1
|
Haiman ZB, Key A, D'Alessandro A, Palsson BO. RBC-GEM: A genome-scale metabolic model for systems biology of the human red blood cell. PLoS Comput Biol 2025; 21:e1012109. [PMID: 40072998 PMCID: PMC11925312 DOI: 10.1371/journal.pcbi.1012109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 03/20/2025] [Accepted: 02/04/2025] [Indexed: 03/14/2025] Open
Abstract
Advancements with cost-effective, high-throughput omics technologies have had a transformative effect on both fundamental and translational research in the medical sciences. These advancements have facilitated a departure from the traditional view of human red blood cells (RBCs) as mere carriers of hemoglobin, devoid of significant biological complexity. Over the past decade, proteomic analyses have identified a growing number of different proteins present within RBCs, enabling systems biology analysis of their physiological functions. Here, we introduce RBC-GEM, one of the most comprehensive, curated genome-scale metabolic reconstructions of a specific human cell type to-date. It was developed through meta-analysis of proteomic data from 29 studies published over the past two decades resulting in an RBC proteome composed of more than 4,600 distinct proteins. Through workflow-guided manual curation, we have compiled the metabolic reactions carried out by this proteome to form a genome-scale metabolic model (GEM) of the RBC. RBC-GEM is hosted on a version-controlled GitHub repository, ensuring adherence to the standardized protocols for metabolic reconstruction quality control and data stewardship principles. RBC-GEM represents a metabolic network is a consisting of 820 genes encoding proteins acting on 1,685 unique metabolites through 2,723 biochemical reactions: a 740% size expansion over its predecessor. We demonstrated the utility of RBC-GEM by creating context-specific proteome-constrained models derived from proteomic data of stored RBCs for 616 blood donors, and classified reactions based on their simulated abundance dependence. This reconstruction as an up-to-date curated GEM can be used for contextualization of data and for the construction of a computational whole-cell models of the human RBC.
Collapse
Affiliation(s)
- Zachary B Haiman
- Department of Bioengineering, University of California San Diego, La Jolla, California, United States of America
| | - Alicia Key
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Angelo D'Alessandro
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Bernhard O Palsson
- Department of Bioengineering, University of California San Diego, La Jolla, California, United States of America
- Department of Pediatrics, University of California San Diego, La Jolla, California, United States of America
- Bioinformatics and Systems Biology Program, University of California, La Jolla, San Diego, California, United States of America
| |
Collapse
|
2
|
Osterman C, Hamlin D, Suter CM, Affleck AJ, Gloss BS, Turner CP, Faull RLM, Stein TD, McKee A, Buckland ME, Curtis MA, Murray HC. Perivascular glial reactivity is a feature of phosphorylated tau lesions in chronic traumatic encephalopathy. Acta Neuropathol 2025; 149:16. [PMID: 39921702 PMCID: PMC11807024 DOI: 10.1007/s00401-025-02854-x] [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: 09/30/2024] [Revised: 01/26/2025] [Accepted: 01/27/2025] [Indexed: 02/10/2025]
Abstract
Chronic traumatic encephalopathy (CTE), a neurodegenerative disease associated with repetitive head injuries, is characterised by perivascular hyperphosphorylated tau (p-tau) accumulations within the depths of cortical sulci. Although the majority of CTE literature focuses on p-tau pathology, other pathological features such as glial reactivity, vascular damage, and axonal damage are relatively unexplored. In this study, we aimed to characterise these other pathological features, specifically in CTE p-tau lesion areas, to better understand the microenvironment surrounding the lesion. We utilised multiplex immunohistochemistry to investigate the distribution of 32 different markers of cytoarchitecture and pathology that are relevant to both traumatic brain injury and neurodegeneration. We qualitatively assessed the multiplex images and measured the percentage area of labelling for each marker in the lesion and non-lesion areas of CTE cases. We identified perivascular glial reactivity as a prominent feature of CTE p-tau lesions, largely driven by increases in astrocyte reactivity compared to non-lesion areas. Furthermore, we identified astrocytes labelled for both NAD(P)H quinone dehydrogenase 1 (NQO1) and L-ferritin, indicating that lesion-associated glial reactivity may be a compensatory response to iron-induced oxidative stress. Our findings demonstrate that perivascular inflammation is a consistent feature of the CTE pathognomonic lesion and may contribute to the pathogenesis of brain injury-related neurodegeneration.
Collapse
Affiliation(s)
- Chelsie Osterman
- Department of Anatomy and Medical Imaging and Centre for Brain Research, Faculty of Medical and Health Science, University of Auckland, 85 Park Road, Grafton, 1023, Auckland, New Zealand
| | - Danica Hamlin
- Department of Anatomy and Medical Imaging and Centre for Brain Research, Faculty of Medical and Health Science, University of Auckland, 85 Park Road, Grafton, 1023, Auckland, New Zealand
| | - Catherine M Suter
- Department of Neuropathology, Royal Prince Alfred Hospital, 94 Mallet St, Camperdown, NSW, 2050, Australia
- School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Camperdown, NSW, 2006, Australia
| | - Andrew J Affleck
- Department of Neuropathology, Royal Prince Alfred Hospital, 94 Mallet St, Camperdown, NSW, 2050, Australia
- School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Camperdown, NSW, 2006, Australia
| | - Brian S Gloss
- Westmead Research Hub, Westmead Institute for Medical Research, Westmead, NSW, Australia
| | - Clinton P Turner
- Department of Anatomy and Medical Imaging and Centre for Brain Research, Faculty of Medical and Health Science, University of Auckland, 85 Park Road, Grafton, 1023, Auckland, New Zealand
- Department of Anatomical Pathology, Pathology and Laboratory Medicine, Auckland City Hospital, 2 Park Road, Grafton, 1023, Auckland, New Zealand
| | - Richard L M Faull
- Department of Anatomy and Medical Imaging and Centre for Brain Research, Faculty of Medical and Health Science, University of Auckland, 85 Park Road, Grafton, 1023, Auckland, New Zealand
| | - Thor D Stein
- Department of Pathology and Laboratory Medicine, VA Boston Healthcare System, Boston, MA, USA
- Department of Pathology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Alzheimer's Disease and CTE Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Bedford Veterans Affairs Medical Center, Bedford, MA, USA
| | - Ann McKee
- Department of Pathology and Laboratory Medicine, VA Boston Healthcare System, Boston, MA, USA
- Department of Pathology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Alzheimer's Disease and CTE Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Bedford Veterans Affairs Medical Center, Bedford, MA, USA
| | - Michael E Buckland
- Department of Neuropathology, Royal Prince Alfred Hospital, 94 Mallet St, Camperdown, NSW, 2050, Australia
- School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Camperdown, NSW, 2006, Australia
| | - Maurice A Curtis
- Department of Anatomy and Medical Imaging and Centre for Brain Research, Faculty of Medical and Health Science, University of Auckland, 85 Park Road, Grafton, 1023, Auckland, New Zealand
| | - Helen C Murray
- Department of Anatomy and Medical Imaging and Centre for Brain Research, Faculty of Medical and Health Science, University of Auckland, 85 Park Road, Grafton, 1023, Auckland, New Zealand.
