1
|
Fehsel K. Metabolic Side Effects from Antipsychotic Treatment with Clozapine Linked to Aryl Hydrocarbon Receptor (AhR) Activation. Biomedicines 2024; 12:2294. [PMID: 39457607 PMCID: PMC11505606 DOI: 10.3390/biomedicines12102294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 09/30/2024] [Accepted: 10/07/2024] [Indexed: 10/28/2024] Open
Abstract
Metabolic syndrome (MetS) is the most common adverse drug reaction from psychiatric pharmacotherapy. Neuroreceptor blockade by the antipsychotic drug clozapine induces MetS in about 30% of patients. Similar to insulin resistance, clozapine impedes Akt kinase activation, leading to intracellular glucose and glutathione depletion. Additional cystine shortage triggers tryptophan degradation to kynurenine, which is a well-known AhR ligand. Ligand-bound AhR downregulates the intracellular iron pool, thereby increasing the risk of mitochondrial dysfunction. Scavenging iron stabilizes the transcription factor HIF-1, which shifts the metabolism toward transient glycolysis. Furthermore, the AhR inhibits AMPK activation, leading to obesity and liver steatosis. Increasing glucose uptake by AMPK activation prevents dyslipidemia and liver damage and, therefore, reduces the risk of MetS. In line with the in vitro results, feeding experiments with rats revealed a disturbed glucose-/lipid-/iron-metabolism from clozapine treatment with hyperglycemia and hepatic iron deposits in female rats and steatosis and anemia in male animals. Decreased energy expenditure from clozapine treatment seems to be the cause of the fast weight gain in the first weeks of treatment. In patients, this weight gain due to neuroleptic treatment correlates with an improvement in psychotic syndromes and can even be used to anticipate the therapeutic effect of the treatment.
Collapse
Affiliation(s)
- Karin Fehsel
- Department of Psychiatry and Psychotherapy, Medical Faculty, Heinrich-Heine-University, Bergische Landstrasse 2, 40629 Duesseldorf, Germany
| |
Collapse
|
2
|
Nomiya H, Sakurai K, Miyamoto Y, Oka M, Yoneda Y, Hikida T, Yamada M. A Kpna1-deficient psychotropic drug-induced schizophrenia model mouse for studying gene-environment interactions. Sci Rep 2024; 14:3376. [PMID: 38336912 PMCID: PMC10858057 DOI: 10.1038/s41598-024-53237-3] [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: 10/13/2023] [Accepted: 01/30/2024] [Indexed: 02/12/2024] Open
Abstract
KPNA1 is a mediator of nucleocytoplasmic transport that is abundantly expressed in the mammalian brain and regulates neuronal differentiation and synaptic function. De novo mutations in Kpna1 have been identified using genome-wide association studies in humans with schizophrenia; however, it remains unclear how KPNA1 contributes to schizophrenia pathogenesis. Recent studies have suggested a complex combination of genetic and environmental factors that are closely related to psychiatric disorders. Here, we found that subchronic administration of phencyclidine, a psychotropic drug, induced vulnerability and behavioral abnormalities consistent with the symptoms of schizophrenia in Kpna1-deficient mice. Microarray assessment revealed that the expression levels of dopamine d1/d2 receptors, an RNA editing enzyme, and a cytoplasmic dynein component were significantly altered in the nucleus accumbens brain region in a gene-environment (G × E) interaction-dependent manner. Our findings demonstrate that Kpna1-deficient mice may be useful as a G × E interaction mouse model for psychiatric disorders and for further investigation into the pathogenesis of such diseases and disorders.
Collapse
Affiliation(s)
- Hirotaka Nomiya
- Department of Cell Biology and Biochemistry, Division of Medicine, Faculty of Medical Sciences, University of Fukui, 23-3 Matsuoka Shimoaizuki, Eiheiji-cho, Yoshida-gun, Fukui, 910-1193, Japan
| | - Koki Sakurai
- Laboratory for Advanced Brain Functions, Institute for Protein Research, Osaka University, 3-2 Yamada-oka, Suita, Osaka, 565-0871, Japan
| | - Yoichi Miyamoto
- Laboratory of Nuclear Transport Dynamics, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), 7-6-8 Saito-Asagi, Ibaraki, Osaka, 567-0085, Japan
| | - Masahiro Oka
- Laboratory of Nuclear Transport Dynamics, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), 7-6-8 Saito-Asagi, Ibaraki, Osaka, 567-0085, Japan
| | - Yoshihiro Yoneda
- The Research Foundation for Microbial Diseases Osaka University, Integrated Life Science Building, Osaka University, 3-1 Yamada-oka, Suita, Osaka, 565-0871, Japan
| | - Takatoshi Hikida
- Laboratory for Advanced Brain Functions, Institute for Protein Research, Osaka University, 3-2 Yamada-oka, Suita, Osaka, 565-0871, Japan.
- Department of Research and Drug Discovery, Medical Innovation Center, Kyoto University Graduate School of Medicine, 53 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto, 606-8397, Japan.
| | - Masami Yamada
- Department of Cell Biology and Biochemistry, Division of Medicine, Faculty of Medical Sciences, University of Fukui, 23-3 Matsuoka Shimoaizuki, Eiheiji-cho, Yoshida-gun, Fukui, 910-1193, Japan.
