1
|
Yang Y, Chen Q, Liu Z, Huang T, Hong Y, Li N, Ai K, Huang Q. Novel reduced heteropolyacid nanoparticles for effective treatment of drug-induced liver injury by manipulating reactive oxygen and nitrogen species and inflammatory signals. J Colloid Interface Sci 2025; 678:174-187. [PMID: 39243718 DOI: 10.1016/j.jcis.2024.08.239] [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: 06/14/2024] [Revised: 08/07/2024] [Accepted: 08/29/2024] [Indexed: 09/09/2024]
Abstract
With the rapid advancements in biomedicine, the use of clinical drugs has surged sharply. However, potential hepatotoxicity limits drug exploitation and widespread usage, posing serious threats to patient health. Hepatotoxic drugs disrupt liver enzyme levels and cause refractory pathological damage, creating a challenge in the application of diverse first-line drugs. The activation and deterioration of reactive oxygen and nitrogen species (RONS) and inflammatory signals are key pathological mechanisms of drug-induced liver injury (DILI). Herein, a novel reduced heteropolyacid nanoparticle (RNP) has been developed, possessing high RONS-scavenging ability, strong anti-inflammatory activity, and excellent biosafety. These features enable it to swiftly restore the redox and immune balance of the liver. Intravenous administration of RNP effectively scavenged RONS storm, reversing liver oxidative stress and restoring normal mitochondrial membrane potential and function. Furthermore, by inhibiting c-Jun-N-terminal kinase phosphorylation, RNP facilitated the restoration of nuclear factor erythroid 2-related factor 2-mediated endogenous antioxidant signaling, ultimately rescuing the liver function and tissue morphology in acetaminophen-induced DILI mice. Crucially, the high biocompatible RNP exhibited superior efficacy in the DILI mouse model compared to the clinical antioxidant N-acetylcysteine. This targeted therapeutic approach, tailored to address the onset and progression of DILI, offers valuable new insights into controlling the condition and restoring liver structure and function.
Collapse
Affiliation(s)
- Yongqi Yang
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410078, China; Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410078, China
| | - Qiaohui Chen
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410078, China; Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410078, China
| | - Zerun Liu
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Ting Huang
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Ying Hong
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Niansheng Li
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410078, China; Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410078, China.
| | - Kelong Ai
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410078, China; Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410078, China
| | - Qiong Huang
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China.
| |
Collapse
|
2
|
Liu M, Ning Z, Cheng Y, Zheng Z, Yang X, Zheng T, Li N, Wu JL. The key to 2,6-dichloro-1,4-benzoquinone reproductive toxicity and green tea detoxification: Covalent binding and competitive binding. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 286:117239. [PMID: 39454356 DOI: 10.1016/j.ecoenv.2024.117239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Revised: 09/24/2024] [Accepted: 10/19/2024] [Indexed: 10/28/2024]
Abstract
Halobenzoquinones (HBQs) are ubiquitous disinfection by-products (DBPs) in chlorinated drinking water with various health risks including reproductive toxicity, while the potential mechanisms are still unclear. Although green tea exhibits common detoxifying properties, its ability to mitigate the toxicity of HBQs still needs to be further deepened and explored. This study attempted to investigate the possible mechanism of the most common HBQ, 2,6-dichloro-1,4-benzoquinone (2,6-DCBQ) induced reproductive toxicity and elucidate the protective effect of green tea using a series of liquid chromatography-tandem mass spectrometry (LC-MS) approaches. Firstly, in vivo experiments showed that 2,6-DCBQ could induce testicular damage in male rats via significantly decreasing sperm-associated Leydig cells and seminiferous tubules. Then, in vitro incubation of 2,6-DCBQ with amino acids suggested that 2,6-DCBQ could bind to proteins via residues of cysteine or lysine and provided five additional modification patterns. Following, proteomics analysis revealed that at least 42 proteins were modified by 2,6-DCBQ, which were mainly enriched in the reproductive system. These results highlighted the significance of covalent protein modification in 2,6-DCBQ reproductive toxicity. Fortunately, we found that catechins (a class of major components of green tea) could competitively bind to 2,6-DCBQ in vivo and in vitro, reducing the amount and type of 2,6-DCBQ-protein adducts, thereby attenuating the reproductive system damage caused by 2,6-DCBQ. This study provides new insights into 2,6-DCBQ-induced reproductive system damage and reveals a new mechanism of green tea detoxification. Moreover, these findings offer potential strategies for alleviating the harmful impacts of environmental toxicants on human health.
Collapse
Affiliation(s)
- Meixian Liu
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, 999078, Macao, China; BayRay Innovation Center, Shenzhen Bay Laboratory, Shenzhen 518132, China
| | - Zhiyuan Ning
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, 999078, Macao, China
| | - Yong Cheng
- Zhejiang Skyherb Biotechnology Inc., Huzhou 313300, China
| | - Zhiyuan Zheng
- Faculty of Pharmaceutical Sciences, Shenzhen University of Advanced Technology, Shenzhen 518107, China; Center for Cancer Immunology, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Xiaoxue Yang
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, 999078, Macao, China
| | - Ting Zheng
- Multi-omics Mass Spectrometry Core, Biomedical Research Core Facilities, Shenzhen Bay Laboratory, Shenzhen 518132, China
| | - Na Li
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, 999078, Macao, China.
| | - Jian-Lin Wu
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, 999078, Macao, China.
| |
Collapse
|
3
|
Offer S, Di Bucchianico S, Czech H, Pardo M, Pantzke J, Bisig C, Schneider E, Bauer S, Zimmermann EJ, Oeder S, Hartner E, Gröger T, Alsaleh R, Kersch C, Ziehm T, Hohaus T, Rüger CP, Schmitz-Spanke S, Schnelle-Kreis J, Sklorz M, Kiendler-Scharr A, Rudich Y, Zimmermann R. The chemical composition of secondary organic aerosols regulates transcriptomic and metabolomic signaling in an epithelial-endothelial in vitro coculture. Part Fibre Toxicol 2024; 21:38. [PMID: 39300536 DOI: 10.1186/s12989-024-00600-x] [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: 02/02/2024] [Accepted: 09/10/2024] [Indexed: 09/22/2024] Open
Abstract
BACKGROUND The formation of secondary organic aerosols (SOA) by atmospheric oxidation reactions substantially contributes to the burden of fine particulate matter (PM2.5), which has been associated with adverse health effects (e.g., cardiovascular diseases). However, the molecular and cellular effects of atmospheric aging on aerosol toxicity have not been fully elucidated, especially in model systems that enable cell-to-cell signaling. METHODS In this study, we aimed to elucidate the complexity of atmospheric aerosol toxicology by exposing a coculture model system consisting of an alveolar (A549) and an endothelial (EA.hy926) cell line seeded in a 3D orientation at the air‒liquid interface for 4 h to model aerosols. Simulation of atmospheric aging was performed on volatile biogenic (β-pinene) or anthropogenic (naphthalene) precursors of SOA condensing on soot particles. The similar physical properties for both SOA, but distinct differences in chemical composition (e.g., aromatic compounds, oxidation state, unsaturated carbonyls) enabled to determine specifically induced toxic effects of SOA. RESULTS In A549 cells, exposure to naphthalene-derived SOA induced stress-related airway remodeling and an early type I immune response to a greater extent. Transcriptomic analysis of EA.hy926 cells not directly exposed to aerosol and integration with metabolome data indicated generalized systemic effects resulting from the activation of early response genes and the involvement of cardiovascular disease (CVD) -related pathways, such as the intracellular signal transduction pathway (PI3K/AKT) and pathways associated with endothelial dysfunction (iNOS; PDGF). Greater induction following anthropogenic SOA exposure might be causative for the observed secondary genotoxicity. CONCLUSION Our findings revealed that the specific effects of SOA on directly exposed epithelial cells are highly dependent on the chemical identity, whereas non directly exposed endothelial cells exhibit more generalized systemic effects with the activation of early stress response genes and the involvement of CVD-related pathways. However, a greater correlation was made between the exposure to the anthropogenic SOA compared to the biogenic SOA. In summary, our study highlights the importance of chemical aerosol composition and the use of cell systems with cell-to-cell interplay on toxicological outcomes.
Collapse
Affiliation(s)
- Svenja Offer
- Joint Mass Spectrometry Center (JMSC) at Comprehensive Molecular Analytics (CMA), Helmholtz Zentrum München, Ingolstädter Landstr. 1, D-85764, Neuherberg, Germany
- Joint Mass Spectrometry Center (JMSC) at Analytical Chemistry, Institute of Chemistry, University of Rostock, Albert-Einstein-Str. 27, D-18059, Rostock, Germany
| | - Sebastiano Di Bucchianico
- Joint Mass Spectrometry Center (JMSC) at Comprehensive Molecular Analytics (CMA), Helmholtz Zentrum München, Ingolstädter Landstr. 1, D-85764, Neuherberg, Germany.
- Joint Mass Spectrometry Center (JMSC) at Analytical Chemistry, Institute of Chemistry, University of Rostock, Albert-Einstein-Str. 27, D-18059, Rostock, Germany.
- Department Life, Light & Matter (LLM), University of Rostock, D-18051, Rostock, Germany.
| | - Hendryk Czech
- Joint Mass Spectrometry Center (JMSC) at Comprehensive Molecular Analytics (CMA), Helmholtz Zentrum München, Ingolstädter Landstr. 1, D-85764, Neuherberg, Germany
- Joint Mass Spectrometry Center (JMSC) at Analytical Chemistry, Institute of Chemistry, University of Rostock, Albert-Einstein-Str. 27, D-18059, Rostock, Germany
| | - Michal Pardo
- Department of Earth and Planetary Sciences, Faculty of Chemistry, Weizmann Institute of Science, 234 Herzl Street, POB 26, Rehovot, ISR-7610001, Israel
| | - Jana Pantzke
- Joint Mass Spectrometry Center (JMSC) at Comprehensive Molecular Analytics (CMA), Helmholtz Zentrum München, Ingolstädter Landstr. 1, D-85764, Neuherberg, Germany
- Joint Mass Spectrometry Center (JMSC) at Analytical Chemistry, Institute of Chemistry, University of Rostock, Albert-Einstein-Str. 27, D-18059, Rostock, Germany
| | - Christoph Bisig
- Joint Mass Spectrometry Center (JMSC) at Comprehensive Molecular Analytics (CMA), Helmholtz Zentrum München, Ingolstädter Landstr. 1, D-85764, Neuherberg, Germany
| | - Eric Schneider
- Joint Mass Spectrometry Center (JMSC) at Analytical Chemistry, Institute of Chemistry, University of Rostock, Albert-Einstein-Str. 27, D-18059, Rostock, Germany
- Department Life, Light & Matter (LLM), University of Rostock, D-18051, Rostock, Germany
| | - Stefanie Bauer
- Joint Mass Spectrometry Center (JMSC) at Comprehensive Molecular Analytics (CMA), Helmholtz Zentrum München, Ingolstädter Landstr. 1, D-85764, Neuherberg, Germany
| | - Elias J Zimmermann
- Joint Mass Spectrometry Center (JMSC) at Comprehensive Molecular Analytics (CMA), Helmholtz Zentrum München, Ingolstädter Landstr. 1, D-85764, Neuherberg, Germany
- Joint Mass Spectrometry Center (JMSC) at Analytical Chemistry, Institute of Chemistry, University of Rostock, Albert-Einstein-Str. 27, D-18059, Rostock, Germany
| | - Sebastian Oeder
- Joint Mass Spectrometry Center (JMSC) at Comprehensive Molecular Analytics (CMA), Helmholtz Zentrum München, Ingolstädter Landstr. 1, D-85764, Neuherberg, Germany
| | - Elena Hartner
- Joint Mass Spectrometry Center (JMSC) at Comprehensive Molecular Analytics (CMA), Helmholtz Zentrum München, Ingolstädter Landstr. 1, D-85764, Neuherberg, Germany
- Joint Mass Spectrometry Center (JMSC) at Analytical Chemistry, Institute of Chemistry, University of Rostock, Albert-Einstein-Str. 27, D-18059, Rostock, Germany
| | - Thomas Gröger
- Joint Mass Spectrometry Center (JMSC) at Comprehensive Molecular Analytics (CMA), Helmholtz Zentrum München, Ingolstädter Landstr. 1, D-85764, Neuherberg, Germany
- Joint Mass Spectrometry Center (JMSC) at Analytical Chemistry, Institute of Chemistry, University of Rostock, Albert-Einstein-Str. 27, D-18059, Rostock, Germany
| | - Rasha Alsaleh
- Institute and Outpatient Clinic of Occupational, Social and Environmental Medicine, Friedrich-Alexander University of Erlangen-Nuremberg, Henkestr. 9-11, D-91054, Erlangen, Germany
| | - Christian Kersch
- Institute and Outpatient Clinic of Occupational, Social and Environmental Medicine, Friedrich-Alexander University of Erlangen-Nuremberg, Henkestr. 9-11, D-91054, Erlangen, Germany
| | - Till Ziehm
- Institute of Energy and Climate Research, Forschungszentrum Jülich GmbH, Troposphere (IEK-8), Wilhelm- Johen-Str, D-52428, Jülich, Germany
| | - Thorsten Hohaus
- Institute of Energy and Climate Research, Forschungszentrum Jülich GmbH, Troposphere (IEK-8), Wilhelm- Johen-Str, D-52428, Jülich, Germany
| | - Christopher P Rüger
- Joint Mass Spectrometry Center (JMSC) at Analytical Chemistry, Institute of Chemistry, University of Rostock, Albert-Einstein-Str. 27, D-18059, Rostock, Germany
- Department Life, Light & Matter (LLM), University of Rostock, D-18051, Rostock, Germany
| | - Simone Schmitz-Spanke
- Institute and Outpatient Clinic of Occupational, Social and Environmental Medicine, Friedrich-Alexander University of Erlangen-Nuremberg, Henkestr. 9-11, D-91054, Erlangen, Germany
| | - Jürgen Schnelle-Kreis
- Joint Mass Spectrometry Center (JMSC) at Comprehensive Molecular Analytics (CMA), Helmholtz Zentrum München, Ingolstädter Landstr. 1, D-85764, Neuherberg, Germany
| | - Martin Sklorz
- Joint Mass Spectrometry Center (JMSC) at Comprehensive Molecular Analytics (CMA), Helmholtz Zentrum München, Ingolstädter Landstr. 1, D-85764, Neuherberg, Germany
| | - Astrid Kiendler-Scharr
- Institute of Energy and Climate Research, Forschungszentrum Jülich GmbH, Troposphere (IEK-8), Wilhelm- Johen-Str, D-52428, Jülich, Germany
| | - Yinon Rudich
- Department of Earth and Planetary Sciences, Faculty of Chemistry, Weizmann Institute of Science, 234 Herzl Street, POB 26, Rehovot, ISR-7610001, Israel
| | - Ralf Zimmermann
- Joint Mass Spectrometry Center (JMSC) at Comprehensive Molecular Analytics (CMA), Helmholtz Zentrum München, Ingolstädter Landstr. 1, D-85764, Neuherberg, Germany
- Joint Mass Spectrometry Center (JMSC) at Analytical Chemistry, Institute of Chemistry, University of Rostock, Albert-Einstein-Str. 27, D-18059, Rostock, Germany
- Department Life, Light & Matter (LLM), University of Rostock, D-18051, Rostock, Germany
| |
Collapse
|
4
|
Xiang Q, Li N, Zhang Y, Wang T, Wang Y, Bian J. GPR116 alleviates acetaminophen-induced liver injury in mice by inhibiting endoplasmic reticulum stress. Cell Mol Life Sci 2024; 81:299. [PMID: 39001944 PMCID: PMC11335223 DOI: 10.1007/s00018-024-05313-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 05/30/2024] [Accepted: 06/10/2024] [Indexed: 07/15/2024]
Abstract
BACKGROUND Acetaminophen (APAP) overdose is a significant contributor to drug-induced liver injury worldwide. G-protein-coupled receptor 116 (GPR116) is an important homeostatic maintenance molecule in the body, but little is known about its role in APAP-induced liver injury (AILI). METHODS GPR116 expression was determined in both human and mouse AILI models. Hepatic function and damage response were analyzed in hepatocyte-specific GPR116 deletion (GPR116△HC) mice undergoing APAP challenge. RNA-sequencing, immunofluorescence confocal, and co-immunoprecipitation (CO-IP) were employed to elucidate the impact and underlying mechanisms of GPR116 in AILI. RESULTS Intrahepatic GPR116 was upregulated in human and mice with AILI. GPR116△HC mice were vulnerable to AILI compared to wild-type mice. Overexpression of GPR116 effectively mitigated AILI in wild-type mice and counteracted the heightened susceptibility of GPR116△HC mice to APAP. Mechanistically, GPR116 inhibits the binding immunoglobulin protein (BiP), a critical regulator of ER function, through its interaction with β-arrestin1, thereby mitigating ER stress during the early stage of AILI. Additionally, the activation of GPR116 by ligand FNDC4 has been shown to confer a protective effect against early hepatotoxicity caused by APAP in murine model. CONCLUSIONS Upregulation of GPR116 on hepatocytes inhibits ER stress by binding to β-arrestin1, protecting mice from APAP-induced hepatotoxicity. GPR116 may serve as a promising therapeutic target for AILI.