| |
Collapse
|
3
|
Lim JJ, Klaassen CD, Cui JY. Deciphering the cell type-specific and zonal distribution of drug-metabolizing enzymes, transporters, and transcription factors in livers of mice using single-cell transcriptomics. Drug Metab Dispos 2025; 53:100029. [PMID: 39919554 DOI: 10.1016/j.dmd.2024.100029] [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: 08/12/2024] [Accepted: 11/08/2024] [Indexed: 02/09/2025] Open
Abstract
The liver contains multiple cell types, including resident cell types and immune cells. The liver is also categorized into 3 zones: periportal (zone 1), midzonal (zone 2), and centrilobular (zone 3). The goal of this study was to characterize the distribution of drug-processing genes (DPGs) in mouse liver using published single-cell and nuclei transcriptomic datasets, which were subjected to zonal deconvolution. Filtering, normalization, clustering, and differential expression analyses were performed using Seurat V5 in R. Hepatocytes were assigned to 3 zones based on known zonal markers and validated with published spatial transcriptomics data. Among the 195 DPGs profiled, most were expressed highest in hepatocytes (61.3%). Interestingly, certain DPGs were expressed most highly in nonparenchymal cells, such as in cholangiocytes (11.2%, eg, carboxylesterase [Ces] 2e, Ces2g), endothelial cells (7.2%, eg, aldo-keto reductase [Akr] 1c19, Akr1e1), Kupffer cells (5.3%, eg, Akr1a1, Akr1b10), stellate cells (5.1%, eg, retinoic acid receptor [Rar] α, Rarβ), myofibroblasts (2.9%, RAR-related orphan receptor [Rar] α), and a few were expressed in immune cell types. In hepatocytes, 72.4% of phase-I enzymes were enriched in zone 3. Phase-II conjugation enzymes such as UDP-glucuronosyltransferases (75%) were enriched in zone 3, whereas sulfotransferases (40%) were enriched in zone 1. Hepatic xenobiotic transporters were enriched in zone 3. The xenobiotic biotransformation-regulating transcription factors were enriched in zone 3 hepatocytes. The enrichment of DPGs in liver cell types, including non-parenchymal cells and zone 1 hepatocytes, may serve as an additional repertoire for xenobiotic biotransformation. SIGNIFICANCE STATEMENT: Our study is among the first to systematically characterize the baseline mRNA enrichment of important drug-processing genes in different cell types and zones in the liver. This finding will aid in further understanding the mechanisms of chemical-induced liver injury with improved resolution and precision.
Collapse
Affiliation(s)
- Joe Jongpyo Lim
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, Washington; Environmental Health and Microbiome Research Center (EHMBRACE), Seattle, Washington
| | - Curtis Dean Klaassen
- Department of Pharmacology, Toxicology, and Therapeutics, School of Medicine, University of Kansas, Kansas City, Kanas.
| | - Julia Yue Cui
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, Washington; Environmental Health and Microbiome Research Center (EHMBRACE), Seattle, Washington.
| |
Collapse
|
4
|
Verbovaya ER, Kadnikov IA, Logvinov IO, Antipova TA, Voronin MV, Seredenin SB. In vitro modelling of Parkinson's disease using 6-OHDA is associated with increased NQO2 activity. Toxicol In Vitro 2024; 101:105940. [PMID: 39271030 DOI: 10.1016/j.tiv.2024.105940] [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: 05/14/2024] [Revised: 08/22/2024] [Accepted: 09/09/2024] [Indexed: 09/15/2024]
Abstract
The pathogenesis of Parkinson's disease (PD) involves abnormalities in the metabolism of catecholamines. The enzyme quinone reductase 2 (NQO2) reduces quinone derivatives of catecholamines, which promotes the formation of reactive oxygen species (ROS), suggesting a role for NQO2 in the development of cellular damage typical of PD. In the present study, we investigated the relationship between 6-hydroxydophamine (6-OHDA) induced cellular damage and NQO2 activity and its levels in SH-SY5Y cell culture to establish an experimental model to evaluate the pharmacological properties of NQO2 inhibitors. Cellular damage was evaluated using the MTT and comet assays. It was shown that oxidative damage of SH-SY5Y cells upon incubation with 6-OHDA for 6, 12 and 24 h was accompanied by an increase in NQO2 activity. The increase in NQO2 protein level in SH-SY5Y cells was observed 24 h after incubation with 6-OHDA at concentrations of 50 and 100 μM. Oxidative damage of SH-SY5Y cells upon 1 h incubation with 6-OHDA is increased in the presence of the selective enzyme co-substrate 1-benzyl-1,4-dihydronicotinamide (BNAH), but is not accompanied by changes in NQO2 activity and protein levels. The data obtained demonstrate the contribution of NQO2 to the cytotoxic mechanism of 6-OHDA action.
Collapse
Affiliation(s)
- Ekaterina R Verbovaya
- Laboratory of Pharmacogenetics, Federal Research Center for Innovator and Emerging Biomedical and Pharmaceutical Technologies, Baltiyskaya Street 8, 125315 Moscow, Russia.
| | - Ilya A Kadnikov
- Laboratory of Pharmacogenetics, Federal Research Center for Innovator and Emerging Biomedical and Pharmaceutical Technologies, Baltiyskaya Street 8, 125315 Moscow, Russia
| | - Ilya O Logvinov
- Laboratory of Molecular Pharmacology, Federal Research Center for Innovator and Emerging Biomedical and Pharmaceutical Technologies, Baltiyskaya Street 8, 125315 Moscow, Russia
| | - Tatyana A Antipova
- Laboratory of Molecular Pharmacology, Federal Research Center for Innovator and Emerging Biomedical and Pharmaceutical Technologies, Baltiyskaya Street 8, 125315 Moscow, Russia
| | - Mikhail V Voronin
- Laboratory of Pharmacogenetics, Federal Research Center for Innovator and Emerging Biomedical and Pharmaceutical Technologies, Baltiyskaya Street 8, 125315 Moscow, Russia
| | - Sergei B Seredenin
- Laboratory of Pharmacogenetics, Federal Research Center for Innovator and Emerging Biomedical and Pharmaceutical Technologies, Baltiyskaya Street 8, 125315 Moscow, Russia
| |
Collapse
|
5
|
Li S, Li X, Lu M, Chen Q, Yao D, Yu X, Li Z, Ge W, Wang N, Jin J, Wang Y, Liao Y, Luo F, Yan J, Chen X, Jiang C, Yue F, Gao D, Tang X, Guo H, Wang Y, Chen X, Xia J, Xu M, Ren S, He C, Hu Z. Homeostatic Shrinkage of Dendritic Spines Requires Melatonin Type 3 Receptor Activation During Sleep. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2400253. [PMID: 39119847 PMCID: PMC11481193 DOI: 10.1002/advs.202400253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 07/25/2024] [Indexed: 08/10/2024]
Abstract
High-frequency oscillatory activity in cognition-related neural circuits during wakefulness consistently induces the growth of dendritic spines and axonal terminals. Although these structural changes are essential for cognitive functions, it is hypothesized that if these newly expanded structures fail to establish functional connections, they may become superfluous. Sleep is believed to facilitate the reduction of such redundant structures to maintain neural homeostasis. However, the mechanisms underlying this pruning process during sleep remain poorly understood. In this study, that melatonin type 3 receptors (MT3Rs) are selectively expressed in the stellate neurons of the medial entorhinal cortex (MEC) is demonstrated, an area where high melatonin levels are detected during sleep. Activation of MT3Rs during sleep initiates the shrinkage of dendritic spines in stellate neurons by downregulating neural network activity and dephosphorylating synaptic proteins in the MEC. This process is disrupted when MT3R expression is knocked down or when MT3Rs are blocked during sleep. Notably, interference with MT3Rs in the MEC during sleep impairs the acquisition of spatial memory but does not affect object memory acquisition following sleep. These findings reveal novel molecular mechanisms involving melatonin and MT3Rs in the regulation of dendritic spine shrinkage during sleep, which is crucial for the acquisition and consolidation of spatial memory.
