown safety in humans, induces NAFLD in rats and promotes hepatic FAO in mice. Therefore, there are no consistent conclusions regarding the effects of TCC on FAO and lipid droplet accumulation. We hypothesized that TCC induces lipid droplet accumulation by inhibiting mtFAO in human hepatocytes. Here, we evaluated mitochondrial respiration in HepaRG cells to investigate the effects of TCC on fatty acid-driven oxidation in cells, electron transport chain parameters, lipid droplet accumulation, and antioxidant genes. The results suggest that TCC increases oxidative stress gene expression (GCLM, p62, HO-1, and NRF2) through lipid droplet accumulation via mtFAO inhibition in HepaRG cells. The results of the present study provide further insights into the effect of TCC on human NAFLD through mtFAO inhibition, and further in vivo studies could be used to validate the mechanisms.
Collapse
Affiliation(s)
- Hitoshi Nakamura
- Global Drug Safety Research and Evaluation, Research, Takeda Pharmaceutical Company Limited
| | - Toshikatsu Matsui
- Global Drug Safety Research and Evaluation, Research, Takeda Pharmaceutical Company Limited
| | - Tadahiro Shinozawa
- Global Drug Safety Research and Evaluation, Research, Takeda Pharmaceutical Company Limited.
| |
Collapse
|
6
|
Pinto M, Silva TB, Sardão VA, Simões R, Albuquerque B, Oliveira PJ, Valente MJ, Remião F, Soares-da-Silva P, Fernandes C, Borges F. Cellular and Mitochondrial Toxicity of Tolcapone, Entacapone, and New Nitrocatechol Derivatives. ACS Pharmacol Transl Sci 2024; 7:1637-1649. [PMID: 38751615 PMCID: PMC11091965 DOI: 10.1021/acsptsci.4c00124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 04/09/2024] [Accepted: 04/11/2024] [Indexed: 05/18/2024]
Abstract
Nitrocatechols are the standard pharmacophore to develop potent tight-binding inhibitors of catechol O-methyltransferase (COMT), which can be used as coadjuvant drugs to manage Parkinson's disease. Tolcapone is the most potent drug of this class, but it has raised safety concerns due to its potential to induce liver damage. Tolcapone-induced hepatotoxicity has been attributed to the nitrocatechol moiety; however, other nitrocatechol-based COMT inhibitors, such as entacapone, are safe and do not damage the liver. There is a knowledge gap concerning which mechanisms and chemical properties govern the toxicity of nitrocatechol-based COMT inhibitors. Using a vast array of cell-based assays, we found that tolcapone-induced toxicity is caused by direct interference with mitochondria that does not depend on bioactivation by P450. Our findings also suggest that (a) lipophilicity is a key property in the toxic potential of nitrocatechols; (b) the presence of a carbonyl group directly attached to the nitrocatechol ring seems to increase the reactivity of the molecule, and (c) the presence of cyano moiety in double bond stabilizes the reactivity decreasing the cytotoxicity. Altogether, the fine balance between lipophilicity and the chemical nature of the C1 substituents of the nitrocatechol ring may explain the difference in the toxicological behavior observed between tolcapone and entacapone.
Collapse
Affiliation(s)
- Miguel Pinto
- CIQUP-IMS
− Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, R. Campo Alegre s/n, Porto 4169-007, Portugal
- Associate
Laboratory i4HB − Institute for Health and Bioeconomy, Faculty
of Pharmacy, University of Porto, R. Jorge de Viterbo Ferreira 228, Porto 4050-313, Portugal
- UCIBIO
− Applied Molecular Biosciences Unit, REQUIMTE. Laboratory
of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, R. Jorge de Viterbo Ferreira 228, Porto 4050-313, Portugal
| | - Tiago Barros Silva
- CIQUP-IMS
− Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, R. Campo Alegre s/n, Porto 4169-007, Portugal
- Associate
Laboratory i4HB − Institute for Health and Bioeconomy, Faculty
of Pharmacy, University of Porto, R. Jorge de Viterbo Ferreira 228, Porto 4050-313, Portugal
- UCIBIO
− Applied Molecular Biosciences Unit, REQUIMTE. Laboratory
of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, R. Jorge de Viterbo Ferreira 228, Porto 4050-313, Portugal
| | - Vilma A. Sardão
- CNC-UC
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra 3004−504, Portugal
- MIA-Portugal
- Multidisciplinary Institute of Aging, University of Coimbra, Coimbra 3004−504, Portugal
| | - Rui Simões
- CNC-UC
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra 3004−504, Portugal
- CIBB
- Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra 3004−504, Portugal
| | - Bárbara Albuquerque
- CIQUP-IMS
− Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, R. Campo Alegre s/n, Porto 4169-007, Portugal
- MedInUP -
Center for Drug Discovery and Innovative Medicines, University of Porto, Porto 4200-319, Portugal
| | - Paulo J. Oliveira
- CNC-UC
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra 3004−504, Portugal
- CIBB
- Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra 3004−504, Portugal
| | - Maria João Valente
- Associate
Laboratory i4HB − Institute for Health and Bioeconomy, Faculty
of Pharmacy, University of Porto, R. Jorge de Viterbo Ferreira 228, Porto 4050-313, Portugal
- UCIBIO
− Applied Molecular Biosciences Unit, REQUIMTE. Laboratory
of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, R. Jorge de Viterbo Ferreira 228, Porto 4050-313, Portugal
| | - Fernando Remião
- Associate
Laboratory i4HB − Institute for Health and Bioeconomy, Faculty
of Pharmacy, University of Porto, R. Jorge de Viterbo Ferreira 228, Porto 4050-313, Portugal
- UCIBIO
− Applied Molecular Biosciences Unit, REQUIMTE. Laboratory
of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, R. Jorge de Viterbo Ferreira 228, Porto 4050-313, Portugal
| | - Patrício Soares-da-Silva
- MedInUP -
Center for Drug Discovery and Innovative Medicines, University of Porto, Porto 4200-319, Portugal
| | - Carlos Fernandes
- CIQUP-IMS
− Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, R. Campo Alegre s/n, Porto 4169-007, Portugal
| | - Fernanda Borges
- CIQUP-IMS
− Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, R. Campo Alegre s/n, Porto 4169-007, Portugal
| |
Collapse
|
7
|
Granberg KL, Sakamaki S, Fuchigami R, Niwa Y, Fujio M, Kato H, Bergström F, Larsson N, Persson M, Villar IC, Fujita T, Sugikawa E, Althage M, Yano N, Yokoyama Y, Kimura J, Lal M, Mochida H. Identification of Novel Series of Potent and Selective Relaxin Family Peptide Receptor 1 (RXFP1) Agonists. J Med Chem 2024; 67:4442-4462. [PMID: 38502780 DOI: 10.1021/acs.jmedchem.3c02183] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/21/2024]
Abstract
Relaxin H2 is a clinically relevant peptide agonist for relaxin family peptide receptor 1 (RXFP1), but a combination of this hormone's short plasma half-life and the need for injectable delivery limits its therapeutic potential. We sought to overcome these limitations through the development of a potent small molecule (SM) RXFP1 agonist. Although two large SM HTS campaigns failed in identifying suitable hit series, we uncovered novel chemical space starting from the only known SM RXFP1 agonist series, represented by ML290. Following a design-make-test-analyze strategy based on improving early dose to man ranking, we discovered compound 42 (AZ7976), a highly selective RXFP1 agonist with sub-nanomolar potency. We used AZ7976, its 10 000-fold less potent enantiomer 43 and recombinant relaxin H2 to evaluate in vivo pharmacology and demonstrate that AZ7976-mediated heart rate increase in rats was a result of RXFP1 agonism. As a result, AZ7976 was selected as lead for continued optimization.
Collapse
Affiliation(s)
- Kenneth L Granberg
- Medicinal Chemistry, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Mölndal 43183, Sweden
| | - Shigeki Sakamaki
- Sohyaku. Innovative Research Division, Mitsubishi Tanabe Pharma Corporation, 1000 Kamoshida-cho, Aoba-ku, Yokohama, Kanagawa 227-0033, Japan
| | - Ryuichi Fuchigami
- Sohyaku. Innovative Research Division, Mitsubishi Tanabe Pharma Corporation, 1000 Kamoshida-cho, Aoba-ku, Yokohama, Kanagawa 227-0033, Japan
| | - Yasuki Niwa
- Sohyaku. Innovative Research Division, Mitsubishi Tanabe Pharma Corporation, 1000 Kamoshida-cho, Aoba-ku, Yokohama, Kanagawa 227-0033, Japan
| | - Masakazu Fujio
- Sohyaku. Innovative Research Division, Mitsubishi Tanabe Pharma Corporation, 1000 Kamoshida-cho, Aoba-ku, Yokohama, Kanagawa 227-0033, Japan
| | - Harutoshi Kato
- Sohyaku. Innovative Research Division, Mitsubishi Tanabe Pharma Corporation, 1000 Kamoshida-cho, Aoba-ku, Yokohama, Kanagawa 227-0033, Japan
| | - Fredrik Bergström
- Drug Metabolism and Pharmacokinetics (DMPK), Cardiovascular, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Mölndal 431 83, Sweden
| | - Niklas Larsson
- Discovery Biology, Discovery Sciences, R&D, AstraZeneca, Gothenburg, Mölndal 431 83, Sweden
| | - Mikael Persson
- Cardiovascular, Renal and Metabolism Safety, Clinical Pharmacology & Safety Sciences, R&D, AstraZeneca, Gothenburg, Mölndal 431 83, Sweden
| | - Inmaculada C Villar
- Regulatory Toxicology & Safety Pharmacology, Clinical Pharmacology & Safety Sciences, R&D, AstraZeneca, Cambridge CB2 0AA, U.K
| | - Takuya Fujita
- Sohyaku. Innovative Research Division, Mitsubishi Tanabe Pharma Corporation, 1000 Kamoshida-cho, Aoba-ku, Yokohama, Kanagawa 227-0033, Japan
| | - Emiko Sugikawa
- Sohyaku. Innovative Research Division, Mitsubishi Tanabe Pharma Corporation, 1000 Kamoshida-cho, Aoba-ku, Yokohama, Kanagawa 227-0033, Japan
| | - Magnus Althage
- Translational Science and Experimental Medicine, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), Biopharmaceuticals R&D, AstraZeneca, Gothenburg, Mölndal 431 83, Sweden
| | - Naoko Yano
- Sohyaku. Innovative Research Division, Mitsubishi Tanabe Pharma Corporation, 1000 Kamoshida-cho, Aoba-ku, Yokohama, Kanagawa 227-0033, Japan
| | - Yoshito Yokoyama
- Sohyaku. Innovative Research Division, Mitsubishi Tanabe Pharma Corporation, 1000 Kamoshida-cho, Aoba-ku, Yokohama, Kanagawa 227-0033, Japan
| | - Junpei Kimura
- Sohyaku. Innovative Research Division, Mitsubishi Tanabe Pharma Corporation, 1000 Kamoshida-cho, Aoba-ku, Yokohama, Kanagawa 227-0033, Japan
| | - Mark Lal
- Bioscience Renal, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), Biopharmaceuticals R&D, AstraZeneca, Gothenburg, Mölndal 431 83, Sweden
| | - Hideki Mochida
- Sohyaku. Innovative Research Division, Mitsubishi Tanabe Pharma Corporation, 1000 Kamoshida-cho, Aoba-ku, Yokohama, Kanagawa 227-0033, Japan
| |
Collapse
|
8
|
Braillard S, Keenan M, Breese KJ, Heppell J, Abbott M, Islam R, Shackleford DM, Katneni K, Crighton E, Chen G, Patil R, Lee G, White KL, Carvalho S, Wall RJ, Chemi G, Zuccotto F, González S, Marco M, Deakyne J, Standing D, Brunori G, Lyon JJ, Castañeda Casado P, Camino I, Martinez MSM, Zulfiqar B, Avery VM, Feijens PB, Van Pelt N, Matheeussen A, Hendrickx S, Maes L, Caljon G, Yardley V, Wyllie S, Charman SA, Chatelain E. DNDI-6174 is a preclinical candidate for visceral leishmaniasis that targets the cytochrome bc 1. Sci Transl Med 2023; 15:eadh9902. [PMID: 38091406 PMCID: PMC7615677 DOI: 10.1126/scitranslmed.adh9902] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 08/12/2023] [Indexed: 12/18/2023]
Abstract
New drugs for visceral leishmaniasis that are safe, low cost, and adapted to the field are urgently required. Despite concerted efforts over the last several years, the number of new chemical entities that are suitable for clinical development for the treatment of Leishmania remains low. Here, we describe the discovery and preclinical development of DNDI-6174, an inhibitor of Leishmania cytochrome bc1 complex activity that originated from a phenotypically identified pyrrolopyrimidine series. This compound fulfills all target candidate profile criteria required for progression into preclinical development. In addition to good metabolic stability and pharmacokinetic properties, DNDI-6174 demonstrates potent in vitro activity against a variety of Leishmania species and can reduce parasite burden in animal models of infection, with the potential to approach sterile cure. No major flags were identified in preliminary safety studies, including an exploratory 14-day toxicology study in the rat. DNDI-6174 is a cytochrome bc1 complex inhibitor with acceptable development properties to enter preclinical development for visceral leishmaniasis.
Collapse
Affiliation(s)
- Stéphanie Braillard
- Drugs for Neglected Diseases initiative (DNDi), Chemin Camille-Vidart 15, 1202 Geneva, Switzerland
| | | | | | - Jacob Heppell
- Epichem Pty Ltd, Perth, Western Australia, Australia
| | | | - Rafiqul Islam
- Epichem Pty Ltd, Perth, Western Australia, Australia
| | - David M. Shackleford
- Centre for Drug Candidate Optimisation, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville 3052, Australia
| | - Kasiram Katneni
- Centre for Drug Candidate Optimisation, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville 3052, Australia
| | - Elly Crighton
- Centre for Drug Candidate Optimisation, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville 3052, Australia
| | - Gong Chen
- Centre for Drug Candidate Optimisation, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville 3052, Australia
| | - Rahul Patil
- Centre for Drug Candidate Optimisation, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville 3052, Australia
| | - Given Lee
- Centre for Drug Candidate Optimisation, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville 3052, Australia
| | - Karen L. White
- Centre for Drug Candidate Optimisation, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville 3052, Australia
| | - Sandra Carvalho
- Wellcome Centre for Anti-infectives Research, School of Life Sciences, University of Dundee, Dow Street, Dundee DD1 5EH, United Kingdom
| | - Richard J. Wall
- Wellcome Centre for Anti-infectives Research, School of Life Sciences, University of Dundee, Dow Street, Dundee DD1 5EH, United Kingdom
| | - Giulia Chemi
- Drug Discovery Unit, Wellcome Centre for Anti-infectives Research, School of Life Sciences, University of Dundee, Dow Street, Dundee DD1 5EH, United Kingdom
| | - Fabio Zuccotto
- Drug Discovery Unit, Wellcome Centre for Anti-infectives Research, School of Life Sciences, University of Dundee, Dow Street, Dundee DD1 5EH, United Kingdom
| | - Silvia González
- Global Health Medicines R&D, GlaxoSmithKline, Tres Cantos, Madrid 28760, Spain
| | - Maria Marco
- Global Health Medicines R&D, GlaxoSmithKline, Tres Cantos, Madrid 28760, Spain
| | | | | | - Gino Brunori
- Global Investigative Safety, GSK, Ware, United Kingdom
| | | | | | | | | | - Bilal Zulfiqar
- Discovery Biology, Griffith University, Nathan, Queensland, Australia 4111
| | - Vicky M. Avery
- Discovery Biology, Griffith University, Nathan, Queensland, Australia 4111
| | - Pim-Bart Feijens
- Laboratory of Microbiology, Parasitology and Hygiene (LMPH), University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium
| | - Natascha Van Pelt
- Laboratory of Microbiology, Parasitology and Hygiene (LMPH), University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium
| | - An Matheeussen
- Laboratory of Microbiology, Parasitology and Hygiene (LMPH), University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium
| | - Sarah Hendrickx
- Laboratory of Microbiology, Parasitology and Hygiene (LMPH), University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium
| | - Louis Maes
- Laboratory of Microbiology, Parasitology and Hygiene (LMPH), University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium
| | - Guy Caljon
- Laboratory of Microbiology, Parasitology and Hygiene (LMPH), University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium
| | - Vanessa Yardley
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, Keppel Street, London WC1E 7HT, United Kingdom
| | - Susan Wyllie
- Wellcome Centre for Anti-infectives Research, School of Life Sciences, University of Dundee, Dow Street, Dundee DD1 5EH, United Kingdom
| | - Susan A. Charman
- Centre for Drug Candidate Optimisation, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville 3052, Australia
| | - Eric Chatelain
- Drugs for Neglected Diseases initiative (DNDi), Chemin Camille-Vidart 15, 1202 Geneva, Switzerland
| |
Collapse
|
9
|
Rosell-Hidalgo A, Eakins J, Walker P, Moore AL, Ghafourian T. Risk Assessment of Psychotropic Drugs on Mitochondrial Function Using In Vitro Assays. Biomedicines 2023; 11:3272. [PMID: 38137493 PMCID: PMC10741027 DOI: 10.3390/biomedicines11123272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Revised: 11/30/2023] [Accepted: 12/04/2023] [Indexed: 12/24/2023] Open
Abstract
Mitochondria are potential targets responsible for some drug- and xenobiotic-induced organ toxicities. However, molecular mechanisms of drug-induced mitochondrial toxicities are mostly unknown. Here, multiple in vitro assays were used to investigate the effects of 22 psychotropic drugs on mitochondrial function. The acute extracellular flux assay identified inhibitors of the electron transport chain (ETC), i.e., aripiprazole, phenytoin, and fluoxetine, an uncoupler (reserpine), substrate inhibitors (quetiapine, carbamazepine, buspirone, and tianeptine), and cytotoxic compounds (chlorpromazine and valproic acid) in HepG2 cells. Using permeabilized HepG2 cells revealed minimum effective concentrations of 66.3, 6730, 44.5, and 72.1 µM for the inhibition of complex-I-linked respiration for quetiapine, valproic acid, buspirone, and fluoxetine, respectively. Assessing complex-II-linked respiration in isolated rat liver mitochondria revealed haloperidol is an ETC inhibitor, chlorpromazine is an uncoupler in basal respiration and an ETC inhibitor under uncoupled respiration (IC50 = 135 µM), while olanzapine causes a mild dissipation of the membrane potential at 50 µM. This research elucidates some mechanisms of drug toxicity and provides some insight into their safety profile for clinical drug decisions.