- Life Science Innovation Center, University of Fukui, 3-9-1, Bunkyo, Fukui-City, Fukui, 910-8507, Japan.
| |
Collapse
|
3
|
Shin EJ, Jeong JH, Nguyen BT, Sharma N, Tran CNK, Nah SY, Lee Y, Byun JK, Ko SK, Kim HC. Ginsenoside Re attenuates 8-OH-DPAT-induced serotonergic behaviors in mice via interactive modulation between PKCδ gene and Nrf2. Drug Chem Toxicol 2023; 46:281-296. [PMID: 35707918 DOI: 10.1080/01480545.2021.2022689] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
It has been recognized that serotonergic blocker showed serious side effects, and that ginsenoside modulated serotonergic system with the safety. However, the effects of ginsenoside on serotonergic impairments remain to be clarified. Thus, we investigated ginsenoside Re (GRe), a major bioactive component in the mountain-cultivated ginseng on (±)-8-hydroxy-dipropylaminotetralin (8-OH-DPAT), a 5-HT1A receptor agonist. In the present study, we observed that the treatment with GRe resulted in significant inhibition of protein kinase C δ (PKCδ) phosphorylation induced by the 5-HT1A receptor agonist (±)-8-hydroxy-dipropylaminotetralin (8-OH-DPAT) in the hypothalamus of the wild-type (WT) mice. The inhibition of GRe was comparable with that of the PKCδ inhibitor rottlerin or the 5-HT1A receptor antagonist WAY100635 (WAY). 8-OH-DPAT-induced significant reduction in nuclear factor erythroid-2-related factor 2 (Nrf2)-related system (i.e., Nrf2 DNA binding activity, γ-glutamylcysteine ligase modifier (GCLm) and γ-glutamylcysteine ligase catalytic (GCLc) mRNA expression, and glutathione (GSH)/oxidized glutathione (GSSG) ratio) was significantly attenuated by GRe, rottlerin, or WAY in WT mice. However, PKCδ gene knockout significantly protected the Nrf2-dependent system from 8-OH-DPAT insult in mice. Increases in 5-hydroxytryptophan (5-HT) turnover rate, overall serotonergic behavioral score, and hypothermia induced by 8-OH-DPAT were significantly attenuated by GRe, rottlerin, or WAY in WT mice. Consistently, PKCδ gene knockout significantly attenuated these parameters in mice. However, GRe or WAY did not provide any additional positive effects on the serotonergic protective potential mediated by PKCδ gene knockout in mice. Therefore, our results suggest that PKCδ is an important mediator for GRe-mediated protective activity against serotonergic impairments/oxidative burden caused by the 5-HT1A receptor.
Collapse
Affiliation(s)
- Eun-Joo Shin
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chuncheon, South Korea
| | - Ji Hoon Jeong
- Department of Global Innovative Drugs, College of Medicine, Graduate School of Chung-Ang University, Chung-Ang University, Seoul, Republic of Korea
| | - Bao-Trong Nguyen
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chuncheon, South Korea
| | - Naveen Sharma
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chuncheon, South Korea.,Department of Global Innovative Drugs, College of Medicine, Graduate School of Chung-Ang University, Chung-Ang University, Seoul, Republic of Korea
| | - Cuong Ngoc Kim Tran
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chuncheon, South Korea
| | - Seung-Yeol Nah
- Ginsentology Research Laboratory and Department of Physiology, College of Veterinary Medicine and Bio/Molecular Informatics Center, Konkuk University, Seoul, Republic of Korea
| | - Yi Lee
- Department of Industrial Plant Science & Technology, Chungbuk National University, Chungju, Republic of Korea
| | - Jae Kyung Byun
- Korea Society of Forest Environmental Research, Namyangju, Republic of Korea
| | - Sung Kwon Ko
- Department of Oriental Medical Food and Nutrition, Semyung University, Jecheon, Republic of Korea
| | - Hyoung-Chun Kim
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chuncheon, South Korea
| |
Collapse
|
4
|
Shin EJ, Nguyen BT, Sharma N, Tran NKC, Nguyen YND, Hwang Y, Park JH, Nah SY, Ko SK, Byun JK, Lee Y, Kim DJ, Jeong JH, Kim HC. Ginsenoside Re mitigates memory impairments in aged GPx-1 KO mice by inhibiting the interplay between PAFR, NFκB, and microgliosis in the hippocampus. Food Chem Toxicol 2023; 173:113627. [PMID: 36682417 DOI: 10.1016/j.fct.2023.113627] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 01/04/2023] [Accepted: 01/17/2023] [Indexed: 01/22/2023]
Abstract
Ginsenoside Re (GRe) upregulates anti-aging klotho by mainly upregulating glutathione peroxidase-1 (GPx-1). However, the anti-aging mechanism of GPx-1 remains elusive. Here we investigated whether the GRe-mediated upregulation of GPx-1 modulates oxidative and proinflammatory insults. GPx-1 gene depletion altered redox homeostasis and platelet-activating factor receptor (PAFR) and nuclear factor kappa B (NFκB) expression, whereas the genetic overexpression of GPx-1 or GRe mitigated this phenomenon in aged mice. Importantly, the NFκB inhibitor pyrrolidine dithiocarbamate (PDTC) did not affect PAFR expression, while PAFR inhibition (i.e., PAFR knockout or ginkgolide B) significantly attenuated NFκB nuclear translocation, suggesting that PAFR could be an upstream molecule for NFκB activation. Iba-1-labeled microgliosis was more underlined in aged GPx-1 KO than in aged WT mice. Triple-labeling immunocytochemistry showed that PAFR and NFκB immunoreactivities were co-localized in Iba-1-positive populations in aged mice, indicating that microglia released these proteins. GRe inhibited triple-labeled immunoreactivity. The microglial inhibitor minocycline attenuated aging-related reduction in phospho-ERK. The effect of minocycline was comparable with that of GRe. GRe, ginkgolide B, PDTC, or minocycline also attenuated aging-evoked memory impairments. Therefore, GRe ameliorated aging-associated memory impairments in the absence of GPx-1 by inactivating oxidative insult, PAFR, NFkB, and microgliosis.