Collapse
Affiliation(s)
- Shiyin Li
- Department of PhysiologyInstitute of Brain and IntelligenceThird Military Medical UniversityChongqing400038China
| | - Xin Li
- Department of PhysiologyInstitute of Brain and IntelligenceThird Military Medical UniversityChongqing400038China
- School of Basic Medical Sciences and IDG/McGovern Institute for Brain ResearchTsinghua UniversityBeijing100084China
| | - Minmin Lu
- Department of PhysiologyInstitute of Brain and IntelligenceThird Military Medical UniversityChongqing400038China
| | - Quanhui Chen
- Department of PhysiologyInstitute of Brain and IntelligenceThird Military Medical UniversityChongqing400038China
| | - Di Yao
- School of Basic Medical SciencesCapital Medical UniversityBeijing100069China
- Chinese Institute for Brain ResearchBeijing102206China
| | - Xiaoqian Yu
- Chinese Institute for Brain ResearchBeijing102206China
| | - Zhen Li
- Chinese Institute for Brain ResearchBeijing102206China
| | - Woo‐ping Ge
- Chinese Institute for Brain ResearchBeijing102206China
| | - Na Wang
- Department of PhysiologyInstitute of Brain and IntelligenceThird Military Medical UniversityChongqing400038China
| | - Jiehua Jin
- Department of PhysiologyInstitute of Brain and IntelligenceThird Military Medical UniversityChongqing400038China
| | - Yaling Wang
- Department of PhysiologyInstitute of Brain and IntelligenceThird Military Medical UniversityChongqing400038China
| | - Yixiang Liao
- Department of PhysiologyInstitute of Brain and IntelligenceThird Military Medical UniversityChongqing400038China
| | - Fenlan Luo
- Department of PhysiologyInstitute of Brain and IntelligenceThird Military Medical UniversityChongqing400038China
| | - Jie Yan
- Department of PhysiologyInstitute of Brain and IntelligenceThird Military Medical UniversityChongqing400038China
| | - Xuedan Chen
- Department of Medical GeneticsCollege of Basic Medical SciencesThird Military Medical UniversityChongqing400038China
| | - Chenggang Jiang
- Department of Sleep and PsychologyChongqing Health Center for Women and ChildrenChongqing401147China
| | - Faguo Yue
- Sleep and Psychology CenterBishan Hospital of Chongqing Medical UniversityChongqing402760China
| | - Dong Gao
- Department of Sleep and PsychologyThe Fifth People's Hospital of ChongqingChongqing400062China
| | - Xiangdong Tang
- Sleep Medicine CenterLaboratory of Anaesthesia and Critical Care MedicineTranslational Neuroscience CenterWest China HospitalSichuan UniversityChengdu610041China
| | - Hong Guo
- Department of Medical GeneticsCollege of Basic Medical SciencesThird Military Medical UniversityChongqing400038China
| | - Yanjiang Wang
- Department of NeurologyDaping HospitalThird Military Medical UniversityChongqing400042China
- Chongqing Institute for Brain and IntelligenceGuangyang Bay LaboratoryChongqing400064China
| | - Xiaowei Chen
- Chongqing Institute for Brain and IntelligenceGuangyang Bay LaboratoryChongqing400064China
- Brain Research CenterInstitute of Brain and IntelligenceThird Military Medical UniversityChongqing400038China
| | - Jianxia Xia
- Department of PhysiologyInstitute of Brain and IntelligenceThird Military Medical UniversityChongqing400038China
| | - Min Xu
- Institute of Neuroscience,Center for Excellence in Brain Science and Intelligence TechnologyChinese Academy of SciencesShanghai200031China
| | - Shuancheng Ren
- Department of PhysiologyInstitute of Brain and IntelligenceThird Military Medical UniversityChongqing400038China
| | - Chao He
- Department of PhysiologyInstitute of Brain and IntelligenceThird Military Medical UniversityChongqing400038China
| | - Zhian Hu
- Department of PhysiologyInstitute of Brain and IntelligenceThird Military Medical UniversityChongqing400038China
- Chongqing Institute for Brain and IntelligenceGuangyang Bay LaboratoryChongqing400064China
| |
Collapse
|
6
|
Duffel MW, Lehmler HJ. Complex roles for sulfation in the toxicities of polychlorinated biphenyls. Crit Rev Toxicol 2024; 54:92-122. [PMID: 38363552 PMCID: PMC11067068 DOI: 10.1080/10408444.2024.2311270] [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: 12/29/2023] [Revised: 01/22/2024] [Accepted: 01/23/2024] [Indexed: 02/17/2024]
Abstract
Polychlorinated biphenyls (PCBs) are persistent organic toxicants derived from legacy pollution sources and their formation as inadvertent byproducts of some current manufacturing processes. Metabolism of PCBs is often a critical component in their toxicity, and relevant metabolic pathways usually include their initial oxidation to form hydroxylated polychlorinated biphenyls (OH-PCBs). Subsequent sulfation of OH-PCBs was originally thought to be primarily a means of detoxication; however, there is strong evidence that it may also contribute to toxicities associated with PCBs and OH-PCBs. These contributions include either the direct interaction of PCB sulfates with receptors or their serving as a localized precursor for OH-PCBs. The formation of PCB sulfates is catalyzed by cytosolic sulfotransferases, and, when transported into the serum, these metabolites may be retained, taken up by other tissues, and subjected to hydrolysis catalyzed by intracellular sulfatase(s) to regenerate OH-PCBs. Dynamic cycling between PCB sulfates and OH-PCBs may lead to further metabolic activation of the resulting OH-PCBs. Ultimate toxic endpoints of such processes may include endocrine disruption, neurotoxicities, and many others that are associated with exposures to PCBs and OH-PCBs. This review highlights the current understanding of the complex roles that PCB sulfates can have in the toxicities of PCBs and OH-PCBs and research on the varied mechanisms that control these roles.