Collapse
Affiliation(s)
- Alicia Rosell-Hidalgo
- Cyprotex Discovery Ltd., No. 24 Mereside, Alderley Park, Macclesfield, Cheshire SK10 4TG, UK; (A.R.-H.); (J.E.)
| | - Julie Eakins
- Cyprotex Discovery Ltd., No. 24 Mereside, Alderley Park, Macclesfield, Cheshire SK10 4TG, UK; (A.R.-H.); (J.E.)
| | - Paul Walker
- Cyprotex Discovery Ltd., No. 24 Mereside, Alderley Park, Macclesfield, Cheshire SK10 4TG, UK; (A.R.-H.); (J.E.)
| | - Anthony L. Moore
- Department of Biochemistry and Biomedicine, School of Life Sciences, University of Sussex, Falmer, Brighton BN1 9QG, UK;
| | - Taravat Ghafourian
- Department of Pharmaceutical Sciences, Barry & Judy Silverman College of Pharmacy, Nova Southeastern University, 3200 South University Drive, Ft. Lauderdale, FL 33328-2018, USA
| |
Collapse
|
10
|
Lee YT, Tan YJ, Oon CE. BZD9L1 Differentially Regulates Sirtuins in Liver-Derived Cells by Inducing Reactive Oxygen Species. Biomedicines 2023; 11:3059. [PMID: 38002059 PMCID: PMC10669747 DOI: 10.3390/biomedicines11113059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 11/04/2023] [Accepted: 11/06/2023] [Indexed: 11/26/2023] Open
Abstract
Growing evidence has highlighted that mitochondrial dysfunction contributes to drug-induced toxicities and leads to drug attrition and post-market withdrawals. The acetylation or deacetylation of mitochondrial proteins can affect mitochondrial functions as the cells adapt to various cellular stresses and other metabolic challenges. SIRTs act as critical deacetylases in modulating mitochondrial function in response to drug toxicity, oxidative stress, reactive oxygen species (ROS), and energy metabolism. We previously showed that a recently characterised SIRT inhibitor (BZD9L1) is non-toxic in rodents in a short-term toxicity evaluation. However, the impact of BZD9L1 on mitochondrial function is unknown. This work aims to determine the effects of BZD9L1 on mitochondrial function in human normal liver and kidney-derived cell lines using the Agilent Seahorse Cell Mito Stress Test to complement our short-term toxicity evaluations in vivo. The Mito Stress assay revealed that BZD9L1 could potentially trigger oxidative stress by inducing ROS, which promotes proton leak and reduces coupling efficiency in liver-derived THLE cells. However, the same was not observed in human kidney-derived HEK293 cells. Interestingly, BZD9L1 had no impact on SIRT3 protein expression in both cell lines but affected SOD2 and its acetylated form at 72 h in THLE cells, indicating that BZD9L1 exerted its effect through SIRT3 activity rather than protein expression. In contrast, BZD9L1 reduced SIRT1 protein expression and impacted the p53 protein differently in both cell lines. Although BZD9L1 did not affect the spare respiratory capacity in vitro, these findings call for further validation of mitochondrial function through assessment of other mitochondrial parameters to evaluate the safety of BZD9L1.
Collapse
Affiliation(s)
| | | | - Chern Ein Oon
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, Gelugor 11800, Penang, Malaysia; (Y.T.L.); (Y.J.T.)
| |
Collapse
|
11
|
Danzi F, Pacchiana R, Mafficini A, Scupoli MT, Scarpa A, Donadelli M, Fiore A. To metabolomics and beyond: a technological portfolio to investigate cancer metabolism. Signal Transduct Target Ther 2023; 8:137. [PMID: 36949046 PMCID: PMC10033890 DOI: 10.1038/s41392-023-01380-0] [Citation(s) in RCA: 79] [Impact Index Per Article: 39.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 02/08/2023] [Accepted: 02/15/2023] [Indexed: 03/24/2023] Open
Abstract
Tumour cells have exquisite flexibility in reprogramming their metabolism in order to support tumour initiation, progression, metastasis and resistance to therapies. These reprogrammed activities include a complete rewiring of the bioenergetic, biosynthetic and redox status to sustain the increased energetic demand of the cells. Over the last decades, the cancer metabolism field has seen an explosion of new biochemical technologies giving more tools than ever before to navigate this complexity. Within a cell or a tissue, the metabolites constitute the direct signature of the molecular phenotype and thus their profiling has concrete clinical applications in oncology. Metabolomics and fluxomics, are key technological approaches that mainly revolutionized the field enabling researchers to have both a qualitative and mechanistic model of the biochemical activities in cancer. Furthermore, the upgrade from bulk to single-cell analysis technologies provided unprecedented opportunity to investigate cancer biology at cellular resolution allowing an in depth quantitative analysis of complex and heterogenous diseases. More recently, the advent of functional genomic screening allowed the identification of molecular pathways, cellular processes, biomarkers and novel therapeutic targets that in concert with other technologies allow patient stratification and identification of new treatment regimens. This review is intended to be a guide for researchers to cancer metabolism, highlighting current and emerging technologies, emphasizing advantages, disadvantages and applications with the potential of leading the development of innovative anti-cancer therapies.
Collapse
Affiliation(s)
- Federica Danzi
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Biochemistry, University of Verona, Verona, Italy
| | - Raffaella Pacchiana
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Biochemistry, University of Verona, Verona, Italy
| | - Andrea Mafficini
- Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | - Maria T Scupoli
- Department of Neurosciences, Biomedicine and Movement Sciences, Biology and Genetics Section, University of Verona, Verona, Italy
| | - Aldo Scarpa
- Department of Diagnostics and Public Health, University of Verona, Verona, Italy
- ARC-NET Research Centre, University and Hospital Trust of Verona, Verona, Italy
| | - Massimo Donadelli
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Biochemistry, University of Verona, Verona, Italy.
| | - Alessandra Fiore
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Biochemistry, University of Verona, Verona, Italy
| |
Collapse
|
12
|
Roth Z'graggen B, Urner M, Beck-Schimmer B, Schläpfer M. Effects of sevoflurane and its metabolite hexafluoroisopropanol on hypoxia/reoxygenation-induced injury and mitochondrial bioenergetics in murine cardiomyocytes. BJA OPEN 2023; 5:100116. [PMID: 37587996 PMCID: PMC10430838 DOI: 10.1016/j.bjao.2022.100116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 11/22/2022] [Indexed: 08/18/2023]
Abstract
Background The volatile anaesthetic sevoflurane protects cardiac tissue from reoxygenation/reperfusion. Mitochondria play an essential role in conditioning. We aimed to investigate how sevoflurane and its primary metabolite hexafluoroisopropanol (HFIP) affect necrosis, apoptosis, and reactive oxygen species formation in cardiomyocytes upon hypoxia/reoxygenation injury. Moreover, we aimed to describe the similarities in the mode of action in a mitochondrial bioenergetics analysis. Methods Murine cardiomyocytes were exposed to hypoxia (0.2% O2 for 6 h), followed by reoxygenation (air with 5% CO2 for 2 h) in the presence or absence sevoflurane 2.2% or HFIP 4 mM. Lactate dehydrogenase (LDH) release (necrosis), caspase activation (apoptosis), reactive oxygen species, mitochondrial membrane potential, and mitochondrial function (Seahorse XF analyser) were measured. Results Hypoxia/reoxygenation increased cell death by 44% (+31 to +55%, P<0.001). Reoxygenation in the presence of sevoflurane 2.2% or HFIP 4 mM increased LDH release only by +18% (+6 to +30%) and 20% (+7 to +32%), respectively. Apoptosis and reactive oxygen species formation were attenuated by sevoflurane and HFIP. Mitochondrial bioenergetics analysis of the two substances was profoundly different. Sevoflurane did not influence oxygen consumption rate (OCR) or extracellular acidification rate (ECAR), whereas HFIP reduced OCR and increased ECAR, an effect similar to oligomycin, an adenosine triphosphate (ATP) synthase inhibitor. When blocking the metabolism of sevoflurane into HFIP, protective effects of sevoflurane - but not of HFIP - on LDH release and caspase were mitigated. Conclusion Together, our data suggest that sevoflurane metabolism into HFIP plays an essential role in cardiomyocyte postconditioning after hypoxia/reoxygenation injury.
Collapse
Affiliation(s)
| | - Martin Urner
- Institute of Physiology, University of Zurich, Zurich, Switzerland
- Interdepartmental Division of Critical Care Medicine and University of Toronto, Toronto, Canada
- Institute of Health Policy, Management, and Evaluation, University of Toronto, Toronto, Canada
| | - Beatrice Beck-Schimmer
- Institute of Physiology, University of Zurich, Zurich, Switzerland
- Institute of Anaesthesiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Martin Schläpfer
- Institute of Physiology, University of Zurich, Zurich, Switzerland
- Institute of Anaesthesiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| |
Collapse
|
13
|
Kim CW, Lee HJ, Ahn D, Go RE, Choi KC. Establishment of a platform for measuring mitochondrial oxygen consumption rate for cardiac mitochondrial toxicity. Toxicol Res 2022; 38:511-522. [PMID: 36277363 PMCID: PMC9532483 DOI: 10.1007/s43188-022-00136-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 04/12/2022] [Accepted: 04/14/2022] [Indexed: 10/18/2022] Open
Abstract
The heart has an abundance of mitochondria since cardiac muscles require copious amounts of energy for providing continuous blood through the circulatory system, thereby implying that myocardial function is largely reliant on mitochondrial energy. Thus, cardiomyocytes are susceptible to mitochondrial dysfunction and are likely targets of mitochondrial toxic drugs. Various methods have been developed to evaluate mitochondrial toxicity by evaluating toxicological mechanisms, but an optimized and standardized assay for cardiomyocytes remains unmet. We have therefore attempted to standardize the evaluation system for determining cardiac mitochondrial toxicity, using AC16 human and H9C2 rat cardiomyocytes. Three clinically administered drugs (acetaminophen, amiodarone, and valproic acid) and two anticancer drugs (doxorubicin and tamoxifen) which are reported to have mitochondrial effects, were applied in this study. The oxygen consumption rate (OCR), which directly reflects mitochondrial function, and changes in mRNA levels of mitochondrial respiratory complex I to complex V, were analyzed. Our results reveal that exposure to all five drugs results in a concentration-dependent decrease in the basal and maximal levels of OCR in AC16 cells and H9C2 cells. In particular, marked reduction in the OCR was observed after treatment with doxorubicin. The reduction in OCR after exposure to mitochondrial toxic drugs was found to be associated with reduced mRNA expression in the mitochondrial respiratory complexes, suggesting that the cardiac mitochondrial toxicity of drugs is majorly due to dysfunction of mitochondrial respiration. Based on the results of this study, we established and standardized a protocol to measure OCR in cardiomyocytes. We expect that this standardized evaluation system for mitochondrial toxicity can be applied as basic data for establishing a screening platform to evaluate cardiac mitochondrial toxicity of drugs, during the developmental stage of new drug discovery.