Collapse
Affiliation(s)
- Eun-Joo Shin
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon, 24341, Republic of Korea
| | - Bao Trong Nguyen
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon, 24341, Republic of Korea
| | - Naveen Sharma
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon, 24341, Republic of Korea; Department of Global Innovative Drugs, Graduate School of Chung-Ang University, College of Medicine, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Ngoc Kim Cuong Tran
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon, 24341, Republic of Korea
| | - Yen Nhi Doan Nguyen
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon, 24341, Republic of Korea
| | - Yeonggwang Hwang
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon, 24341, Republic of Korea
| | - Jung Hoon Park
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon, 24341, Republic of Korea
| | - Seung-Yeol Nah
- Ginsentology Research Laboratory and Department of Physiology, College of Veterinary Medicine, Konkuk University, Seoul, 05029, Republic of Korea
| | - Sung Kwon Ko
- Department of Oriental Medical Food & Nutrition, Semyung University, Jecheon, 27136, Republic of Korea
| | - Jae Kyung Byun
- Korea Society of Forest Environmental Research, Namyanju, 12106, Republic of Korea
| | - Yi Lee
- Department of Industrial Plant Science & Technology, Chungbuk National University, Chungju, 28644, Republic of Korea
| | - Dae-Joong Kim
- Department of Anatomy and Cell Biology, Medical School, Kangwon National University, Chunchon, 24341, Republic of Korea
| | - Ji Hoon Jeong
- Department of Global Innovative Drugs, Graduate School of Chung-Ang University, College of Medicine, Chung-Ang University, Seoul, 06974, Republic of Korea.
| | - Hyoung-Chun Kim
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon, 24341, Republic of Korea.
| |
Collapse
|
5
|
Nguyen BT, Shin EJ, Jeong JH, Sharma N, Tran NKC, Nguyen YND, Kim DJ, Wie MB, Lee Y, Byun JK, Ko SK, Nah SY, Kim HC. Mountain-cultivated ginseng protects against cognitive impairments in aged GPx-1 knockout mice via activation of Nrf2/ChAT/ERK signaling pathway. J Ginseng Res 2023. [DOI: 10.1016/j.jgr.2023.01.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
|
6
|
Hu J, Tian J, Yuan T, Yin Q, Yin J. The critical role of nanoparticle sizes in the interactions between gold nanoparticles and ABC transporters in zebrafish embryos. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2022; 251:106286. [PMID: 36084499 DOI: 10.1016/j.aquatox.2022.106286] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 07/18/2022] [Accepted: 08/30/2022] [Indexed: 06/15/2023]
Abstract
Despite the increasing evidences for adenosine triphosphate-binding cassette (ABC transporters)-mediated efflux of nanoparticles, the universality of these phenomena and the determining factors for the process remained to be clarified. This paper aimed to systemically investigate the role of nanoparticle size in the interactions between adenosine triphosphate-binding cassette (ABC transporters) and gold nanoparticles (AuNPs, 3 nm, 19 nm, and 84 nm, named as Au-3, Au-19, and Au-84) in zebrafish embryos. The results showed that all the three AuNPs induced significant toxicity as reflected by delayed hatching of embryos, decreased glutathione (GSH) contents, and increased reactive oxygen species (ROS) levels. Under the hindrance of embryo chorions, smaller AuNPs could more easily accumulate in the embryos, causing higher toxicity. Addition of transporter inhibitors enhanced the accumulation and toxicity of Au-3 and Au-19, and these nanoparticles induced the expressions of abcc2 and abcb4, indicating a fact that Au-3 and Au-19 were the potential substrates of ABC transporters, but these phenomena were barely found for Au-84. On the contrary, Au-84 suppressed the gene expressions of various ABC transporters like abcc1, abcg5, and abcg8. With specific suppressors, transcription factors like nuclear factor-erythroid 2-related factor-2 (Nrf2) and pregnane X receptor (Pxr) were found to be important in the induction of ABC transporters by AuNPs. After all, these results revealed a vital role of nanoparticle sizes in the interactions between ABC transporters and AuNPs in zebrafish embryos, and the critical size could be around 19 nm. Such information would be beneficial in assessing the environmental risk of nanoparticles, as well as their interactions with other chemical toxicants.
Collapse
Affiliation(s)
- Jia Hu
- School of Biology & Basic Medical Sciences, Medical College, Soochow University, Suzhou, Jiangsu 215123, China
| | - Jingjing Tian
- CAS Key Lab of Bio-Medical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, Jiangsu 215163, China; Jinan Guo Ke Medical Technology Development Co. Ltd., Jinan, China
| | - Tongkuo Yuan
- CAS Key Lab of Bio-Medical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, Jiangsu 215163, China; Jinan Guo Ke Medical Technology Development Co. Ltd., Jinan, China
| | - Qingqing Yin
- Department of Geriatric Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China
| | - Jian Yin
- CAS Key Lab of Bio-Medical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, Jiangsu 215163, China; Jinan Guo Ke Medical Technology Development Co. Ltd., Jinan, China.
| |
Collapse
|
7
|
Hwang Y, Kim HC, Shin EJ. BKM120 alters the migration of doublecortin-positive cells in the dentate gyrus of mice. Pharmacol Res 2022; 179:106226. [PMID: 35460881 DOI: 10.1016/j.phrs.2022.106226] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 03/28/2022] [Accepted: 04/15/2022] [Indexed: 11/16/2022]
Abstract
BKM120 is an inhibitor of class I phosphoinositide 3-kinases and its anti-cancer effects have been demonstrated in various solid cancer models. BKM120 is highly brain permeable and has been reported to induce mood disturbances in clinical trials. Therefore, we examined whether BKM120 produces anxiety- and depression-like behaviors in mice, as with patients receiving BKM120 in clinical trials. In this study, repeated BKM120 treatment (2.0 or 5.0mg/kg, i.p., five times at 12-h interval) significantly induced anxiety- and depression-like behaviors in mice. Although abnormal changes in hippocampal neurogenesis have been suggested to, at least in part, associated with the pathogenesis of depression and anxiety, BKM120 did not affect the incorporation of 5-bromo-2'-deoxyuridine or the expression of doublecortin (DCX); however, it significantly enhanced the radial migration of DCX-positive cells in the dentate gyrus. BKM120-induced changes in migration were not accompanied by obvious neuronal damage in the hippocampus. Importantly, BKM120-induced anxiety- and depression-like behaviors were positively correlated with the extent of DCX-positive cell migration. Concomitantly, p-Akt expression was significantly decreased in the dentate gyrus. Moreover, the expression of p-c-Jun N-terminal kinase (JNK), p-DCX, and Ras homolog family member A (RhoA)-GTP decreased significantly, particularly in aberrantly migrated DCX-positive cells. Together, the results suggest that repeated BKM120 treatment enhances the radial migration of DCX-positive cells and induces anxiety- and depression-like behaviors by regulating the activity of Akt, JNK, DCX, and RhoA in the dentate gyrus. It also suggests that the altered migration of adult-born neurons in the dentate gyrus plays a role in mood disturbances.