Collapse
Affiliation(s)
- Michael W. Duffel
- Department of Pharmaceutical Sciences & Experimental Therapeutics, College of Pharmacy, The University of Iowa, Iowa City, Iowa, 52242, United States
| | - Hans-Joachim Lehmler
- Department of Occupational and Environmental Health, College of Public Health, The University of Iowa, Iowa City, Iowa, 52242, United States
| |
Collapse
|
7
|
Janda E, Boutin JA, De Lorenzo C, Arbitrio M. Polymorphisms and Pharmacogenomics of NQO2: The Past and the Future. Genes (Basel) 2024; 15:87. [PMID: 38254976 PMCID: PMC10815803 DOI: 10.3390/genes15010087] [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: 11/30/2023] [Revised: 12/31/2023] [Accepted: 01/05/2024] [Indexed: 01/24/2024] Open
Abstract
The flavoenzyme N-ribosyldihydronicotinamide (NRH):quinone oxidoreductase 2 (NQO2) catalyzes two-electron reductions of quinones. NQO2 contributes to the metabolism of biogenic and xenobiotic quinones, including a wide range of antitumor drugs, with both toxifying and detoxifying functions. Moreover, NQO2 activity can be inhibited by several compounds, including drugs and phytochemicals such as flavonoids. NQO2 may play important roles that go beyond quinone metabolism and include the regulation of oxidative stress, inflammation, and autophagy, with implications in carcinogenesis and neurodegeneration. NQO2 is a highly polymorphic gene with several allelic variants, including insertions (I), deletions (D) and single-nucleotide (SNP) polymorphisms located mainly in the promoter, but also in other regulatory regions and exons. This is the first systematic review of the literature reporting on NQO2 gene variants as risk factors in degenerative diseases or drug adverse effects. In particular, hypomorphic 29 bp I alleles have been linked to breast and other solid cancer susceptibility as well as to interindividual variability in response to chemotherapy. On the other hand, hypermorphic polymorphisms were associated with Parkinson's and Alzheimer's disease. The I and D promoter variants and other NQO2 polymorphisms may impact cognitive decline, alcoholism and toxicity of several nervous system drugs. Future studies are required to fill several gaps in NQO2 research.
Collapse
Affiliation(s)
- Elzbieta Janda
- Laboratory of Cellular and Molecular Toxicology, Department of Health Science, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy
| | - Jean A. Boutin
- Laboratory of Neuroendocrine Endocrine and Germinal Differentiation and Communication (NorDiC), Université de Rouen Normandie, INSERM, UMR 1239, 76000 Rouen, France;
| | - Carlo De Lorenzo
- Laboratory of Cellular and Molecular Toxicology, Department of Health Science, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy
| | - Mariamena Arbitrio
- Institute for Biomedical Research and Innovation (IRIB), National Research Council of Italy (CNR), 88100 Catanzaro, Italy
| |
Collapse
|
8
|
Yuhan L, Khaleghi Ghadiri M, Gorji A. Impact of NQO1 dysregulation in CNS disorders. J Transl Med 2024; 22:4. [PMID: 38167027 PMCID: PMC10762857 DOI: 10.1186/s12967-023-04802-3] [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: 07/08/2023] [Accepted: 12/12/2023] [Indexed: 01/05/2024] Open
Abstract
NAD(P)H Quinone Dehydrogenase 1 (NQO1) plays a pivotal role in the regulation of neuronal function and synaptic plasticity, cellular adaptation to oxidative stress, neuroinflammatory and degenerative processes, and tumorigenesis in the central nervous system (CNS). Impairment of the NQO1 activity in the CNS can result in abnormal neurotransmitter release and clearance, increased oxidative stress, and aggravated cellular injury/death. Furthermore, it can cause disturbances in neural circuit function and synaptic neurotransmission. The abnormalities of NQO1 enzyme activity have been linked to the pathophysiological mechanisms of multiple neurological disorders, including Parkinson's disease, Alzheimer's disease, epilepsy, multiple sclerosis, cerebrovascular disease, traumatic brain injury, and brain malignancy. NQO1 contributes to various dimensions of tumorigenesis and treatment response in various brain tumors. The precise mechanisms through which abnormalities in NQO1 function contribute to these neurological disorders continue to be a subject of ongoing research. Building upon the existing knowledge, the present study reviews current investigations describing the role of NQO1 dysregulations in various neurological disorders. This study emphasizes the potential of NQO1 as a biomarker in diagnostic and prognostic approaches, as well as its suitability as a target for drug development strategies in neurological disorders.
Collapse
Affiliation(s)
- Li Yuhan
- Epilepsy Research Center, Münster University, Münster, Germany
- Department of Breast Surgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | | | - Ali Gorji
- Epilepsy Research Center, Münster University, Münster, Germany.
- Department of Neurosurgery, Münster University, Münster, Germany.
- Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran, Iran.
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
9
|
Vallucci M, Boutin JA, Janda E, Blandel F, Musgrove R, Di Monte D, Ferry G, Michel PP, Hirsch EC. The specific NQO2 inhibitor, S29434, only marginally improves the survival of dopamine neurons in MPTP-intoxicated mice. J Neural Transm (Vienna) 2024; 131:1-11. [PMID: 37851107 DOI: 10.1007/s00702-023-02709-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Accepted: 10/02/2023] [Indexed: 10/19/2023]
Abstract
Over the years, evidence has accumulated on a possible contributive role of the cytosolic quinone reductase NQO2 in models of dopamine neuron degeneration induced by parkinsonian toxin, but most of the data have been obtained in vitro. For this reason, we asked the question whether NQO2 is involved in the in vivo toxicity of MPTP, a neurotoxin classically used to model Parkinson disease-induced neurodegeneration. First, we show that NQO2 is expressed in mouse substantia nigra dopaminergic cell bodies and in human dopaminergic SH-SY5Y cells as well. A highly specific NQO2 inhibitor, S29434, was able to reduce MPTP-induced cell death in a co-culture system of SH-SY5Y cells with astrocytoma U373 cells but was inactive in SH-SY5Y monocultures. We found that S29434 only marginally prevents substantia nigra tyrosine hydroxylase+ cell loss after MPTP intoxication in vivo. The compound produced a slight increase of dopaminergic cell survival at day 7 and 21 following MPTP treatment, especially with 1.5 and 3 mg/kg dosage regimen. The rescue effect did not reach statistical significance (except for one experiment at day 7) and tended to decrease with the 4.5 mg/kg dose, at the latest time point. Despite the lack of robust protective activity of the inhibitor of NQO2 in the mouse MPTP model, we cannot rule out a possible role of the enzyme in parkinsonian degeneration, particularly because it is substantially expressed in dopaminergic neurons.
Collapse
Affiliation(s)
- Maeva Vallucci
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute -ICM, INSERM, CNRS, Paris, France
| | - Jean A Boutin
- Laboratory of Neuroendocrine Endocrine and Germinal Differentiation and Communication (NorDiC), Univ Rouen Normandie, Inserm, NorDiC, UMR 1239, 76000, Rouen, France.