Collapse
Affiliation(s)
- Qian Xiang
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai, 200433, China
- Department of Anesthesiology, Peking University Third Hospital, Beijing, 100191, China
| | - Na Li
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Yan Zhang
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Ting Wang
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Ying Wang
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Jinjun Bian
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai, 200433, China.
| |
Collapse
|
5
|
Shreya S, Grosset CF, Jain BP. Unfolded Protein Response Signaling in Liver Disorders: A 2023 Updated Review. Int J Mol Sci 2023; 24:14066. [PMID: 37762367 PMCID: PMC10531763 DOI: 10.3390/ijms241814066] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 09/04/2023] [Accepted: 09/12/2023] [Indexed: 09/29/2023] Open
Abstract
Endoplasmic reticulum (ER) is the site for synthesis and folding of secreted and transmembrane proteins. Disturbance in the functioning of ER leads to the accumulation of unfolded and misfolded proteins, which finally activate the unfolded protein response (UPR) signaling. The three branches of UPR-IRE1 (Inositol requiring enzyme 1), PERK (Protein kinase RNA-activated (PKR)-like ER kinase), and ATF6 (Activating transcription factor 6)-modulate the gene expression pattern through increased expression of chaperones and restore ER homeostasis by enhancing ER protein folding capacity. The liver is a central organ which performs a variety of functions which help in maintaining the overall well-being of our body. The liver plays many roles in cellular physiology, blood homeostasis, and detoxification, and is the main site at which protein synthesis occurs. Disturbance in ER homeostasis is triggered by calcium level imbalance, change in redox status, viral infection, and so on. ER dysfunction and subsequent UPR signaling participate in various hepatic disorders like metabolic (dysfunction) associated fatty liver disease, liver cancer, viral hepatitis, and cholestasis. The exact role of ER stress and UPR signaling in various liver diseases is not fully understood and needs further investigation. Targeting UPR signaling with drugs is the subject of intensive research for therapeutic use in liver diseases. The present review summarizes the role of UPR signaling in liver disorders and describes why UPR regulators are promising therapeutic targets.
Collapse
Affiliation(s)
- Smriti Shreya
- Gene Expression and Signaling Lab, Department of Zoology, Mahatma Gandhi Central University, Motihari 845401, Bihar, India;
| | - Christophe F. Grosset
- MIRCADE Team, U1312, Bordeaux Institute in Oncology, BRIC, Université de Bordeaux, 146 Rue Léo Saignat, F-33000 Bordeaux, France
| | - Buddhi Prakash Jain
- Gene Expression and Signaling Lab, Department of Zoology, Mahatma Gandhi Central University, Motihari 845401, Bihar, India;
| |
Collapse
|
6
|
Kato Y, Sakanishi A, Matsuda K, Hattori M, Kaneko I, Nishikawa M, Ikushiro S. Covalent adduction of serotonin-derived quinones to the SARS-CoV-2 main protease expressed in a cultured cell. Free Radic Biol Med 2023; 206:74-82. [PMID: 37391098 PMCID: PMC10300202 DOI: 10.1016/j.freeradbiomed.2023.06.018] [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: 04/25/2023] [Revised: 06/18/2023] [Accepted: 06/20/2023] [Indexed: 07/02/2023]
Abstract
The SARS-CoV-2 main protease is an essential molecule for viral replication and is often targeted by medications to treat the infection. In this study, we investigated the possible inhibitory action of endogenous quinones on the enzyme. Recombinant SARS-CoV-2 main protease was exposed to tryptamine-4,5-dione (TD) or quinone from 5-hydroxyindoleacetic acid (Q5HIAA). As a result, the protease activity was considerably decreased in a dose-dependent manner. The IC50 values of the quinones toward the enzyme were approximately 0.28 μM (TD) and 0.49 μM (Q5HIAA). Blot analyses using specific antibodies to quinone-modified proteins revealed that quinones were adducted to the enzyme at concentrations as low as 0.12 μM. Intact mass analyses showed that one or two quinone molecules were covalently adducted onto the main protease. Chymotrypsin-digested main protease analyses revealed that the quinones bind to thiol residues at the enzyme's active site. When TD or Q5HIAA were exposed to cultured cells expressing the viral enzyme, quinone-modified enzyme was identified in the cell lysate, suggesting that even extracellularly generated quinones could react with the viral enzyme expressed in an infected cell. Thus, these endogenous quinones could act as inhibitors of the viral enzyme.
Collapse
Affiliation(s)
- Yoji Kato
- School of Human Science and Environment, University of Hyogo, Himeji, Hyogo, 670-0092, Japan; Research Institute for Food and Nutritional Sciences, University of Hyogo, Himeji, Hyogo, 670-0092, Japan.
| | - Asahi Sakanishi
- School of Human Science and Environment, University of Hyogo, Himeji, Hyogo, 670-0092, Japan
| | - Kaoru Matsuda
- School of Human Science and Environment, University of Hyogo, Himeji, Hyogo, 670-0092, Japan
| | - Mai Hattori
- School of Human Science and Environment, University of Hyogo, Himeji, Hyogo, 670-0092, Japan
| | - Ichiro Kaneko
- School of Human Science and Environment, University of Hyogo, Himeji, Hyogo, 670-0092, Japan; Research Institute for Food and Nutritional Sciences, University of Hyogo, Himeji, Hyogo, 670-0092, Japan
| | - Miyu Nishikawa
- Department of Biotechnology, Faculty of Engineering, Toyama Prefectural University, Imizu, Toyama, 939-0398, Japan
| | - Shinichi Ikushiro
- Department of Biotechnology, Faculty of Engineering, Toyama Prefectural University, Imizu, Toyama, 939-0398, Japan
| |
Collapse
|
7
|
Ganapathy AS, Saha K, Wang A, Arumugam P, Dharmaprakash V, Yochum G, Koltun W, Nighot M, Perdew G, Thompson TA, Ma T, Nighot P. Alpha-tocopherylquinone differentially modulates claudins to enhance intestinal epithelial tight junction barrier via AhR and Nrf2 pathways. Cell Rep 2023; 42:112705. [PMID: 37393618 PMCID: PMC10528852 DOI: 10.1016/j.celrep.2023.112705] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 04/25/2023] [Accepted: 06/12/2023] [Indexed: 07/04/2023] Open
Abstract
Defects in intestinal epithelial tight junctions (TJs) allow paracellular permeation of noxious luminal antigens and are important pathogenic factors in inflammatory bowel disease (IBD). We show that alpha-tocopherylquinone (TQ), a quinone-structured oxidation product of vitamin E, consistently enhances the intestinal TJ barrier by increasing barrier-forming claudin-3 (CLDN3) and reducing channel-forming CLDN2 in Caco-2 cell monolayers (in vitro), mouse models (in vivo), and surgically resected human colons (ex vivo). TQ reduces colonic permeability and ameliorates colitis symptoms in multiple colitis models. TQ, bifunctionally, activates both aryl hydrocarbon receptor (AhR) and nuclear factor erythroid 2-related factor 2 (Nrf2) pathways. Genetic deletion studies reveal that TQ-induced AhR activation transcriptionally increases CLDN3 via xenobiotic response element (XRE) in the CLDN3 promoter. Conversely, TQ suppresses CLDN2 expression via Nrf2-mediated STAT3 inhibition. TQ offers a naturally occurring, non-toxic intervention for enhancement of the intestinal TJ barrier and adjunct therapeutics to treat intestinal inflammation.
Collapse
Affiliation(s)
| | - Kushal Saha
- Division of Gastroenterology and Hepatology, Department of Medicine, Pennsylvania State College of Medicine, Hershey, PA 17033, USA
| | - Alexandra Wang
- Division of Gastroenterology and Hepatology, Department of Medicine, Pennsylvania State College of Medicine, Hershey, PA 17033, USA
| | - Priya Arumugam
- Division of Gastroenterology and Hepatology, Department of Medicine, Pennsylvania State College of Medicine, Hershey, PA 17033, USA
| | - Viszwapriya Dharmaprakash
- Division of Gastroenterology and Hepatology, Department of Medicine, Pennsylvania State College of Medicine, Hershey, PA 17033, USA
| | - Gregory Yochum
- Division of Colon and Rectal Surgery, Department of Surgery, Pennsylvania State College of Medicine, Hershey, PA 17033, USA
| | - Walter Koltun
- Division of Colon and Rectal Surgery, Department of Surgery, Pennsylvania State College of Medicine, Hershey, PA 17033, USA
| | - Meghali Nighot
- Division of Gastroenterology and Hepatology, Department of Medicine, Pennsylvania State College of Medicine, Hershey, PA 17033, USA
| | - Gary Perdew
- Department of Veterinary and Biomedical Sciences and the Center for Molecular Toxicology and Carcinogenesis, The Pennsylvania State University, University Park, PA 16802, USA
| | - Todd A Thompson
- University of New Mexico College of Pharmacy, Albuquerque, NM 87131, USA
| | - Thomas Ma
- Division of Gastroenterology and Hepatology, Department of Medicine, Pennsylvania State College of Medicine, Hershey, PA 17033, USA
| | - Prashant Nighot
- Division of Gastroenterology and Hepatology, Department of Medicine, Pennsylvania State College of Medicine, Hershey, PA 17033, USA.
| |
Collapse
|
8
|
Zhang J, Wei Z, Qi X, Hou X, Liu D, He J. Integrative proteomics, phosphoproteomics and acetylation proteomics analyses of acute pancreatitis in rats. Int J Med Sci 2023; 20:888-900. [PMID: 37324185 PMCID: PMC10266050 DOI: 10.7150/ijms.81658] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 04/27/2023] [Indexed: 06/17/2023] Open
Abstract
Acute pancreatitis (AP) is a common acute abdominalgia of the digestive tract. When the disease progresses to severe acute pancreatitis (SAP), the complications and mortality rate greatly increase. Determining the key factors and pathways underlying AP and SAP will help elucidate the pathological processes involved in disease progression and will be beneficial for identifying potential therapeutic targets. We conducted an integrative proteomics, phosphoproteomics and acetylation proteomics analysis of pancreas samples collected from normal, AP and SAP rat models. We identified 9582 proteins, 3130 phosphorylated modified proteins, and 1677 acetylated modified proteins across all samples. The differentiated expression proteins and KEGG pathway analysis suggested the pronounced enrichment of key pathways based on the following group comparisons: AP versus normal, SAP versus normal, and SAP versus AP. Integrative proteomics and phosphoproteomics analyses revealed 985 jointly detected proteins in the comparison of AP and normal samples, 911 proteins in the comparison of SAP and normal samples, and 910 proteins in the comparison of SAP and AP samples. Based on proteomics and acetylation proteomics analyses, we found that 984 proteins were jointly detected in the comparison of AP and normal samples, 990 proteins in SAP and normal samples, and 728 proteins in SAP and AP samples. Thus, our study offers a valuable resource to understand the proteomic and protein modification atlas in AP.
Collapse
Affiliation(s)
| | | | | | | | | | - Jun He
- ✉ Corresponding author: Jun He, PhD, Department of General Surgery, Second Xiangya Hospital, Central South University, 139 Renmin Road, Changsha, Hunan 410011, China. Email address:
| |
Collapse
|
9
|
Huang Q, Chen Y, Zhang Z, Xue Z, Hua Z, Luo X, Li Y, Lu C, Lu A, Liu Y. The endoplasmic reticulum participated in drug metabolic toxicity. Cell Biol Toxicol 2022; 38:945-961. [PMID: 35040016 DOI: 10.1007/s10565-021-09689-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 12/07/2021] [Indexed: 01/25/2023]
Abstract
Covalent binding of reactive metabolites formed by drug metabolic activation with biological macromolecules is considered to be an important mechanism of drug metabolic toxicity. Recent studies indicate that the endoplasmic reticulum (ER) could play an important role in drug toxicity by participating in the metabolic activation of drugs and could be a primarily attacked target by reactive metabolites. In this article, we summarize the generation and mechanism of reactive metabolites in ER stress and their associated cell death and inflammatory cascade, as well as the systematic modulation of unfolded protein response (UPR)-mediated adaptive pathways.
Collapse
Affiliation(s)
- Qingcai Huang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Youwen Chen
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Zhengjia Zhang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Zeyu Xue
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Zhenglai Hua
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Xinyi Luo
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Yang Li
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Cheng Lu
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Aiping Lu
- School of Chinese Medicine, Hong Kong Baptist University, Kowloon, Hong Kong, China
| | - Yuanyan Liu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China.
| |
Collapse
|
10
|
Khallouki F, Saber S, Bouddine T, Hajji L, Elbouhali B, Silvente-Poirot S, Poirot M. In vitro and In vivo oxidation and cleavage products of tocols: From chemical tuners to “VitaminEome” therapeutics. A narrative review. FOOD BIOSCI 2022. [DOI: 10.1016/j.fbio.2022.101839] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
11
|
Comparative study on the in vitro digestibility of chicken protein after different modifications. Food Chem 2022; 385:132652. [PMID: 35278732 DOI: 10.1016/j.foodchem.2022.132652] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Revised: 02/09/2022] [Accepted: 03/05/2022] [Indexed: 11/24/2022]
Abstract
The effects of tea polyphenols (TPPs) and ultrasound treatment (UDT) on the digestibility of chicken myofibrillar protein (MPN) in anenhanced oxidation system were investigated. As observed, the original aggregates of MPN were much lower in the UDT-assisted group than in the control protein group, and the difference widened after the incorporation of TPPs. The covalent structures of the UDT-assisted oxidation groups were verified via mass spectrometry and amino acid (AAD) measurements. The peptide abundance increased after the UDT-assisted covalent reaction and most of these peptides were derived from the structural proteins of MPNs according to the results of nano-LC-ESI-MS/MS. Digestion kinetic analysis showed that the digestion level of the EGCG-treated group was better than that of the other treated groups, regardless of the UDT-assisted covalent reaction. Overall, the combination of EGCG oxidation and UDT may be an efficient way to promote the nutritional value of the final MPN products.
Collapse
|
12
|
Liu Q, Tang Q, Jing X, Zhang J, Xia Y, Yan J, Xu Y, Li J, Li Y, He J, Mo L. Mesencephalic astrocyte-derived neurotrophic factor protects against paracetamol -induced liver injury by inhibiting PERK-ATF4-CHOP signaling pathway. Biochem Biophys Res Commun 2022; 602:163-169. [PMID: 35278889 DOI: 10.1016/j.bbrc.2022.02.059] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 02/16/2022] [Indexed: 02/08/2023]
Abstract
Paracetamol (APAP), an over-the-counter drug, is normally safe within the therapeutic dose range but can cause irreversible liver damage after an overdose. Mesencephalic astrocyte-derived neurotrophic factor (MANF) is an endoplasmic reticulum (ER) stress protein and plays a crucial role in metabolic disease. However, the role of MANF in APAP-induced acute hepatotoxicity is still unknown. We used hepatocyte-specific MANF-knockout mice and hepatocyte-specific MANF transgenic mice to investigate the role of hepatocyte-derived MANF in APAP-induced acute liver injury. MANF deficiency was associated with a decreased expression of detoxification enzymes, aggravated glutathione depletion and apoptosis in hepatocytes. Mechanistically, MANF knockout significantly increased PERK-eIF2α-ATF4-CHOP signaling pathway. Blockade of PERK abolished MANF deficiency-over-induced hepatotoxicity after APAP administration. Conversely, hepatocyte-specific MANF overexpression attenuated APAP-induced hepatotoxicity by downregulating the PERK-eIF2α-ATF4-CHOP signaling pathway. Thus, hepatocyte-derived MANF may play a protective role in APAP-induced hepatotoxicity.
Collapse
Affiliation(s)
- Qinhui Liu
- Laboratory of Clinical Pharmacy and Adverse Drug Reaction, West China Hospital of Sichuan University, Chengdu, China
| | - Qin Tang
- Laboratory of Clinical Pharmacy and Adverse Drug Reaction, West China Hospital of Sichuan University, Chengdu, China
| | - Xiandan Jing
- Laboratory of Clinical Pharmacy and Adverse Drug Reaction, West China Hospital of Sichuan University, Chengdu, China
| | - Jinhang Zhang
- Laboratory of Clinical Pharmacy and Adverse Drug Reaction, West China Hospital of Sichuan University, Chengdu, China
| | - Yan Xia
- Laboratory of Clinical Pharmacy and Adverse Drug Reaction, West China Hospital of Sichuan University, Chengdu, China
| | - Jiamin Yan
- Laboratory of Clinical Pharmacy and Adverse Drug Reaction, West China Hospital of Sichuan University, Chengdu, China
| | - Ying Xu
- Laboratory of Clinical Pharmacy and Adverse Drug Reaction, West China Hospital of Sichuan University, Chengdu, China
| | - Jiahui Li
- Laboratory of Clinical Pharmacy and Adverse Drug Reaction, West China Hospital of Sichuan University, Chengdu, China
| | - Yanping Li
- Laboratory of Clinical Pharmacy and Adverse Drug Reaction, West China Hospital of Sichuan University, Chengdu, China
| | - Jinhan He
- Laboratory of Clinical Pharmacy and Adverse Drug Reaction, West China Hospital of Sichuan University, Chengdu, China
| | - Li Mo
- Center of Gerontology and Geriatrics, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China.
| |
Collapse
|
13
|
Terenzi A, La Franca M, van Schoonhoven S, Panchuk R, Martínez Á, Heffeter P, Gober R, Pirker C, Vician P, Kowol CR, Stoika R, Salassa L, Rohr J, Berger W. Landomycins as glutathione-depleting agents and natural fluorescent probes for cellular Michael adduct-dependent quinone metabolism. Commun Chem 2021; 4:162. [PMID: 36697631 PMCID: PMC9814637 DOI: 10.1038/s42004-021-00600-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 11/03/2021] [Indexed: 01/28/2023] Open
Abstract
Landomycins are angucyclines with promising antineoplastic activity produced by Streptomyces bacteria. The aglycone landomycinone is the distinctive core, while the oligosaccharide chain differs within derivatives. Herein, we report that landomycins spontaneously form Michael adducts with biothiols, including reduced cysteine and glutathione, both cell-free or intracellularly involving the benz[a]anthraquinone moiety of landomycinone. While landomycins generally do not display emissive properties, the respective Michael adducts exerted intense blue fluorescence in a glycosidic chain-dependent manner. This allowed label-free tracking of the short-lived nature of the mono-SH-adduct followed by oxygen-dependent evolution with addition of another SH-group. Accordingly, hypoxia distinctly stabilized the fluorescent mono-adduct. While extracellular adduct formation completely blocked the cytotoxic activity of landomycins, intracellularly it led to massively decreased reduced glutathione levels. Accordingly, landomycin E strongly synergized with glutathione-depleting agents like menadione but exerted reduced activity under hypoxia. Summarizing, landomycins represent natural glutathione-depleting agents and fluorescence probes for intracellular anthraquinone-based angucycline metabolism.