Collapse
Affiliation(s)
- Cho-Won Kim
- Laboratory of Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk 28644 Republic of Korea
| | - Hee-Jin Lee
- Laboratory of Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk 28644 Republic of Korea
| | - Dohee Ahn
- Laboratory of Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk 28644 Republic of Korea
| | - Ryeo-Eun Go
- Laboratory of Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk 28644 Republic of Korea
| | - Kyung-Chul Choi
- Laboratory of Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk 28644 Republic of Korea
| |
Collapse
|
14
|
Pinho SA, Costa CF, Deus CM, Pinho SLC, Miranda‐Santos I, Afonso G, Bagshaw O, Stuart JA, Oliveira PJ, Cunha‐Oliveira T. Mitochondrial and metabolic remodelling in human skin fibroblasts in response to glucose availability. FEBS J 2022; 289:5198-5217. [DOI: 10.1111/febs.16413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 02/01/2022] [Accepted: 02/24/2022] [Indexed: 11/29/2022]
Affiliation(s)
- Sónia A. Pinho
- CNC ‐ Center for Neuroscience and Cell Biology CIBB ‐ Centre for Innovative Biomedicine and Biotechnology University of Coimbra Portugal
- IIIUC ‐ Institute for Interdisciplinary Research University of Coimbra Portugal
- PhD Programme in Experimental Biology and Biomedicine (PDBEB) Institute for Interdisciplinary Research (IIIUC) University of Coimbra Portugal
| | - Cláudio F. Costa
- CNC ‐ Center for Neuroscience and Cell Biology CIBB ‐ Centre for Innovative Biomedicine and Biotechnology University of Coimbra Portugal
- IIIUC ‐ Institute for Interdisciplinary Research University of Coimbra Portugal
| | - Cláudia M. Deus
- CNC ‐ Center for Neuroscience and Cell Biology CIBB ‐ Centre for Innovative Biomedicine and Biotechnology University of Coimbra Portugal
- IIIUC ‐ Institute for Interdisciplinary Research University of Coimbra Portugal
| | - Sonia L. C. Pinho
- CNC ‐ Center for Neuroscience and Cell Biology CIBB ‐ Centre for Innovative Biomedicine and Biotechnology University of Coimbra Portugal
- IIIUC ‐ Institute for Interdisciplinary Research University of Coimbra Portugal
- CIVG‐ Vasco da Gama Research Center Vasco da Gama University School Portugal
| | - Inês Miranda‐Santos
- CNC ‐ Center for Neuroscience and Cell Biology CIBB ‐ Centre for Innovative Biomedicine and Biotechnology University of Coimbra Portugal
- IIIUC ‐ Institute for Interdisciplinary Research University of Coimbra Portugal
| | - Gonçalo Afonso
- CNC ‐ Center for Neuroscience and Cell Biology CIBB ‐ Centre for Innovative Biomedicine and Biotechnology University of Coimbra Portugal
- IIIUC ‐ Institute for Interdisciplinary Research University of Coimbra Portugal
| | - Olivia Bagshaw
- Department of Biological Sciences Brock University St. Catharines ON Canada
| | - Jeffrey A. Stuart
- Department of Biological Sciences Brock University St. Catharines ON Canada
| | - Paulo J. Oliveira
- CNC ‐ Center for Neuroscience and Cell Biology CIBB ‐ Centre for Innovative Biomedicine and Biotechnology University of Coimbra Portugal
- IIIUC ‐ Institute for Interdisciplinary Research University of Coimbra Portugal
| | - Teresa Cunha‐Oliveira
- CNC ‐ Center for Neuroscience and Cell Biology CIBB ‐ Centre for Innovative Biomedicine and Biotechnology University of Coimbra Portugal
- IIIUC ‐ Institute for Interdisciplinary Research University of Coimbra Portugal
| |
Collapse
|
15
|
Seal S, Carreras-Puigvert J, Trapotsi MA, Yang H, Spjuth O, Bender A. Integrating cell morphology with gene expression and chemical structure to aid mitochondrial toxicity detection. Commun Biol 2022; 5:858. [PMID: 35999457 PMCID: PMC9399120 DOI: 10.1038/s42003-022-03763-5] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 07/25/2022] [Indexed: 12/05/2022] Open
Abstract
Mitochondrial toxicity is an important safety endpoint in drug discovery. Models based solely on chemical structure for predicting mitochondrial toxicity are currently limited in accuracy and applicability domain to the chemical space of the training compounds. In this work, we aimed to utilize both -omics and chemical data to push beyond the state-of-the-art. We combined Cell Painting and Gene Expression data with chemical structural information from Morgan fingerprints for 382 chemical perturbants tested in the Tox21 mitochondrial membrane depolarization assay. We observed that mitochondrial toxicants differ from non-toxic compounds in morphological space and identified compound clusters having similar mechanisms of mitochondrial toxicity, thereby indicating that morphological space provides biological insights related to mechanisms of action of this endpoint. We further showed that models combining Cell Painting, Gene Expression features and Morgan fingerprints improved model performance on an external test set of 244 compounds by 60% (in terms of F1 score) and improved extrapolation to new chemical space. The performance of our combined models was comparable with dedicated in vitro assays for mitochondrial toxicity. Our results suggest that combining chemical descriptors with biological readouts enhances the detection of mitochondrial toxicants, with practical implications in drug discovery.
Collapse
Affiliation(s)
- Srijit Seal
- Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Rd, Cambridge, CB2 1EW, UK
| | - Jordi Carreras-Puigvert
- Department of Pharmaceutical Biosciences and Science for Life Laboratory, Uppsala University, Box 591, SE-75124, Uppsala, Sweden
| | - Maria-Anna Trapotsi
- Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Rd, Cambridge, CB2 1EW, UK
| | - Hongbin Yang
- Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Rd, Cambridge, CB2 1EW, UK
| | - Ola Spjuth
- Department of Pharmaceutical Biosciences and Science for Life Laboratory, Uppsala University, Box 591, SE-75124, Uppsala, Sweden.
| | - Andreas Bender
- Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Rd, Cambridge, CB2 1EW, UK.
| |
Collapse
|
16
|
Jin P, Jiang J, Zhou L, Huang Z, Nice EC, Huang C, Fu L. Mitochondrial adaptation in cancer drug resistance: prevalence, mechanisms, and management. J Hematol Oncol 2022; 15:97. [PMID: 35851420 PMCID: PMC9290242 DOI: 10.1186/s13045-022-01313-4] [Citation(s) in RCA: 104] [Impact Index Per Article: 34.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 06/29/2022] [Indexed: 02/08/2023] Open
Abstract
Drug resistance represents a major obstacle in cancer management, and the mechanisms underlying stress adaptation of cancer cells in response to therapy-induced hostile environment are largely unknown. As the central organelle for cellular energy supply, mitochondria can rapidly undergo dynamic changes and integrate cellular signaling pathways to provide bioenergetic and biosynthetic flexibility for cancer cells, which contributes to multiple aspects of tumor characteristics, including drug resistance. Therefore, targeting mitochondria for cancer therapy and overcoming drug resistance has attracted increasing attention for various types of cancer. Multiple mitochondrial adaptation processes, including mitochondrial dynamics, mitochondrial metabolism, and mitochondrial apoptotic regulatory machinery, have been demonstrated to be potential targets. However, recent increasing insights into mitochondria have revealed the complexity of mitochondrial structure and functions, the elusive functions of mitochondria in tumor biology, and the targeting inaccessibility of mitochondria, which have posed challenges for the clinical application of mitochondrial-based cancer therapeutic strategies. Therefore, discovery of both novel mitochondria-targeting agents and innovative mitochondria-targeting approaches is urgently required. Here, we review the most recent literature to summarize the molecular mechanisms underlying mitochondrial stress adaptation and their intricate connection with cancer drug resistance. In addition, an overview of the emerging strategies to target mitochondria for effectively overcoming chemoresistance is highlighted, with an emphasis on drug repositioning and mitochondrial drug delivery approaches, which may accelerate the application of mitochondria-targeting compounds for cancer therapy.
Collapse
Affiliation(s)
- Ping Jin
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, People's Republic of China
| | - Jingwen Jiang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, People's Republic of China
| | - Li Zhou
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, People's Republic of China
| | - Zhao Huang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, People's Republic of China
| | - Edouard C Nice
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, 3800, Australia
| | - Canhua Huang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, People's Republic of China.
| | - Li Fu
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Pharmacology and International Cancer Center, Shenzhen University Health Science Center, Shenzhen, 518060, Guangdong, People's Republic of China.
| |
Collapse
|
17
|
Biale G, La Nasa J, Mattonai M, Corti A, Castelvetro V, Modugno F. Seeping plastics: Potentially harmful molecular fragments leaching out from microplastics during accelerated ageing in seawater. WATER RESEARCH 2022; 219:118521. [PMID: 35526427 DOI: 10.1016/j.watres.2022.118521] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 04/21/2022] [Accepted: 04/26/2022] [Indexed: 06/14/2023]
Abstract
Microplastics are the particulate plastic debris found almost everywhere as environmental contaminants. They are not chemically stable persistent pollutants, but reactive materials. In fact, synthetic polymers exposed to the environment undergo chemical and physical degradation processes which lead not only to mechanical but also molecular fragmentation, releasing compounds that are potentially harmful for the environment and human health. We carried out accelerated photo-oxidative ageing of four reference microplastics (low- and high-density polyethylene, polypropylene, and polystyrene) directly in artificial seawater. We then made a characterization at the molecular level along with a quantification of the chemical species leached into water. Gas chromatography/mass spectrometry analyses performed after selective extraction and derivatization enabled us to identify more than 60 different compounds. Analysis of the leachates from the three polyolefins revealed that the main degradation products were mono- and dicarboxylic acids, along with linear and branched hydroxy acids. The highest amount of leached degradation species was observed for polystyrene, with benzoic acid and phenol derivatives as the most abundant, along with oligomeric styrene derivatives. The results from reference microplastics were then compared with those obtained by analyzing leachates in artificial seawater from aged plastic debris collected in a natural environment. The differences observed between the reference and the environmental plastic leachates mainly concerned the relative abundances of the chemical species detected, with the environmental samples showing higher amounts of dicarboxylic acids and oxidized species.
Collapse
Affiliation(s)
- Greta Biale
- Department of Chemistry and Industrial Chemistry, University of Pisa, Pisa, Italy
| | - Jacopo La Nasa
- Department of Chemistry and Industrial Chemistry, University of Pisa, Pisa, Italy; National Interuniversity Consortium of Materials Science and Technology (INSTM), Florence, Italy.
| | - Marco Mattonai
- Department of Chemistry and Industrial Chemistry, University of Pisa, Pisa, Italy
| | - Andrea Corti
- Department of Chemistry and Industrial Chemistry, University of Pisa, Pisa, Italy; CISUP Center for the Integration of Scientific Instruments of the University of Pisa, Pisa, Italy
| | - Valter Castelvetro
- Department of Chemistry and Industrial Chemistry, University of Pisa, Pisa, Italy; CISUP Center for the Integration of Scientific Instruments of the University of Pisa, Pisa, Italy
| | - Francesca Modugno
- Department of Chemistry and Industrial Chemistry, University of Pisa, Pisa, Italy; CISUP Center for the Integration of Scientific Instruments of the University of Pisa, Pisa, Italy
| |
Collapse
|
18
|
An Engineered Protein-Based Building Block (Albumin Methacryloyl) for Fabrication of a 3D In Vitro Cryogel Model. Gels 2022; 8:gels8070404. [PMID: 35877489 PMCID: PMC9324498 DOI: 10.3390/gels8070404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 06/15/2022] [Accepted: 06/22/2022] [Indexed: 11/25/2022] Open
Abstract
Drug-induced liver injury (DILI) is a leading cause of attrition in drug development or withdrawal; current animal experiments and traditional 2D cell culture systems fail to precisely predict the liver toxicity of drug candidates. Hence, there is an urgent need for an alternative in vitro model that can mimic the liver microenvironments and accurately detect human-specific drug hepatotoxicity. Here, for the first time we propose the fabrication of an albumin methacryloyl cryogel platform inspired by the liver’s microarchitecture via emulating the mechanical properties and extracellular matrix (ECM) cues of liver. Engineered crosslinkable albumin methacryloyl is used as a protein-based building block for fabrication of albumin cryogel in vitro models that can have potential applications in 3D cell culture and drug screening. In this work, protein modification, cryogelation, and liver ECM coating were employed to engineer highly porous three-dimensional cryogels with high interconnectivity, liver-like stiffness, and liver ECM as artificial liver constructs. The resulting albumin-based cryogel in vitro model provided improved cell–cell and cell–material interactions and consequently displayed excellent liver functional gene expression, being conducive to detection of fialuridine (FIAU) hepatotoxicity.
Collapse
|
19
|
Segovia-Zafra A, Di Zeo-Sánchez DE, López-Gómez C, Pérez-Valdés Z, García-Fuentes E, Andrade RJ, Lucena MI, Villanueva-Paz M. Preclinical models of idiosyncratic drug-induced liver injury (iDILI): Moving towards prediction. Acta Pharm Sin B 2021; 11:3685-3726. [PMID: 35024301 PMCID: PMC8727925 DOI: 10.1016/j.apsb.2021.11.013] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 11/07/2021] [Accepted: 11/10/2021] [Indexed: 02/08/2023] Open
Abstract
Idiosyncratic drug-induced liver injury (iDILI) encompasses the unexpected harms that prescription and non-prescription drugs, herbal and dietary supplements can cause to the liver. iDILI remains a major public health problem and a major cause of drug attrition. Given the lack of biomarkers for iDILI prediction, diagnosis and prognosis, searching new models to predict and study mechanisms of iDILI is necessary. One of the major limitations of iDILI preclinical assessment has been the lack of correlation between the markers of hepatotoxicity in animal toxicological studies and clinically significant iDILI. Thus, major advances in the understanding of iDILI susceptibility and pathogenesis have come from the study of well-phenotyped iDILI patients. However, there are many gaps for explaining all the complexity of iDILI susceptibility and mechanisms. Therefore, there is a need to optimize preclinical human in vitro models to reduce the risk of iDILI during drug development. Here, the current experimental models and the future directions in iDILI modelling are thoroughly discussed, focusing on the human cellular models available to study the pathophysiological mechanisms of the disease and the most used in vivo animal iDILI models. We also comment about in silico approaches and the increasing relevance of patient-derived cellular models.
Collapse
Affiliation(s)
- Antonio Segovia-Zafra
- Unidad de Gestión Clínica de Gastroenterología, Servicio de Farmacología Clínica, Instituto de Investigación Biomédica de Málaga-IBIMA, Hospital Universitario Virgen de la Victoria, Universidad de Málaga, Málaga 29071, Spain
- Centro de Investigación Biomédica en Red en el Área Temática de Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid 28029, Spain
| | - Daniel E. Di Zeo-Sánchez
- Unidad de Gestión Clínica de Gastroenterología, Servicio de Farmacología Clínica, Instituto de Investigación Biomédica de Málaga-IBIMA, Hospital Universitario Virgen de la Victoria, Universidad de Málaga, Málaga 29071, Spain
| | - Carlos López-Gómez
- Unidad de Gestión Clínica de Aparato Digestivo, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Universitario Virgen de la Victoria, Málaga 29010, Spain
| | - Zeus Pérez-Valdés
- Unidad de Gestión Clínica de Gastroenterología, Servicio de Farmacología Clínica, Instituto de Investigación Biomédica de Málaga-IBIMA, Hospital Universitario Virgen de la Victoria, Universidad de Málaga, Málaga 29071, Spain
| | - Eduardo García-Fuentes
- Unidad de Gestión Clínica de Aparato Digestivo, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Universitario Virgen de la Victoria, Málaga 29010, Spain
| | - Raúl J. Andrade
- Unidad de Gestión Clínica de Gastroenterología, Servicio de Farmacología Clínica, Instituto de Investigación Biomédica de Málaga-IBIMA, Hospital Universitario Virgen de la Victoria, Universidad de Málaga, Málaga 29071, Spain
- Centro de Investigación Biomédica en Red en el Área Temática de Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid 28029, Spain
| | - M. Isabel Lucena
- Unidad de Gestión Clínica de Gastroenterología, Servicio de Farmacología Clínica, Instituto de Investigación Biomédica de Málaga-IBIMA, Hospital Universitario Virgen de la Victoria, Universidad de Málaga, Málaga 29071, Spain
- Centro de Investigación Biomédica en Red en el Área Temática de Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid 28029, Spain
- Platform ISCIII de Ensayos Clínicos, UICEC-IBIMA, Málaga 29071, Spain
| | - Marina Villanueva-Paz
- Unidad de Gestión Clínica de Gastroenterología, Servicio de Farmacología Clínica, Instituto de Investigación Biomédica de Málaga-IBIMA, Hospital Universitario Virgen de la Victoria, Universidad de Málaga, Málaga 29071, Spain
| |
Collapse
|
20
|
van der Stel W, Yang H, Vrijenhoek NG, Schimming JP, Callegaro G, Carta G, Darici S, Delp J, Forsby A, White A, le Dévédec S, Leist M, Jennings P, Beltman JB, van de Water B, Danen EHJ. Mapping the cellular response to electron transport chain inhibitors reveals selective signaling networks triggered by mitochondrial perturbation. Arch Toxicol 2021; 96:259-285. [PMID: 34642769 PMCID: PMC8748354 DOI: 10.1007/s00204-021-03160-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 09/09/2021] [Indexed: 12/24/2022]
Abstract
Mitochondrial perturbation is a key event in chemical-induced organ toxicities that is incompletely understood. Here, we studied how electron transport chain (ETC) complex I, II, or III (CI, CII and CIII) inhibitors affect mitochondrial functionality, stress response activation, and cell viability using a combination of high-content imaging and TempO-Seq in HepG2 hepatocyte cells. CI and CIII inhibitors perturbed mitochondrial membrane potential (MMP) and mitochondrial and cellular ATP levels in a concentration- and time-dependent fashion and, under conditions preventing a switch to glycolysis attenuated cell viability, whereas CII inhibitors had no effect. TempO-Seq analysis of changes in mRNA expression pointed to a shared cellular response to CI and CIII inhibition. First, to define specific ETC inhibition responses, a gene set responsive toward ETC inhibition (and not to genotoxic, oxidative, or endoplasmic reticulum stress) was identified using targeted TempO-Seq in HepG2. Silencing of one of these genes, NOS3, exacerbated the impact of CI and CIII inhibitors on cell viability, indicating its functional implication in cellular responses to mitochondrial stress. Then by monitoring dynamic responses to ETC inhibition using a HepG2 GFP reporter panel for different classes of stress response pathways and applying pathway and gene network analysis to TempO-Seq data, we looked for downstream cellular events of ETC inhibition and identified the amino acid response (AAR) as being triggered in HepG2 by ETC inhibition. Through in silico approaches we provide evidence indicating that a similar AAR is associated with exposure to mitochondrial toxicants in primary human hepatocytes. Altogether, we (i) unravel quantitative, time- and concentration-resolved cellular responses to mitochondrial perturbation, (ii) identify a gene set associated with adaptation to exposure to active ETC inhibitors, and (iii) show that ER stress and an AAR accompany ETC inhibition in HepG2 and primary hepatocytes.