| | - Elzbieta Janda
- Department of Health Sciences, Campus Germaneto, Magna Graecia University, 88100, Catanzaro, Italy
| | - Florence Blandel
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute -ICM, INSERM, CNRS, Paris, France
| | - Ruth Musgrove
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Australia
| | - Donato Di Monte
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Gilles Ferry
- Institut de R&D, Servier Paris-Saclay, 91190, Gif-Sur-Yvette, France
- Gilles Ferry Consulting, Les Issambres, France
| | - Patrick P Michel
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute -ICM, INSERM, CNRS, Paris, France
| | - Etienne C Hirsch
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute -ICM, INSERM, CNRS, Paris, France
| |
Collapse
|
10
|
Janda E, Parafati M, Martino C, Crupi F, George William JN, Reybier K, Arbitrio M, Mollace V, Boutin JA. Autophagy and neuroprotection in astrocytes exposed to 6-hydroxydopamine is negatively regulated by NQO2: relevance to Parkinson's disease. Sci Rep 2023; 13:21624. [PMID: 38062122 PMCID: PMC10703796 DOI: 10.1038/s41598-023-44666-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 10/11/2023] [Indexed: 12/18/2023] Open
Abstract
Dopaminergic degeneration is a central feature of Parkinson's disease (PD), but glial dysfunction may accelerate or trigger neuronal death. In fact, astrocytes play a key role in the maintenance of the blood-brain barrier and detoxification. 6-hydroxydopamine (6OHDA) is used to induce PD in rodent models due to its specific toxicity to dopaminergic neurons, but its effect on astrocytes has been poorly investigated. Here, we show that 6OHDA dose-dependently impairs autophagy in human U373 cells and primary murine astrocytes in the absence of cell death. LC3II downregulation was observed 6 to 48 h after treatment. Interestingly, 6OHDA enhanced NRH:quinone oxidoreductase 2 (NQO2) expression and activity in U373 cells, even if 6OHDA turned out not to be its substrate. Autophagic flux was restored by inhibition of NQO2 with S29434, which correlated with a partial reduction in oxidative stress in response to 6OHDA in human and murine astrocytes. NQO2 inhibition also increased the neuroprotective capability of U373 cells, since S29434 protected dopaminergic SHSY5Y cells from 6OHDA-induced cell death when cocultured with astrocytes. The toxic effects of 6OHDA on autophagy were attenuated by silencing NQO2 in human cells and primary astrocytes from NQO2-/- mice. Finally, the analysis of Gene Expression Omnibus datasets showed elevated NQO2 gene expression in the blood cells of early-stage PD patients. These data support a toxifying function of NQO2 in dopaminergic degeneration via negative regulation of autophagy and neuroprotection in astrocytes, suggesting a potential pharmacological target in PD.
Collapse
Affiliation(s)
- Elzbieta Janda
- Laboratory of Cellular and Molecular Toxicology, Department of Health Science, University "Magna Græcia" of Catanzaro, 88100, Catanzaro, Italy.
| | - Maddalena Parafati
- Laboratory of Cellular and Molecular Toxicology, Department of Health Science, University "Magna Græcia" of Catanzaro, 88100, Catanzaro, Italy
- Department of Pharmacodynamics, University of Florida, Gainesville, FL 32611, USA
| | - Concetta Martino
- Laboratory of Cellular and Molecular Toxicology, Department of Health Science, University "Magna Græcia" of Catanzaro, 88100, Catanzaro, Italy
| | - Francesco Crupi
- Laboratory of Cellular and Molecular Toxicology, Department of Health Science, University "Magna Græcia" of Catanzaro, 88100, Catanzaro, Italy
| | | | - Karine Reybier
- UMR 152 Pharma-Dev, Université de Toulouse III, IRD, UPS, 31400, Toulouse, France
| | - Mariamena Arbitrio
- Institute for Biomedical Research and Innovation (IRIB), National Research Council of Italy (CNR), 88100, Catanzaro, Italy.
| | - Vincenzo Mollace
- Laboratory of Cellular and Molecular Toxicology, Department of Health Science, University "Magna Græcia" of Catanzaro, 88100, Catanzaro, Italy
| | - Jean A Boutin
- Laboratory of Neuroendocrine Endocrine and Germinal Differentiation and Communication (NorDiC), Univ Rouen Normandie, Inserm, NorDiC UMR 1239, 76000, Rouen, France
| |
Collapse
|
11
|
Chen G, Chen W, Xu J, Ma G, Hu X, Chen G. The current trend and challenges of developing red yeast rice-based food supplements for hypercholesterolemia. JOURNAL OF FUTURE FOODS 2023; 3:312-329. [DOI: 10.1016/j.jfutfo.2023.03.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/06/2024]
|
12
|
3-Arylidene-2-oxindoles as Potent NRH:Quinone Oxidoreductase 2 Inhibitors. Molecules 2023; 28:molecules28031174. [PMID: 36770840 PMCID: PMC9920986 DOI: 10.3390/molecules28031174] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 01/19/2023] [Accepted: 01/21/2023] [Indexed: 01/26/2023] Open
Abstract
The enzyme NRH:quinone oxidoreductase 2 (NQO2) plays an important role in the pathogenesis of various diseases such as neurodegenerative disorders, malaria, glaucoma, COVID-19 and cancer. NQO2 expression is known to be increased in some cancer cell lines. Since 3-arylidene-2-oxindoles are widely used in the design of new anticancer drugs, such as kinase inhibitors, it was interesting to study whether such structures have additional activity towards NQO2. Herein, we report the synthesis and study of 3-arylidene-2-oxindoles as novel NRH:quinone oxidoreductase inhibitors. It was demonstrated that oxindoles with 6-membered aryls in the arylidene moiety were obtained predominantly as E-isomers while for some 5-membered aryls, the Z-isomers prevailed. The most active compounds inhibited NQO2 with an IC50 of 0.368 µM. The presence of a double bond in the oxindoles was crucial for NQO2 inhibition activity. There was no correlation between NQO2 inhibition activity of the synthesized compounds and their cytotoxic effect on the A549 cell line.
Collapse
|
13
|
Aljohani GF, Abolibda TZ, Alhilal M, Al-Humaidi JY, Alhilal S, Ahmed HA, Gomha SM. Novel thiadiazole-thiazole hybrids: synthesis, molecular docking, and cytotoxicity evaluation against liver cancer cell lines. JOURNAL OF TAIBAH UNIVERSITY FOR SCIENCE 2022. [DOI: 10.1080/16583655.2022.2135805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Affiliation(s)
- Ghadah F. Aljohani
- Chemistry Department, College of Science, Taibah University, Al-Madinah Al Munawarah, Saudi Arabia
| | - Tariq Z. Abolibda
- Chemistry Department, Faculty of Science, Islamic University of Madinah, Madinah, Saudi Arabia
| | - Mohammad Alhilal
- Department of Nursing, Faculty of Health Sciences, Mardin Artuklu University, Mardin, Turkey
| | - Jehan Y. Al-Humaidi
- Department of Chemistry, College of Science, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Suzan Alhilal
- Department of Chemistry, Faculty of Science, Albaath University, Erzurum, Turkey
| | - Hoda A. Ahmed
- Department of Chemistry, Faculty of Science, Cairo University, Cairo, Egypt
| | - Sobhi M. Gomha
- Chemistry Department, Faculty of Science, Islamic University of Madinah, Madinah, Saudi Arabia
- Department of Chemistry, Faculty of Science, Cairo University, Cairo, Egypt
| |
Collapse
|
14
|
Nephrotoxicity evaluation and proteomic analysis in kidneys of rats exposed to thioacetamide. Sci Rep 2022; 12:6837. [PMID: 35477741 PMCID: PMC9046159 DOI: 10.1038/s41598-022-11011-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 04/18/2022] [Indexed: 11/18/2022] Open
Abstract
Thioacetamide (TAA) was administered orally at 0, 10, and 30 mg/kg body weight (BW) daily to Sprague–Dawley rats aged 6–7 weeks for 28 consecutive days. Nephrotoxicity and proteomics were evaluated in the kidneys of rats exposed to TAA. The BW decreased, however, the relative kidneys weight increased. No significant histopathologic abnormalities were found in the kidneys. The numbers of monocytes and platelets were significantly increased. However, the mean corpuscular volume and hematocrit values were decreased significantly in rats exposed to 30 mg/kg BW TAA. The expression levels of Kim-1 and NGAL were increased 4 to 5-fold in the kidneys, resulting in significant nephrotoxicity. Proteomic analysis was conducted and a total of 5221 proteins spots were resolved. Of these, 3 and 21 protein spots were up- and downregulated, respectively. The validation of seven proteins was performed by Western blot analysis. The expression level of ASAP2 was significantly upregulated, whereas RGS14, MAP7Dl, IL-3Rα, Tmod1, NQO2, and MUP were reduced. Sixteen isoforms of MUP were found by the 2DE immunoblot assay and were significantly downregulated with increasing exposure to TAA. MUP isoforms were compared in the liver, kidneys, and urine of untreated rats and a total of 43 isoforms were found.