Collapse
Affiliation(s)
- Alessio Terenzi
- grid.10776.370000 0004 1762 5517Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, Viale delle Scienze, Ed. 17, 90128 Palermo, Italy
| | - Mery La Franca
- grid.10776.370000 0004 1762 5517Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, Viale delle Scienze, Ed. 17, 90128 Palermo, Italy ,grid.22937.3d0000 0000 9259 8492Institute of Cancer Research and Comprehensive Cancer Center, Medical University Vienna, Spitalgasse 23, 1090 Vienna, Austria
| | - Sushilla van Schoonhoven
- grid.22937.3d0000 0000 9259 8492Institute of Cancer Research and Comprehensive Cancer Center, Medical University Vienna, Spitalgasse 23, 1090 Vienna, Austria
| | - Rostyslav Panchuk
- grid.466769.cDepartment of Regulation of Cell Proliferation and Apoptosis, Institute of Cell Biology, Drahomanov St., 14/16, Lviv, 79005 Ukraine
| | - Álvaro Martínez
- grid.452382.a0000 0004 1768 3100Donostia International Physics Center and Polimero eta Material Aurreratuak: Fisika, Kimika eta Teknologia, Kimika Fakultatea, Euskal Herriko Unibertsitatea UPV/EHU, Paseo Manuel de Lardizabal 4, Donostia, 20018 Spain
| | - Petra Heffeter
- grid.22937.3d0000 0000 9259 8492Institute of Cancer Research and Comprehensive Cancer Center, Medical University Vienna, Spitalgasse 23, 1090 Vienna, Austria ,grid.22937.3d0000 0000 9259 8492Research Cluster “Translational Cancer Therapy Research”, University of Vienna and Medical University of Vienna, Vienna, Austria
| | - Redding Gober
- grid.266539.d0000 0004 1936 8438College of Pharmacy, University of Kentucky, South Limestone Str. 789, Lexington, 40536-0596 USA
| | - Christine Pirker
- grid.22937.3d0000 0000 9259 8492Institute of Cancer Research and Comprehensive Cancer Center, Medical University Vienna, Spitalgasse 23, 1090 Vienna, Austria
| | - Petra Vician
- grid.22937.3d0000 0000 9259 8492Institute of Cancer Research and Comprehensive Cancer Center, Medical University Vienna, Spitalgasse 23, 1090 Vienna, Austria
| | - Christian R. Kowol
- grid.22937.3d0000 0000 9259 8492Research Cluster “Translational Cancer Therapy Research”, University of Vienna and Medical University of Vienna, Vienna, Austria ,grid.10420.370000 0001 2286 1424Institute of Inorganic Chemistry, Faculty of Chemistry, University of Vienna, Waehringer Straße 42, 1090 Vienna, Austria
| | - Rostyslav Stoika
- grid.466769.cDepartment of Regulation of Cell Proliferation and Apoptosis, Institute of Cell Biology, Drahomanov St., 14/16, Lviv, 79005 Ukraine
| | - Luca Salassa
- grid.452382.a0000 0004 1768 3100Donostia International Physics Center and Polimero eta Material Aurreratuak: Fisika, Kimika eta Teknologia, Kimika Fakultatea, Euskal Herriko Unibertsitatea UPV/EHU, Paseo Manuel de Lardizabal 4, Donostia, 20018 Spain ,grid.424810.b0000 0004 0467 2314Ikerbasque, Basque Foundation for Science, Bilbao, 48011 Spain
| | - Jürgen Rohr
- grid.266539.d0000 0004 1936 8438College of Pharmacy, University of Kentucky, South Limestone Str. 789, Lexington, 40536-0596 USA
| | - Walter Berger
- Institute of Cancer Research and Comprehensive Cancer Center, Medical University Vienna, Spitalgasse 23, 1090, Vienna, Austria. .,Research Cluster "Translational Cancer Therapy Research", University of Vienna and Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
14
|
Hurben AK, Erber LN, Tretyakova NY, Doran TM. Proteome-Wide Profiling of Cellular Targets Modified by Dopamine Metabolites Using a Bio-Orthogonally Functionalized Catecholamine. ACS Chem Biol 2021; 16:2581-2594. [PMID: 34726906 PMCID: PMC9872492 DOI: 10.1021/acschembio.1c00629] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Selective death of midbrain dopaminergic neurons is a hallmark pathology of Parkinson's disease (PD), but the molecular mechanisms that initiate the cascade of events resulting in neurodegeneration in PD remain unclear. Compelling evidence suggests that dysregulation of dopamine (DA) induces neuronal stress and damage responses that are operative processes in striatal degeneration preceding PD-like symptoms. Improper DA sequestration to vesicles raises cytosolic DA levels, which is rapidly converted into electrophilic dopaquinone species (DQs) that react readily with protein nucleophiles forming covalent modifications that alter the native structure and function of proteins. These so-called DA-protein adducts (DPAs) have been reported to play a role in neurotoxicity, and their abundance with respect to neurodegeneration has been linked to clinical and pathological features of PD that suggest that they play a causal role in PD pathogenesis. Therefore, characterizing DPAs is a critical first step in understanding the susceptibility of midbrain dopaminergic neurons during PD. To help achieve this goal, we report here a novel DA-mimetic (DAyne) containing a biorthogonal alkyne handle that exhibits a reactivity profile similar to DA in aqueous buffers. By linking DPAs formed with DAyne to a fluorescent reporter molecule, DPAs were visualized in fixed cells and within lysates. DAyne enabled global mapping of cellular proteins affected by DQ modification and their bioactive pathways through enrichment. Our proteomic profiling of DPAs in neuronal SH-SY5Y cells indicates that proteins susceptible to DPA formation are extant throughout the proteome, potentially influencing several diverse biological pathways involved in PD such as endoplasmic reticulum (ER) stress, cytoskeletal instability, proteotoxicity, and clathrin function. We validated that a protein involved in the ER stress pathway, protein disulfide isomerase 3 (PDIA3), which was enriched in our chemoproteomic analysis, is functionally inhibited by DA, providing evidence that dysregulated cellular DA may induce or exacerbate ER stress. Thus, DAyne provided new mechanistic insights into DA toxicity that may be observed during PD by enabling characterization of DPAs generated reproducibly at physiologically relevant quinone exposures. We anticipate our design and application of this reactivity-based probe will be generally applicable for clarifying mechanisms of metabolic quinone toxicity.
Collapse
Affiliation(s)
- Alexander K. Hurben
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Luke N. Erber
- Department of Medicinal Chemistry and Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Natalia Y. Tretyakova
- Department of Medicinal Chemistry and Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Todd M. Doran
- Department of Medicinal Chemistry and Institute for Translational Neuroscience, University of Minnesota, Minneapolis, Minnesota 55455, United States
| |
Collapse
|
15
|
Saito Y. Diverse cytoprotective actions of vitamin E isoforms- role as peroxyl radical scavengers and complementary functions with selenoproteins. Free Radic Biol Med 2021; 175:121-129. [PMID: 34481936 DOI: 10.1016/j.freeradbiomed.2021.08.234] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 08/25/2021] [Accepted: 08/26/2021] [Indexed: 02/05/2023]
Abstract
Vitamin E, a generic term for tocopherol (T) and tocotrienol (T3), is one of the most potent lipid-soluble antioxidants in the body. It is classified into T and T3 based on the difference in the side chain structure. T and T3 have four isoforms: α-, β-, γ-, and δ, which have different chroman rings. Both T and T3 exhibit a similar ability to scavenge free radicals, and the extent of this ability depends on the difference in the chroman structure. However, they display unique cytoprotective activities in cultured cells depending on the difference in the side chain structure. The cytoprotective effects of vitamin E have received much attention in the prevention of ferroptosis, which is a distinct form of cell death involving iron-dependent lipid peroxidation. This review focuses on the cytoprotective actions of vitamin E isoforms against oxidative stress, particularly the difference between T and T3 and its relation to cellular uptake and distribution. Moreover, the molecular mechanism for cytoprotection of vitamin E oxidation products is explained, and the complementary role of vitamin E and selenoproteins to prevent lipid peroxidation and ferroptosis is described. Furthermore, the evaluation of vitamin E's radical scavenging activity in vivo using oxidative stress markers is discussed, particularly based on kinetic data and the physiological molar ratio of vitamin E to substrates, and the limited role of vitamin E as a peroxyl radical scavenger is described. The future directions and unresolved issues related to vitamin E and lipid peroxidation are also discussed.
Collapse
Affiliation(s)
- Yoshiro Saito
- Laboratory of Molecular Biology and Metabolism, Graduate School of Pharmaceutical Sciences, Tohoku University C301, 6-3 Aoba, Aramaki, Aoba-ku, Sendai, Miyagi 980-8578, Japan.
| |
Collapse
|
16
|
Berthelet J, Michail C, Bui LC, Le Coadou L, Sirri V, Wang L, Dulphy N, Dupret JM, Chomienne C, Guidez F, Rodrigues-Lima F. The benzene hematotoxic and reactive metabolite 1,4-benzoquinone impairs the activity of the histone methyltransferase SETD2 and causes aberrant H3K36 trimethylation (H3K36me3). Mol Pharmacol 2021; 100:283-294. [PMID: 34266924 DOI: 10.1124/molpharm.121.000303] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 06/28/2021] [Indexed: 11/22/2022] Open
Abstract
Human SETD2 is the unique histone methyltransferase that generates H3K36me3, an epigenetic mark that plays a key role in normal hematopoiesis. Interestingly, recurrent-inactivating mutations of SETD2 and aberrant H3K36 trimethylation (H3K36me3) are increasingly reported to be involved in hematopoietic malignancies. Benzene (BZ) is an ubiquitous environmental pollutant and carcinogen that causes leukemia. The leukemogenic properties of BZ depend on its biotransformation in the bone marrow into oxidative metabolites in particular 1,4-benzoquinone (BQ). This hematotoxic metabolite can form DNA and protein adducts that result in the damage and the alteration of cellular processes. Recent studies suggest that BZ-depend leukemogenesis could depend on epigenetic perturbations notably aberrant histone methylation. We investigated whether H3K36 trimethylation by SETD2 could be impacted by BZ and its hematotoxic metabolites. Herein, we show that BQ, the major leukemogenic metabolite of BZ, inhibits irreversibly the human histone methyltransferase SETD2 resulting in decreased H3K36 trimethylation (H3K36me3). Our mechanistic studies further indicate that the BQ-dependent inactivation of SETD2 is due to covalent binding of BQ to reactive Zn-finger cysteines within the catalytic domain of the enzyme. The formation of these quinoprotein adducts results in loss of enzyme activity and protein cross-links/oligomers. Experiments conducted in hematopoietic cells confirm that exposure to BQ results in the formation of SETD2 cross-links/oligomers and concomitant loss of H3K36me3 in cells. Taken together, our data indicate that BQ, a major hematotoxic metabolite of BZ could contribute to BZ-dependent leukemogenesis by perturbing the functions of SETD2, an histone lysine methyltransferase of hematopoietic relevance. Significance Statement Benzoquinone is a major leukemogenic metabolite of benzene. Dysregulation of histone methyltransferase is involved in hematopoietic malignancies. We found that benzoquinone irreversibly impairs SETD2, a histone H3K36 methyltransferase that plays a key role in hematopoiesis. Benzoquinone forms covalent adducts on Zn-finger cysteines within the catalytic site leading to loss of activity, protein cross-links/oligomers and concomitant decrease of H3K36me3 histone mark. Our data provide evidence that a leukemogenic metabolite of benzene can impair a key epigenetic enzyme.
Collapse
Affiliation(s)
| | | | | | | | | | - Li Wang
- The First Affiliated Hospital of Chongqing Medical University, China
| | | | | | | | | | | |
Collapse
|
17
|
de Almeida PDO, Dos Santos Barbosa Jobim G, Dos Santos Ferreira CC, Rocha Bernardes L, Dias RB, Schlaepfer Sales CB, Valverde LDF, Rocha CAG, Soares MBP, Bezerra DP, de Carvalho da Silva F, Cardoso MFDC, Ferreira VF, Brito LF, Pires de Sousa L, de Vasconcellos MC, Lima ES. A new synthetic antitumor naphthoquinone induces ROS-mediated apoptosis with activation of the JNK and p38 signaling pathways. Chem Biol Interact 2021; 343:109444. [PMID: 33939975 DOI: 10.1016/j.cbi.2021.109444] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 12/11/2020] [Accepted: 03/09/2021] [Indexed: 02/07/2023]
Abstract
Quinones are plant-derived secondary metabolites that present diverse pharmacological properties, including antibacterial, antifungal, antiviral, anti-inflammatory, antipyretic and anticancer activities. In the present study, we evaluated the cytotoxic effect of a new naphthoquinone 6b,7-dihydro-5H-cyclopenta [b]naphtho [2,1-d]furan-5,6 (9aH)-dione) (CNFD) in different tumor cell lines. CNFD displayed cytotoxic activity against different tumor cell lines, especially in MCF-7 human breast adenocarcinoma cells, which showed IC50 values of 3.06 and 0.98 μM for 24 and 48 h incubation, respectively. In wound-healing migration assays, CNFD promoted inhibition of cell migration. We have found typical hallmarks of apoptosis, such as cell shrinkage, chromatin condensation, phosphatidylserine exposure, increase of caspases-9 and-3 activation, increase of internucleosomal DNA fragmentation without affecting the cell membrane permeabilization, increase of ROS production, and loss of mitochondrial membrane potential induced by CNFD. Moreover, gene expression experiments indicated that CNFD increased the expression of the genes CDKN1A, FOS, MAX, and RAC1 and decreased the levels of mRNA transcripts of several genes, including CCND1, CDK2, SOS1, RHOA, GRB2, EGFR and KRAS. The CNFD treatment of MCF-7 cells induced the phosphorylation of c-jun N-terminal kinase (JNK) and p38 mitogen-activated protein kinases (MAPKs) and inactivation of extracellular signal-regulated protein kinase 1/2 (ERK1/2). In a study using melanoma cells in a murine model in vivo, CNFD induced a potent anti-tumor activity. Herein, we describe, for the first time, the cytotoxicity and anti-tumor activity of CNFD and sequential mechanisms of apoptosis in MCF-7 cells. CNFD seems to be a promising candidate for anti-tumor therapy.
Collapse
Affiliation(s)
- Patricia D O de Almeida
- Laboratory of Biological Activity, Faculty of Pharmaceutical Sciences, Federal University of Amazonas - UFAM, Manaus, Amazonas, 69077-000, Brazil
| | - Gleyce Dos Santos Barbosa Jobim
- Laboratory of Biological Activity, Faculty of Pharmaceutical Sciences, Federal University of Amazonas - UFAM, Manaus, Amazonas, 69077-000, Brazil
| | - Caio César Dos Santos Ferreira
- Laboratory of Biological Activity, Faculty of Pharmaceutical Sciences, Federal University of Amazonas - UFAM, Manaus, Amazonas, 69077-000, Brazil
| | - Lucas Rocha Bernardes
- Laboratory of Biological Activity, Faculty of Pharmaceutical Sciences, Federal University of Amazonas - UFAM, Manaus, Amazonas, 69077-000, Brazil
| | - Rosane B Dias
- Laboratory of Pathology and Molecular Biology, Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, Bahia, 40296-710, Brazil
| | - Caroline B Schlaepfer Sales
- Laboratory of Pathology and Molecular Biology, Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, Bahia, 40296-710, Brazil; Department of Biomorphology, Institute of Health Sciences, Federal University of Bahia - UFBA, Salvador, Bahia, 40110-902, Brazil
| | - Ludmila de F Valverde
- Laboratory of Pathology and Molecular Biology, Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, Bahia, 40296-710, Brazil
| | - Clarissa A G Rocha
- Laboratory of Pathology and Molecular Biology, Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, Bahia, 40296-710, Brazil
| | - Milena B P Soares
- Laboratory of Tissue Engineering and Immunopharmacology, Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, Bahia, 40296-710, Brazil
| | - Daniel P Bezerra
- Laboratory of Tissue Engineering and Immunopharmacology, Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, Bahia, 40296-710, Brazil
| | - Fernando de Carvalho da Silva
- Laboratory of Carbohydrate and Nucleotide Synthesis, Department of Organic Chemistry, Federal Fluminense University - UFF, Niterói, Rio de Janeiro, 24020-141, Brazil
| | - Mariana Filomena do Carmo Cardoso
- Laboratory of Carbohydrate and Nucleotide Synthesis, Department of Organic Chemistry, Federal Fluminense University - UFF, Niterói, Rio de Janeiro, 24020-141, Brazil
| | - Vitor Francisco Ferreira
- Laboratory of Carbohydrate and Nucleotide Synthesis, Department of Organic Chemistry, Federal Fluminense University - UFF, Niterói, Rio de Janeiro, 24020-141, Brazil
| | - Larissa F Brito
- Laboratory of Signaling in Inflammation, Department of Clinical and Toxicological Analysis, Faculty of Pharmacy, Federal University of Minas Gerais - UFMG, Belo Horizonte, Minas Gerais, 31270-901, Brazil
| | - Lirlândia Pires de Sousa
- Laboratory of Signaling in Inflammation, Department of Clinical and Toxicological Analysis, Faculty of Pharmacy, Federal University of Minas Gerais - UFMG, Belo Horizonte, Minas Gerais, 31270-901, Brazil
| | - Marne C de Vasconcellos
- Laboratory of Biological Activity, Faculty of Pharmaceutical Sciences, Federal University of Amazonas - UFAM, Manaus, Amazonas, 69077-000, Brazil
| | - Emerson S Lima
- Laboratory of Biological Activity, Faculty of Pharmaceutical Sciences, Federal University of Amazonas - UFAM, Manaus, Amazonas, 69077-000, Brazil.
| |
Collapse
|
18
|
Lipid peroxidation products as a mediator of toxicity and adaptive response - The regulatory role of selenoprotein and vitamin E. Arch Biochem Biophys 2021; 703:108840. [PMID: 33744199 DOI: 10.1016/j.abb.2021.108840] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 01/14/2021] [Accepted: 03/08/2021] [Indexed: 02/07/2023]
Abstract
Lipid peroxidation and its products have been investigated extensively and their biological importance, particularly in relation to physiological and pathophysiological conditions, has received considerable attention. Lipids are oxidized by three distinct mechanisms, i.e., enzymatic oxidation, nonenzymatic, free radical-mediated oxidation, and nonenzymatic, nonradical-mediated oxidation, which respectively yield specific products. Lipid hydroperoxides are the major primary products formed and are reduced to the corresponding hydroxides by antioxidative enzymes such as selenoproteins, and/or undergo secondary oxidation, generating various products with electrophilic properties, such as 4-hydroxy-2-nonenal. Lipid peroxidation induces a loss of fine structure and natural function of lipids, and can produce cytotoxicity and/or novel biological activity. This review broadly discusses the mechanisms of lipid peroxidation and its products, its utility as a biomarker for oxidative stress, the biological effects of lipid peroxidation products, including their action as a mediator of the adaptive response, and the role of the antioxidant system, particularly selenoproteins and vitamin E, in preventing lipid peroxidation and ferroptosis.