Collapse
Affiliation(s)
- Wanda van der Stel
- Division of Drug Discovery and Safety, Leiden Academic Centre of Drug Research, Leiden University, Leiden, The Netherlands
| | - Huan Yang
- Division of Drug Discovery and Safety, Leiden Academic Centre of Drug Research, Leiden University, Leiden, The Netherlands
| | - Nanette G Vrijenhoek
- Division of Drug Discovery and Safety, Leiden Academic Centre of Drug Research, Leiden University, Leiden, The Netherlands
| | - Johannes P Schimming
- Division of Drug Discovery and Safety, Leiden Academic Centre of Drug Research, Leiden University, Leiden, The Netherlands
| | - Giulia Callegaro
- Division of Drug Discovery and Safety, Leiden Academic Centre of Drug Research, Leiden University, Leiden, The Netherlands
| | - Giada Carta
- Division Molecular and Computational Toxicology, Vrije University Amsterdam, Amsterdam, The Netherlands
| | - Salihanur Darici
- Division of Drug Discovery and Safety, Leiden Academic Centre of Drug Research, Leiden University, Leiden, The Netherlands
| | - Johannes Delp
- Chair for In Vitro Toxicology and Biomedicine, Department Inaugurated by the Doerenkamp-Zbinden Foundation, University of Konstanz, Konstanz, Germany
| | - Anna Forsby
- Department of Biochemistry and Biophysics, Stockholm University, Stockholm, Sweden
| | | | - Sylvia le Dévédec
- Division of Drug Discovery and Safety, Leiden Academic Centre of Drug Research, Leiden University, Leiden, The Netherlands
| | - Marcel Leist
- Chair for In Vitro Toxicology and Biomedicine, Department Inaugurated by the Doerenkamp-Zbinden Foundation, University of Konstanz, Konstanz, Germany
| | - Paul Jennings
- Division Molecular and Computational Toxicology, Vrije University Amsterdam, Amsterdam, The Netherlands
| | - Joost B Beltman
- Division of Drug Discovery and Safety, Leiden Academic Centre of Drug Research, Leiden University, Leiden, The Netherlands
| | - Bob van de Water
- Division of Drug Discovery and Safety, Leiden Academic Centre of Drug Research, Leiden University, Leiden, The Netherlands.
| | - Erik H J Danen
- Division of Drug Discovery and Safety, Leiden Academic Centre of Drug Research, Leiden University, Leiden, The Netherlands.
| |
Collapse
|
21
|
Host bioenergetic parameters reveal cytotoxicity of anti-tuberculosis drugs undetected using conventional viability assays. Antimicrob Agents Chemother 2021; 65:e0093221. [PMID: 34339269 PMCID: PMC8448146 DOI: 10.1128/aac.00932-21] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
High attrition rates in tuberculosis (TB) drug development have been largely attributed to safety, which is likely due to the use of endpoint assays measuring cell viability to detect drug cytotoxicity. In drug development for cancer, metabolic, and neurological disorders and for antibiotics, cytotoxicity is increasingly being assessed using extracellular flux (XF) analysis, which measures cellular bioenergetic metabolism in real time. Here, we adopt the XF platform to investigate the cytotoxicity of drugs currently used in TB treatment on the bioenergetic metabolism of HepG2 cells, THP-1 macrophages, and human monocyte-derived macrophages (hMDMs). We found that the XF analysis reveals earlier drug-induced effects on the cells’ bioenergetic metabolism prior to cell death, measured by conventional viability assays. Furthermore, each cell type has a distinct response to drug treatment, suggesting that more than one cell type should be considered to examine cytotoxicity in TB drug development. Interestingly, chemically unrelated drugs with different modes of action on Mycobacterium tuberculosis have similar effects on the bioenergetic parameters of the cells, thus discouraging the prediction of potential cytotoxicity based on chemical structure and mode of action of new chemical entities. The clustering of the drug-induced effects on the hMDM bioenergetic parameters are reflected in the clustering of the effects of the drugs on cytokine production in hMDMs, demonstrating concurrence between the effects of the drugs on the metabolism and functioning of the macrophages. These findings can be used as a benchmark to establish XF analysis as a new tool to assay cytotoxicity in TB drug development.
Collapse
|
22
|
Lin YT, Lin KH, Huang CJ, Wei AC. MitoTox: a comprehensive mitochondrial toxicity database. BMC Bioinformatics 2021; 22:369. [PMID: 34266386 PMCID: PMC8283953 DOI: 10.1186/s12859-021-04285-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2021] [Accepted: 07/06/2021] [Indexed: 11/22/2022] Open
Abstract
Background Mitochondria play essential roles in regulating cellular functions. Some drug treatments and molecular interventions have been reported to have off-target effects damaging mitochondria and causing severe side effects. The development of a database for the management of mitochondrial toxicity-related molecules and their targets is important for further analyses. Results To correlate chemical, biological and mechanistic information on clinically relevant mitochondria-related toxicity, a comprehensive mitochondrial toxicity database (MitoTox) was developed. MitoTox is an electronic repository that integrates comprehensive information about mitochondria-related toxins and their targets. Information and data related to mitochondrial toxicity originate from various sources, including scientific journals and other electronic databases. These resources were manually verified and extracted into MitoTox. The database currently contains over 1400 small-molecule compounds, 870 mitochondrial targets, and more than 4100 mitochondrial toxin-target associations. Each MitoTox data record contains over 30 fields, including biochemical properties, therapeutic classification, target proteins, toxicological data, mechanistic information, clinical side effects, and references. Conclusions MitoTox provides a fully searchable database with links to references and other databases. Potential applications of MitoTox include toxicity classification, prediction, reference and education. MitoTox is available online at http://www.mitotox.org.
Collapse
Affiliation(s)
- Yu-Te Lin
- Graduate Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, Taipei, Taiwan
| | - Ko-Hong Lin
- Department of Electrical Engineering, National Taiwan University, Taipei, Taiwan
| | - Chi-Jung Huang
- Graduate Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, Taipei, Taiwan
| | - An-Chi Wei
- Graduate Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, Taipei, Taiwan. .,Department of Electrical Engineering, National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
23
|
George JW, Mattingly JE, Roland NJ, Small CM, Lamberty BG, Fox HS, Stauch KL. Physiologically Relevant Concentrations of Dolutegravir, Emtricitabine, and Efavirenz Induce Distinct Metabolic Alterations in HeLa Epithelial and BV2 Microglial Cells. Front Immunol 2021; 12:639378. [PMID: 34093527 PMCID: PMC8173175 DOI: 10.3389/fimmu.2021.639378] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 05/04/2021] [Indexed: 11/13/2022] Open
Abstract
Microglia, the resident brain phagocytes, likely play a key role in human immunodeficiency virus (HIV) infection of the central nervous system (CNS) and subsequent neuropathogenesis; however, the nature of the infection-induced changes that yield damaging CNS effects and the stimuli that provoke microglial activation remains elusive, especially in the current era of using antiretroviral (ARV) drugs for ARV therapy (ART). Altered microglial metabolism can modulate cellular functionality and pathogenicity in neurological disease. While HIV infection itself alters brain energy metabolism, the effect of ARV drugs, particularly those currently used in treatment, on metabolism is understudied. Dolutegravir (DTG) and emtricitabine (FTC) combination, together with tenofovir (TAF or TDF), is one of the recommended first line treatments for HIV. Despite the relatively good tolerability and safety profile of FTC, a nucleoside reverse transcriptase inhibitor, and DTG, an integrase inhibitor, adverse side effects have been reported and highlight a need to understand off-target effects of these medications. We hypothesized that similar to previous ART regimen drugs, DTG and FTC side effects involve mitochondrial dysfunction. To increase detection of ARV-induced mitochondrial effects, highly glycolytic HeLa epithelial cells were forced to rely on oxidative phosphorylation by substituting galactose for glucose in the growth media. We assessed ATP levels, resazurin oxidation-reduction (REDOX), and mitochondrial membrane potential following 24-hour exposure (to approximate effects of one dose equivalent) to DTG, FTC, and efavirenz (EFV, a known mitotoxic ARV drug). Further, since microglia support productive HIV infection, act as latent HIV cellular reservoirs, and when dysfunctional likely contribute to HIV-associated neurocognitive disorders, the experiments were repeated using BV2 microglial cells. In HeLa cells, FTC decreased mitochondrial REDOX activity, while DTG, similar to EFV, impaired both mitochondrial ATP generation and REDOX activity. In contrast to HeLa cells, DTG increased cellular ATP generation and mitochondrial REDOX activity in BV2 cells. Bioenergetic analysis revealed that DTG, FTC, and EFV elevated BV2 cell mitochondrial respiration. DTG and FTC exposure induced distinct mitochondrial functional changes in HeLa and BV2 cells. These findings suggest cell type-specific metabolic changes may contribute to the toxic side effects of these ARV drugs.
Collapse
Affiliation(s)
- Joseph W George
- Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, NE, United States
| | - Jane E Mattingly
- Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, NE, United States
| | - Nashanthea J Roland
- Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, NE, United States
| | - Cassandra M Small
- Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, NE, United States
| | - Benjamin G Lamberty
- Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, NE, United States
| | - Howard S Fox
- Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, NE, United States
| | - Kelly L Stauch
- Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, NE, United States
| |
Collapse
|
24
|
Ivashchenko AA, Mitkin OD, Jones JC, Nikitin AV, Koryakova AG, Karapetian RN, Kravchenko DV, Mochalov SV, Ryakhovskiy AA, Aladinskiy V, Leneva IA, Falynskova IN, Glubokova EA, Govorkova EA, Ivachtchenko AV. Synthesis, inhibitory activity and oral dosing formulation of AV5124, the structural analogue of influenza virus endonuclease inhibitor baloxavir. J Antimicrob Chemother 2021; 76:1010-1018. [PMID: 33367751 PMCID: PMC7953317 DOI: 10.1093/jac/dkaa524] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 10/26/2020] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND The development and clinical implementation of the cap-dependent endonuclease (CEN) inhibitor baloxavir marboxil was a breakthrough in influenza therapy, but it was associated with the emergence of drug-resistant variants. OBJECTIVES To design and synthesize structural analogues of CEN inhibitors and evaluate their safety, pharmacokinetics and antiviral potency in vitro and in vivo. METHODS The drug candidate AV5124 and its active metabolite AV5116 were synthesized based on pharmacophore modelling. Stability in plasma and microsomes, plasma protein binding, cytotoxicity and antiviral activities were assessed in vitro. Pharmacokinetics after IV or oral administration were analysed in CD-1 mice. Acute toxicity and protective efficacy against lethal A(H1N1)pdm09 influenza virus challenge were examined in BALB/c mice. RESULTS Pharmacophore model-assisted, 3D molecular docking predicted key supramolecular interactions of the metal-binding group and bulky hydrophobic group of AV5116 with the CEN binding site (Protein Data Bank code: 6FS6) that are essential for high antiviral activity. AV5116 inhibited influenza virus polymerase complexes in cell-free assays and replication of oseltamivir-susceptible and -resistant influenza A and B viruses at nanomolar concentrations. Notably, AV5116 was equipotent or more potent than baloxavir acid (BXA) against WT (I38-WT) viruses and viruses with reduced BXA susceptibility carrying an I38T polymerase acidic (PA) substitution. AV5116 exhibited low cytotoxicity in Madin-Darby canine kidney cells and lacked mitochondrial toxicity, resulting in favourable selective indices. Treatment with 20 or 50 mg/kg AV5124 prevented death in 60% and 100% of animals, respectively. CONCLUSIONS Overall, AV5124 and A5116 are promising inhibitors of the influenza virus CEN and warrant further development as potent anti-influenza agents.
Collapse
Affiliation(s)
- Andrei A Ivashchenko
- Chemical Diversity Research Institute, Rabochaya St. 2a, Khimki, Moscow Region 141401, Russia
- ChemDiv, 12760 High Bluff Drive, Ste. 370 San Diego, CA 92130, USA
- Moscow Institute of Physics and Technology (State University), Institutskiy Lane, Dolgoprudny, Moscow Region 141700, Russia
| | - Oleg D Mitkin
- Chemical Diversity Research Institute, Rabochaya St. 2a, Khimki, Moscow Region 141401, Russia
| | - Jeremy C Jones
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Alexander V Nikitin
- Chemical Diversity Research Institute, Rabochaya St. 2a, Khimki, Moscow Region 141401, Russia
| | - Angela G Koryakova
- Chemical Diversity Research Institute, Rabochaya St. 2a, Khimki, Moscow Region 141401, Russia
| | - Ruben N Karapetian
- Chemical Diversity Research Institute, Rabochaya St. 2a, Khimki, Moscow Region 141401, Russia
| | - Dmitry V Kravchenko
- Chemical Diversity Research Institute, Rabochaya St. 2a, Khimki, Moscow Region 141401, Russia
| | - Stephan V Mochalov
- Chemical Diversity Research Institute, Rabochaya St. 2a, Khimki, Moscow Region 141401, Russia
| | - Alexey A Ryakhovskiy
- Chemical Diversity Research Institute, Rabochaya St. 2a, Khimki, Moscow Region 141401, Russia
| | - Vladimir Aladinskiy
- Moscow Institute of Physics and Technology (State University), Institutskiy Lane, Dolgoprudny, Moscow Region 141700, Russia
| | - Irina A Leneva
- I. Mechnikov Research Institute for Vaccines and Sera, 5a Malyy Kazennyy lane, Moscow 105064, Russia
| | - Irina N Falynskova
- I. Mechnikov Research Institute for Vaccines and Sera, 5a Malyy Kazennyy lane, Moscow 105064, Russia
| | - Ekaterina A Glubokova
- I. Mechnikov Research Institute for Vaccines and Sera, 5a Malyy Kazennyy lane, Moscow 105064, Russia
| | - Elena A Govorkova
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Alexandre V Ivachtchenko
- Chemical Diversity Research Institute, Rabochaya St. 2a, Khimki, Moscow Region 141401, Russia
- ChemDiv, 12760 High Bluff Drive, Ste. 370 San Diego, CA 92130, USA
- ASAVI LLC, 1835 E. Hallandale Beach Blvd, #442, Hallandale Beach, FL 33009, USA
| |
Collapse
|
25
|
Santos CS, Macedo F, Kowaltowski AJ, Bertotti M, Unwin PR, Marques da Cunha F, Meloni GN. Unveiling the contribution of the reproductive system of individual Caenorhabditis elegans on oxygen consumption by single-point scanning electrochemical microscopy measurements. Anal Chim Acta 2021; 1146:88-97. [PMID: 33461723 PMCID: PMC7836392 DOI: 10.1016/j.aca.2020.12.030] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 12/11/2020] [Accepted: 12/17/2020] [Indexed: 01/03/2023]
Abstract
Metabolic analysis in animals is usually either evaluated as whole-body measurements or in isolated tissue samples. To reveal tissue specificities in vivo, this study uses scanning electrochemical microscopy (SECM) to provide localized oxygen consumption rates (OCRs) in different regions of single adult Caenorhabditis elegans individuals. This is achieved by measuring the oxygen reduction current at the SECM tip electrode and using a finite element method model of the experiment that defines oxygen concentration and flux at the surface of the organism. SECM mapping measurements uncover a marked heterogeneity of OCR along the worm, with high respiration rates at the reproductive system region. To enable sensitive and quantitative measurements, a self-referencing approach is adopted, whereby the oxygen reduction current at the SECM tip is measured at a selected point on the worm and in bulk solution (calibration). Using genetic and pharmacological approaches, our SECM measurements indicate that viable eggs in the reproductive system are the main contributors in the total oxygen consumption of adult Caenorhabditis elegans. The finding that large regional differences in OCR exist within the animal provides a new understanding of oxygen consumption and metabolic measurements, paving the way for tissue-specific metabolic analyses and toxicity evaluation within single organisms.