Collapse
Affiliation(s)
- Yeonggwang Hwang
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Hyoung-Chun Kim
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chuncheon 24341, Republic of Korea.
| | - Eun-Joo Shin
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chuncheon 24341, Republic of Korea.
| |
Collapse
|
8
|
Shin EJ, Nguyen BT, Jeong JH, Hoai Nguyen BC, Tran NKC, Sharma N, Kim DJ, Nah SY, Lichtstein D, Nabeshima T, Kim HC. Ouabain inhibitor rostafuroxin attenuates dextromethorphan-induced manic potential. Food Chem Toxicol 2021; 158:112657. [PMID: 34740715 DOI: 10.1016/j.fct.2021.112657] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 10/29/2021] [Accepted: 10/31/2021] [Indexed: 02/08/2023]
Abstract
Dextromethorphan (DM) abuse produces mania-like symptoms in humans. ERK/Akt signaling activation involved in manic potential can be attenuated by the inhibition of ouabain-like cardiac steroids. In this study, increased phosphorylations of ERK/Akt and hyperlocomotion induced by DM (30 mg/kg, i.p./day × 7) were significantly protected by the ouabain inhibitor rostafuroxin (ROSTA), suggesting that DM induces the manic potential. ROSTA significantly attenuated DM-induced protein kinase C δ (PKCδ) phosphorylation, GluN2B (i.e., MDA receptor subunit) expression, and phospho-PKCδ/GluN2B interaction. DM instantly upregulated the nuclear factor erythroid-2-related factor 2 (Nrf2)-dependent system. However, DM reduced Nrf2 nuclear translocation, Nrf2 DNA binding activity, γ-glutamylcysteine mRNA expression, and subsequent GSH/GSSG level and enhanced oxidative parameters following 1-h of administration. ROSTA, PKCδ inhibitor rottlerin, and GluN2B inhibitor traxoprodil significantly attenuated DM-induced alterations in Nrf2-related redox parameters and locomotor activity induced by DM in wild-type mice. Importantly, in PKCδ knockout mice, DM failed to alter the above parameters. Further, ROSTA and traxoprodil also failed to enhance PKCδ depletion effect, suggesting that PKCδ is a critical target for the anti-manic potential of ROSTA or GluN2B antagonism. Our results suggest that ROSTA inhibits DM-induced manic potential by attenuating ERK/Akt activation, GluN2B/PKCδ signalings, and Nrf2-dependent system.
Collapse
Affiliation(s)
- Eun-Joo Shin
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon, 24341, Republic of Korea
| | - Bao-Trong Nguyen
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon, 24341, Republic of Korea
| | - Ji Hoon Jeong
- Department of Global Innovative Drugs, Graduate School of Chung-Ang University, College of Medicine, Chung-Ang University, Seoul, 06974, Republic of Korea.
| | - Bao-Chau Hoai Nguyen
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon, 24341, Republic of Korea
| | - Ngoc Kim Cuong Tran
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon, 24341, Republic of Korea
| | - Naveen Sharma
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon, 24341, Republic of Korea; Department of Global Innovative Drugs, Graduate School of Chung-Ang University, College of Medicine, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Dae-Joong Kim
- Department of Anatomy and Cell Biology, Medical School, Kangwon National University, Chunchon, 24341, Republic of Korea
| | - Seung-Yeol Nah
- Ginsentology Research Laboratory and Department of Physiology, College of Veterinary Medicine and Bio/Molecular Informatics Center, Konkuk University, Seoul, 05029, Republic of Korea
| | - David Lichtstein
- Walter and Greta Stiel Chair in Heart Studies, Dean, Faculty of Medicine 2013-2017, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Toshitaka Nabeshima
- Advanced Diagnostic System Research Laboratory, Fujita Health University Graduate School of Health Science, Toyoake, 470-1192, Japan
| | - Hyoung-Chun Kim
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon, 24341, Republic of Korea.
| |
Collapse
|
9
|
Wang X, Liu J, Dai Z, Sui Y. Andrographolide improves PCP-induced schizophrenia-like behaviors through blocking interaction between NRF2 and KEAP1. J Pharmacol Sci 2021; 147:9-17. [PMID: 34294378 DOI: 10.1016/j.jphs.2021.05.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Revised: 05/02/2021] [Accepted: 05/17/2021] [Indexed: 12/15/2022] Open
Abstract
Schizophrenia is one of the foremost psychological illness around the world, and recent evidence shows that inflammation and oxidative stress may play a critical role in the etiology of schizophrenia. Andrographolide is a diterpenoid lactone from Andrographis paniculate, which has shown anti-inflammation and anti-oxidative effects. In this study, we explored whether andrographolide can improve schizophrenia-like behaviors through its inhibition of inflammation and oxidative stress in Phencyclidine (PCP)-induced mouse model of schizophrenia. We found that abnormal behavioral including locomotor activity, forced swimming and novel object recognition were ameliorated following andrographolide administration (5 mg/kg and 10 mg/kg). Andrographolide inhibited PCP-induced production of inflammatory cytokines, decreased p-p65, p-IκBα, p-p38 and p-ERK1/2 in the prefrontal cortex. Andrographolide significantly declined the level of MDA and GSH, as well as elevated the activity of SOD, CAT and GCH-px. In addition, andrographolide increased expression of NRF-2, HO-1 and NQO-1, promoted nuclear translocation of NRF-2 through blocking the interaction between NRF-2 and KEAP1, which may be associated with directly binding to NRF-2. Furthermore, antioxidative effects and anti-schizophrenia-like behaviors of andrographolide were compromised by the application of NRF-2 inhibitor ML385. In conclusion, these results suggested that andrographolide improved oxidative stress and schizophrenia-like behaviors induced by PCP through increasing NRF-2 pathway.