Collapse
|
19
|
Gao J, Song J, Ye J, Duan X, Dionysiou DD, Yadav JS, Nadagouda MN, Yang L, Luo S. Comparative toxicity reduction potential of UV/sodium percarbonate and UV/hydrogen peroxide treatments for bisphenol A in water: An integrated analysis using chemical, computational, biological, and metabolomic approaches. WATER RESEARCH 2021; 190:116755. [PMID: 33383346 DOI: 10.1016/j.watres.2020.116755] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2020] [Revised: 12/13/2020] [Accepted: 12/14/2020] [Indexed: 06/12/2023]
Abstract
Bisphenol A (BPA) is a common industrial chemical with significant adverse impacts on biological systems as an environmental contaminant. UV/hydrogen peroxide (UV/H2O2) is a well-established technology for BPA treatment in water while UV/sodium percarbonate (UV/SPC) is an emerging technology with unclear biological impacts of treated effluent. Therefore, in this study, the toxicity evaluation of BPA solution treated with UV/H2O2 and UV/SPC was preformed and compared based on transformation products (TPs) profile, quantitative structure-activity relationship (QSAR), Escherichia coli (E. coli) toxicity assays, and metabolomic analysis. TPs with hydroxylation, double-ring split, and single-ring cleavage were generated from BPA during the treatments with both technologies, but TPs with quinonation were specifically detected in UV/H2O2 treated solution at the UV dose of 1470 mJ cm-2. QSAR prediction based on TPs profile (excluding benzoquinone TPs) suggested that UV/H2O2 and UV/SPC treatments of BPA may increase matrix toxicity due to the formation of multi-hydroxylated TPs; however decreased bioaccumulation potential of all TPs may mitigate the increase of toxicity by reducing the chance of TPs to reach the concentration of toxicity threshold. In vivo assays with E. coli showed inhibited cell growth, arrested cell cycle, and increased cell death in BPA solution treated with UV/H2O2 at the UV dose of 1470 mJ cm-2. Metabolomic analysis indicated that BPA solution treated with UV/H2O2 at UV dose of 1470 mJ cm-2 impacted E. coli metabolism differently than other solutions with unique inhibition on glycerolipid metabolism. Moreover, BPA interfered in various metabolic pathways including alanine, aspartate and glutamate metabolism, starch and sucrose metabolism, pentose phosphate pathway, and lysine degradation, which were mitigated after the treatments. UV/SPC showed advantage over UV/H2O2 of attenuated impact on butanoate metabolism with UV irradiation. This study has generated valuable data for better understanding of biological impacts of BPA and its solutions treated with UV/H2O2 or UV/SPC, thus providing insights for their application prospect for water and wastewater treatment.
Collapse
Affiliation(s)
- Jiong Gao
- Environmental Engineering and Science Program, University of Cincinnati, Cincinnati, OH 45221, United States
| | - Jie Song
- Guangdong Key Laboratory of Environmental Pollution and Health, School of Environment, Jinan University, Guangzhou, Guangdong 510632, China
| | - Jinshao Ye
- Guangdong Key Laboratory of Environmental Pollution and Health, School of Environment, Jinan University, Guangzhou, Guangdong 510632, China.
| | - Xiaodi Duan
- Environmental Engineering and Science Program, University of Cincinnati, Cincinnati, OH 45221, United States.
| | - Dionysios D Dionysiou
- Environmental Engineering and Science Program, University of Cincinnati, Cincinnati, OH 45221, United States.
| | - Jagjit S Yadav
- Division of Environmental Genetics and Molecular Toxicology, Department of Environmental and Public Health Sciences, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, United States
| | - Mallikarjuna N Nadagouda
- Department of Mechanical and Materials Engineering, Wright State University, Dayton, OH 45324, United States
| | - Lixia Yang
- College of Environmental and Chemical Engineering, Nanchang Hangkong University, Nanchang, Jiangxi 330036, China
| | - Shenglian Luo
- College of Environmental and Chemical Engineering, Nanchang Hangkong University, Nanchang, Jiangxi 330036, China
| |
Collapse
|
20
|
Zhang W, Berthelet J, Michail C, Bui LC, Gou P, Liu R, Duval R, Renault J, Dupret JM, Guidez F, Chomienne C, Rodrigues Lima F. Human CREBBP acetyltransferase is impaired by etoposide quinone, an oxidative and leukemogenic metabolite of the anticancer drug etoposide through modification of redox-sensitive zinc-finger cysteine residues. Free Radic Biol Med 2021; 162:27-37. [PMID: 33278510 DOI: 10.1016/j.freeradbiomed.2020.11.027] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 11/22/2020] [Accepted: 11/23/2020] [Indexed: 12/25/2022]
Abstract
Etoposide is an extensively prescribed anticancer drug that, unfortunately, causes therapy-related leukemia. The mechanisms by which etoposide induces secondary hematopoietic malignancies are poorly documented. However, etoposide-related leukemogenesis is known to depend on oxidative metabolites of etoposide, notably etoposide quinone, that can react with protein cysteine residues such as in topoisomerases II. CREBBP is a major histone acetyltransferase that functions mainly as a transcriptional co-activator. This epigenetic enzyme is considered as a tumor suppressor that plays a major role in hematopoiesis. Genetic alterations affecting CREBBP activity are highly common in hematopoietic malignancies. We report here that CREBBP is impaired by etoposide quinone. Molecular and kinetic analyses show that this inhibition occurs through the rapid and covalent (kinhib = 16.102 M-1. s-1) adduction of etoposide quinone with redox sensitive cysteine residues within the RING and PHD Zn2+-fingers of CREBBP catalytic core leading to subsequent release of Zn2+. In agreement with these findings, experiments conducted in cells and in mice treated with etoposide showed irreversible inhibition of endogenous CREBBP activity and decreased H3K18 and H3K27 acetylation. As shown for topoisomerases II, our work thus suggests that the leukemogenic metabolite etoposide quinone can impair the epigenetic CREBBP acetyltransferase through reaction with redox sensitive cysteine residues.
Collapse
Affiliation(s)
- Wenchao Zhang
- Université de Paris, BFA, UMR 8251, CNRS, F-75013, Paris, France
| | - Jérémy Berthelet
- Université de Paris, BFA, UMR 8251, CNRS, F-75013, Paris, France; Université de Paris, CEDC, UMR 7216, CNRS, F-75013, Paris, France
| | | | - Linh-Chi Bui
- Université de Paris, BFA, UMR 8251, CNRS, F-75013, Paris, France
| | - Panhong Gou
- Université de Paris, Institut de Recherche Saint-Louis, UMRS 1131, INSERM, F-75010, Paris, France
| | - Rongxing Liu
- Université de Paris, BFA, UMR 8251, CNRS, F-75013, Paris, France
| | - Romain Duval
- Université de Paris, BIGR, UMRS 1134, INSERM, F-75015, Paris, France
| | - Justine Renault
- Université de Paris, BFA, UMR 8251, CNRS, F-75013, Paris, France
| | | | - Fabien Guidez
- Université de Paris, Institut de Recherche Saint-Louis, UMRS 1131, INSERM, F-75010, Paris, France
| | - Christine Chomienne
- Université de Paris, Institut de Recherche Saint-Louis, UMRS 1131, INSERM, F-75010, Paris, France; Service de Biologie Cellulaire, Assistance Publique des Hôpitaux de Paris (AP-HP), Hôpital Saint Louis, Paris, France
| | | |
Collapse
|
21
|
Aeschimann W, Kammer S, Staats S, Schneider P, Schneider G, Rimbach G, Cascella M, Stocker A. Engineering of a functional γ-tocopherol transfer protein. Redox Biol 2020; 38:101773. [PMID: 33197771 PMCID: PMC7677715 DOI: 10.1016/j.redox.2020.101773] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 10/22/2020] [Accepted: 10/27/2020] [Indexed: 11/28/2022] Open
Abstract
α-tocopherol transfer protein (TTP) was previously reported to self-aggregate into 24-meric spheres (α-TTPS) and to possess transcytotic potency across mono-layers of human umbilical vein endothelial cells (HUVECs). In this work, we describe the characterisation of a functional TTP variant with its vitamer selectivity shifted towards γ-tocopherol. The shift was obtained by introducing an alanine to leucine substitution into the substrate-binding pocket at position 156 through site directed mutagenesis. We report here the X-ray crystal structure of the γ-tocopherol specific particle (γ-TTPS) at 2.24 Å resolution. γ-TTPS features full functionality compared to its α-tocopherol specific parent including self-aggregation potency and transcytotic activity in trans-well experiments using primary HUVEC cells. The impact of the A156L mutation on TTP function is quantified in vitro by measuring the affinity towards γ-tocopherol through micro-differential scanning calorimetry and by determining its ligand-transfer activity. Finally, cell culture experiments using adherently grown HUVEC cells indicate that the protomers of γ-TTP, in contrast to α-TTP, do not counteract cytokine-mediated inflammation at a transcriptional level. Our results suggest that the A156L substitution in TTP is fully functional and has the potential to pave the way for further experiments towards the understanding of α-tocopherol homeostasis in humans.
Collapse
Affiliation(s)
- Walter Aeschimann
- University of Bern, Department of Chemistry and Biochemistry, Bern, 3012, Switzerland
| | - Stephan Kammer
- University of Bern, Department of Chemistry and Biochemistry, Bern, 3012, Switzerland
| | - Stefanie Staats
- University of Kiel, Institute of Human Nutrition and Food Science, Kiel, 24118, Germany
| | - Petra Schneider
- Institute of Pharmaceutical Sciences, ETH Zürich, Vladimir-Prelog-Weg 4, 8093, Zürich, Switzerland
| | - Gisbert Schneider
- Institute of Pharmaceutical Sciences, ETH Zürich, Vladimir-Prelog-Weg 4, 8093, Zürich, Switzerland
| | - Gerald Rimbach
- University of Kiel, Institute of Human Nutrition and Food Science, Kiel, 24118, Germany
| | - Michele Cascella
- University of Oslo, Department of Chemistry and Hylleraas Centre for Quantum Molecular Sciences, PO Box 1033 Blindern, 0315, Oslo, Norway
| | - Achim Stocker
- University of Bern, Department of Chemistry and Biochemistry, Bern, 3012, Switzerland.
| |
Collapse
|
22
|
Proteome-wide effects of naphthalene-derived secondary organic aerosol in BEAS-2B cells are caused by short-lived unsaturated carbonyls. Proc Natl Acad Sci U S A 2020; 117:25386-25395. [PMID: 32989125 DOI: 10.1073/pnas.2001378117] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Exposure to air pollution causes adverse health outcomes, but the toxicity mechanisms remain unclear. Here, we investigated the dynamic toxicities of naphthalene-derived secondary organic aerosol (NSOA) in a human bronchial epithelial cell line (BEAS-2B) and identified the chemical components responsible for toxicities. The chemical composition of NSOA was found to vary with six simulated atmospheric aging conditions (C1-C6), as characterized by high-resolution mass spectrometry and ion mobility mass spectrometry. Global proteome profiling reveals dynamic evolution in toxicity: Stronger proteome-wide impacts were detected in fresh NSOA, but the effects declined along with atmospheric aging. While Nrf2-regulated proteins (e.g., NQO1) were significantly up-regulated, the majority (78 to 97%) of proteins from inflammation and other pathways were down-regulated by NSOA exposure (e.g., Rho GTPases). This pattern is distinct from the reactive oxygen species (ROS)-mediated toxicity pathway, and an alternative cysteine reaction pathway was revealed by the decreased abundance of proteins (e.g., MT1X) prone to posttranslational thiol modification. This pathway was further validated by observing decreased Nrf2 response in reporter cells, after preincubating NSOA with cysteine. Ethynyl-naphthalene probe was employed to confirm the alkylation of cellular proteome thiols on the proteome-wide level by fresh NSOA via in-gel fluorescence imaging. Nontarget analysis identified several unsaturated carbonyls, including naphthoquinones and hydroxylated naphthoquinones, as the toxic components responsible for cysteine reactivity. Our study provides insights into the dynamic toxicities of NSOA during atmospheric aging and identifies short-lived unsaturated carbonyls as the predominant toxic components at the posttranslational level.
Collapse
|
23
|
Zhang J, Duan D, Song ZL, Liu T, Hou Y, Fang J. Small molecules regulating reactive oxygen species homeostasis for cancer therapy. Med Res Rev 2020; 41:342-394. [PMID: 32981100 DOI: 10.1002/med.21734] [Citation(s) in RCA: 93] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Revised: 07/27/2020] [Accepted: 08/22/2020] [Indexed: 12/13/2022]
Abstract
Elevated intracellular reactive oxygen species (ROS) and antioxidant defense systems have been recognized as one of the hallmarks of cancer cells. Compared with normal cells, cancer cells exhibit increased ROS to maintain their malignant phenotypes and are more dependent on the "redox adaptation" mechanism. Thus, there are two apparently contradictory but virtually complementary therapeutic strategies for the regulation of ROS to prevent or treat cancer. The first strategy, that is, chemoprevention, is to prevent or reduce intracellular ROS either by suppressing ROS production pathways or by employing antioxidants to enhance ROS clearance, which protects normal cells from malignant transformation and inhibits the early stage of tumorigenesis. The second strategy is the ROS-mediated anticancer therapy, which stimulates intracellular ROS to a toxicity threshold to activate ROS-induced cell death pathways. Therefore, targeting the regulation of intracellular ROS-related pathways by small-molecule candidates is considered to be a promising treatment for tumors. We herein first briefly introduce the source and regulation of ROS, and then focus on small molecules that regulate ROS-related pathways and show efficacy in cancer therapy from the perspective of pharmacophores. Finally, we discuss several challenges in developing cancer therapeutic agents based on ROS regulation and propose the direction of future development.
Collapse
Affiliation(s)
- Junmin Zhang
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, and School of Pharmacy, Lanzhou University, Lanzhou, China
| | - Dongzhu Duan
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, and School of Pharmacy, Lanzhou University, Lanzhou, China.,Shaanxi Key Laboratory of Phytochemistry, Baoji University of Arts and Sciences, Baoji, China
| | - Zi-Long Song
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, and School of Pharmacy, Lanzhou University, Lanzhou, China
| | - Tianyu Liu
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, and School of Pharmacy, Lanzhou University, Lanzhou, China
| | - Yanan Hou
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, and School of Pharmacy, Lanzhou University, Lanzhou, China
| | - Jianguo Fang
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, and School of Pharmacy, Lanzhou University, Lanzhou, China
| |
Collapse
|
24
|
The critical role of PPARα in the binary switch between life and death induced by endoplasmic reticulum stress. Cell Death Dis 2020; 11:691. [PMID: 32826849 PMCID: PMC7443130 DOI: 10.1038/s41419-020-02811-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 07/13/2020] [Accepted: 07/14/2020] [Indexed: 11/08/2022]
Abstract
Endoplasmic reticulum stress (ER stress) just like a double-edged sword depending on different conditions in the development of multiple hepatic diseases. But the molecular mechanisms of functional conversion during ER stress have not been fully elucidated. In this study, we aim to illustrate the role of PPARα and the subtle mechanism in the functional conversion of ER stress. Tunicamycin (TM) and thapsigargin (TG), as ER stress inducers, were used to induce ER stress in AML12 cells. During the ER stress, qRT-PCR and immunoblotting was used to measure the expression levels of GRP78 and CHOP which show a gradually increasing trend, while PPARα and autophagy was significantly activated in the early stage but was inhibited in the late stage. Moreover, PPARα inhibition by siRNA promoted cell injury in the mild-ER stress and PPARα activation by WY-14643 reduced cell apoptosis in the serious ER stress. In the mild-ER stress with PPARα knocked down, activation of autophagy by rapamycin significantly improved cell survival, in the serious ER stress with PPARα activation, inhibition of autophagy by 3-MA aggravate cell injury. In addition, in the mild-ER stress with PPARα knocked down, CHOP knocked down by siRNA reduced cell apoptosis, in the serious ER stress activated PPARα, CHOP over-expression mediated by lentiviral vector contributed to serious cell injury. Furthermore, C57BL/6 mice was used to induce ER stress with TM intraperitoneal injection, PPARα and autophagy was upregulated in the mild-ER stress while downregulated in the serious ER stress, measured by qRT-PCR and immunoblotting, further confirmed the finding in vitro. Our results firstly demonstrated that PPARα is a key molecule in the functional conversion of ER stress: protective effects in the mild ER stress was mediated by PPARα-autophagy pathway and destructive effects in the serious ER stress was mediated by PPARα-CHOP pathway.