Collapse
|
15
|
Boutin JA. [Melatonin: A short clarification for the over-enthusiasts]. Med Sci (Paris) 2022; 38:89-95. [PMID: 35060893 DOI: 10.1051/medsci/2021115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Melatonin is a naturally occurring molecule derived from tryptophan. Melatonin is a key player in relaying the circadian rhythm between our environment and our body. It has also a key role in rhythming the seasons (more production during long nights and less during short ones) as well as in the reproduction cycles of the mammals. Melatonin is often and surprisingly presented as a molecule with multiple therapeutic properties that can fix (or help to fix) many health issues, such as diseases (cancer, ageing, virus-induced affections including COVID-19, etc…) or toxicological situations (metals, venoms, chemical such as adriamycin [doxorubicin], methotrexate or paclitaxel). The mechanistics behind those wonders is still missing and this is puzzling. In the present commentary, the main well-established biological properties are presented and briefly discussed with the aim of delineating the borders between facts and wishful thinking.
Collapse
Affiliation(s)
- Jean A Boutin
- PHARMADEV, Pharmacochimie et biologie pour le développement, UUM 152, Faculté de pharmacie, Rue des Maraîchers, 31000 Toulouse, France
| |
Collapse
|
16
|
Calamini B, Ferry G, Boutin JA. Cloning, Expression, Purification, Crystallization, and X-Ray Structural Determination of the Human NQO2 in Complex with Melatonin. Methods Mol Biol 2022; 2550:291-304. [PMID: 36180700 DOI: 10.1007/978-1-0716-2593-4_31] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Melatonin (N-acetyl-5-methoxytryptamine) is a neurohormone which possesses a wide range of biological effects. The effects mediated by melatonin are in part attributed to the antioxidant properties of the molecule, which may act as scavenger of free radicals, and also to the binding of melatonin to its protein targets. For a long time, melatonin had been described as a ligand of a putative "receptor" present in the mammalian brain. Several studies were thus carried out with the goal of clarifying the nature of this melatonin "receptor," which led to the discovery of MT3 as the third melatonin binding site. This binding site was confirmed independently by several groups, and it was eventually demonstrated that MT3 was the enzyme quinone reductase 2 (NQO2). Among the different approaches used to validate that MT3 was indeed NQO2, the co-crystallization of NQO2 with melatonin was key in demonstrating the exact binding site and mode of melatonin to the enzyme and led to a clear understanding of the residues important for protein binding and inhibition. In this chapter, we described the details for the cloning, expression, and purification of the human enzyme NQO2. We also describe a detailed protocol for the crystallization of melatonin with this protein.
Collapse
Affiliation(s)
- Barbara Calamini
- Sanofi Strasbourg R&D Center, Strasbourg, France
- Eli Lilly and Company, Cambridge, MA, USA
| | - Gilles Ferry
- Pole d'expertise Biotechnologie, Chimie & Biologie, Institut de Recherches Servier, Croissy-sur-Seine, France
| | - Jean A Boutin
- Pole d'expertise Biotechnologie, Chimie & Biologie, Institut de Recherches Servier, Croissy-sur-Seine, France.
- Pharma-Dev, UMR 152, Eli Lilly and Company, Toulouse, France.
| |
Collapse
|
17
|
Ferry G, Boutin JA. Measurement of NQO2 Catalytic Activity and of Its Inhibition by Melatonin. Methods Mol Biol 2022; 2550:315-321. [PMID: 36180702 DOI: 10.1007/978-1-0716-2593-4_33] [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] [Indexed: 06/16/2023]
Abstract
The third melatonin binding site MT3 turned out to be an enzyme, NQO2 (E.C. 1.6.99.2). Its catalytic activity is inhibited by melatonin with an IC50 in the 50-100 μM range. Some of the functions of melatonin at pharmacological concentrations (1 μM and above) might be explained by this inhibition capacity of melatonin at NQO2. In order to determine precisely these parameters, it is required to comprehend the basic enzymology of this enzyme. In the following chapter, we present the basic conditions of measuring NQO2 catalytic activities and inhibition.
Collapse
Affiliation(s)
- Gilles Ferry
- Pole d'expertise Biotechnologie, Chimie & Biologie, Institut de Recherches Servier, Croissy-sur-Seine, France
| | - Jean A Boutin
- Pole d'expertise Biotechnologie, Chimie & Biologie, Institut de Recherches Servier, Croissy-sur-Seine, France.
- Pharma-Dev, UMR 152, Université de Toulouse, Toulouse, France.
| |
Collapse
|
18
|
Boutin JA, Stojko J, Ferry G, Cianferani S. Measuring the NQO2: Melatonin Complex by Native Nano-Electrospray Ionization Mass Spectrometry. Methods Mol Biol 2022; 2550:323-328. [PMID: 36180703 DOI: 10.1007/978-1-0716-2593-4_34] [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] [Indexed: 06/16/2023]
Abstract
Melatonin exerts its effects through a series of target proteins/receptors and enzymes. Its antioxidant capacity might be due to its capacity to inhibit a quinone reductase (NQO2) at high concentration (50 μM). Demonstrating the existence of a complex between a compound and a protein is often not easy. It requires either that the compound is an inhibitor-and the complex translates by an inhibition of the catalytic activity-or the compound is radiolabeled-and the complex translates in standard binding approaches, such as in receptology. Outside these two cases, the detection of the protein:small molecule complexes by mass spectrometry has recently been made possible, thanks to the development of so-called native mass spectrometry. Using this approach, one can measure masses corresponding to an intact noncovalent complex between a compound and its target, usually after titration or competition experiments. In the present chapter, we detail the characterization of NQO2:melatonin interaction using native mass spectrometry.
Collapse
Affiliation(s)
- Jean A Boutin
- Pole d'expertise Biotechnologie, Chimie & Biologie, Institut de Recherches Servier, Croissy-sur-Seine, France.