Collapse
Affiliation(s)
- Xiying Wang
- Department of Psychiatry, The Affiliated Nanjing Brain Hospital of Nanjing Medical University, Nanjing, China.
| | - Jia Liu
- Department of Clinical Pharmacy, The Affiliated Nanjing Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Zhiping Dai
- Department of Psychiatry, The Affiliated Nanjing Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Yuxiu Sui
- Department of Psychiatry, The Affiliated Nanjing Brain Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
10
|
Sharma N, Shin EJ, Pham DT, Sharma G, Dang DK, Duong CX, Kang SW, Nah SY, Jang CG, Lei XG, Nabeshima T, Bing G, Jeong JH, Kim HC. GPx-1-encoded adenoviral vector attenuates dopaminergic impairments induced by methamphetamine in GPx-1 knockout mice through modulation of NF-κB transcription factor. Food Chem Toxicol 2021; 154:112313. [PMID: 34082047 DOI: 10.1016/j.fct.2021.112313] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 05/24/2021] [Accepted: 05/27/2021] [Indexed: 01/10/2023]
Abstract
We suggested that selenium-dependent glutathione peroxidase (GPx) plays a protective role against methamphetamine (MA)-induced dopaminergic toxicity. We focused on GPx-1, a major selenium-dependent enzyme and constructed a GPx-1 gene-encoded adenoviral vector (Ad-GPx-1) to delineate the role of GPx-1 in MA-induced dopaminergic neurotoxicity. Exposure to Ad-GPx-1 significantly induced GPx activity and GPx-1 protein levels in GPx-1-knockout (GPx-1-KO) mice. MA-induced dopaminergic impairments [i.e., hyperthermia; increased nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) DNA-binding activity; and decreased dopamine levels, TH activity, and behavioral activity] were more pronounced in GPx-1-KO mice than in WT mice. In contrast, exposure to Ad-GPx-1 significantly attenuated MA-induced dopaminergic loss in GPx-1-KO mice. The protective effect exerted by Ad-GPx-1 was comparable to that exerted by pyrrolidine dithiocarbamate (PDTC), an NF-κB inhibitor against MA insult. Consistently, GPx-1 overexpression significantly attenuated MA dopaminergic toxicity in mice. PDTC did not significantly impact the protective effect of GPx-1 overexpression, suggesting that interaction between NF-κB and GPx-1 is critical for dopaminergic protection. Thus, NF-κB is a potential therapeutic target for GPx-1-mediated dopaminergic protective activity. This study for the first time demonstrated that Ad-GPx-1 rescued dopaminergic toxicity in vivo following MA insult. Furthermore, GPx-1-associated therapeutic interventions may be important against dopaminergic toxicity.
Collapse
Affiliation(s)
- Naveen Sharma
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon, 24341, South Korea; Department of Global Innovative Drugs, Graduate School of Chung-Ang University, College of Medicine, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Eun-Joo Shin
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon, 24341, South Korea
| | - Duc Toan Pham
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon, 24341, South Korea
| | - Garima Sharma
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon, 24341, South Korea
| | - Duy-Khanh Dang
- Pharmacy Faculty, Can Tho University of Medicine and Pharmacy, Can Tho City, 900000, Viet Nam
| | - Chu Xuan Duong
- Pharmacy Faculty, Can Tho University of Medicine and Pharmacy, Can Tho City, 900000, Viet Nam
| | - Sang Won Kang
- Department of Life Science, College of Natural Science, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Seung-Yeol Nah
- Ginsentology Research Laboratory and Department of Physiology, College of Veterinary Medicine and Bio/Molecular Informatics Center, Konkuk University, Seoul, 05029, Republic of Korea
| | - Choon-Gon Jang
- Department of Pharmacology, School of Pharmacy, Sungkyunkwan University, Suwon, 440-746, Republic of Korea
| | - Xin Gen Lei
- Department of Animal Science, Cornell University, Ithaca, NY, 14853, USA
| | - Toshitaka Nabeshima
- Advanced Diagnostic System Research Laboratory, Fujita Health University Graduate School of Health Science, Toyoake, 470-1192, Japan
| | - Guoying Bing
- Anatomy and Neurobiology, University of Kentucky Medical Center, Medical Center MN208 800 Rose Strees, Lexington, KY, 40536, USA
| | - Ji Hoon Jeong
- Department of Global Innovative Drugs, Graduate School of Chung-Ang University, College of Medicine, Chung-Ang University, Seoul, 06974, Republic of Korea.
| | - Hyoung-Chun Kim
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon, 24341, South Korea.