Collapse
|
25
|
Obaidi I, Cassidy H, Ibáñez Gaspar V, McCaul J, Higgins M, Halász M, Reynolds AL, Kennedy BN, McMorrow T. Curcumin Sensitizes Kidney Cancer Cells to TRAIL-Induced Apoptosis via ROS Mediated Activation of JNK-CHOP Pathway and Upregulation of DR4. BIOLOGY 2020; 9:E92. [PMID: 32370057 PMCID: PMC7284747 DOI: 10.3390/biology9050092] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 04/24/2020] [Accepted: 04/25/2020] [Indexed: 12/14/2022]
Abstract
Tumour necrosis factor-related apoptosis-inducing ligand (TRAIL), is a selective anticancer cytokine capable of exerting a targeted therapy approach. Disappointingly, recent research has highlighted the development of TRAIL resistance in cancer cells, thus minimising its usefulness in clinical settings. However, several recent studies have demonstrated that cancer cells can be sensitised to TRAIL through the employment of a combinatorial approach, utilizing TRAIL in conjunction with other natural or synthetic anticancer agents. In the present study, the chemo-sensitising effect of curcumin on TRAIL-induced apoptosis in renal carcinoma cells (RCC) was investigated. The results indicate that exposure of kidney cancer ACHN cells to curcumin sensitised the cells to TRAIL, with the combination treatment of TRAIL and curcumin synergistically targeting the cancer cells without affecting the normal renal proximal tubular epithelial cells (RPTEC/TERT1) cells. Furthermore, this combination treatment was shown to induce caspase-dependent apoptosis, inhibition of the proteasome, induction of ROS, upregulation of death receptor 4 (DR4), alterations in mitogen-activated protein kinase (MAPK) signalling and induction of endoplasmic reticulum stress. An in vivo zebrafish embryo study demonstrated the effectiveness of the combinatorial regime to inhibit tumour formation without affecting zebrafish embryo viability or development. Overall, the results arising from this study demonstrate that curcumin has the ability to sensitise TRAIL-resistant ACHN cells to TRAIL-induced apoptosis.
Collapse
Affiliation(s)
- Ismael Obaidi
- NIBRT|National Institute for Bioprocessing, Research and Training, Foster Avenue, Mount Merrion, Blackrock, Co., A94 X099 Dublin, Ireland
- UCD Centre for Toxicology, School of Biomedical and Biomolecular Sciences, Conway Institute, University College Dublin, 4 Dublin, Ireland; (H.C.); (V.I.G.); (J.M.); (M.H.); (A.L.R.); (B.N.K.)
- College of Pharmacy, University of Babylon, Babylon 51002, Iraq
| | - Hilary Cassidy
- UCD Centre for Toxicology, School of Biomedical and Biomolecular Sciences, Conway Institute, University College Dublin, 4 Dublin, Ireland; (H.C.); (V.I.G.); (J.M.); (M.H.); (A.L.R.); (B.N.K.)
- Systems Biology Ireland, School of Medicine, University College Dublin, Belfield, 4 Dublin, Ireland;
| | - Verónica Ibáñez Gaspar
- UCD Centre for Toxicology, School of Biomedical and Biomolecular Sciences, Conway Institute, University College Dublin, 4 Dublin, Ireland; (H.C.); (V.I.G.); (J.M.); (M.H.); (A.L.R.); (B.N.K.)
| | - Jasmin McCaul
- UCD Centre for Toxicology, School of Biomedical and Biomolecular Sciences, Conway Institute, University College Dublin, 4 Dublin, Ireland; (H.C.); (V.I.G.); (J.M.); (M.H.); (A.L.R.); (B.N.K.)
| | - Michael Higgins
- UCD Centre for Toxicology, School of Biomedical and Biomolecular Sciences, Conway Institute, University College Dublin, 4 Dublin, Ireland; (H.C.); (V.I.G.); (J.M.); (M.H.); (A.L.R.); (B.N.K.)
| | - Melinda Halász
- Systems Biology Ireland, School of Medicine, University College Dublin, Belfield, 4 Dublin, Ireland;
| | - Alison L. Reynolds
- UCD Centre for Toxicology, School of Biomedical and Biomolecular Sciences, Conway Institute, University College Dublin, 4 Dublin, Ireland; (H.C.); (V.I.G.); (J.M.); (M.H.); (A.L.R.); (B.N.K.)
- UCD School of Veterinary Medicine, Rm 232, University College Dublin, Belfield, 4 Dublin, Ireland
| | - Breandan N. Kennedy
- UCD Centre for Toxicology, School of Biomedical and Biomolecular Sciences, Conway Institute, University College Dublin, 4 Dublin, Ireland; (H.C.); (V.I.G.); (J.M.); (M.H.); (A.L.R.); (B.N.K.)
| | - Tara McMorrow
- UCD Centre for Toxicology, School of Biomedical and Biomolecular Sciences, Conway Institute, University College Dublin, 4 Dublin, Ireland; (H.C.); (V.I.G.); (J.M.); (M.H.); (A.L.R.); (B.N.K.)
| |
Collapse
|
26
|
Zheng L, Jin J, Shi L, Huang J, Chang M, Wang X, Zhang H, Jin Q. Gamma tocopherol, its dimmers, and quinones: Past and future trends. Crit Rev Food Sci Nutr 2020; 60:3916-3930. [DOI: 10.1080/10408398.2020.1711704] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Liyou Zheng
- National Engineering Research Center for Functional Food, Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, School of Food Science and Technology, Jiangnan University, Jiangsu, P. R. China
| | - Jun Jin
- National Engineering Research Center for Functional Food, Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, School of Food Science and Technology, Jiangnan University, Jiangsu, P. R. China
| | - Longkai Shi
- National Engineering Research Center for Functional Food, Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, School of Food Science and Technology, Jiangnan University, Jiangsu, P. R. China
| | - Jianhua Huang
- National Engineering Research Center for Functional Food, Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, School of Food Science and Technology, Jiangnan University, Jiangsu, P. R. China
| | - Ming Chang
- National Engineering Research Center for Functional Food, Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, School of Food Science and Technology, Jiangnan University, Jiangsu, P. R. China
| | - Xingguo Wang
- National Engineering Research Center for Functional Food, Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, School of Food Science and Technology, Jiangnan University, Jiangsu, P. R. China
| | - Hui Zhang
- National Engineering Research Center for Functional Food, Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, School of Food Science and Technology, Jiangnan University, Jiangsu, P. R. China
| | - Qingzhe Jin
- National Engineering Research Center for Functional Food, Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, School of Food Science and Technology, Jiangnan University, Jiangsu, P. R. China
| |
Collapse
|
27
|
Analysis of the Mechanisms of Action of Naphthoquinone-Based Anti-Acute Myeloid Leukemia Chemotherapeutics. Molecules 2019; 24:molecules24173121. [PMID: 31466259 PMCID: PMC6749238 DOI: 10.3390/molecules24173121] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2019] [Revised: 08/24/2019] [Accepted: 08/25/2019] [Indexed: 01/02/2023] Open
Abstract
Acute myeloid leukemia (AML) is a neoplastic disorder resulting from clonal proliferation of poorly differentiated immature myeloid cells. Distinct genetic and epigenetic aberrations are key features of AML that account for its variable response to standard therapy. Irrespective of their oncogenic mutations, AML cells produce elevated levels of reactive oxygen species (ROS). They also alter expression and activity of antioxidant enzymes to promote cell proliferation and survival. Subsequently, selective targeting of redox homeostasis in a molecularly heterogeneous disease, such as AML, has been an appealing approach in the development of novel anti-leukemic chemotherapeutics. Naphthoquinones are able to undergo redox cycling and generate ROS in cancer cells, which have made them excellent candidates for testing against AML cells. In addition to inducing oxidative imbalance in AML cells, depending on their structure, naphthoquinones negatively affect other cellular apparatus causing neoplastic cell death. Here we provide an overview of the anti-AML activities of naphthoquinone derivatives, as well as analysis of their mechanism of action, including induction of reduction-oxidation imbalance, alteration in mitochondrial transmembrane potential, Bcl-2 modulation, initiation of DNA damage, and modulation of MAPK and STAT3 activity, alterations in the unfolded protein response and translocation of FOX-related transcription factors to the nucleus.
Collapse
|
28
|
Torres S, Baulies A, Insausti-Urkia N, Alarcón-Vila C, Fucho R, Solsona-Vilarrasa E, Núñez S, Robles D, Ribas V, Wakefield L, Grompe M, Lucena MI, Andrade RJ, Win S, Aung TA, Kaplowitz N, García-Ruiz C, Fernández-Checa JC. Endoplasmic Reticulum Stress-Induced Upregulation of STARD1 Promotes Acetaminophen-Induced Acute Liver Failure. Gastroenterology 2019; 157:552-568. [PMID: 31029706 DOI: 10.1053/j.gastro.2019.04.023] [Citation(s) in RCA: 82] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 04/11/2019] [Accepted: 04/20/2019] [Indexed: 01/14/2023]
Abstract
BACKGROUND & AIMS Acetaminophen (APAP) overdose is a major cause of acute liver failure (ALF). Mitochondrial SH3BP5 (also called SAB) and phosphorylation of c-Jun N-terminal kinase (JNK) mediate the hepatotoxic effects of APAP. We investigated the involvement of steroidogenic acute regulatory protein (STARD1), a mitochondrial cholesterol transporter, in this process and sensitization by valproic acid (VPA), which depletes glutathione and stimulates steroidogenesis. METHODS Nonfasted C57BL/6J mice (control) and mice with liver-specific deletion of STARD1 (Stard1ΔHep), SAB (SabΔHep), or JNK1 and JNK2 (Jnk1+2ΔHep) were given VPA with or without APAP. Liver tissues were collected and analyzed by histology and immunohistochemistry and for APAP metabolism, endoplasmic reticulum (ER) stress, and mitochondrial function. Adult human hepatocytes were transplanted into Fah-/-/Rag2-/-/Il2rg-/-/NOD (FRGN) mice to create mice with humanized livers. RESULTS Administration of VPA before administration of APAP increased the severity of liver damage in control mice. The combination of VPA and APAP increased expression of CYP2E1, formation of NAPQI-protein adducts, and depletion of glutathione from liver tissues of control mice, resulting in ER stress and the upregulation of STARD1. Livers from control mice given VPA and APAP accumulated cholesterol in the mitochondria and had sustained mitochondrial depletion of glutathione and mitochondrial dysfunction. Inhibition of ER stress, by administration of tauroursodeoxycholic acid to control mice, prevented upregulation of STARD1 in liver and protected the mice from hepatoxicity following administration of VPA and APAP. Administration of N-acetylcysteine to control mice prevented VPA- and APAP-induced ER stress and liver injury. Stard1ΔHep mice were resistant to induction of ALF by VPA and APAP, despite increased mitochondrial levels of glutathione and phosphorylated JNK; we made similar observations in fasted Stard1ΔHep mice given APAP alone. SabΔHep mice or Jnk1+2ΔHep mice did not develop ALF following administration of VPA and APAP. The ability of VPA to increase the severity of APAP-induced liver damage was observed in FRGN mice with humanized liver. CONCLUSIONS In studies of mice, we found that upregulation of STARD1 following ER stress mediates APAP hepatoxicity via SH3BP5 and phosphorylation of JNK1 and JNK2.
Collapse
Affiliation(s)
- Sandra Torres
- Cell Death and Proliferation, IIBB-CSIC, Barcelona, Spain; Liver Unit, Hospital Cínic, IDIBAPS and CIBEREHD, Barcelona, Spain
| | - Anna Baulies
- Cell Death and Proliferation, IIBB-CSIC, Barcelona, Spain; Liver Unit, Hospital Cínic, IDIBAPS and CIBEREHD, Barcelona, Spain
| | - Naroa Insausti-Urkia
- Cell Death and Proliferation, IIBB-CSIC, Barcelona, Spain; Liver Unit, Hospital Cínic, IDIBAPS and CIBEREHD, Barcelona, Spain
| | - Cristina Alarcón-Vila
- Cell Death and Proliferation, IIBB-CSIC, Barcelona, Spain; Liver Unit, Hospital Cínic, IDIBAPS and CIBEREHD, Barcelona, Spain
| | - Raquel Fucho
- Cell Death and Proliferation, IIBB-CSIC, Barcelona, Spain; Liver Unit, Hospital Cínic, IDIBAPS and CIBEREHD, Barcelona, Spain
| | - Estel Solsona-Vilarrasa
- Cell Death and Proliferation, IIBB-CSIC, Barcelona, Spain; Liver Unit, Hospital Cínic, IDIBAPS and CIBEREHD, Barcelona, Spain
| | - Susana Núñez
- Cell Death and Proliferation, IIBB-CSIC, Barcelona, Spain; Liver Unit, Hospital Cínic, IDIBAPS and CIBEREHD, Barcelona, Spain
| | - David Robles
- Cell Death and Proliferation, IIBB-CSIC, Barcelona, Spain; Liver Unit, Hospital Cínic, IDIBAPS and CIBEREHD, Barcelona, Spain
| | - Vicent Ribas
- Cell Death and Proliferation, IIBB-CSIC, Barcelona, Spain; Liver Unit, Hospital Cínic, IDIBAPS and CIBEREHD, Barcelona, Spain
| | | | - Markus Grompe
- Oregon Health and Science University, Portland, Oregon
| | - M Isabel Lucena
- Unidad de Gestión Clínica de Aparato Digestivo, Servicio de Farmacología Clínica, Instituto de Investigación Biomédica de Málaga-IBIMA, CIBEREHD, Hospital Universitario Virgen de la Victoria, Universidad de Málaga, Málaga, Spain
| | - Raul J Andrade
- Unidad de Gestión Clínica de Aparato Digestivo, Servicio de Farmacología Clínica, Instituto de Investigación Biomédica de Málaga-IBIMA, CIBEREHD, Hospital Universitario Virgen de la Victoria, Universidad de Málaga, Málaga, Spain
| | - Sanda Win
- USC Research Center for Liver Disease, USC Keck School of Medicine, Los Angeles, California
| | - Tin A Aung
- USC Research Center for Liver Disease, USC Keck School of Medicine, Los Angeles, California
| | - Neil Kaplowitz
- USC Research Center for Liver Disease, USC Keck School of Medicine, Los Angeles, California
| | - Carmen García-Ruiz
- Cell Death and Proliferation, IIBB-CSIC, Barcelona, Spain; Liver Unit, Hospital Cínic, IDIBAPS and CIBEREHD, Barcelona, Spain; USC Research Center for ALPD, Keck School of Medicine, Los Angeles, California
| | - Jose C Fernández-Checa
- Cell Death and Proliferation, IIBB-CSIC, Barcelona, Spain; Liver Unit, Hospital Cínic, IDIBAPS and CIBEREHD, Barcelona, Spain; USC Research Center for ALPD, Keck School of Medicine, Los Angeles, California.
| |
Collapse
|
29
|
Carcamo-Noriega EN, Sathyamoorthi S, Banerjee S, Gnanamani E, Mendoza-Trujillo M, Mata-Espinosa D, Hernández-Pando R, Veytia-Bucheli JI, Possani LD, Zare RN. 1,4-Benzoquinone antimicrobial agents against Staphylococcus aureus and Mycobacterium tuberculosis derived from scorpion venom. Proc Natl Acad Sci U S A 2019; 116:12642-12647. [PMID: 31182590 PMCID: PMC6600905 DOI: 10.1073/pnas.1812334116] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Two 1,4-benzoquinone derivatives, found in the venom of the scorpion Diplocentrus melici following exposure to air, have been isolated, characterized, synthesized, and assessed for antimicrobial activities. Initially a white, viscous liquid, the extracted venom colors within minutes under ambient conditions. From this colored mixture, two compounds, one red, the other blue, were isolated and purified using chromatography. After a variety of NMR and mass spectrometry experiments, the red compound was determined to be 3,5- dimethoxy-2-(methylthio)cyclohexa-2,5-diene-1,4-dione, and the blue compound was determined to be 5-methoxy-2,3- bis(methylthio)cyclohexa-2,5-diene-1,4-dione. Because extremely small amounts of these compounds were isolated from the scorpion venom, we developed laboratory syntheses from commercially available precursors, allowing us to produce sufficient quantities for crystallization and biological assays. The red benzoquinone is effective against Staphylococcus aureus [minimum inhibitory concentration (MIC) = 4 µg/mL], while the blue benzoquinone is active against Mycobacterium tuberculosis (MIC = 4 µg/mL) and even against a multidrug-resistant (MDR) strain with nearly equal effectiveness. The bactericidal effects of both benzoquinones show comparable activity to commercially available antibiotics used against these pathogens and were cytotoxic to neoplastic cell lines, suggesting their potential as lead compounds for the development of novel antimicrobial and anticancer drugs. Importantly, the blue benzoquinone was also effective in vivo with mouse models of MDR tuberculosis infection. After treatment for 2 mo, four mice with late-stage active MDR tuberculosis had a significant decrease in pulmonary bacillary loads and tissue damage. Healthy mice served as negative controls and tolerated treatment well, without adverse side effects.