Collapse
Affiliation(s)
- Carla S Santos
- Departamento de Química Fundamental, Av. Professor Lineu Prestes, 748, 05508-000, São Paulo, SP, Brazil.
| | - Felipe Macedo
- Departamento de Bioquímica, Escola Paulista de Medicina, Universidade Federal de São Paulo, Rua três de Maio, 100, 04044-020, São Paulo, Brazil
| | - Alicia J Kowaltowski
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, Av. Professor Lineu Prestes, 748, 05508-000, São Paulo, SP, Brazil
| | - Mauro Bertotti
- Departamento de Química Fundamental, Av. Professor Lineu Prestes, 748, 05508-000, São Paulo, SP, Brazil
| | - Patrick R Unwin
- Department of Chemistry, University of Warwick, Coventry, CV4 7AL, United Kingdom; Bio-Electrical Engineering Innovation Hub, University of Warwick, Coventry, CV4 7AL, United Kingdom
| | - Fernanda Marques da Cunha
- Departamento de Bioquímica, Escola Paulista de Medicina, Universidade Federal de São Paulo, Rua três de Maio, 100, 04044-020, São Paulo, Brazil
| | - Gabriel N Meloni
- Department of Chemistry, University of Warwick, Coventry, CV4 7AL, United Kingdom; Bio-Electrical Engineering Innovation Hub, University of Warwick, Coventry, CV4 7AL, United Kingdom.
| |
Collapse
|
26
|
Jones SW, Penman SL, French NS, Park BK, Chadwick AE. Investigating dihydroorotate dehydrogenase inhibitor mediated mitochondrial dysfunction in hepatic in vitro models. Toxicol In Vitro 2021; 72:105096. [PMID: 33460737 DOI: 10.1016/j.tiv.2021.105096] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 12/17/2020] [Accepted: 01/12/2021] [Indexed: 01/13/2023]
Abstract
Inhibition of dihydroorotate dehydrogenase (DHODH), the rate-limiting enzymatic step in de novo pyrimidine synthesis, has broad immunosuppressive effects in vivo and shows promise as a therapeutic target for the treatment of malignancies, viral infections and auto-immune diseases. Whilst there are numerous DHODH inhibitors under development, leflunomide and teriflunomide are the only FDA approved compounds on the market, each of which have been issued with black-box warnings for hepatotoxicity. Mitochondrial dysfunction is a putative mechanism by which teriflunomide and leflunomide elicit their hepatotoxic effects, however it is as yet unclear whether this is shared by other nascent DHODH inhibitors. The present study aimed to evaluate the propensity for DHODH inhibitors to mediate mitochondrial dysfunction in two hepatic in vitro models. Initial comparisons of cytotoxicity and ATP content in HepaRG® cells primed for oxidative metabolism, in tandem with mechanistic evaluations by extracellular flux analysis identified multifactorial toxicity and moderate indications of respiratory chain dysfunction or uncoupling. Further investigations using HepG2 cells, a hepatic line with limited capability for phase I xenobiotic metabolism, identified leflunomide and brequinar as positive mitochondrial toxicants. Taken together, biotransformation of some DHODH inhibitor species may play a role in mediating or masking hepatic mitochondrial liabilities.
Collapse
Affiliation(s)
- Samantha W Jones
- Department of Molecular and Clinical Pharmacology, MRC Centre for Drug Safety Science, University of Liverpool, Ashton street Liverpool, L69 3GE, UK
| | - Sophie L Penman
- Department of Molecular and Clinical Pharmacology, MRC Centre for Drug Safety Science, University of Liverpool, Ashton street Liverpool, L69 3GE, UK
| | - Neil S French
- Department of Molecular and Clinical Pharmacology, MRC Centre for Drug Safety Science, University of Liverpool, Ashton street Liverpool, L69 3GE, UK
| | - B Kevin Park
- Department of Molecular and Clinical Pharmacology, MRC Centre for Drug Safety Science, University of Liverpool, Ashton street Liverpool, L69 3GE, UK
| | - Amy E Chadwick
- Department of Molecular and Clinical Pharmacology, MRC Centre for Drug Safety Science, University of Liverpool, Ashton street Liverpool, L69 3GE, UK.
| |
Collapse
|
27
|
Anderson CC, Marentette JO, Rauniyar AK, Prutton KM, Khatri M, Matheson C, Reisz JA, Reigan P, D'Alessandro A, Roede JR. Maneb alters central carbon metabolism and thiol redox status in a toxicant model of Parkinson's disease. Free Radic Biol Med 2021; 162:65-76. [PMID: 33279619 PMCID: PMC7889696 DOI: 10.1016/j.freeradbiomed.2020.11.028] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 11/16/2020] [Accepted: 11/23/2020] [Indexed: 11/22/2022]
Abstract
The dithiocarbamate fungicide maneb (MB) has attracted interest due to increasing concern of the negative health effects of pesticides, as well as its association with Parkinson's disease (PD). Our laboratory has previously reported distinct phenotypic changes of neuroblastoma cells exposed to acute, sub-toxic levels of MB, including decreased mitochondrial respiration, altered lactate dynamics, and metabolic stress. In this study, we aimed to further define the specific molecular mechanisms of MB toxicity through the comparison of several thiol-containing compounds and their effects on cellular energy metabolism and thiol redox nodes. Extracellular flux analyses and stable isotope labeled tracer metabolomics were employed to evaluate alterations in energy metabolism of SK-N-AS human neuroblastoma cells after acute exposure of an array of compounds, including dithiocarbamates (maneb, nabam, zineb) and other thiol-containing small molecules (glutathione, N-acetylcysteine). These studies revealed MB and its methylated form (MeDTC) as unique toxicants with significant alterations to mitochondrial respiration, proliferation, and glycolysis. We observed MB to significantly impact cellular thiol redox status by oxidizing cellular glutathione and altering the thiol redox status of peroxiredoxin 3 (Prx3, mitochondrial) after acute exposure. Redox Western blotting revealed a MB-specific modification of cellular Prx3, strengthening the argument that MB can preferentially target mitochondrial enzymes containing reactive cysteine thiols. Further, stable isotope tracer metabolomics confirmed our energetics assessments, and demonstrated that MB exposure results in acute derangement of central carbon metabolism. Specifically, we observed shunting of cellular glucose into the pentose-phosphate pathway and reduction of TCA intermediates derived from glucose and glutamine. Also, we report novel lactate utilization for TCA enrichment and glutathione synthesis after MB exposure. In summary, our results further confirm that MB exerts its toxic effects via thiol modification, and significantly transforms central carbon metabolism.
Collapse
Affiliation(s)
- Colin C Anderson
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, Aurora, CO, 80045, USA
| | - John O Marentette
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, Aurora, CO, 80045, USA
| | - Abhishek K Rauniyar
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, Aurora, CO, 80045, USA
| | - Kendra M Prutton
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, Aurora, CO, 80045, USA
| | - Meera Khatri
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, Aurora, CO, 80045, USA
| | - Chris Matheson
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, Aurora, CO, 80045, USA
| | - Julie A Reisz
- Department of Biochemistry and Molecular Genetics, School of Medicine, University of Colorado, Aurora, CO, 80045, USA
| | - Philip Reigan
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, Aurora, CO, 80045, USA
| | - Angelo D'Alessandro
- Department of Biochemistry and Molecular Genetics, School of Medicine, University of Colorado, Aurora, CO, 80045, USA
| | - James R Roede
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, Aurora, CO, 80045, USA.
| |
Collapse
|
28
|
Preston S, Garcia-Bustos J, Hall LG, Martin SD, Le TG, Kundu A, Ghoshal A, Nguyen NH, Jiao Y, Ruan B, Xue L, Huang F, Chang BCH, McGee SL, Wells TNC, Palmer MJ, Jabbar A, Gasser RB, Baell JB. 1-Methyl-1 H-pyrazole-5-carboxamide Derivatives Exhibit Unexpected Acute Mammalian Toxicity. J Med Chem 2020; 64:840-844. [PMID: 33352050 DOI: 10.1021/acs.jmedchem.0c01793] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
A series of 1-methyl-1H-pyrazole-5-carboxamides were synthesized as potent inhibitors of the parasitic nematode of sheep, Haemonchus contortus. These compounds did not show overt cytotoxicity to a range of mammalian cell lines under standard in vitro culture conditions, had high selectivity indices, and were progressed to an acute toxicity study in a rodent model. Strikingly, acute toxicity was observed in mice. Experiments measuring cellular respiration showed a dose-dependent inhibition of mitochondrial respiration. Under these conditions, potent cytotoxicity was observed for these compounds in rat hepatocytes suggesting that the potent acute mammalian toxicity of this chemotype is most likely associated with respiratory inhibition. In contrast, parasite toxicity was not correlated to acute toxicity or cytotoxicity in respiring cells. This paper highlights the importance of identifying an appropriate in vitro predictor of in vivo toxicity early on in the drug discovery pipeline, in particular assessment for in vitro mitochondrial toxicity.
Collapse
Affiliation(s)
- Sarah Preston
- School of Health and Life Sciences, Federation University, Ballarat, Victoria 3353, Australia
| | - Jose Garcia-Bustos
- Department of Veterinary Biosciences, Melbourne Veterinary School, Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Liam G Hall
- Metabolic Research Unit, Metabolic Reprogramming Laboratory, School of Medicine, Faculty of Health, Deakin University, Waurn Ponds, Victoria 3216, Australia
| | - Sheree D Martin
- Metabolic Research Unit, Metabolic Reprogramming Laboratory, School of Medicine, Faculty of Health, Deakin University, Waurn Ponds, Victoria 3216, Australia
| | - Thuy G Le
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Abhijit Kundu
- TCG Lifesciences Private Limited, Block BN, Plot 7, Salt-lake Electronics Complex, Sector V, Kolkata 700091, West Bengal, India
| | - Atanu Ghoshal
- TCG Lifesciences Private Limited, Block BN, Plot 7, Salt-lake Electronics Complex, Sector V, Kolkata 700091, West Bengal, India
| | - Nghi H Nguyen
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Yaqing Jiao
- Department of Veterinary Biosciences, Melbourne Veterinary School, Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Banfeng Ruan
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Lian Xue
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Fei Huang
- School of Pharmaceutical Sciences, Nanjing Tech University, No. 30 South Puzhu Road, Nanjing 211816, People's Republic of China
| | | | - Sean L McGee
- Metabolic Research Unit, Metabolic Reprogramming Laboratory, School of Medicine, Faculty of Health, Deakin University, Waurn Ponds, Victoria 3216, Australia
| | | | | | - Abdul Jabbar
- Department of Veterinary Biosciences, Melbourne Veterinary School, Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Robin B Gasser
- Department of Veterinary Biosciences, Melbourne Veterinary School, Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Jonathan B Baell
- School of Pharmaceutical Sciences, Nanjing Tech University, No. 30 South Puzhu Road, Nanjing 211816, People's Republic of China.,Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| |
Collapse
|
29
|
HepG2 (C3A) spheroids show higher sensitivity compared to HepaRG spheroids for drug-induced liver injury (DILI). Toxicol Appl Pharmacol 2020; 408:115279. [DOI: 10.1016/j.taap.2020.115279] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 09/25/2020] [Accepted: 10/11/2020] [Indexed: 12/20/2022]
|
30
|
Matsui T, Miyamoto N, Saito F, Shinozawa T. Molecular Profiling of Human Induced Pluripotent Stem Cell-Derived Cells and their Application for Drug Safety Study. Curr Pharm Biotechnol 2020; 21:807-828. [PMID: 32321398 DOI: 10.2174/1389201021666200422090952] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 12/10/2019] [Accepted: 03/02/2020] [Indexed: 12/13/2022]
Abstract
Drug-induced toxicity remains one of the leading causes of discontinuation of the drug candidate and post-marketing withdrawal. Thus, early identification of the drug candidates with the potential for toxicity is crucial in the drug development process. With the recent discovery of human- Induced Pluripotent Stem Cells (iPSC) and the establishment of the differentiation protocol of human iPSC into the cell types of interest, the differentiated cells from human iPSC have garnered much attention because of their potential applicability in toxicity evaluation as well as drug screening, disease modeling and cell therapy. In this review, we expanded on current information regarding the feasibility of human iPSC-derived cells for the evaluation of drug-induced toxicity with a focus on human iPSCderived hepatocyte (iPSC-Hep), cardiomyocyte (iPSC-CMs) and neurons (iPSC-Neurons). Further, we CSAHi, Consortium for Safety Assessment using Human iPS Cells, reported our gene expression profiling data with DNA microarray using commercially available human iPSC-derived cells (iPSC-Hep, iPSC-CMs, iPSC-Neurons), their relevant human tissues and primary cultured human cells to discuss the future direction of the three types of human iPSC-derived cells.
Collapse
Affiliation(s)
- Toshikatsu Matsui
- Consortium for Safety Assessment using Human iPS Cells (CSAHi), Japan
| | - Norimasa Miyamoto
- Consortium for Safety Assessment using Human iPS Cells (CSAHi), Japan
| | - Fumiyo Saito
- Consortium for Safety Assessment using Human iPS Cells (CSAHi), Japan
| | | |
Collapse
|
31
|
Ivashchenko AA, Mitkin OD, Jones JC, Nikitin AV, Koryakova AG, Ryakhovskiy A, Karapetian RN, Kravchenko DV, Aladinskiy V, Leneva IA, Falynskova IN, Glubokova EA, Govorkova EA, Ivachtchenko AV. Non-rigid Diarylmethyl Analogs of Baloxavir as Cap-Dependent Endonuclease Inhibitors of Influenza Viruses. J Med Chem 2020; 63:9403-9420. [PMID: 32787099 DOI: 10.1021/acs.jmedchem.0c00565] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
4-Substituted 2,4-dioxobutanoic acids inhibit influenza virus cap-dependent endonuclease (CEN) activity. Baloxavir marboxil, 4, is approved for treating influenza virus infections. We describe here the synthesis and biological evaluation of active compounds, 5a-5g, and their precursors (6a, 6b, 6d, and 6e) with flexible bulky hydrophobic groups instead of the rigid polyheterocyclic moieties. In silico docking confirmed the ability of 5a-5g to bind to the active site of influenza A CEN (PDB code: 6FS6) like baloxavir acid, 3. These novel compounds inhibited polymerase complex activity, inhibited virus replication in cells, prevented death in a lethal influenza A virus mouse challenge model, and dramatically lowered viral lung titers. 5a and 5e potently inhibited different influenza genera in vitro. Precursors 6a and 6d demonstrated impressive mouse oral bioavailability with 6a, providing effective in vivo protection. Thus, these novel compounds are potent CEN inhibitors with in vitro and in vivo activity comparable to baloxavir.