- PHARMADEV (Pharmacochimie et biologie pour le développement), Faculté de Pharmacie, Toulouse, France.
| | - Johann Stojko
- Pole d'expertise Biotechnologie, Chimie & Biologie, Institut de Recherches Servier, Croissy-sur-Seine, France
| | - Gilles Ferry
- Pole d'expertise Biotechnologie, Chimie & Biologie, Institut de Recherches Servier, Croissy-sur-Seine, France
| | - Sarah Cianferani
- Laboratoire de Spectrométrie de Masse BioOrganique, Université de Strasbourg, CNRS, Strasbourg, France
- Infrastructure Nationale de Protéomique ProFI, FR2048 CNRS, Strasbourg, France
| |
Collapse
|
19
|
Voronin MV, Kadnikov IA, Zainullina LF, Logvinov IO, Verbovaya ER, Antipova TA, Vakhitova YV, Seredenin SB. Neuroprotective Properties of Quinone Reductase 2 Inhibitor M-11, a 2-Mercaptobenzimidazole Derivative. Int J Mol Sci 2021; 22:13061. [PMID: 34884863 PMCID: PMC8658107 DOI: 10.3390/ijms222313061] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 11/29/2021] [Accepted: 11/29/2021] [Indexed: 01/03/2023] Open
Abstract
The ability of NQO2 to increase the production of free radicals under enhanced generation of quinone derivatives of catecholamines is considered to be a component of neurodegenerative disease pathogenesis. The present study aimed to investigate the neuroprotective mechanisms of original NQO2 inhibitor M-11 (2-[2-(3-oxomorpholin-4-il)-ethylthio]-5-ethoxybenzimidazole hydrochloride) in a cellular damage model using NQO2 endogenous substrate adrenochrome (125 µM) and co-substrate BNAH (100 µM). The effects of M-11 (10-100 µM) on the reactive oxygen species (ROS) generation, apoptosis and lesion of nuclear DNA were evaluated using flow cytometry and single-cell gel electrophoresis assay (comet assay). Results were compared with S29434, the reference inhibitor of NQO2. It was found that treatment of HT-22 cells with M-11 results in a decline of ROS production triggered by incubation of cells with NQO2 substrate and co-substrate. Pre-incubation of HT-22 cells with compounds M-11 or S29434 results in a decrease of DNA damage and late apoptotic cell percentage reduction. The obtained results provide a rationale for further development of the M-11 compound as a potential neuroprotective agent.
Collapse
Affiliation(s)
- Mikhail V. Voronin
- Department of Pharmacogenetics, Federal State Budgetary Institution “Research Zakusov Institute of Pharmacology”, Baltiyskaya Street 8, 125315 Moscow, Russia; (L.F.Z.); (I.O.L.); (E.R.V.); (T.A.A.)
| | - Ilya A. Kadnikov
- Department of Pharmacogenetics, Federal State Budgetary Institution “Research Zakusov Institute of Pharmacology”, Baltiyskaya Street 8, 125315 Moscow, Russia; (L.F.Z.); (I.O.L.); (E.R.V.); (T.A.A.)
| | | | | | | | | | - Yulia V. Vakhitova
- Department of Pharmacogenetics, Federal State Budgetary Institution “Research Zakusov Institute of Pharmacology”, Baltiyskaya Street 8, 125315 Moscow, Russia; (L.F.Z.); (I.O.L.); (E.R.V.); (T.A.A.)
| | - Sergei B. Seredenin
- Department of Pharmacogenetics, Federal State Budgetary Institution “Research Zakusov Institute of Pharmacology”, Baltiyskaya Street 8, 125315 Moscow, Russia; (L.F.Z.); (I.O.L.); (E.R.V.); (T.A.A.)
| |
Collapse
|
20
|
Fukami T, Yokoi T, Nakajima M. Non-P450 Drug-Metabolizing Enzymes: Contribution to Drug Disposition, Toxicity, and Development. Annu Rev Pharmacol Toxicol 2021; 62:405-425. [PMID: 34499522 DOI: 10.1146/annurev-pharmtox-052220-105907] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Most clinically used drugs are metabolized in the body via oxidation, reduction, or hydrolysis reactions, which are considered phase I reactions. Cytochrome P450 (P450) enzymes, which primarily catalyze oxidation reactions, contribute to the metabolism of over 50% of clinically used drugs. In the last few decades, the function and regulation of P450s have been extensively studied, whereas the characterization of non-P450 phase I enzymes is still incomplete. Recent studies suggest that approximately 30% of drug metabolism is carried out by non-P450 enzymes. This review summarizes current knowledge of non-P450 phase I enzymes, focusing on their roles in controlling drug efficacy and adverse reactions as an important aspect of drug development. Expected final online publication date for the Annual Review of Pharmacology and Toxicology, Volume 62 is January 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Tatsuki Fukami
- Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences, and WPI Nano Life Science Institute (WPI-NanoLSI), Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan;
| | - Tsuyoshi Yokoi
- Department of Drug Safety Sciences, Nagoya University Graduate School of Medicine, Showa-ku, Nagoya 466-8550, Japan
| | - Miki Nakajima
- Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences, and WPI Nano Life Science Institute (WPI-NanoLSI), Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan;
| |
Collapse
|
21
|
Petronek MS, Stolwijk JM, Murray SD, Steinbach EJ, Zakharia Y, Buettner GR, Spitz DR, Allen BG. Utilization of redox modulating small molecules that selectively act as pro-oxidants in cancer cells to open a therapeutic window for improving cancer therapy. Redox Biol 2021; 42:101864. [PMID: 33485837 PMCID: PMC8113052 DOI: 10.1016/j.redox.2021.101864] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Revised: 01/08/2021] [Accepted: 01/10/2021] [Indexed: 02/07/2023] Open
Abstract
There is a rapidly growing body of literature supporting the notion that differential oxidative metabolism in cancer versus normal cells represents a metabolic frailty that can be exploited to open a therapeutic window into cancer therapy. These cancer cell-specific metabolic frailties may be amenable to manipulation with non-toxic small molecule redox active compounds traditionally thought to be antioxidants. In this review we describe the potential mechanisms and clinical applicability in cancer therapy of four small molecule redox active agents: melatonin, vitamin E, selenium, and vitamin C. Each has shown the potential to have pro-oxidant effects in cancer cells while retaining antioxidant activity in normal cells. This dichotomy can be exploited to improve responses to radiation and chemotherapy by opening a therapeutic window based on a testable biochemical rationale amenable to confirmation with biomarker studies during clinical trials. Thus, the unique pro-oxidant/antioxidant properties of melatonin, vitamin E, selenium, and vitamin C have the potential to act as effective adjuvants to traditional cancer therapies, thereby improving cancer patient outcomes.