| |
Collapse
|
11
|
Yuan T, Sun J, Tian J, Hu J, Yin H, Yin J. Involvement of ABC transporters in the detoxification of non-substrate nanoparticles in lung and cervical cancer cells. Toxicology 2021; 455:152762. [PMID: 33766574 DOI: 10.1016/j.tox.2021.152762] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 03/10/2021] [Accepted: 03/18/2021] [Indexed: 02/06/2023]
Abstract
This paper aimed to systemically investigate the role of adenosine triphosphate-binding cassette (ABC transporters) in the detoxification of non-substrate nanoparticles including titanium dioxide (n-TiO2, 5-10 nm) and gold (AuNPs, 3 nm, 15 nm, and 80 nm, named as Au-3, Au-15 and Au-80) in human lung cancer (A549) and human cervical cancer (HeLa) cells. All these nanoparticles were of larger hydrophilic diameters than the channel sizes of ABC transporters, thus should not be the substrates of membrane proteins. After 24-h treatment, they induced significant cytotoxicity as reflected by the reduction in cell viability and glutathione (GSH) contents, as well as the increase in reactive oxygen species (ROS) level. At median-lethal concentrations (10 mg/L n-TiO2, 2 mg/L Au-3, 5 mg/L Au-15, and 10 mg/L Au-80 for A549 cells; 20 mg/L n-TiO2, 2 mg/L Au-3, 5 mg/L Au-15, and 10 mg/L Au-80 for Hela cells), all the nanoparticles significantly induced the gene expressions and activities of ABC transporters including P-glycoprotein (PGP) and multidrug resistance associated protein 1 (MRP1). Addition of transporter inhibitors enhanced the ROS levels produced by nanoparticles, but didn't alter their death-inducing effects and intracellular accumulations. With specific suppressors, transcription factors like nuclear factor-erythroid 2-related factor-2 (NRF2) and pregnane X receptor (PXR) were proved to be important in the induction of ABC transporters by nanoparticles. After all, this paper revealed a damage-dependent modulation of ABC transporters by non-substrate nanoparticles. The up-regulated ABC transporters could help in reducing the oxidative stress produced by nanoparticles. Such information should be useful in assessing the environmental risk of nanoparticles, as well as their interactions with other chemical toxicants or drugs.
Collapse
Affiliation(s)
- Tongkuo Yuan
- CAS Key Lab of Bio-Medical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, Jiangsu 215163, PR China; Jinan Guo Ke Medical Technology Development Co., Ltd, PR China
| | - Jiaojiao Sun
- CAS Key Lab of Bio-Medical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, Jiangsu 215163, PR China; University of Science and Technology of China, Hefei, Anhui 230026, PR China
| | - Jingjing Tian
- CAS Key Lab of Bio-Medical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, Jiangsu 215163, PR China; Academy for Engineering & Technology, Fudan University, Shanghai 200433, PR China
| | - Jia Hu
- School of Biology & Basic Medical Sciences, Medical College, Soochow University, Suzhou, Jiangsu 215123, PR China
| | - Huancai Yin
- CAS Key Lab of Bio-Medical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, Jiangsu 215163, PR China; Jinan Guo Ke Medical Technology Development Co., Ltd, PR China; University of Science and Technology of China, Hefei, Anhui 230026, PR China.
| | - Jian Yin
- CAS Key Lab of Bio-Medical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, Jiangsu 215163, PR China; Jinan Guo Ke Medical Technology Development Co., Ltd, PR China; University of Science and Technology of China, Hefei, Anhui 230026, PR China.
| |
Collapse
|
12
|
Sharma G, Shin EJ, Sharma N, Nah SY, Mai HN, Nguyen BT, Jeong JH, Lei XG, Kim HC. Glutathione peroxidase-1 and neuromodulation: Novel potentials of an old enzyme. Food Chem Toxicol 2021; 148:111945. [PMID: 33359022 DOI: 10.1016/j.fct.2020.111945] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Revised: 12/15/2020] [Accepted: 12/18/2020] [Indexed: 12/14/2022]
Abstract
Glutathione peroxidase (GPx) acts in co-ordination with other signaling molecules to exert its own antioxidant role. We have demonstrated the protective effects of GPx,/GPx-1, a selenium-dependent enzyme, on various neurodegenerative disorders (i.e., Parkinson's disease, Alzheimer's disease, cerebral ischemia, and convulsive disorders). In addition, we summarized the recent findings indicating that GPx-1 might play a role as a neuromodulator in neuropsychiatric conditions, such as, stress, bipolar disorder, schizophrenia, and drug intoxication. In this review, we attempted to highlight the mechanistic scenarios mediated by the GPx/GPx-1 gene in impacting these neurodegenerative and neuropsychiatric disorders, and hope to provide new insights on the therapeutic interventions against these disorders.
Collapse
Affiliation(s)
- Garima Sharma
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon, 24341, Republic of Korea
| | - Eun-Joo Shin
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon, 24341, Republic of Korea
| | - Naveen Sharma
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon, 24341, Republic of Korea; Department of Global Innovative Drugs, Graduate School of Chung-Ang University, College of Medicine, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Seung-Yeol Nah
- Ginsentology Research Laboratory and Department of Physiology, College of Veterinary Medicine and Bio/Molecular Informatics Center, Konkuk University, Seoul, 05029, Republic of Korea
| | - Huynh Nhu Mai
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon, 24341, Republic of Korea; Pharmacy Faculty, Can Tho University of Medicine and Pharmacy, Can Tho City, 900000, Viet Nam
| | - Bao Trong Nguyen
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon, 24341, Republic of Korea
| | - Ji Hoon Jeong
- Department of Global Innovative Drugs, Graduate School of Chung-Ang University, College of Medicine, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Xin Gen Lei
- Department of Animal Science, Cornell University, Ithaca, NY, 14853, USA
| | - Hyoung-Chun Kim
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon, 24341, Republic of Korea.