Collapse
Affiliation(s)
- Andrei A Ivashchenko
- Chemical Diversity Research Institute, Rabochaya Street 2a, Khimki, Moscow Region 141401, Russia.,ChemDiv, 6605 Nancy Ridge Drive, San Diego, California 92121, United States.,Moscow Institute of Physics and Technology (State University), 9 Institutskiy Lane, Dolgoprudny City, Moscow Region 141700, Russia
| | - Oleg D Mitkin
- Chemical Diversity Research Institute, Rabochaya Street 2a, Khimki, Moscow Region 141401, Russia
| | - Jeremy C Jones
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, United States
| | - Alexander V Nikitin
- Chemical Diversity Research Institute, Rabochaya Street 2a, Khimki, Moscow Region 141401, Russia
| | - Angela G Koryakova
- Chemical Diversity Research Institute, Rabochaya Street 2a, Khimki, Moscow Region 141401, Russia
| | - Alexey Ryakhovskiy
- Chemical Diversity Research Institute, Rabochaya Street 2a, Khimki, Moscow Region 141401, Russia
| | - Ruben N Karapetian
- Chemical Diversity Research Institute, Rabochaya Street 2a, Khimki, Moscow Region 141401, Russia
| | - Dmitry V Kravchenko
- Chemical Diversity Research Institute, Rabochaya Street 2a, Khimki, Moscow Region 141401, Russia
| | - Vladimir Aladinskiy
- Moscow Institute of Physics and Technology (State University), 9 Institutskiy Lane, Dolgoprudny City, Moscow Region 141700, Russia
| | - Irina A Leneva
- I. Mechnikov Research Institute for Vaccines and Sera, 5a Malyy Kazennyy Lane, Moscow 105064, Russia
| | - Irina N Falynskova
- I. Mechnikov Research Institute for Vaccines and Sera, 5a Malyy Kazennyy Lane, Moscow 105064, Russia
| | - Ekaterina A Glubokova
- I. Mechnikov Research Institute for Vaccines and Sera, 5a Malyy Kazennyy Lane, Moscow 105064, Russia
| | - Elena A Govorkova
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, United States
| | - Alexandre V Ivachtchenko
- Chemical Diversity Research Institute, Rabochaya Street 2a, Khimki, Moscow Region 141401, Russia.,ChemDiv, 6605 Nancy Ridge Drive, San Diego, California 92121, United States.,ASAVI LLC, 1835 E. Hallandale Beach Blvd, #442, Hallandale Beach, Florida 33009, United States
| |
Collapse
|
32
|
van der Stel W, Carta G, Eakins J, Darici S, Delp J, Forsby A, Bennekou SH, Gardner I, Leist M, Danen EHJ, Walker P, van de Water B, Jennings P. Multiparametric assessment of mitochondrial respiratory inhibition in HepG2 and RPTEC/TERT1 cells using a panel of mitochondrial targeting agrochemicals. Arch Toxicol 2020; 94:2707-2729. [PMID: 32607615 PMCID: PMC7395062 DOI: 10.1007/s00204-020-02792-5] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 05/20/2020] [Indexed: 12/21/2022]
Abstract
Evidence is mounting for the central role of mitochondrial dysfunction in several pathologies including metabolic diseases, accelerated ageing, neurodegenerative diseases and in certain xenobiotic-induced organ toxicity. Assessing mitochondrial perturbations is not trivial and the outcomes of such investigations are dependent on the cell types used and assays employed. Here we systematically investigated the effect of electron transport chain (ETC) inhibitors on multiple mitochondrial-related parameters in two human cell types, HepG2 and RPTEC/TERT1. Cells were exposed to a broad range of concentrations of 20 ETC-inhibiting agrochemicals and capsaicin, consisting of inhibitors of NADH dehydrogenase (Complex I, CI), succinate dehydrogenase (Complex II, CII) and cytochrome bc1 complex (Complex III, CIII). A battery of tests was utilised, including viability assays, lactate production, mitochondrial membrane potential (MMP) and the Seahorse bioanalyser, which simultaneously measures extracellular acidification rate [ECAR] and oxygen consumption rate [OCR]. CI inhibitors caused a potent decrease in OCR, decreased mitochondrial membrane potential, increased ECAR and increased lactate production in both cell types. Twenty-fourhour exposure to CI inhibitors decreased viability of RPTEC/TERT1 cells and 3D spheroid-cultured HepG2 cells in the presence of glucose. CI inhibitors decreased 2D HepG2 viability only in the absence of glucose. CII inhibitors had no notable effects in intact cells up to 10 µM. CIII inhibitors had similar effects to the CI inhibitors. Antimycin A was the most potent CIII inhibitor, with activity in the nanomolar range. The proposed CIII inhibitor cyazofamid demonstrated a mitochondrial uncoupling signal in both cell types. The study presents a comprehensive example of a mitochondrial assessment workflow and establishes measurable key events of ETC inhibition.
Collapse
Affiliation(s)
- Wanda van der Stel
- Division of Drug Discovery and Safety, Leiden Academic Centre of Drug Research, Leiden University, Einsteinweg 55, 2333 CC Leiden, The Netherlands
| | - Giada Carta
- Division of Molecular and Computational Toxicology, Department of Chemistry and Pharmaceutical Sciences, AIMMS, Vrije Universiteit Amsterdam, De
Boelelaan, 1108, 1081 HZ Amsterdam, The Netherlands
| | - Julie Eakins
- Cyprotex Discovery Ltd, Alderley Park, Macclesfield, Cheshire, UK
| | - Salihanur Darici
- Division of Drug Discovery and Safety, Leiden Academic Centre of Drug Research, Leiden University, Einsteinweg 55, 2333 CC Leiden, The Netherlands
| | | | - Anna Forsby
- Department of Biochemistry and Biophysics, Stockholm University, Stockholm, Sweden
| | | | | | | | - Erik H. J. Danen
- Division of Drug Discovery and Safety, Leiden Academic Centre of Drug Research, Leiden University, Einsteinweg 55, 2333 CC Leiden, The Netherlands
| | - Paul Walker
- Cyprotex Discovery Ltd, Alderley Park, Macclesfield, Cheshire, UK
| | - Bob van de Water
- Division of Drug Discovery and Safety, Leiden Academic Centre of Drug Research, Leiden University, Einsteinweg 55, 2333 CC Leiden, The Netherlands
| | - Paul Jennings
- Division of Molecular and Computational Toxicology, Department of Chemistry and Pharmaceutical Sciences, AIMMS, Vrije Universiteit Amsterdam, De
Boelelaan, 1108, 1081 HZ Amsterdam, The Netherlands
| |
Collapse
|
33
|
Hatherell S, Baltazar MT, Reynolds J, Carmichael PL, Dent M, Li H, Ryder S, White A, Walker P, Middleton AM. Identifying and Characterizing Stress Pathways of Concern for Consumer Safety in Next-Generation Risk Assessment. Toxicol Sci 2020; 176:11-33. [PMID: 32374857 PMCID: PMC7357173 DOI: 10.1093/toxsci/kfaa054] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Many substances for which consumer safety risk assessments need to be conducted are not associated with specific toxicity modes of action, but rather exhibit nonspecific toxicity leading to cell stress. In this work, a cellular stress panel is described, consisting of 36 biomarkers representing mitochondrial toxicity, cell stress, and cell health, measured predominantly using high content imaging. To evaluate the panel, data were generated for 13 substances at exposures consistent with typical use-case scenarios. These included some that have been shown to cause adverse effects in a proportion of exposed humans and have a toxicological mode-of-action associated with cellular stress (eg, doxorubicin, troglitazone, and diclofenac), and some that are not associated with adverse effects due to cellular stress at human-relevant exposures (eg, caffeine, niacinamide, and phenoxyethanol). For each substance, concentration response data were generated for each biomarker at 3 timepoints. A Bayesian model was then developed to quantify the evidence for a biological response, and if present, a credibility range for the estimated point of departure (PoD) was determined. PoDs were compared with the plasma Cmax associated with the typical substance exposures, and indicated a clear differentiation between "low" risk and "high" risk chemical exposure scenarios. Developing robust methods to characterize the in vitro bioactivity of xenobiotics is an important part of non-animal safety assessment. The results presented in this work show that the cellular stress panel can be used, together with other new approach methodologies, to identify chemical exposures that are protective of consumer health.
Collapse
Affiliation(s)
- Sarah Hatherell
- Unilever Safety and Environmental Assurance Centre, Colworth Science Park, Sharnbrook, Bedfordshire MK44 1LQ, UK
| | - Maria T Baltazar
- Unilever Safety and Environmental Assurance Centre, Colworth Science Park, Sharnbrook, Bedfordshire MK44 1LQ, UK
| | - Joe Reynolds
- Unilever Safety and Environmental Assurance Centre, Colworth Science Park, Sharnbrook, Bedfordshire MK44 1LQ, UK
| | - Paul L Carmichael
- Unilever Safety and Environmental Assurance Centre, Colworth Science Park, Sharnbrook, Bedfordshire MK44 1LQ, UK
| | - Matthew Dent
- Unilever Safety and Environmental Assurance Centre, Colworth Science Park, Sharnbrook, Bedfordshire MK44 1LQ, UK
| | - Hequn Li
- Unilever Safety and Environmental Assurance Centre, Colworth Science Park, Sharnbrook, Bedfordshire MK44 1LQ, UK
| | | | - Andrew White
- Unilever Safety and Environmental Assurance Centre, Colworth Science Park, Sharnbrook, Bedfordshire MK44 1LQ, UK
| | - Paul Walker
- Cyprotex Discovery Ltd, Macclesfield, Cheshire SK10 4TG, UK
| | - Alistair M Middleton
- Unilever Safety and Environmental Assurance Centre, Colworth Science Park, Sharnbrook, Bedfordshire MK44 1LQ, UK
| |
Collapse
|
34
|
Norman BH. Drug Induced Liver Injury (DILI). Mechanisms and Medicinal Chemistry Avoidance/Mitigation Strategies. J Med Chem 2020; 63:11397-11419. [PMID: 32511920 DOI: 10.1021/acs.jmedchem.0c00524] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Adverse drug reactions (ADRs) are a common cause of attrition in drug discovery and development and drug-induced liver injury (DILI) is a leading cause of preclinical and clinical drug terminations. This perspective outlines many of the known DILI mechanisms and assessment methods used to evaluate and mitigate DILI risk. Literature assessments and retrospective analyses using verified DILI-associated drugs from the Liver Tox Knowledge Base (LTKB) have been used to derive the predictive value of each end point, along with combination approaches of multiple methods. In vitro assays to assess inhibition of the bile salt export pump (BSEP), mitotoxicity, reactive metabolite (RM) formation, and hepatocyte cytolethality, along with physicochemical properties and clinical dose provide useful DILI predictivity. This Perspective also highlights some of the strategies used by medicinal chemists to reduce DILI risk during the optimization of drug candidates.
Collapse
Affiliation(s)
- Bryan H Norman
- Norman Drug Discovery Training and Consulting, LLC, 8540 Bluefin Circle, Indianapolis, Indiana 46236, United States
| |
Collapse
|
35
|
Analysis of the Potential for N 4-Hydroxycytidine To Inhibit Mitochondrial Replication and Function. Antimicrob Agents Chemother 2020; 64:AAC.01719-19. [PMID: 31767721 PMCID: PMC6985706 DOI: 10.1128/aac.01719-19] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Accepted: 11/18/2019] [Indexed: 02/06/2023] Open
Abstract
N4-Hydroxycytidine (NHC) is an antiviral ribonucleoside analog that acts as a competitive alternative substrate for virally encoded RNA-dependent RNA polymerases. It exhibits measurable levels of cytotoxicity, with 50% cytotoxic concentration values ranging from 7.5 μM in CEM cells and up to >100 μM in other cell lines. N4-Hydroxycytidine (NHC) is an antiviral ribonucleoside analog that acts as a competitive alternative substrate for virally encoded RNA-dependent RNA polymerases. It exhibits measurable levels of cytotoxicity, with 50% cytotoxic concentration values ranging from 7.5 μM in CEM cells and up to >100 μM in other cell lines. The mitochondrial DNA-dependent RNA polymerase (POLRMT) has been shown to incorporate some nucleotide analogs into mitochondrial RNAs, resulting in substantial mitochondrial toxicity. NHC was tested in multiple assays intended to determine its potential to cause mitochondrial toxicity. NHC showed similar cytotoxicity in HepG2 cells incubated in a glucose-free and glucose-containing media, suggesting that NHC does not impair mitochondrial function in this cell line based on the Crabtree effect. We demonstrate that the 5′-triphosphate of NHC can be used by POLRMT for incorporation into nascent RNA chain but does not cause immediate chain termination. In PC-3 cells treated with NHC, the 50% inhibitory concentrations of mitochondrial protein expression inhibition were 2.7-fold lower than those for nuclear-encoded protein expression, but this effect did not result in selective mitochondrial toxicity. A 14-day incubation of HepG2 cells with NHC had no effect on mitochondrial DNA copy number or extracellular lactate levels. In CEM cells treated with NHC at 10 μM, a slight decrease (by ∼20%) in mitochondrial DNA copy number and a corresponding slight increase in extracellular lactate levels were detected, but these effects were not enhanced by an increase in NHC treatment concentration. In summary, the results indicate that mitochondrial impairment by NHC is not the main contributor to the compound’s observed cytotoxicity in these cell lines.
Collapse
|
36
|
Longo DM, Woodhead JL, Walker P, Herédi-Szabó K, Mogyorósi K, Wolenski FS, Dragan YP, Mosedale M, Siler SQ, Watkins PB, Howell BA. Quantitative Systems Toxicology Analysis of In Vitro Mechanistic Assays Reveals Importance of Bile Acid Accumulation and Mitochondrial Dysfunction in TAK-875-Induced Liver Injury. Toxicol Sci 2020; 167:458-467. [PMID: 30289550 PMCID: PMC6358270 DOI: 10.1093/toxsci/kfy253] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
TAK-875 (fasiglifam), a GPR40 agonist in development for the treatment of type 2 diabetes (T2D), was voluntarily terminated in Phase III trials due to adverse liver effects. The potential mechanisms of TAK-875 toxicity were explored by combining in vitro experiments with quantitative systems toxicology (QST) using DILIsym, a mathematical representation of drug-induced liver injury. In vitro assays revealed that bile acid transporters were inhibited by both TAK-875 and its metabolite, TAK-875-Glu. Experimental data indicated that human bile salt export pump (BSEP) inhibition by TAK-875 was mixed whereas sodium taurocholate co-transporting polypeptide (NTCP) inhibition by TAK-875 was competitive. Furthermore, experimental data demonstrated that both TAK-875 and TAK-875-Glu inhibit mitochondrial electron transport chain (ETC) enzymes. These mechanistic data were combined with a physiologically based pharmacokinetic (PBPK) model constructed within DILIsym to estimate liver exposure of TAK-875 and TAK-875-Glu. In a simulated population (SimPops) constructed to reflect T2D patients, 16/245 (6.5%) simulated individuals developed alanine aminotransferase (ALT) elevations, an incidence similar to that observed with 200 mg daily dosing in clinical trials. Determining the mode of bile acid transporter inhibition (Ki) was critical to accurate predictions. In addition, simulations conducted on a sensitive subset of individuals (SimCohorts) revealed that when either BSEP or ETC inhibition was inactive, ALT elevations were not predicted to occur, suggesting that the two mechanisms operate synergistically to produce the observed clinical response. These results demonstrate how utilizing QST methods to interpret in vitro experimental results can lead to an improved understanding of the clinically relevant mechanisms underlying drug-induced toxicity.