Collapse
Affiliation(s)
- Edson Norberto Carcamo-Noriega
- Department of Molecular Medicine and Bioprocesses, Instituto de Biotecnologia, Universidad Nacional Autonoma de Mexico, 62210 Morelos, Mexico
| | | | - Shibdas Banerjee
- Department of Chemistry, Stanford University, Stanford, CA 94305
- Department of Chemistry, Indian Institute of Science Education and Research Tirupati, Tirupati 517507, India
| | | | - Monserrat Mendoza-Trujillo
- Section of Experimental Pathology, Department of Pathology, Instituto Nacional de Ciencias Médicas y Nutrición "Salvador Zubirán," 14080 Mexico City, Mexico
| | - Dulce Mata-Espinosa
- Section of Experimental Pathology, Department of Pathology, Instituto Nacional de Ciencias Médicas y Nutrición "Salvador Zubirán," 14080 Mexico City, Mexico
| | - Rogelio Hernández-Pando
- Section of Experimental Pathology, Department of Pathology, Instituto Nacional de Ciencias Médicas y Nutrición "Salvador Zubirán," 14080 Mexico City, Mexico
| | - José Ignacio Veytia-Bucheli
- Department of Molecular Medicine and Bioprocesses, Instituto de Biotecnologia, Universidad Nacional Autonoma de Mexico, 62210 Morelos, Mexico
| | - Lourival D Possani
- Department of Molecular Medicine and Bioprocesses, Instituto de Biotecnologia, Universidad Nacional Autonoma de Mexico, 62210 Morelos, Mexico;
| | - Richard N Zare
- Department of Chemistry, Stanford University, Stanford, CA 94305;
| |
Collapse
|
30
|
Abstract
Acetaminophen (APAP) is one of the most popular and safe pain medications worldwide. However, due to its wide availability, it is frequently implicated in intentional or unintentional overdoses where it can cause severe liver injury and even acute liver failure (ALF). In fact, APAP toxicity is responsible for 46% of all ALF cases in the United States. Early mechanistic studies in mice demonstrated the formation of a reactive metabolite, which is responsible for hepatic glutathione depletion and initiation of the toxicity. This insight led to the rapid introduction of N-acetylcysteine as a clinical antidote. However, more recently, substantial progress was made in further elucidating the detailed mechanisms of APAP-induced cell death. Mitochondrial protein adducts trigger a mitochondrial oxidant stress, which requires amplification through a MAPK cascade that ultimately results in activation of c-jun N-terminal kinase (JNK) in the cytosol and translocation of phospho-JNK to the mitochondria. The enhanced oxidant stress is responsible for the membrane permeability transition pore opening and the membrane potential breakdown. The ensuing matrix swelling causes the release of intermembrane proteins such as endonuclease G, which translocate to the nucleus and induce DNA fragmentation. These pathophysiological signaling mechanisms can be additionally modulated by removing damaged mitochondria by autophagy and replacing them by mitochondrial biogenesis. Importantly, most of the mechanisms have been confirmed in human hepatocytes and indirectly through biomarkers in plasma of APAP overdose patients. The extensive necrosis caused by APAP overdose leads to a sterile inflammatory response. Although recruitment of inflammatory cells is necessary for removal of cell debris in preparation for regeneration, these cells have the potential to aggravate the injury. This review touches on the newest insight into the intracellular mechanisms of APAP-induced cells death and the resulting inflammatory response. Furthermore, it discusses the translation of these findings to humans and the emergence of new therapeutic interventions.
Collapse
Affiliation(s)
- Anup Ramachandran
- Department of Pharmacology, Toxicology & Therapeutics, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Hartmut Jaeschke
- Department of Pharmacology, Toxicology & Therapeutics, University of Kansas Medical Center, Kansas City, KS 66160, USA
| |
Collapse
|
31
|
Novel Curcumin Inspired Bis-Chalcone Promotes Endoplasmic Reticulum Stress and Glioblastoma Neurosphere Cell Death. Cancers (Basel) 2019; 11:cancers11030357. [PMID: 30871215 PMCID: PMC6468769 DOI: 10.3390/cancers11030357] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 03/04/2019] [Accepted: 03/07/2019] [Indexed: 01/11/2023] Open
Abstract
Glioblastoma (GBM) has a dismal prognosis and successful elimination of GBM stem cells (GSCs) is a high-priority as these cells are responsible for tumor regrowth following therapy and ultimately patient relapse. Natural products and their derivatives continue to be a source for the development of effective anticancer drugs and have been shown to effectively target pathways necessary for cancer stem cell self-renewal and proliferation. We generated a series of curcumin inspired bis-chalcones and examined their effect in multiple patient-derived GSC lines. Of the 19 compounds synthesized, four analogs robustly induced GSC death in six separate GSC lines, with a half maximal inhibitory concentration (IC50) ranging from 2.7–5.8 μM and significantly reduced GSC neurosphere formation at sub-cytotoxic levels. Structural analysis indicated that the presence of a methoxy group at position 3 of the lateral phenylic appendages was important for activity. Pathway and drug connectivity analysis of gene expression changes in response to treatment with the most active bis-chalcone 4j (the 3,4,5 trimethoxy substituted analog) suggested that the mechanism of action was the induction of endoplasmic reticulum (ER) stress and unfolded protein response (UPR) mediated cell death. This was confirmed by Western blot analysis in which 4j induced robust increases in CHOP, p-jun and caspase 12. The UPR is believed to play a significant role in GBM pathogenesis and resistance to therapy and as such represents a promising therapeutic target.
Collapse
|
32
|
Aivazidis S, Anderson CC, Roede JR. Toxicant-mediated redox control of proteostasis in neurodegeneration. CURRENT OPINION IN TOXICOLOGY 2019; 13:22-34. [PMID: 31602419 PMCID: PMC6785977 DOI: 10.1016/j.cotox.2018.12.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Disruption in redox signaling and control of cellular processes has emerged as a key player in many pathologies including neurodegeneration. As protein aggregations are a common hallmark of several neuronal pathologies, a firm understanding of the interplay between redox signaling, oxidative and free radical stress, and proteinopathies is required to sort out the complex mechanisms in these diseases. Fortunately, models of toxicant-induced neurodegeneration can be utilized to evaluate and report mechanistic alterations in the proteostasis network (PN). The epidemiological links between environmental toxicants and neurological disease gives further credence into characterizing the toxicant-mediated PN disruptions observed in these conditions. Reviewed here are examples of mechanistic interaction between oxidative or free radical stress and PN alterations. Additionally, investigations into toxicant-mediated PN disruptions, specifically focusing on environmental metals and pesticides, are discussed. Finally, we emphasize the need to distinguish whether the presence of protein aggregations are contributory to phenotypes related to neurodegeneration, or if they are a byproduct of PN deficiencies.
Collapse
Affiliation(s)
- Stefanos Aivazidis
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Colin C Anderson
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - James R Roede
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| |
Collapse
|
33
|
Grushevskaya HV, Krylova NG. Carbon Nanotubes as A High-Performance Platform for Target Delivery of Anticancer Quinones. Curr Pharm Des 2019; 24:5207-5218. [PMID: 30652640 DOI: 10.2174/1381612825666190117095132] [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: 12/02/2018] [Accepted: 01/11/2019] [Indexed: 12/20/2022]
Abstract
BACKGROUND In spite of considerable efforts of researchers the cancer deseases remain to be incurable and a percentage of cancer deseases in the structure of mortality increases every year. At that, high systemic toxicity of antitumor drugs hampers their effective use. Because of this fact, the development of nanosystems for targeted delivery of antitumor drugs is one of the leading problem in nanomedicine and nanopharmacy. OBJECTIVE To critically examine the modern strategies for carbon nanotubes (CNTs)-based delivery of anticancer quinones and to summarize the mechanisms which can provide high effectiveness and multifunctionality of the CNT-based quinone delivery platform. RESULTS Quinones, including anthracycline antibiotics - doxorubicin and daunorubicin, are among the most prospective group of natural and syntetic compounds which exhibit high antitumor activity against different type of tumors. In this review, we focus on the possibilities of using CNTs for targeted delivery of antitumor compounds with quinoid moiety which is ordinarily characterized by high specific interaction with DNA molecules. Quinones can be non-covalently adsorbed on CNT surface due to their aromatic structure and π-conjugated system of double bonds. The characteristic features of doxorubicine-CNT complex are high loading efficiency, pH-dependent release in acidic tumor microenviroment, enough stability in biological fluid. Different types of CNT functionalization, targeting strategies and designs for multifunctional CNT-based doxorubicine delivery platform are disscussed. CONCLUSION Nanosystems based on functionalized CNTs are very promising platform for quinone delivery resulting in significant enhancement of cancer treatment efficiency. Functionalization of CNTs with the polymeric shell, especially DNA-based shells, can provide the greatest affinity and mimicry with biological structures.
Collapse
Affiliation(s)
- H V Grushevskaya
- Physics Department, Belarusian State University, 4 Nezavisimosti Ave., Minsk 220030, Belarus
| | - N G Krylova
- Physics Department, Belarusian State University, 4 Nezavisimosti Ave., Minsk 220030, Belarus
| |
Collapse
|
34
|
Melnikov F, Botta D, White CC, Schmuck SC, Winfough M, Schaupp CM, Gallagher EP, Brooks BW, Williams ES, Coish P, Anastas PT, Voutchkova-Kostal A, Kostal J, Kavanagh TJ. Kinetics of Glutathione Depletion and Antioxidant Gene Expression as Indicators of Chemical Modes of Action Assessed in Vitro in Mouse Hepatocytes with Enhanced Glutathione Synthesis. Chem Res Toxicol 2019; 32:421-436. [PMID: 30547568 DOI: 10.1021/acs.chemrestox.8b00259] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Here we report a vertically integrated in vitro - in silico study that aims to elucidate the molecular initiating events involved in the induction of oxidative stress (OS) by seven diverse chemicals (cumene hydroperoxide, t-butyl hydroperoxide, hydroquinone, t-butyl hydroquinone, bisphenol A, Dinoseb, and perfluorooctanoic acid). To that end, we probe the relationship between chemical properties, cell viability, glutathione (GSH) depletion, and antioxidant gene expression. Concentration-dependent effects on cell viability were assessed by MTT assay in two Hepa-1 derived mouse liver cell lines: a control plasmid vector transfected cell line (Hepa-V), and a cell line with increased glutamate-cysteine ligase (GCL) activity and GSH content (CR17). Changes to intracellular GSH content and mRNA expression levels for the Nrf2-driven antioxidant genes Gclc, Gclm, heme oxygenase-1 ( Hmox1), and NADPH quinone oxidoreductase-1 ( Nqo1) were monitored after sublethal exposure to the chemicals. In silico models of covalent and redox reactivity were used to rationalize differences in activity of quinones and peroxides. Our findings show CR17 cells were generally more resistant to chemical toxicity and showed markedly attenuated induction of OS biomarkers; however, differences in viability effects between the two cell lines were not the same for all chemicals. The results highlight the vital role of GSH in protecting against oxidative stress-inducing chemicals as well as the importance of probing molecular initiating events in order to identify chemicals with lower potential to cause oxidative stress.
Collapse
Affiliation(s)
- Fjodor Melnikov
- Yale School of Forestry and Environmental Sciences , Yale University , New Haven , Connecticut 06520 , United States
| | - Dianne Botta
- Department of Environmental and Occupational Health Sciences , University of Washington , Seattle , Washington 98195 , United States
| | - Collin C White
- Department of Environmental and Occupational Health Sciences , University of Washington , Seattle , Washington 98195 , United States
| | - Stefanie C Schmuck
- Department of Environmental and Occupational Health Sciences , University of Washington , Seattle , Washington 98195 , United States
| | - Matthew Winfough
- Department of Chemistry , George Washington University , Washington , D.C. 20052 , United States
| | - Christopher M Schaupp
- Department of Environmental and Occupational Health Sciences , University of Washington , Seattle , Washington 98195 , United States
| | - Evan P Gallagher
- Department of Environmental and Occupational Health Sciences , University of Washington , Seattle , Washington 98195 , United States
| | - Bryan W Brooks
- Department of Environmental Science , Baylor University , Waco , Texas 76798 , United States
| | - Edward Spencer Williams
- Department of Environmental Science , Baylor University , Waco , Texas 76798 , United States
| | - Philip Coish
- Yale School of Forestry and Environmental Sciences , Yale University , New Haven , Connecticut 06520 , United States
| | - Paul T Anastas
- Yale School of Forestry and Environmental Sciences , Yale University , New Haven , Connecticut 06520 , United States.,School of Public Health , Yale University , New Haven , Connecticut 06520 , United States
| | | | - Jakub Kostal
- Department of Chemistry , George Washington University , Washington , D.C. 20052 , United States
| | - Terrance J Kavanagh
- Department of Environmental and Occupational Health Sciences , University of Washington , Seattle , Washington 98195 , United States
| |
Collapse
|
35
|
Żur J, Wojcieszyńska D, Hupert-Kocurek K, Marchlewicz A, Guzik U. Paracetamol - toxicity and microbial utilization. Pseudomonas moorei KB4 as a case study for exploring degradation pathway. CHEMOSPHERE 2018; 206:192-202. [PMID: 29751245 DOI: 10.1016/j.chemosphere.2018.04.179] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Revised: 04/25/2018] [Accepted: 04/30/2018] [Indexed: 06/08/2023]
Abstract
Paracetamol, a widely used analgesic and antipyretic drug, is currently one of the most emerging pollutants worldwide. Besides its wide prevalence in the literature only several bacterial strains able to degrade this compound have been described. In this study, we isolated six new bacterial strains able to remove paracetamol. The isolated strains were identified as the members of Pseudomonas, Bacillus, Acinetobacter and Sphingomonas genera and characterized phenotypically and biochemically using standard methods. From the isolated strains, Pseudomonas moorei KB4 was able to utilize 50 mg L-1 of paracetamol. As the main degradation products, p-aminophenol and hydroquinone were identified. Based on the measurements of specific activity of acyl amidohydrolase, deaminase and hydroquinone 1,2-dioxygenase and the results of liquid chromatography analyses, we proposed a mechanism of paracetamol degradation by KB4 strain under co-metabolic conditions with glucose. Additionally, toxicity bioassays and the influence of various environmental factors, including pH, temperature, heavy metals at no-observed-effective-concentrations, and the presence of aromatic compounds on the efficiency and mechanism of paracetamol degradation by KB4 strain were determined. This comprehensive study about paracetamol biodegradation will be helpful in designing a treatment systems of wastewaters contaminated with paracetamol.
Collapse
Affiliation(s)
- Joanna Żur
- Department of Biochemistry, Faculty of Biology and Environmental Protection, University of Silesia in Katowice, Jagiellońska 28, 40-032 Katowice, Poland.
| | - Danuta Wojcieszyńska
- Department of Biochemistry, Faculty of Biology and Environmental Protection, University of Silesia in Katowice, Jagiellońska 28, 40-032 Katowice, Poland.
| | - Katarzyna Hupert-Kocurek
- Department of Biochemistry, Faculty of Biology and Environmental Protection, University of Silesia in Katowice, Jagiellońska 28, 40-032 Katowice, Poland.
| | - Ariel Marchlewicz
- Department of Biochemistry, Faculty of Biology and Environmental Protection, University of Silesia in Katowice, Jagiellońska 28, 40-032 Katowice, Poland.
| | - Urszula Guzik
- Department of Biochemistry, Faculty of Biology and Environmental Protection, University of Silesia in Katowice, Jagiellońska 28, 40-032 Katowice, Poland.
| |
Collapse
|
36
|
Autophagy and acetaminophen-induced hepatotoxicity. Arch Toxicol 2018; 92:2153-2161. [PMID: 29876591 DOI: 10.1007/s00204-018-2237-5] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Accepted: 06/04/2018] [Indexed: 12/15/2022]
Abstract
Acetaminophen (APAP) is a widely used analgesic and antipyretic drug. APAP overdose can induce acute liver injury in humans, which is responsible for approximately 50% of total cases of acute liver failure in the United States and some European countries. Currently, the metabolism of APAP in the body has been extensively investigated; however, the exact mechanisms for APAP hepatotoxicity are not well understood. Recent studies have shown that mitochondrial dysfunction, oxidative stress and inflammatory responses play a critical role in the pathogenesis of APAP hepatotoxicity. Autophagy is a catabolic machinery aimed at recycling cellular components and damaged organelles in response to a variety of stimuli, such as nutrient deprivation and toxic stress. Increasing evidence supports that autophagy is involved in the pathophysiological process of APAP-induced liver injury. In this review, we summarized the changes of autophagy in the liver following APAP intoxication and discussed the role and its possible mechanisms of autophagy in APAP hepatotoxicity. Furthermore, this review highlights the crosstalk between mitophagy, oxidative stress and inflammation in APAP-induced liver injury and presents some possible molecular mechanisms by which activated autophagy protects against APAP-induced liver injury.
Collapse
|
37
|
Yan M, Huo Y, Yin S, Hu H. Mechanisms of acetaminophen-induced liver injury and its implications for therapeutic interventions. Redox Biol 2018; 17:274-283. [PMID: 29753208 PMCID: PMC6006912 DOI: 10.1016/j.redox.2018.04.019] [Citation(s) in RCA: 362] [Impact Index Per Article: 60.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Revised: 04/18/2018] [Accepted: 04/18/2018] [Indexed: 02/06/2023] Open
Abstract
Acetaminophen (APAP) overdose is the leading cause of drug-induced acute liver failure in many developed countries. Mitochondrial oxidative stress is considered to be the predominant cellular event in APAP-induced liver injury. Accordingly, N-acetyl cysteine, a known scavenger of reactive oxygen species (ROS), is recommended as an effective clinical antidote against APAP-induced acute liver injury (AILI) when it is given at an early phase; however, the narrow therapeutic window limits its use. Hence, the development of novel therapeutic approaches that can offer broadly protective effects against AILI is clearly needed. To this end, it is necessary to better understand the mechanisms of APAP hepatotoxicity. Up to now, in addition to mitochondrial oxidative stress, many other cellular processes, including phase I/phase II metabolism, endoplasmic reticulum stress, autophagy, sterile inflammation, microcirculatory dysfunction, and liver regeneration, have been identified to be involved in the pathogenesis of AILI, providing new targets for developing more effective therapeutic interventions against APAP-induced liver injury. In this review, we summarize intracellular and extracellular events involved in APAP hepatotoxicity, along with emphatic discussions on the possible therapeutic approaches targeting these different cellular events.