| |
Collapse
|
13
|
Shin EJ, Lee SH, Sharma N, Nguyen BT, Chung YH, Kang SW, Nah SY, Lee YJ, Nabeshima T, Jeong JH, Kim HC. An adenoviral vector encoded with the GPx-1 gene attenuates memory impairments induced by β-amyloid (1-42) in GPx-1 KO mice via activation of M1 mAChR-mediated signalling. Free Radic Res 2020; 55:11-25. [PMID: 33222572 DOI: 10.1080/10715762.2020.1854455] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
In the present study, we examined whether glutathione peroxidase-1 (GPx-1), a major H2O2 scavenger in the brain, affects memory deficits induced by Aβ (1-42) in mice. Treatment with 400 pmol/5 μl Aβ (1-42) (i.c.v.) resulted in a reduction of GPx-1 expression in wild-type (WT) mice. An Aβ (1-42)-induced reduction in acetylcholine (ACh) level was observed in the hippocampus. Treatment with Aβ (1-42) consistently resulted in reduced expression and activity of choline acetyltransferase (ChAT) and in an increase in expression and activity of acetylcholinesterase (AChE). Upon examining each of the muscarinic acetylcholine receptors (mAChRs) and nicotinic AChRs, we noted that Aβ (1-42) treatment selectively reduced the levels of M1 mAChR. In addition, Aβ (1-42) induced a significant reduction in phospho-cAMP response element-binding protein (p-CREB) and brain-derived neurotrophic factor (BDNF) expression. The cholinergic impairments induced by Aβ (1-42) were more pronounced in GPx-1 knockout mice than in WT mice. Importantly, an adenoviral vector encoded with the GPx-1 gene (Ad-GPx-1) significantly rescued Aβ (1-42)-induced cholinergic impairments in GPx-1 knockout mice. In addition, M1 mAChR antagonist dicyclomine significantly counteracted Ad-GPx-1-mediated increases in p-CREB and BDNF expression, as well as memory-enhancing effects in GPx-1 knockout mice, thus indicating that M1 mAChR might be a critical mediator for the rescue effects of Ad-GPx-1. Combined, our results suggest that GPx-1 gene protected against Aβ (1-42)-induced memory impairments via activation of M1 mAChR-dependent CREB/BDNF signalling.
Collapse
Affiliation(s)
- Eun-Joo Shin
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon, Republic of Korea
| | - Sung Hoon Lee
- Department of Pharmacology, College of Pharmacy, Chung-Ang University, Seoul, Republic of Korea
| | - Naveen Sharma
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon, Republic of Korea
| | - Bao Trong Nguyen
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon, Republic of Korea
| | - Yoon Hee Chung
- Department of Anatomy, College of Medicine, Chung-Ang University, Seoul, Korea
| | - Sang Won Kang
- Department of Life Science, College of Natural science, Ewha Womans University, Seoul, Republic of Korea
| | - Seung-Yeol Nah
- Ginsentology Research Laboratory and Department of Physiology, College of Veterinary Medicine and Bio/Molecular Informatics Center, Konkuk University, Seoul, Republic of Korea
| | - Yu Jeung Lee
- Clinical Pharmacy, College of Pharmacy, Kangwon National University, Chunchon, Republic of Korea
| | - Toshitaka Nabeshima
- Advanced Diagnostic System Research Laboratory, Fujita Health University Graduate School of Health Sciences, Aichi, Japan
| | - Ji Hoon Jeong
- Department of Global Innovative Drugs, Graduate School of Chung-Ang University, College of Medicine, Chung-Ang University, Seoul, Republic of Korea
| | - Hyoung-Chun Kim
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon, Republic of Korea
| |
Collapse
|
14
|
Mai HN, Pham DT, Chung YH, Sharma N, Cheong JH, Yun J, Nah SY, Jeong JH, Gen Lei X, Shin EJ, Nabeshima T, Kim HC. Glutathione peroxidase-1 knockout potentiates behavioral sensitization induced by cocaine in mice via σ-1 receptor-mediated ERK signaling: A comparison with the case of glutathione peroxidase-1 overexpressing transgenic mice. Brain Res Bull 2020; 164:107-120. [PMID: 32822804 DOI: 10.1016/j.brainresbull.2020.08.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 08/01/2020] [Accepted: 08/11/2020] [Indexed: 02/07/2023]
Abstract
We demonstrated that the gene of glutathione peroxidase-1 (GPx-1), a major antioxidant enzyme, is a potential protectant against the neurotoxicity and conditioned place preference induced by cocaine. Because the sigma (σ)-1 receptor is implicated in cocaine-induced drug dependence, we investigated whether the GPx-1 gene modulates the σ-1 receptor in the behavioral sensitization induced by cocaine. Cocaine-induced behavioral sensitization was more pronounced in GPx-1 knockout (KO) than wild-type (WT) mice and was less pronounced in GPx-1 overexpressing transgenic (GPx-1 TG) than non-TG mice. Cocaine treatment significantly enhanced the oxidative burden and reduced the GSH levels in the striatum of WT, GPx-1 KO, and non-TG mice but not in that of GPx-1 TG mice. In addition, cocaine significantly increased the nuclear translocation, its DNA binding activity of nuclear factor erythroid-2-related factor 2 (Nrf2) as well as the mRNA expression of γ-glutamylcysteine (GCL). The genetic depletion of GPx-1 inhibited the Nrf2-related glutathione system, whereas the genetic overexpression of GPx-1 activated this system against behavioral sensitization. BD1047, a σ-1 receptor antagonist, and U0126, an ERK inhibitor significantly induced the Nrf2-related antioxidant potential against behavioral sensitization. Unlike BD1047, U0126 did not affect the cocaine-induced σ-1 receptor immunoreactivity, suggesting that the σ-1 receptor is an upstream molecule for ERK signaling. Importantly, BD1047 and U0126 failed to affect the σ-1 receptor immunoreactivity and ERK phosphorylation induced by cocaine in GPx-1 TG mice. Our results suggest that GPx-1 is a critical mediator for the attenuation of cocaine-induced behavioral sensitization via modulating σ-1 receptor-mediated ERK activation by the induction of the Nrf2-related system.