Collapse
Affiliation(s)
- Diane M Longo
- DILIsym Services, Inc., Research Triangle Park, North Carolina 27709
| | | | | | | | | | - Francis S Wolenski
- Takeda Pharmaceuticals International, Inc., Cambridge, Massachusetts 02139
| | - Yvonne P Dragan
- Takeda Pharmaceuticals International, Inc., Cambridge, Massachusetts 02139
| | - Merrie Mosedale
- UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599.,UNC Institute for Drug Safety Sciences, University of North Carolina at Chapel Hill, Research Triangle Park, North Carolina 27709
| | - Scott Q Siler
- DILIsym Services, Inc., Research Triangle Park, North Carolina 27709
| | - Paul B Watkins
- DILIsym Services, Inc., Research Triangle Park, North Carolina 27709.,UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599.,UNC Institute for Drug Safety Sciences, University of North Carolina at Chapel Hill, Research Triangle Park, North Carolina 27709
| | - Brett A Howell
- DILIsym Services, Inc., Research Triangle Park, North Carolina 27709
| |
Collapse
|
37
|
Generaux G, Lakhani VV, Yang Y, Nadanaciva S, Qiu L, Riccardi K, Di L, Howell BA, Siler SQ, Watkins PB, Barton HA, Aleo MD, Shoda LKM. Quantitative systems toxicology (QST) reproduces species differences in PF-04895162 liver safety due to combined mitochondrial and bile acid toxicity. Pharmacol Res Perspect 2019; 7:e00523. [PMID: 31624633 PMCID: PMC6785660 DOI: 10.1002/prp2.523] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2019] [Revised: 08/17/2019] [Accepted: 08/19/2019] [Indexed: 01/15/2023] Open
Abstract
Many compounds that appear promising in preclinical species, fail in human clinical trials due to safety concerns. The FDA has strongly encouraged the application of modeling in drug development to improve product safety. This study illustrates how DILIsym, a computational representation of liver injury, was able to reproduce species differences in liver toxicity due to PF-04895162 (ICA-105665). PF-04895162, a drug in development for the treatment of epilepsy, was terminated after transaminase elevations were observed in healthy volunteers (NCT01691274). Liver safety concerns had not been raised in preclinical safety studies. DILIsym, which integrates in vitro data on mechanisms of hepatotoxicity with predicted in vivo liver exposure, reproduced clinical hepatotoxicity and the absence of hepatotoxicity observed in the rat. Simulated differences were multifactorial. Simulated liver exposure was greater in humans than rats. The simulated human hepatotoxicity was demonstrated to be due to the interaction between mitochondrial toxicity and bile acid transporter inhibition; elimination of either mechanism from the simulations abrogated injury. The bile acid contribution occurred despite the fact that the IC50 for bile salt export pump (BSEP) inhibition by PF-04895162 was higher (311 µmol/L) than that has been generally thought to contribute to hepatotoxicity. Modeling even higher PF-04895162 liver exposures than were measured in the rat safety studies aggravated mitochondrial toxicity but did not result in rat hepatotoxicity due to insufficient accumulation of cytotoxic bile acid species. This investigative study highlights the potential for combined in vitro and computational screening methods to identify latent hepatotoxic risks and paves the way for similar and prospective studies.
Collapse
Affiliation(s)
- Grant Generaux
- DILIsym Services Inc.Research Triangle ParkNorth Carolina
| | | | - Yuching Yang
- DILIsym Services Inc.Research Triangle ParkNorth Carolina
- Present address:
Division of PharmacometricsOffice of Clinical PharmacologyOffice of Translational SciencesCenter for Drug Evaluation and ResearchFood and Drug Administration Food and Drug AdministrationSilver SpringMaryland
| | - Sashi Nadanaciva
- Compound Safety PredictionWorldwide Medicinal ChemistryPfizer Inc.GrotonConnecticut
| | - Luping Qiu
- Investigative ToxicologyDrug Safety Research and DevelopmentPfizer Inc.GrotonConnecticut
| | - Keith Riccardi
- Pharmacokinetics, Dynamics and MetabolismMedicinal SciencesPfizer Inc.GrotonConnecticut
| | - Li Di
- Pharmacokinetics, Dynamics and MetabolismMedicinal SciencesPfizer Inc.GrotonConnecticut
| | | | - Scott Q. Siler
- DILIsym Services Inc.Research Triangle ParkNorth Carolina
| | - Paul B. Watkins
- UNC Eshelman School of PharmacyUniversity of North Carolina at Chapel HillChapel HillNorth Carolina
- UNC Institute for Drug Safety SciencesUniversity of North Carolina at Chapel HillChapel HillNorth Carolina
| | - Hugh A. Barton
- Translational Modeling and SimulationBiomedicine DesignPfizer, Inc.GrotonConnecticut
| | - Michael D. Aleo
- Investigative ToxicologyDrug Safety Research and DevelopmentPfizer Inc.GrotonConnecticut
| | | |
Collapse
|
38
|
Managing the challenge of drug-induced liver injury: a roadmap for the development and deployment of preclinical predictive models. Nat Rev Drug Discov 2019; 19:131-148. [DOI: 10.1038/s41573-019-0048-x] [Citation(s) in RCA: 89] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/26/2019] [Indexed: 12/13/2022]
|
39
|
Battista C, Yang K, Stahl SH, Mettetal JT, Watkins PB, Siler SQ, Howell BA. Using Quantitative Systems Toxicology to Investigate Observed Species Differences in CKA-Mediated Hepatotoxicity. Toxicol Sci 2019; 166:123-130. [PMID: 30060248 PMCID: PMC6204762 DOI: 10.1093/toxsci/kfy191] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
CKA, a chemokine receptor antagonist intended for treating inflammatory conditions, produced dose-dependent hepatotoxicity in rats but advanced into the clinic where single doses of CKA up to 600 mg appeared safe in humans. Because existing toxicological platforms used during drug development are not perfectly predictive, a quantitative systems toxicology model investigated the hepatotoxic potential of CKA in humans and rats through in vitro assessments of CKA on mitochondrial respiration, oxidative stress, and bile acid transporters. DILIsym predicted that single doses of CKA caused serum ALT >3xULN in a subset of the simulated rat population, while single doses in a simulated human population did not produce serum ALT elevations. Species differences were largely attributed to differences in liver exposure, but increased sensitivity to inhibition of mitochondrial respiration in the rat also contributed. We conclude that mechanistic modeling can elucidate species differences in the hepatotoxic potential of drug candidates.
Collapse
Affiliation(s)
- Christina Battista
- DILIsym Services, Inc., Research Triangle Park, North Carolina.,Division of Pharmacotherapy and Experimental Therapeutics, UNC Institute for Drug Safety Sciences, UNC Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Kyunghee Yang
- DILIsym Services, Inc., Research Triangle Park, North Carolina
| | - Simone H Stahl
- Safety and ADME Translational Sciences, Drug Safety and Metabolism, IMED Biotech Unit, Astra Zeneca R&D, Cambridge CB4 0WG, UK
| | - Jerome T Mettetal
- Safety and ADME Translational Sciences, Drug Safety and Metabolism, IMED Biotech Unit, Astra Zeneca R&D, Waltham, Massachusetts
| | - Paul B Watkins
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Institute for Drug Safety Sciences, UNC Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Scott Q Siler
- DILIsym Services, Inc., Research Triangle Park, North Carolina
| | - Brett A Howell
- DILIsym Services, Inc., Research Triangle Park, North Carolina.,DILIsym Services, Inc., Six Davis Drive, PO BOX 12317, Research Triangle Park, NC 27709
| |
Collapse
|
40
|
Anderson CC, Aivazidis S, Kuzyk CL, Jain A, Roede JR. Acute Maneb Exposure Significantly Alters Both Glycolysis and Mitochondrial Function in Neuroblastoma Cells. Toxicol Sci 2019; 165:61-73. [PMID: 29767788 DOI: 10.1093/toxsci/kfy116] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The pesticides paraquat (PQ) and maneb (MB) have been described as environmental risk factors for Parkinson's disease (PD), with mechanisms associated with mitochondrial dysfunction and reactive oxygen species generation. A combined exposure of PQ and MB in murine models and neuroblastoma cells has been utilized to further advance understanding of the PD phenotype. MB acts as a redox modulator through alkylation of protein thiols and has been previously characterized to inhibit complex III of the electron transport chain and uncouple the mitochondrial proton gradient. The purpose of this study was to analyze ATP-linked respiration and glycolysis in human neuroblastoma cells utilizing the Seahorse extracellular flux platform. Employing an acute, subtoxic exposure of MB, this investigation revealed a MB-mediated decrease in mitochondrial oxygen consumption at baseline and maximal respiration, with inhibition of ATP synthesis and coupling efficiency. Additionally, MB-treated cells showed an increase in nonmitochondrial respiration and proton leak. Further investigation into mitochondrial fuel flex revealed an elimination of fuel flexibility across all 3 major substrates, with a decrease in pyruvate capacity as well as glutamine dependency. Analyses of glycolytic function showed a substantial decrease in glycolytic acidification caused by lactic acid export. This inhibition of glycolytic parameters was also observed after titrating the MB dose as low as 6 μM, and appears to be dependent on the dithiocarbamate functional group, with manganese possibly potentiating the effect. Further studies into cellular ATP and NAD levels revealed a drastic decrease in cells treated with MB. In summary, MB significantly impacted both aerobic and anaerobic energy production; therefore, further characterization of MB's effect on cellular energetics may provide insight into the specificity of PD to dopaminergic neurons.
Collapse
Affiliation(s)
- Colin C Anderson
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Aurora, Colorado 80045
| | - Stefanos Aivazidis
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Aurora, Colorado 80045
| | - Crystal L Kuzyk
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Aurora, Colorado 80045
| | - Abhilasha Jain
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Aurora, Colorado 80045
| | - James R Roede
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Aurora, Colorado 80045
| |
Collapse
|
41
|
Delp J, Funke M, Rudolf F, Cediel A, Bennekou SH, van der Stel W, Carta G, Jennings P, Toma C, Gardner I, van de Water B, Forsby A, Leist M. Development of a neurotoxicity assay that is tuned to detect mitochondrial toxicants. Arch Toxicol 2019; 93:1585-1608. [PMID: 31190196 DOI: 10.1007/s00204-019-02473-y] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 05/07/2019] [Indexed: 12/18/2022]
Abstract
Many neurotoxicants affect energy metabolism in man, but currently available test methods may still fail to predict mito- and neurotoxicity. We addressed this issue using LUHMES cells, i.e., human neuronal precursors that easily differentiate into mature neurons. Within the NeuriTox assay, they have been used to screen for neurotoxicants. Our new approach is based on culturing the cells in either glucose or galactose (Glc-Gal-NeuriTox) as the main carbohydrate source during toxicity testing. Using this Glc-Gal-NeuriTox assay, 52 mitochondrial and non-mitochondrial toxicants were tested. The panel of chemicals comprised 11 inhibitors of mitochondrial respiratory chain complex I (cI), 4 inhibitors of cII, 8 of cIII, and 2 of cIV; 8 toxicants were included as they are assumed to be mitochondrial uncouplers. In galactose, cells became more dependent on mitochondrial function, which made them 2-3 orders of magnitude more sensitive to various mitotoxicants. Moreover, galactose enhanced the specific neurotoxicity (destruction of neurites) compared to a general cytotoxicity (plasma membrane lysis) of the toxicants. The Glc-Gal-NeuriTox assay worked particularly well for inhibitors of cI and cIII, while the toxicity of uncouplers and non-mitochondrial toxicants did not differ significantly upon glucose ↔ galactose exchange. As a secondary assay, we developed a method to quantify the inhibition of all mitochondrial respiratory chain functions/complexes in LUHMES cells. The combination of the Glc-Gal-NeuriTox neurotoxicity screening assay with the mechanistic follow up of target site identification allowed both, a more sensitive detection of neurotoxicants and a sharper definition of the mode of action of mitochondrial toxicants.
Collapse
Affiliation(s)
- Johannes Delp
- Chair for In Vitro Toxicology and Biomedicine, Department of Inaugurated by the Doerenkamp-Zbinden Foundation, University of Konstanz, Universitaetsstr. 10, 78457, Constance, Germany
- Cooperative Doctorate College InViTe, University of Konstanz, Constance, Germany
| | - Melina Funke
- Chair for In Vitro Toxicology and Biomedicine, Department of Inaugurated by the Doerenkamp-Zbinden Foundation, University of Konstanz, Universitaetsstr. 10, 78457, Constance, Germany
| | - Franziska Rudolf
- Chair for In Vitro Toxicology and Biomedicine, Department of Inaugurated by the Doerenkamp-Zbinden Foundation, University of Konstanz, Universitaetsstr. 10, 78457, Constance, Germany
| | - Andrea Cediel
- Swetox Unit for Toxicological Sciences, Karolinska Institutet, Stockholm, Sweden
| | | | - Wanda van der Stel
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands
| | - Giada Carta
- Division of Molecular and Computational Toxicology, Amsterdam Institute for Molecules, Medicines and Systems, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Paul Jennings
- Division of Molecular and Computational Toxicology, Amsterdam Institute for Molecules, Medicines and Systems, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Cosimo Toma
- Laboratory of Environmental Chemistry and Toxicology, Department of Environmental Health Sciences, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via la Masa 19, 20156, Milan, Italy
| | | | - Bob van de Water
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands
| | - Anna Forsby
- Swetox Unit for Toxicological Sciences, Karolinska Institutet, Stockholm, Sweden
- Department of Biochemistry and Biophysics, Stockholm University, Stockholm, Sweden
| | - Marcel Leist
- Chair for In Vitro Toxicology and Biomedicine, Department of Inaugurated by the Doerenkamp-Zbinden Foundation, University of Konstanz, Universitaetsstr. 10, 78457, Constance, Germany.
| |
Collapse
|
42
|
Wang X, Cirit M, Wishnok JS, Griffith LG, Tannenbaum SR. Analysis of an Integrated Human Multiorgan Microphysiological System for Combined Tolcapone Metabolism and Brain Metabolomics. Anal Chem 2019; 91:8667-8675. [DOI: 10.1021/acs.analchem.9b02224] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
43
|
Sousa ML, Preto M, Vasconcelos V, Linder S, Urbatzka R. Antiproliferative Effects of the Natural Oxadiazine Nocuolin A Are Associated With Impairment of Mitochondrial Oxidative Phosphorylation. Front Oncol 2019; 9:224. [PMID: 31001482 PMCID: PMC6456697 DOI: 10.3389/fonc.2019.00224] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Accepted: 03/13/2019] [Indexed: 11/20/2022] Open
Abstract
Natural products are interesting sources for drug discovery. The natural product oxadiazine Nocuolin A (NocA) was previously isolated from the cyanobacterial strain Nodularia sp. LEGE 06071 and here we examined its cytotoxic effects against different strains of the colon cancer cell line HCT116 and the immortalized epithelial cell line hTERT RPE-1. NocA was cytotoxic against colon cancer cells and immortalized cells under conditions of exponential growth but was only weakly active against non-proliferating immortalized cells. NocA induced apoptosis by mechanism(s) resistant to overexpression of BCL family members. Interestingly, NocA affected viability and induced apoptosis of HCT116 cells grown as multicellular spheroids. Analysis of transcriptome profiles did not match signatures to any known compounds in CMap but indicated stress responses and induction of cell starvation. Evidence for autophagy was observed, and a decrease in various mitochondrial respiration parameter within 1 h of treatment. These results are consistent with previous findings showing that nutritionally compromised cells in spheroids are sensitive to impairment of mitochondrial energy production due to limited metabolic plasticity. We conclude that the antiproliferative effects of NocA are associated with effects on mitochondrial oxidative phosphorylation.
Collapse
Affiliation(s)
- Maria Lígia Sousa
- Faculty of Sciences of University of Porto, Porto, Portugal.,Interdisciplinary Centre of Marine and Environmental Research, Porto, Portugal
| | - Marco Preto
- Interdisciplinary Centre of Marine and Environmental Research, Porto, Portugal
| | - Vítor Vasconcelos
- Faculty of Sciences of University of Porto, Porto, Portugal.,Interdisciplinary Centre of Marine and Environmental Research, Porto, Portugal
| | - Stig Linder
- Department of Oncology and Pathology, Cancer Centre Karolinska, Karolinska Institute, Stockholm, Sweden.,Department of Medical and Health Sciences, Linköping University, Linköping, Sweden
| | - Ralph Urbatzka
- Interdisciplinary Centre of Marine and Environmental Research, Porto, Portugal
| |
Collapse
|
44
|
Sarni AR, Baroni L. Milk and Parkinson disease: Could galactose be the missing link. MEDITERRANEAN JOURNAL OF NUTRITION AND METABOLISM 2019. [DOI: 10.3233/mnm-180234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Affiliation(s)
| | - Luciana Baroni
- Primary Care Unit, Northern District, Local Health Unit 2 Marca Trevigiana, Treviso, Italy
| |
Collapse
|
45
|
Burgos-Aceves MA, Cohen A, Paolella G, Lepretti M, Smith Y, Faggio C, Lionetti L. Modulation of mitochondrial functions by xenobiotic-induced microRNA: From environmental sentinel organisms to mammals. THE SCIENCE OF THE TOTAL ENVIRONMENT 2018; 645:79-88. [PMID: 30015121 DOI: 10.1016/j.scitotenv.2018.07.109] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 07/09/2018] [Accepted: 07/09/2018] [Indexed: 06/08/2023]
Abstract
Mitochondria play a crucial role in energetic metabolism, signaling pathways, and overall cell viability. They are in the first line in facing cellular energy requirements in stress conditions, such as in response to xenobiotic exposure. Recently, a novel regulatory key role of microRNAs (miRNAs) in important signaling pathways in mitochondria has been proposed. Consequently, alteration in miRNAs expression by xenobiotics could outcome into mitochondrial dysfunction, reactive oxygen species overexpression, and liberation of apoptosis or necrosis activating proteins. The aim of this review is to show the highlights about mitochondria-associated miRNAs in cellular processes exposed to xenobiotic stress in different cell types involved in detoxification processes or sensitive to environmental hazards in marine sentinel organisms and mammals.