Collapse
Affiliation(s)
- M S Petronek
- Department of Radiation Oncology, University of Iowa, Iowa City, IA, USA
| | - J M Stolwijk
- Department of Radiation Oncology, University of Iowa, Iowa City, IA, USA
| | - S D Murray
- Department of Cancer Biology, University of Iowa, Iowa City, IA, USA
| | - E J Steinbach
- Department of Radiation Oncology, University of Iowa, Iowa City, IA, USA
| | - Y Zakharia
- Division of Hematology and Oncology, Department of Internal Medicine, University of Iowa, Iowa City, IA, USA
| | - G R Buettner
- Department of Radiation Oncology, University of Iowa, Iowa City, IA, USA
| | - D R Spitz
- Department of Radiation Oncology, University of Iowa, Iowa City, IA, USA
| | - B G Allen
- Department of Radiation Oncology, University of Iowa, Iowa City, IA, USA.
| |
Collapse
|
22
|
Apigenin and Luteolin Regulate Autophagy by Targeting NRH-Quinone Oxidoreductase 2 in Liver Cells. Antioxidants (Basel) 2021; 10:antiox10050776. [PMID: 34068281 PMCID: PMC8153271 DOI: 10.3390/antiox10050776] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 05/04/2021] [Accepted: 05/07/2021] [Indexed: 12/15/2022] Open
Abstract
Dietary flavonoids stimulate autophagy and prevent liver dysfunction, but the upstream signaling pathways triggered by these compounds are not well understood. Certain polyphenols bind directly to NRH-quinone oxidoreductase 2 (NQO2) and inhibit its activity. NQO2 is highly expressed in the liver, where it participates in quinone metabolism, but recent evidence indicates that it may also play a role in the regulation of oxidative stress and autophagy. Here, we addressed a potential role of NQO2 in autophagy induction by flavonoids. The pro-autophagic activity of seven flavonoid aglycons correlated perfectly with their ability to inhibit NQO2 activity, and flavones such as apigenin and luteolin showed the strongest activity in all assays. The silencing of NQO2 strongly reduced flavone-induced autophagic flux, although it increased basal LC3-II levels in HepG2 cells. Both flavones induced AMP kinase (AMPK) activation, while its reduction by AMPK beta (PRKAB1) silencing inhibited flavone-induced autophagy. Interestingly, the depletion of NQO2 levels by siRNA increased the basal AMPK phosphorylation but abrogated its further increase by apigenin. Thus, NQO2 contributes to the negative regulation of AMPK activity and autophagy, while its targeting by flavones releases pro-autophagic signals. These findings imply that NQO2 works as a flavone receptor mediating autophagy and may contribute to other hepatic effects of flavonoids.
Collapse
|
23
|
Chhour M, Perio P, Gayon R, Ternet-Fontebasso H, Ferry G, Nepveu F, Boutin JA, Sudor J, Reybier K. Association of NQO2 With UDP-Glucuronosyltransferases Reduces Menadione Toxicity in Neuroblastoma Cells. Front Pharmacol 2021; 12:660641. [PMID: 34040527 PMCID: PMC8142080 DOI: 10.3389/fphar.2021.660641] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 04/22/2021] [Indexed: 11/13/2022] Open
Abstract
The balance between detoxification and toxicity is linked to enzymes of the drug metabolism Phase I (cytochrome P450 or oxidoreductases) and phase II conjugating enzymes (such as the UGTs). After the reduction of quinones, the product of the reaction, the quinols-if not conjugated-re-oxidizes spontaneously to form the substrate quinone with the concomitant production of the toxic reactive oxygen species (ROS). Herein, we documented the modulation of the toxicity of the quinone menadione on a genetically modified neuroblastoma model cell line that expresses both the quinone oxidoreductase 2 (NQO2, E.C. 1.10.5.1) alone or together with the conjugation enzyme UDP-glucuronosyltransferase (UGT1A6, E.C. 2.4.1.17), one of the two UGT isoenzymes capable to conjugate menadione. As previously shown, NQO2 enzymatic activity is concomitant to massive ROS production, as previously shown. The quantification of ROS produced by the menadione metabolism was probed by electron-paramagnetic resonance (EPR) on cell homogenates, while the production of superoxide was measured by liquid chromatography coupled to mass spectrometry (LC-MS) on intact cells. In addition, the dysregulation of the redox homeostasis upon the cell exposure to menadione was studied by fluorescence measurements. Both EPR and LCMS studies confirmed a significant increase in the ROS production in the NQO2 overexpressing cells due to the fast reduction of quinone into quinol that can re-oxidize to form superoxide radicals. However, the effect of NQO2 inhibition was drastically different between cells overexpressing only NQO2 vs. both NQO2 and UGT. Whereas NQO2 inhibition decreases the amount of superoxide in the first case by decreasing the amount of quinol formed, it increased the toxicity of menadione in the cells co-expressing both enzymes. Moreover, for the cells co-expressing QR2 and UGT the homeostasis dysregulation was lower in presence of menadione than for the its counterpart expressing only QR2. Those results confirmed that the cooperation of the two enzymes plays a fundamental role during the cells' detoxification process. The fluorescence measurements of the variation of redox homeostasis of each cell line and the detection of a glucuronide form of menadiol in the cells co-expressing NQO2 and UGT1A6 enzymes further confirmed our findings.
Collapse
Affiliation(s)
- Monivan Chhour
- Pharma-Dev UMR 152, Université de Toulouse, IRD, UPS, Toulouse, France
| | - Pierre Perio
- Pharma-Dev UMR 152, Université de Toulouse, IRD, UPS, Toulouse, France
| | - Regis Gayon
- Flash Therapeutics, Parc Technologique du Canal, Toulouse, France
| | | | - Gilles Ferry
- Biotechnologie, Pharmacologie Moléculaire et Cellulaire, Institut de Recherches Servier, Croissy-sur-Seine, France
| | - Françoise Nepveu
- Pharma-Dev UMR 152, Université de Toulouse, IRD, UPS, Toulouse, France
| | - Jean A Boutin
- Pharma-Dev UMR 152, Université de Toulouse, IRD, UPS, Toulouse, France.,Biotechnologie, Pharmacologie Moléculaire et Cellulaire, Institut de Recherches Servier, Croissy-sur-Seine, France
| | - Jan Sudor
- Pharma-Dev UMR 152, Université de Toulouse, IRD, UPS, Toulouse, France
| | - Karine Reybier
- Pharma-Dev UMR 152, Université de Toulouse, IRD, UPS, Toulouse, France
| |
Collapse
|
24
|
Ross D, Siegel D. The diverse functionality of NQO1 and its roles in redox control. Redox Biol 2021; 41:101950. [PMID: 33774477 PMCID: PMC8027776 DOI: 10.1016/j.redox.2021.101950] [Citation(s) in RCA: 250] [Impact Index Per Article: 62.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 03/15/2021] [Accepted: 03/16/2021] [Indexed: 12/20/2022] Open
Abstract
In this review, we summarize the multiple functions of NQO1, its established roles in redox processes and potential roles in redox control that are currently emerging. NQO1 has attracted interest due to its roles in cell defense and marked inducibility during cellular stress. Exogenous substrates for NQO1 include many xenobiotic quinones. Since NQO1 is highly expressed in many solid tumors, including via upregulation of Nrf2, the design of compounds activated by NQO1 and NQO1-targeted drug delivery have been active areas of research. Endogenous substrates have also been proposed and of relevance to redox stress are ubiquinone and vitamin E quinone, components of the plasma membrane redox system. Established roles for NQO1 include a superoxide reductase activity, NAD+ generation, interaction with proteins and their stabilization against proteasomal degradation, binding and regulation of mRNA translation and binding to microtubules including the mitotic spindles. We also summarize potential roles for NQO1 in regulation of glucose and insulin metabolism with relevance to diabetes and the metabolic syndrome, in Alzheimer's disease and in aging. The conformation and molecular interactions of NQO1 can be modulated by changes in the pyridine nucleotide redox balance suggesting that NQO1 may function as a redox-dependent molecular switch.
Collapse
Affiliation(s)
- David Ross
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA.
| | - David Siegel
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| |
Collapse
|