Collapse
Affiliation(s)
- Mingzhu Yan
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing Key Laboratory for Food Non-thermal Processing, National Engineering Research Centre for Fruit and Vegetable Processing, College of Food Science and Nutritional Engineering, China Agricultural University, No. 17 Qinghua East Road, Haidian District, Beijing 100083, China
| | - Yazhen Huo
- State Key Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Shutao Yin
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing Key Laboratory for Food Non-thermal Processing, National Engineering Research Centre for Fruit and Vegetable Processing, College of Food Science and Nutritional Engineering, China Agricultural University, No. 17 Qinghua East Road, Haidian District, Beijing 100083, China
| | - Hongbo Hu
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing Key Laboratory for Food Non-thermal Processing, National Engineering Research Centre for Fruit and Vegetable Processing, College of Food Science and Nutritional Engineering, China Agricultural University, No. 17 Qinghua East Road, Haidian District, Beijing 100083, China.
| |
Collapse
|
38
|
Zhang Y, den Braver-Sewradj SP, den Braver MW, Hiemstra S, Vermeulen NPE, van de Water B, Commandeur JNM, Vos JC. Glutathione S-Transferase P1 Protects Against Amodiaquine Quinoneimines-Induced Cytotoxicity but Does Not Prevent Activation of Endoplasmic Reticulum Stress in HepG2 Cells. Front Pharmacol 2018; 9:388. [PMID: 29720942 PMCID: PMC5915463 DOI: 10.3389/fphar.2018.00388] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Accepted: 04/04/2018] [Indexed: 12/15/2022] Open
Abstract
Formation of the reactive amodiaquine quinoneimine (AQ-QI) and N-desethylamodiaquine quinoneimine (DEAQ-QI) plays an important role in the toxicity of the anti-malaria drug amodiaquine (AQ). Glutathione conjugation protects against AQ-induced toxicity and GSTP1 is able to conjugate its quinoneimine metabolites AQ-QI and DEA-QI with glutathione. In this study, HepG2 cells transiently transfected with the human GSTP1 construct were utilized to investigate the protective effect of GSTP1 in a cellular context. HepG2 cells were exposed to synthesized QIs, which bypasses the need for intracellular bioactivation of AQ or DEAQ. Exposure was accompanied by decreased cell viability, increased caspase 3 activity, and decreased intracellular GSH levels. Using high-content imaging-based BAC-GFP reporters, it was shown that AQ-QI and DEAQ-QI specifically activated the endoplasmic reticulum (ER) stress response. In contrast, oxidative stress, DNA damage, or inflammatory stress responses were not activated. Overexpression of GSTP1 resulted in a two-fold increase in GSH-conjugation of the QIs, attenuated QI-induced cytotoxicity especially under GSH-depletion condition, abolished QIs-induced apoptosis but did not significantly inhibit the activation of the ER stress response. In conclusion, these results indicate a protective role of GSTP1 by increasing enzymatic detoxification of AQ-QI and DEAQ-QI and suggest a second protective mechanism by interfering with ER stress induced apoptosis.
Collapse
Affiliation(s)
- Yongjie Zhang
- Division of Molecular Toxicology, Department of Chemistry and Pharmaceutical Sciences, Amsterdam Institute for Molecules, Medicines and Systems, Vrije Universiteit Amsterdam, Amsterdam, Netherlands.,Clinical Pharmacokinetics Research Laboratory, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Shalenie P den Braver-Sewradj
- Division of Molecular Toxicology, Department of Chemistry and Pharmaceutical Sciences, Amsterdam Institute for Molecules, Medicines and Systems, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Michiel W den Braver
- Division of Molecular Toxicology, Department of Chemistry and Pharmaceutical Sciences, Amsterdam Institute for Molecules, Medicines and Systems, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Steven Hiemstra
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, Leiden, Netherlands
| | - Nico P E Vermeulen
- Division of Molecular Toxicology, Department of Chemistry and Pharmaceutical Sciences, Amsterdam Institute for Molecules, Medicines and Systems, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Bob van de Water
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, Leiden, Netherlands
| | - Jan N M Commandeur
- Division of Molecular Toxicology, Department of Chemistry and Pharmaceutical Sciences, Amsterdam Institute for Molecules, Medicines and Systems, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - J C Vos
- Division of Molecular Toxicology, Department of Chemistry and Pharmaceutical Sciences, Amsterdam Institute for Molecules, Medicines and Systems, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
39
|
Saito Y. DJ-1 as a Biomarker of Parkinson's Disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1037:149-171. [PMID: 29147908 DOI: 10.1007/978-981-10-6583-5_10] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Parkinson's disease is a progressive, age-related, neurodegenerative disorder, and oxidative stress is an important mediator in its pathogenesis. DJ-1 has been identified as a causative gene of a familial form of Parkinson's disease, PARK7, and plays a significant role in antioxidative defense, protecting cells from oxidative stress. A cysteine residue of DJ-1 at position 106 (Cys-106) is preferentially oxidized under oxidative stress. This reactive Cys-106 plays a critical role in the biological function of DJ-1, which could act as a sensor of oxidative stress by regulating antioxidative defense depending on Cys-106 oxidation. Thus, the levels of Cys-106-oxidized DJ-1 (oxDJ-1) could be a possible biomarker of oxidative stress. This chapter focuses on the properties of DJ-1 and oxDJ-1 levels as a biomarker of Parkinson's disease. In particular, the usability of these biomarkers to prevent and treat this neurodegenerative disease is discussed. Further, this section deals with the importance of identifying a biomarker of early-phase Parkinson's disease. Finally, this chapter summarizes the features of oxDJ-1 levels in the brain and blood as a biomarker candidate for early-phase Parkinson's disease based on our results using oxDJ-1-specific antibodies.
Collapse
Affiliation(s)
- Yoshiro Saito
- Department of Medical Life Systems, Faculty of Life and Medical Sciences, Doshisha University, 1-3 Miyakodani, Tatara, Kyotanabe, Kyoto, 610-0394, Japan.
| |
Collapse
|
40
|
Inhibition of Cyanobacterial Growth on a Municipal Wastewater Sidestream Is Impacted by Temperature. mSphere 2018; 3:mSphere00538-17. [PMID: 29507895 PMCID: PMC5830474 DOI: 10.1128/msphere.00538-17] [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: 11/13/2017] [Accepted: 02/07/2018] [Indexed: 01/21/2023] Open
Abstract
Cyanobacteria are viewed as promising platforms to produce fuels and/or high-value chemicals as part of so-called “biorefineries.” Their integration into wastewater treatment systems is particularly interesting because removal of the nitrogen and phosphorus in many wastewater streams is an expensive but necessary part of wastewater treatment. In this study, we evaluated strategies for cultivating Synechococcus sp. strain PCC 7002 on media comprised of two wastewater streams, i.e., treated secondary effluent supplemented with the liquid fraction extracted from sludge following anaerobic digestion. This strain is commonly used for metabolic engineering to produce a variety of valuable chemical products and product precursors (e.g., lactate). However, initial attempts to grow PCC 7002 under otherwise-standard conditions of light and temperature failed. We thus systematically evaluated alternative cultivation conditions and then used multiple methods to dissect the apparent toxicity of the media under standard cultivation conditions. Sidestreams in wastewater treatment plants can serve as concentrated sources of nutrients (i.e., nitrogen and phosphorus) to support the growth of photosynthetic organisms that ultimately serve as feedstock for production of fuels and chemicals. However, other chemical characteristics of these streams may inhibit growth in unanticipated ways. Here, we evaluated the use of liquid recovered from municipal anaerobic digesters via gravity belt filtration as a nutrient source for growing the cyanobacterium Synechococcus sp. strain PCC 7002. The gravity belt filtrate (GBF) contained high levels of complex dissolved organic matter (DOM), which seemed to negatively influence cells. We investigated the impact of GBF on physiological parameters such as growth rate, membrane integrity, membrane composition, photosystem composition, and oxygen evolution from photosystem II. At 37°C, we observed an inverse correlation between GBF concentration and membrane integrity. Radical production was also detected upon exposure to GBF at 37°C. However, the dose-dependent relationship between the GBF concentration and the lack of membrane integrity was abolished at 27°C. Immediate resuspension of strains in high levels of GBF showed markedly reduced oxygen evolution rates relative to those seen with the control. Taken together, the data indicate that one mechanism responsible for GBF toxicity to Synechococcus is the interruption of photosynthetic electron flow and subsequent phenomena. We hypothesize that this is likely due to the presence of phenolic compounds within the DOM. IMPORTANCE Cyanobacteria are viewed as promising platforms to produce fuels and/or high-value chemicals as part of so-called “biorefineries.” Their integration into wastewater treatment systems is particularly interesting because removal of the nitrogen and phosphorus in many wastewater streams is an expensive but necessary part of wastewater treatment. In this study, we evaluated strategies for cultivating Synechococcus sp. strain PCC 7002 on media comprised of two wastewater streams, i.e., treated secondary effluent supplemented with the liquid fraction extracted from sludge following anaerobic digestion. This strain is commonly used for metabolic engineering to produce a variety of valuable chemical products and product precursors (e.g., lactate). However, initial attempts to grow PCC 7002 under otherwise-standard conditions of light and temperature failed. We thus systematically evaluated alternative cultivation conditions and then used multiple methods to dissect the apparent toxicity of the media under standard cultivation conditions.
Collapse
|
41
|
Rondeau G, Abedinpour P, Chrastina A, Pelayo J, Borgstrom P, Welsh J. Differential gene expression induced by anti-cancer agent plumbagin is mediated by androgen receptor in prostate cancer cells. Sci Rep 2018; 8:2694. [PMID: 29426892 PMCID: PMC5807367 DOI: 10.1038/s41598-018-20451-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Accepted: 01/15/2018] [Indexed: 12/24/2022] Open
Abstract
Treatment of mice harboring PTEN-P2 tumors in the prostate or on prostate tissue in vivo with 5-hydroxy-2-methyl-1,4-naphthoquinone, also known as plumbagin, results in tumor regression in castrated mice, but not in intact mice. This suggested that dihydrotestosterone (DHT) production in the testes may prevent cell death due to plumbagin treatment, but the underlying mechanism is not understood. We performed RNA-seq analysis on cells treated with combinations of plumbagin and DHT, and analyzed differential gene expression, to gain insight into the interactions between androgen and plumbgin. DHT and plumbagin synergize to alter the expression of many genes that are not differentially regulated by either single agent when used alone. These experiments revealed that, for many genes, increases in mRNAs caused by DHT are sharply down-regulated by plumbagin, and that many transcripts change in response to plumbagin in a DHT-dependent manner. This suggests that androgen receptor mediates some of the effects of plumbagin on gene expression.
Collapse
Affiliation(s)
- Gaelle Rondeau
- Vaccine Research Institute of San Diego, 3030 Bunker Hill Street, Suite 200, San Diego, CA, 92109, USA
| | - Parisa Abedinpour
- Vaccine Research Institute of San Diego, 3030 Bunker Hill Street, Suite 200, San Diego, CA, 92109, USA
| | - Adrian Chrastina
- Vaccine Research Institute of San Diego, 3030 Bunker Hill Street, Suite 200, San Diego, CA, 92109, USA
| | - Jennifer Pelayo
- Vaccine Research Institute of San Diego, 3030 Bunker Hill Street, Suite 200, San Diego, CA, 92109, USA
| | - Per Borgstrom
- Vaccine Research Institute of San Diego, 3030 Bunker Hill Street, Suite 200, San Diego, CA, 92109, USA
- Pellficure Pharmaceuticals, Inc., 2325 Camino del Collado, La Jolla, CA, 92037, USA
| | - John Welsh
- Vaccine Research Institute of San Diego, 3030 Bunker Hill Street, Suite 200, San Diego, CA, 92109, USA.
| |
Collapse
|
42
|
Chowdhury A, Choudhury A, Chakraborty S, Ghosh A, Banerjee V, Ganguly S, Bhaduri G, Banerjee R, Das K, Chatterjee IB. p-Benzoquinone-induced aggregation and perturbation of structure and chaperone function of α-crystallin is a causative factor of cigarette smoke-related cataractogenesis. Toxicology 2017; 394:11-18. [PMID: 29196190 DOI: 10.1016/j.tox.2017.11.018] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Revised: 11/15/2017] [Accepted: 11/27/2017] [Indexed: 02/08/2023]
Abstract
Cigarette smoking is a significant risk factor for cataract. However, the mechanism by which cigarette smoke (CS) causes cataract remains poorly understood. We had earlier shown that in CS-exposed guinea pig, p-benzoquinone (p-BQ) derived from CS in the lungs is carried by the circulatory system to distant organs and induces various smoke-related pathogeneses. Here, we observed that CS exposure caused accumulation of the p-BQ-protein adduct in the eye lens of guinea pigs. We also observed accumulation of the p-BQ-protein adduct in resected lens from human smokers with cataract. No such accumulation was observed in the lens of never smokers. p-BQ is a strong arylating agent that forms Michael adducts with serum albumin and haemoglobin resulting in alterations of structure and function. A major protein in the mammalian eye lens is αA-crystallin, which is a potent molecular chaperone. αA-crystallin plays a key role in maintaining the integrity and transparency of the lens. SDS-PAGE indicated that p-BQ induced aggregation of αA-crystallin. Various biophysical techniques including UV-vis spectroscopy, fluorescence spectroscopy, FT-IR, bis-ANS titration suggested a perturbation of structure and chaperone function of αA-crystallin upon p-BQ modification. Our results indicate that p-BQ is a causative agent involved in the modification of αA-crystallin and pathogenesis of CS-induced cataract. Our findings would educate public about the impacts of smoking on eye health and help to discourage them from smoking. The study might also help scientists to develop new drugs for the intervention of CS-induced cataract at an early stage.
Collapse
Affiliation(s)
- Aritra Chowdhury
- Department of Biotechnology and Dr. B. C. Guha Centre for Genetic Engineering & Biotechnology, Calcutta University College of Science, Kolkata 700019, India
| | - Aparajita Choudhury
- Department of Biotechnology and Dr. B. C. Guha Centre for Genetic Engineering & Biotechnology, Calcutta University College of Science, Kolkata 700019, India
| | - Shruti Chakraborty
- Department of Biotechnology and Dr. B. C. Guha Centre for Genetic Engineering & Biotechnology, Calcutta University College of Science, Kolkata 700019, India
| | - Arunava Ghosh
- Department of Biotechnology and Dr. B. C. Guha Centre for Genetic Engineering & Biotechnology, Calcutta University College of Science, Kolkata 700019, India
| | - Victor Banerjee
- Department of Chemistry, Bose Institute, 93/1 A.P.C. Road, Kolkata, 700 009, India
| | - Shinjini Ganguly
- Department of Biotechnology and Dr. B. C. Guha Centre for Genetic Engineering & Biotechnology, Calcutta University College of Science, Kolkata 700019, India
| | - Gautam Bhaduri
- Regional Institute of Opthalmology, Medical College, Kolkata, India
| | - Rajat Banerjee
- Department of Biotechnology and Dr. B. C. Guha Centre for Genetic Engineering & Biotechnology, Calcutta University College of Science, Kolkata 700019, India.
| | - Kalipada Das
- Department of Chemistry, Bose Institute, 93/1 A.P.C. Road, Kolkata, 700 009, India.
| | - Indu B Chatterjee
- Department of Biotechnology and Dr. B. C. Guha Centre for Genetic Engineering & Biotechnology, Calcutta University College of Science, Kolkata 700019, India.
| |
Collapse
|
43
|
Udagawa O, Furuyama A, Imai K, Fujitani Y, Hirano S. Effects of diesel exhaust-derived secondary organic aerosol (SOA) on oocytes: Potential risks to meiotic maturation. Reprod Toxicol 2017; 75:56-64. [PMID: 29158200 DOI: 10.1016/j.reprotox.2017.11.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Revised: 09/09/2017] [Accepted: 11/16/2017] [Indexed: 11/16/2022]
Abstract
Particulate air pollution (PM 2.5) is a worldwide concern. Growing epidemiological evidence has shown pathophysiological effects of PM 2.5, not only on cardiovascular system but also on reproductive performance. The composition and physicochemical properties of PM 2.5 vary depending on the emission sources, climate conditions, and complex chemical reactions in the air. These factors make it difficult to understand the cause and mechanistic details of the adverse health effects of PM 2.5. Here, we show potential impacts of PM 2.5 on oocyte maturation in mice by utilizing diesel exhaust-derived secondary organic aerosol (SOA), a major component of urban PM 2.5. We found that the SOA destabilized microtubules of mouse oocytes and p-benzoquinone is one of the candidates for the microtubule-destabilizing compounds. We propose that some biologically reactive components of PM 2.5 should be prioritized for the regulation of atmospheric quality.