Collapse
Affiliation(s)
- Mario Alberto Burgos-Aceves
- Department of Chemistry and Biology, University of Salerno, via Giovanni Paolo II, 132, 84084 Fisciano, SA, Italy
| | - Amit Cohen
- Genomic Data Analysis Unit, The Hebrew University of Jerusalem-Hadassah Medical School, P.O. Box 12272, Jerusalem 91120, Israel
| | - Gaetana Paolella
- Department of Chemistry and Biology, University of Salerno, via Giovanni Paolo II, 132, 84084 Fisciano, SA, Italy
| | - Marilena Lepretti
- Department of Chemistry and Biology, University of Salerno, via Giovanni Paolo II, 132, 84084 Fisciano, SA, Italy
| | - Yoav Smith
- Genomic Data Analysis Unit, The Hebrew University of Jerusalem-Hadassah Medical School, P.O. Box 12272, Jerusalem 91120, Israel
| | - Caterina Faggio
- Department of Chemical, Biological, Pharmaceutical, and Environmental Sciences, University of Messina, Viale F. Stagno d'Alcontres, 31, 98166 Messina, Italy.
| | - Lillà Lionetti
- Department of Chemistry and Biology, University of Salerno, via Giovanni Paolo II, 132, 84084 Fisciano, SA, Italy
| |
Collapse
|
46
|
Rana P, Aleo MD, Gosink M, Will Y. Evaluation of in Vitro Mitochondrial Toxicity Assays and Physicochemical Properties for Prediction of Organ Toxicity Using 228 Pharmaceutical Drugs. Chem Res Toxicol 2018; 32:156-167. [DOI: 10.1021/acs.chemrestox.8b00246] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- Payal Rana
- Drug Safety Research & Development, Pfizer, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Michael D. Aleo
- Drug Safety Research & Development, Pfizer, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Mark Gosink
- Drug Safety Research & Development, Pfizer, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Yvonne Will
- Drug Safety Research & Development, Pfizer, Eastern Point Road, Groton, Connecticut 06340, United States
| |
Collapse
|
47
|
Kamalian L, Douglas O, Jolly CE, Snoeys J, Simic D, Monshouwer M, Williams DP, Kevin Park B, Chadwick AE. The utility of HepaRG cells for bioenergetic investigation and detection of drug-induced mitochondrial toxicity. Toxicol In Vitro 2018; 53:136-147. [PMID: 30096366 DOI: 10.1016/j.tiv.2018.08.001] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Revised: 07/03/2018] [Accepted: 08/05/2018] [Indexed: 12/12/2022]
Abstract
The importance of mitochondrial toxicity in drug-induced liver injury is well established. The bioenergetic phenotype of the HepaRG cell line was defined in order to assess their suitability as a model of mitochondrial hepatotoxicity. Bioenergetic phenotyping categorised the HepaRG cells as less metabolically active when measured beside the more energetic HepG2 cells. However, inhibition of mitochondrial ATP synthase induced an increase in glycolytic activity of both HepaRG and HepG2 cells suggesting an active Crabtree Effect in both cell lines. The suitability of HepaRG cells for the acute metabolic modification assay as a screen for mitotoxicity was confirmed using a panel of compounds, including both positive and negative mitotoxic compounds. Seahorse respirometry studies demonstrated that a statistically significant decrease in spare respiratory capacity is the first indication of mitochondrial dysfunction. Furthermore, based upon comparing changes in respiratory parameters to those of the positive controls, rotenone and carbonyl cyanide m-chlorophenyl hydrazone, compounds were categorised into two mechanistic groups; inhibitors or uncouplers of the electron transport chain. Overall, the findings from this study have demonstrated that HepaRG cells, despite having different resting bioenergetic phenotype to HepG2 cells are a suitable model to detect drug-induced mitochondrial toxicity with similar detection rates to HepG2 cells.
Collapse
Affiliation(s)
- Laleh Kamalian
- MRC Centre for Drug Safety Science, The Department of Clinical and Molecular Pharmacology, The University of Liverpool, Ashton Street, Liverpool L69 3GE, United Kingdom.
| | - Oisin Douglas
- MRC Centre for Drug Safety Science, The Department of Clinical and Molecular Pharmacology, The University of Liverpool, Ashton Street, Liverpool L69 3GE, United Kingdom
| | - Carol E Jolly
- MRC Centre for Drug Safety Science, The Department of Clinical and Molecular Pharmacology, The University of Liverpool, Ashton Street, Liverpool L69 3GE, United Kingdom.
| | - Jan Snoeys
- Pharmacokinetics Dynamics and Metabolism, Janssen Research and Development, Beerse, Belgium.
| | - Damir Simic
- Mechanistic and Investigative Toxicology, Janssen Research and Development, Spring House, PA, USA.
| | - Mario Monshouwer
- Pharmacokinetics Dynamics and Metabolism, Janssen Research and Development, Beerse, Belgium
| | - Dominic P Williams
- Innovative Medicines and Early Development
- Drug Safety and Metabolism
- Translational Safety, Darwin Building 310, Cambridge Science Park, Milton Rd, Cambridge, CB4 0FZ, United Kingdom.
| | - B Kevin Park
- MRC Centre for Drug Safety Science, The Department of Clinical and Molecular Pharmacology, The University of Liverpool, Ashton Street, Liverpool L69 3GE, United Kingdom.
| | - Amy E Chadwick
- MRC Centre for Drug Safety Science, The Department of Clinical and Molecular Pharmacology, The University of Liverpool, Ashton Street, Liverpool L69 3GE, United Kingdom.
| |
Collapse
|
48
|
Li E, Bolser DG, Kroll KJ, Brockmeier EK, Falciani F, Denslow ND. Comparative toxicity of three phenolic compounds on the embryo of fathead minnow, Pimephales promelas. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2018; 201:66-72. [PMID: 29879596 DOI: 10.1016/j.aquatox.2018.05.024] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Revised: 05/25/2018] [Accepted: 05/26/2018] [Indexed: 06/08/2023]
Abstract
Phenols are classified as polar narcotics, which are thought to cause toxicity by non-specific mechanisms, possibly by disrupting membrane structure and function. Here we test three phenolic chemicals, phenol, 2,4-dichlorphenol and pentachlorophenol on embryo development, heartbeat rate and mitochondrial respiration in fathead minnow (Pimephales promelas). While these chemicals have been used on isolated mitochondria, they have not yet been used to verify respiration in intact embryos. Mitochondrial respiration in intact embryos was measured after optimizing the Seahorse XFe24 Extracellular Flux Analyzer. Heartbeat rate and mitochondrial respiration patterns of fathead minnow embryos at different developmental stages were also characterized. Exposures of embryos at developmental stage 20 occurred for 24 h with five concentrations of each phenolic compound ranging from 0.85 to 255 μM for phenol, 0.49 to 147 μM for 2,4-dichlorophenol and 0.3 to 90 μM for pentachlorophenol. Exposure to phenol at the concentrations tested had no effects on development, heartbeat or mitochondrial respiration. However, both 2,4-dichlorophenol and pentachlorophenol showed dose-dependent effects on development, heartbeat rate, and mitochondrial respiration, with the effects occurring at lower concentrations of pentachlorophenol, compared to 2,4-dichlorophenol, highlighting the higher toxicity of the more chlorinated phenols. Both 2,4-dichlorophenol and pentachlorophenol decreased basal mitochondrial respiration of embryos and ATP production. These results indicate that higher chlorinated phenolic chemicals cause developmental toxicity in fathead minnow embryos by decreasing mitochondrial respiration and heartbeat rate.
Collapse
Affiliation(s)
- Erchao Li
- Department of Physiological Sciences and Center for Environmental and Human Toxicology, University of Florida, Gainesville, FL 32611, USA; College of Marine Sciences, Hainan University, Haikou, Hainan 570228, China
| | - Derek G Bolser
- Department of Physiological Sciences and Center for Environmental and Human Toxicology, University of Florida, Gainesville, FL 32611, USA
| | - Kevin J Kroll
- Department of Physiological Sciences and Center for Environmental and Human Toxicology, University of Florida, Gainesville, FL 32611, USA
| | - Erica K Brockmeier
- Institute of Integrative Biology, University of Liverpool, Liverpool, United Kingdom
| | - Francesco Falciani
- Institute of Integrative Biology, University of Liverpool, Liverpool, United Kingdom
| | - Nancy D Denslow
- Department of Physiological Sciences and Center for Environmental and Human Toxicology, University of Florida, Gainesville, FL 32611, USA.
| |
Collapse
|
49
|
In vitro screening of cell bioenergetics to assess mitochondrial dysfunction in drug development. Toxicol In Vitro 2018; 52:374-383. [PMID: 30030051 DOI: 10.1016/j.tiv.2018.07.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2018] [Revised: 05/28/2018] [Accepted: 07/15/2018] [Indexed: 12/17/2022]
Abstract
Drug-induced mitochondrial toxicity is considered as a common cellular mechanism that can induce a variety of organ toxicities. In the present manuscript, 17 in vitro mitochondrial toxic drugs, reported to induce Drug-Induced Liver Injury (DILI) and 6 non-mitochondrial toxic drugs (3 with DILI and 3 without DILI concern), were tested in HepG2 cells using a bioenergetics system. The 17 mitochondrial toxic drugs represent a wide variety of mitochondrial dysfunctions as well as DILI and include 4 pairs of drugs which are structurally related but associated with different DILI concerns in human. Cell bioenergetics were measured using the XF96e analyzer which simultaneous monitor oxygen consumption rate (OCR) and extracellular acidification rate (ECAR), indirect measurements of oxidative phosphorylation and glycolysis, respectively. OCR associated with ATP production, maximal respiration, proton leak and spare respiratory capacity, were also assessed. Duplicate experiments resulted in a sensitivity of 82% (14/17) and specificity of 83% (5/6). The addition of stressors improved specificity considerably. Cut-offs, statistics and rules are clearly discussed to facilitate the use of this assay for screening purposes. Overall, the authors consider that this assay should be part of the battery of safety screening assays at early stages of drug development.
Collapse
|
50
|
Le Guillou D, Bucher S, Begriche K, Hoët D, Lombès A, Labbe G, Fromenty B. Drug-Induced Alterations of Mitochondrial DNA Homeostasis in Steatotic and Nonsteatotic HepaRG Cells. J Pharmacol Exp Ther 2018; 365:711-726. [PMID: 29669730 DOI: 10.1124/jpet.117.246751] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Accepted: 04/06/2018] [Indexed: 12/19/2022] Open
Abstract
Although mitochondriotoxicity plays a major role in drug-induced hepatotoxicity, alteration of mitochondrial DNA (mtDNA) homeostasis has been described only with a few drugs. Because it requires long drug exposure, this mechanism of toxicity cannot be detected with investigations performed in isolated liver mitochondria or cultured cells exposed to drugs for several hours or a few days. Thus, a first aim of this study was to determine whether a 2-week treatment with nine hepatotoxic drugs could affect mtDNA homeostasis in HepaRG cells. Previous investigations with these drugs showed rapid toxicity on oxidative phosphorylation but did not address the possibility of delayed toxicity secondary to mtDNA homeostasis impairment. The maximal concentration used for each drug induced about 10% cytotoxicity. Two other drugs, zalcitabine and linezolid, were used as positive controls for their respective effects on mtDNA replication and translation. Another goal was to determine whether drug-induced mitochondriotoxicity could be modulated by lipid overload mimicking nonalcoholic fatty liver. Among the nine drugs, imipramine and ritonavir induced mitochondrial effects suggesting alteration of mtDNA translation. Ritonavir toxicity was stronger in nonsteatotic cells. None of the nine drugs decreased mtDNA levels. However, increased mtDNA was observed with five drugs, especially in nonsteatotic cells. The mtDNA levels could not be correlated with the expression of key factors involved in mitochondrial biogenesis, such as peroxisome proliferator-activated receptor-γ coactivator 1α (PGC1α), PGC1β, and AMP-activated protein kinase α-subunit. Hence, drug-induced impairment of mtDNA translation might not be rare, and increased mtDNA levels could be a frequent adaptive response to slight energy shortage. Nevertheless, this adaptation could be impaired by lipid overload.
Collapse
Affiliation(s)
- Dounia Le Guillou
- INSERM, INRA, Université de Rennes, UBL, Nutrition Metabolisms and Cancer (NuMeCan), Rennes, France (D.L.G., S.B., K.B., B.F.); Sanofi, Investigative Toxicology, Alfortville, France (D.H., G.L.); and INSERM, UMR 1016, Institut Cochin, Université Paris V René Descartes, Paris, France (A.L.)
| | - Simon Bucher
- INSERM, INRA, Université de Rennes, UBL, Nutrition Metabolisms and Cancer (NuMeCan), Rennes, France (D.L.G., S.B., K.B., B.F.); Sanofi, Investigative Toxicology, Alfortville, France (D.H., G.L.); and INSERM, UMR 1016, Institut Cochin, Université Paris V René Descartes, Paris, France (A.L.)
| | - Karima Begriche
- INSERM, INRA, Université de Rennes, UBL, Nutrition Metabolisms and Cancer (NuMeCan), Rennes, France (D.L.G., S.B., K.B., B.F.); Sanofi, Investigative Toxicology, Alfortville, France (D.H., G.L.); and INSERM, UMR 1016, Institut Cochin, Université Paris V René Descartes, Paris, France (A.L.)
| | - Delphine Hoët
- INSERM, INRA, Université de Rennes, UBL, Nutrition Metabolisms and Cancer (NuMeCan), Rennes, France (D.L.G., S.B., K.B., B.F.); Sanofi, Investigative Toxicology, Alfortville, France (D.H., G.L.); and INSERM, UMR 1016, Institut Cochin, Université Paris V René Descartes, Paris, France (A.L.)
| | - Anne Lombès
- INSERM, INRA, Université de Rennes, UBL, Nutrition Metabolisms and Cancer (NuMeCan), Rennes, France (D.L.G., S.B., K.B., B.F.); Sanofi, Investigative Toxicology, Alfortville, France (D.H., G.L.); and INSERM, UMR 1016, Institut Cochin, Université Paris V René Descartes, Paris, France (A.L.)
| | - Gilles Labbe
- INSERM, INRA, Université de Rennes, UBL, Nutrition Metabolisms and Cancer (NuMeCan), Rennes, France (D.L.G., S.B., K.B., B.F.); Sanofi, Investigative Toxicology, Alfortville, France (D.H., G.L.); and INSERM, UMR 1016, Institut Cochin, Université Paris V René Descartes, Paris, France (A.L.)
| | - Bernard Fromenty
- INSERM, INRA, Université de Rennes, UBL, Nutrition Metabolisms and Cancer (NuMeCan), Rennes, France (D.L.G., S.B., K.B., B.F.); Sanofi, Investigative Toxicology, Alfortville, France (D.H., G.L.); and INSERM, UMR 1016, Institut Cochin, Université Paris V René Descartes, Paris, France (A.L.)
| |
Collapse
|