Collapse
Affiliation(s)
- Osamu Udagawa
- Center for Health & Environmental Risk Research, National Institute for Environmental Studies, Tsukuba, Ibaraki, Japan.
| | - Akiko Furuyama
- Center for Health & Environmental Risk Research, National Institute for Environmental Studies, Tsukuba, Ibaraki, Japan
| | - Koji Imai
- Center for Health & Environmental Risk Research, National Institute for Environmental Studies, Tsukuba, Ibaraki, Japan
| | - Yuji Fujitani
- Center for Health & Environmental Risk Research, National Institute for Environmental Studies, Tsukuba, Ibaraki, Japan
| | - Seishiro Hirano
- Center for Health & Environmental Risk Research, National Institute for Environmental Studies, Tsukuba, Ibaraki, Japan
| |
Collapse
|
44
|
Comitato R, Ambra R, Virgili F. Tocotrienols: A Family of Molecules with Specific Biological Activities. Antioxidants (Basel) 2017; 6:antiox6040093. [PMID: 29156559 PMCID: PMC5745503 DOI: 10.3390/antiox6040093] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Revised: 11/08/2017] [Accepted: 11/16/2017] [Indexed: 01/15/2023] Open
Abstract
Vitamin E is a generic term frequently used to group together eight different molecules, namely: α-, β-, γ- and δ-tocopherol and the corresponding tocotrienols. The term tocopherol and eventually Vitamin E and its related activity was originally based on the capacity of countering foetal re-absorption in deficient rodents or the development of encephalomalacia in chickens. In humans, Vitamin E activity is generally considered to be solely related to the antioxidant properties of the tocolic chemical structure. In recent years, several reports have shown that specific activities exist for each different tocotrienol form. In this short review, tocotrienol ability to inhibit cancer cell growth and induce apoptosis thanks to specific mechanisms, not shared by tocopherols, such as the binding to Estrogen Receptor-β (ERβ) and the triggering of endoplasmic reticulum (EndoR) stress will be described. The neuroprotective activity will also be presented and discussed. We propose that available studies strongly indicate that specific forms of tocotrienols have a distinct mechanism and biological activity, significantly different from tocopherol and more specifically from α-tocopherol. We therefore suggest not pooling them together within the broad term “Vitamin E” on solely the basis of their putative antioxidant properties. This option implies obvious consequences in the assessment of dietary Vitamin E adequacy and, probably more importantly, on the possibility of evaluating a separate biological variable, determinant in the relationship between diet and health.
Collapse
Affiliation(s)
- Raffaella Comitato
- Council for Agricultural Research and Economics, Research Centre for Food and Nutrition (CREA-AN) via Ardeatina 546, 00178 Rome, Italy.
| | - Roberto Ambra
- Council for Agricultural Research and Economics, Research Centre for Food and Nutrition (CREA-AN) via Ardeatina 546, 00178 Rome, Italy.
| | - Fabio Virgili
- Council for Agricultural Research and Economics, Research Centre for Food and Nutrition (CREA-AN) via Ardeatina 546, 00178 Rome, Italy.
| |
Collapse
|
45
|
Liu Z, Wang Y, Wang Y, Dong W, Xia X, Song E, Song Y. Effect of Subcellular Translocation of Protein Disulfide Isomerase on Tetrachlorobenzoquinone-Induced Signaling Shift from Endoplasmic Reticulum Stress to Apoptosis. Chem Res Toxicol 2017; 30:1804-1814. [DOI: 10.1021/acs.chemrestox.7b00118] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Zixuan Liu
- Key Laboratory of Luminescence
and Real-Time Analytical Chemistry (Southwest University), Ministry
of Education, College of Pharmaceutical Sciences, Southwest University, Chongqing, People’s Republic of China 400715
| | - Yawen Wang
- Key Laboratory of Luminescence
and Real-Time Analytical Chemistry (Southwest University), Ministry
of Education, College of Pharmaceutical Sciences, Southwest University, Chongqing, People’s Republic of China 400715
| | - Yuxin Wang
- Key Laboratory of Luminescence
and Real-Time Analytical Chemistry (Southwest University), Ministry
of Education, College of Pharmaceutical Sciences, Southwest University, Chongqing, People’s Republic of China 400715
| | - Wenjing Dong
- Key Laboratory of Luminescence
and Real-Time Analytical Chemistry (Southwest University), Ministry
of Education, College of Pharmaceutical Sciences, Southwest University, Chongqing, People’s Republic of China 400715
| | - Xiaomin Xia
- Key Laboratory of Luminescence
and Real-Time Analytical Chemistry (Southwest University), Ministry
of Education, College of Pharmaceutical Sciences, Southwest University, Chongqing, People’s Republic of China 400715
| | - Erqun Song
- Key Laboratory of Luminescence
and Real-Time Analytical Chemistry (Southwest University), Ministry
of Education, College of Pharmaceutical Sciences, Southwest University, Chongqing, People’s Republic of China 400715
| | - Yang Song
- Key Laboratory of Luminescence
and Real-Time Analytical Chemistry (Southwest University), Ministry
of Education, College of Pharmaceutical Sciences, Southwest University, Chongqing, People’s Republic of China 400715
| |
Collapse
|
46
|
Aeschimann W, Staats S, Kammer S, Olieric N, Jeckelmann JM, Fotiadis D, Netscher T, Rimbach G, Cascella M, Stocker A. Self-assembled α-Tocopherol Transfer Protein Nanoparticles Promote Vitamin E Delivery Across an Endothelial Barrier. Sci Rep 2017; 7:4970. [PMID: 28694484 PMCID: PMC5504013 DOI: 10.1038/s41598-017-05148-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Accepted: 05/24/2017] [Indexed: 01/16/2023] Open
Abstract
Vitamin E is one of the most important natural antioxidants, protecting polyunsaturated fatty acids in the membranes of cells. Among different chemical isoforms assimilated from dietary regimes, RRR-α-tocopherol is the only one retained in higher animals. This is possible thanks to α-Tocopherol Transfer Protein (α-TTP), which extracts α-tocopherol from endosomal compartments in liver cells, facilitating its distribution into the body. Here we show that, upon binding to its substrate, α-TTP acquires tendency to aggregation into thermodynamically stable high molecular weight oligomers. Determination of the structure of such aggregates by X-ray crystallography revealed a spheroidal particle formed by 24 protein monomers. Oligomerization is triggered by refolding of the N-terminus. Experiments with cultured cell monolayers demonstrate that the same oligomers are efficiently transported through an endothelial barrier (HUVEC) and not through an epithelial one (Caco-2). Discovery of a human endogenous transport protein with intrinsic capability of crossing endothelial tissues opens to new ways of drug delivery into the brain or other tissues protected by endothelial barriers.
Collapse
Affiliation(s)
- Walter Aeschimann
- University of Bern, Department of Chemistry and Biochemistry, Bern, Switzerland
| | - Stefanie Staats
- University of Kiel, Institute of Human Nutrition and Food Science, Kiel, Germany
| | - Stephan Kammer
- University of Bern, Department of Chemistry and Biochemistry, Bern, Switzerland
| | | | - Jean-Marc Jeckelmann
- University of Bern, Institute of Biochemistry and Molecular Medicine, Bern, Switzerland
| | - Dimitrios Fotiadis
- University of Bern, Institute of Biochemistry and Molecular Medicine, Bern, Switzerland
| | | | - Gerald Rimbach
- University of Kiel, Institute of Human Nutrition and Food Science, Kiel, Germany
| | - Michele Cascella
- University of Oslo, Department of Chemistry and Centre for Theoretical and Computational Chemistry (CTCC), Oslo, Norway.
| | - Achim Stocker
- University of Bern, Department of Chemistry and Biochemistry, Bern, Switzerland.
| |
Collapse
|
47
|
Scherz LF, Abdel-Rahman EA, Ali SS, Schlüter AD, Abdel-Rahman MA. Design, synthesis and cytotoxic activity of water-soluble quinones with dibromo- p-benzoquinone cores and amino oligo(ethylene glycol) side chains against MCF-7 breast cancer cells. MEDCHEMCOMM 2017; 8:662-672. [PMID: 30108784 DOI: 10.1039/c6md00728g] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/31/2016] [Accepted: 02/11/2017] [Indexed: 11/21/2022]
Abstract
A series of novel quinones was synthesized by reacting tetrabromo-p-benzoquinone with amino oligo(ethylene glycol) dendrons of generation numbers g = 0-2. According to the performed shake-flask experiments, their aqueous solubility (S = 18 mg l-1-1.6 g ml-1) and partition coefficients (log Poct/wat = 2.53-0.21) can be tuned in a wide range as a function of g. In vitro cytotoxicity assays of tetrabromo-p-benzoquinone and its derivatives against MCF-7 human breast cancer cells showed a concentration- and generation-specific biological activity with IC50-values as low as 0.8 μM. Further investigations revealed a considerable selectivity against cancer cells, as indicated by a weak cytotoxicity against human skin fibroblast cells (>80% survival) within the studied range of concentrations. The results demonstrate that these novel amino oligo(ethylene glycol) dendrons depict versatile tools to ameliorate physical and pharmacological characteristics of extremely hydrophobic molecules and make them susceptible to biological applications.
Collapse
Affiliation(s)
- Leon F Scherz
- Institute of Polymers , Department of Materials , ETH Zürich , 8093 Zürich , Switzerland
| | - Engy A Abdel-Rahman
- Pharmacology Department , Faculty of Medicine , Assiut University , 71515 Assiut , Egypt.,Center for Aging and Associated Diseases , Helmy Institute of Medical Science , Zewail City of Science and Technology , 12588 Giza , Egypt
| | - Sameh S Ali
- Center for Aging and Associated Diseases , Helmy Institute of Medical Science , Zewail City of Science and Technology , 12588 Giza , Egypt
| | - A Dieter Schlüter
- Institute of Polymers , Department of Materials , ETH Zürich , 8093 Zürich , Switzerland
| | - Mona A Abdel-Rahman
- Chemistry Department , Polymer Lab. 109 , Faculty of Science , Assiut University , 71516 Assiut , Egypt .
| |
Collapse
|
48
|
Bolton JL, Dunlap T. Formation and Biological Targets of Quinones: Cytotoxic versus Cytoprotective Effects. Chem Res Toxicol 2016; 30:13-37. [PMID: 27617882 PMCID: PMC5241708 DOI: 10.1021/acs.chemrestox.6b00256] [Citation(s) in RCA: 258] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Quinones represent a class of toxicological intermediates, which can create a variety of hazardous effects in vivo including, acute cytotoxicity, immunotoxicity, and carcinogenesis. In contrast, quinones can induce cytoprotection through the induction of detoxification enzymes, anti-inflammatory activities, and modification of redox status. The mechanisms by which quinones cause these effects can be quite complex. The various biological targets of quinones depend on their rate and site of formation and their reactivity. Quinones are formed through a variety of mechanisms from simple oxidation of catechols/hydroquinones catalyzed by a variety of oxidative enzymes and metal ions to more complex mechanisms involving initial P450-catalyzed hydroxylation reactions followed by two-electron oxidation. Quinones are Michael acceptors, and modification of cellular processes could occur through alkylation of crucial cellular proteins and/or DNA. Alternatively, quinones are highly redox active molecules which can redox cycle with their semiquinone radical anions leading to the formation of reactive oxygen species (ROS) including superoxide, hydrogen peroxide, and ultimately the hydroxyl radical. Production of ROS can alter redox balance within cells through the formation of oxidized cellular macromolecules including lipids, proteins, and DNA. This perspective explores the varied biological targets of quinones including GSH, NADPH, protein sulfhydryls [heat shock proteins, P450s, cyclooxygenase-2 (COX-2), glutathione S-transferase (GST), NAD(P)H:quinone oxidoreductase 1, (NQO1), kelch-like ECH-associated protein 1 (Keap1), IκB kinase (IKK), and arylhydrocarbon receptor (AhR)], and DNA. The evidence strongly suggests that the numerous mechanisms of quinone modulations (i.e., alkylation versus oxidative stress) can be correlated with the known pathology/cytoprotection of the parent compound(s) that is best described by an inverse U-shaped dose-response curve.
Collapse
Affiliation(s)
- Judy L Bolton
- Department of Medicinal Chemistry and Pharmacognosy (M/C 781), College of Pharmacy, University of Illinois at Chicago , 833 S. Wood Street, Chicago, Illinois 60612-7231, United States
| | - Tareisha Dunlap
- Department of Medicinal Chemistry and Pharmacognosy (M/C 781), College of Pharmacy, University of Illinois at Chicago , 833 S. Wood Street, Chicago, Illinois 60612-7231, United States
| |
Collapse
|
49
|
Wu FL, Liu WY, Van Poucke S, Braddock M, Jin WM, Xiao J, Li XK, Zheng MH. Targeting endoplasmic reticulum stress in liver disease. Expert Rev Gastroenterol Hepatol 2016; 10:1041-52. [PMID: 27093595 DOI: 10.1080/17474124.2016.1179575] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
INTRODUCTION The accumulation of unfolded protein in the endoplasmic reticulum (ER) initiates an unfolded protein response (UPR) via three signal transduction cascades, which involve protein kinase RNA-like ER kinase (PERK), inositol requiring enzyme-1α (IRE1α) and activating transcription factor-6α (ATF6α). An ER stress response is observed in nearly all physiologies related to acute and chronic liver disease and therapeutic targeting of the mechanisms implicated in UPR signaling have attracted considerable attention. AREAS COVERED This review focuses on the correlation between ER stress and liver disease and the possible targets which may drive the potential for novel therapeutic intervention. Expert Commentary: We describe pathways which are involved in UPR signaling and their potential correlation with various liver diseases and underlying mechanisms which may present opportunities for novel therapeutic strategies are discussed.
Collapse
Affiliation(s)
- Fa-Ling Wu
- a Department of Hepatology, Liver Research Center , the First Affiliated Hospital of Wenzhou Medical University , Wenzhou , China.,b Institute of Hepatology , Wenzhou Medical University , Wenzhou , China
| | - Wen-Yue Liu
- c Department of Endocrinology , the First Affiliated Hospital of Wenzhou Medical University , Wenzhou , China
| | - Sven Van Poucke
- d Department of Anesthesiology, Intensive Care, Emergency Medicine and Pain Therapy , Ziekenhuis Oost-Limburg , Genk , Belgium
| | - Martin Braddock
- e Global Medicines Development , AstraZeneca R&D , Alderley Park , UK
| | - Wei-Min Jin
- f Department of Infection Diseases , People Hospital of Wencheng County , Wenzhou , China
| | - Jian Xiao
- g Institute of Biology Science , Wenzhou University , Wenzhou , China.,h School of Pharmacy , Wenzhou Medical University , Wenzhou , China
| | - Xiao-Kun Li
- g Institute of Biology Science , Wenzhou University , Wenzhou , China.,h School of Pharmacy , Wenzhou Medical University , Wenzhou , China
| | - Ming-Hua Zheng
- a Department of Hepatology, Liver Research Center , the First Affiliated Hospital of Wenzhou Medical University , Wenzhou , China.,b Institute of Hepatology , Wenzhou Medical University , Wenzhou , China
| |
Collapse
|
50
|
Tang CB, Zhang WG, Wang YS, Xing LJ, Xu XL, Zhou GH. Identification of Rosmarinic Acid-Adducted Sites in Meat Proteins in a Gel Model under Oxidative Stress by Triple TOF MS/MS. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2016; 64:6466-76. [PMID: 27486909 DOI: 10.1021/acs.jafc.6b02438] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Triple TOF MS/MS was used to identify adducts between rosmarinic acid (RosA)-derived quinones and meat proteins in a gel model under oxidative stress. Seventy-five RosA-modified peptides responded to 67 proteins with adduction of RosA. RosA conjugated with different amino acids in proteins, and His, Arg, and Lys adducts with RosA were identified for the first time in meat. A total of 8 peptides containing Cys, 14 peptides containing His, 48 peptides containing Arg, 64 peptides containing Lys, and 5 peptides containing N-termini that which participated in adduction reaction with RosA were identified, respectively. Seventy-seven adduction sites were subdivided into all adducted proteins including 2 N-terminal adduction sites, 3 Cys adduction sites, 4 His adduction sites, 29 Arg adduction sites, and 39 Lys adduction sites. Site occupancy analyses showed that approximately 80.597% of the proteins carried a single RosA-modified site, 14.925% retained two sites, 1.492% contained three sites, and the rest 2.985% had four or more sites. Large-scale triple TOF MS/MS mapping of RosA-adducted sites reveals the adduction regulations of quinone and different amino acids as well as the adduction ratios, which clarify phenol-protein adductions and pave the way for industrial meat processing and preservation.
Collapse
Affiliation(s)
- Chang-Bo Tang
- Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, Key Laboratory of Meat Processing and Quality Control, Ministry of Education, Key Laboratory of Animal Products Processing, Ministry of Agriculture, College of Food Science and Technology, Nanjing Agricultural University , Nanjing 210095, China
- Department of Food Nutrition and Detection, College of Education and Humanity, Suzhou Vocational University , Suzhou 215104, China
| | - Wan-Gang Zhang
- Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, Key Laboratory of Meat Processing and Quality Control, Ministry of Education, Key Laboratory of Animal Products Processing, Ministry of Agriculture, College of Food Science and Technology, Nanjing Agricultural University , Nanjing 210095, China
| | - Yao-Song Wang
- College of Light Industry Science and Engineering, Nanjing Forestry University , Nanjing 210037, China
| | - Lu-Juan Xing
- Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, Key Laboratory of Meat Processing and Quality Control, Ministry of Education, Key Laboratory of Animal Products Processing, Ministry of Agriculture, College of Food Science and Technology, Nanjing Agricultural University , Nanjing 210095, China
| | - Xing-Lian Xu
- Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, Key Laboratory of Meat Processing and Quality Control, Ministry of Education, Key Laboratory of Animal Products Processing, Ministry of Agriculture, College of Food Science and Technology, Nanjing Agricultural University , Nanjing 210095, China
| | - Guang-Hong Zhou
- Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, Key Laboratory of Meat Processing and Quality Control, Ministry of Education, Key Laboratory of Animal Products Processing, Ministry of Agriculture, College of Food Science and Technology, Nanjing Agricultural University , Nanjing 210095, China
| |
Collapse
|