Collapse
Affiliation(s)
- Huynh Nhu Mai
- Neuropsychopharmacology and Toxicology Program, BK21 PLUS Project, College of Pharmacy, Kangwon National University, Chunchon, 24341, Republic of Korea; Pharmacy Faculty, Can Tho University of Medicine and Pharmacy, Can Tho City, 900000, Viet Nam
| | - Duc Toan Pham
- Neuropsychopharmacology and Toxicology Program, BK21 PLUS Project, College of Pharmacy, Kangwon National University, Chunchon, 24341, Republic of Korea
| | - Yoon Hee Chung
- Department of Anatomy, College of Medicine, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Naveen Sharma
- Neuropsychopharmacology and Toxicology Program, BK21 PLUS Project, College of Pharmacy, Kangwon National University, Chunchon, 24341, Republic of Korea
| | - Jae Hoon Cheong
- Department of Pharmacy, Sahmyook University, Seoul, 01795, Republic of Korea
| | - Jaesuk Yun
- College of Pharmacy, Chungbuk National University, Chungbuk, 28160, Republic of Korea
| | - Seung-Yeol Nah
- Ginsentology Research Laboratory and Department of Physiology, College of Veterinary Medicine and Bio/Molecular Informatics Center, Konkuk University, Seoul, 05029, Republic of Korea
| | - Ji Hoon Jeong
- Department of Pharmacology, College of Medicine, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Xin Gen Lei
- Department of Animal Science, Cornell University, Ithaca, NY 14853, United States
| | - Eun-Joo Shin
- Neuropsychopharmacology and Toxicology Program, BK21 PLUS Project, College of Pharmacy, Kangwon National University, Chunchon, 24341, Republic of Korea.
| | - Toshitaka Nabeshima
- Advanced Diagnostic System Research Laboratory, Fujita Health University Graduate School of Health Science, Aichi, 470-1192, Japan
| | - Hyoung-Chun Kim
- Neuropsychopharmacology and Toxicology Program, BK21 PLUS Project, College of Pharmacy, Kangwon National University, Chunchon, 24341, Republic of Korea.
| |
Collapse
|
15
|
Shin EJ, Chung YH, Sharma N, Nguyen BT, Lee SH, Kang SW, Nah SY, Wie MB, Nabeshima T, Jeong JH, Kim HC. Glutathione Peroxidase-1 Knockout Facilitates Memory Impairment Induced by β-Amyloid (1-42) in Mice via Inhibition of PKC βII-Mediated ERK Signaling; Application with Glutathione Peroxidase-1 Gene-Encoded Adenovirus Vector. Neurochem Res 2020; 45:2991-3002. [PMID: 33064252 DOI: 10.1007/s11064-020-03147-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 09/16/2020] [Accepted: 10/07/2020] [Indexed: 12/20/2022]
Abstract
A growing body evidence suggests that selenium (Se) deficiency is associated with an increased risk of developing Alzheimer's disease (AD). Se-dependent glutathione peroxidase-1 (GPx-1) of a major antioxidant enzyme, and the most abundant isoform of GPx in the brain. In the present study, we investigated whether GPx-1 is protective against memory impairments induced by beta-amyloid (Aβ) (1-42) in mice. As the alteration of protein kinase C (PKC)-mediated ERK activation was recognized in the early stage of AD, we examined whether the GPx-1 gene modulates Aβ (1-42)-induced changes in PKC and ERK levels. We observed that Aβ (1-42) treatment (400 pmol, i.c.v.) significantly decreased PKC βII expression in the hippocampus of mice. Aβ (1-42)-induced neurotoxic changes [i.e., oxidative stress (i.e., reactive oxygen species, 4-hydroxy-2-noneal, and protein carbonyl), reduced PKC βII and phospho-ERK expressions, and memory impairment under Y-maze and passive avoidance test] were more pronounced in GPx-1 knockout than in wild type mice. Importantly, exposure to a GPx-1 gene-encoded adenovirus vector (Adv-GPx-1) significantly increased GPx-1 mRNA and GPx activity in the hippocampus of GPx-1 knockout mice. Adv-GPx-1 exposure also significantly blocked the neurotoxic changes induced by Aβ (1-42) in GPx-1 knockout mice. Treatment with ERK inhibitor U0126 did not significantly change Adv-GPx-1-mediated attenuation in PKC βII expression. In contrast, treatment with PKC inhibitor chelerythrine (CHE) reversed Adv-GPx-1-mediated attenuation in ERK phosphorylation, suggesting that PKC βII-mediated ERK signaling is important for Adv-GPx-1-mediated potentials against Aβ (1-42) insult. Our results suggest that treatment with the antioxidant gene GPx-1 rescues Aβ (1-42)-induced memory impairment via activating PKC βII-mediated ERK signaling.
Collapse
Affiliation(s)
- Eun-Joo Shin
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon, 24341, Republic of Korea
| | - Yoon Hee Chung
- Department of Anatomy, College of Medicine, Chung-Ang University, Seoul, Seoul, 06974, Republic of Korea
| | - Naveen Sharma
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon, 24341, Republic of Korea.,Department of Global Innovative Drugs, Graduate School of Chung-Ang University, College of Medicine, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Bao Trong Nguyen
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon, 24341, Republic of Korea
| | - Sung Hoon Lee
- Department of Pharmacology, College of Pharmacy, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Sang Won Kang
- Department of Life Science, College of Natural Science, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Seung-Yeol Nah
- Ginsentology Research Laboratory and Department of Physiology, College of Veterinary Medicine and Bio/Molecular Informatics Center, Konkuk University, Seoul, 05029, Republic of Korea
| | - Myung Bok Wie
- Department of Veterinary Toxicology, College of Veterinary Medicine and Institute of Veterinary Science, Kangwon National University, Chuncheon, 24341, Korea
| | - Toshitaka Nabeshima
- Advanced Diagnostic System Research Laboratory, Fujita Health University Graduate School of Health Sciences, Aichi, 470-1192, Japan
| | - Ji Hoon Jeong
- Department of Global Innovative Drugs, Graduate School of Chung-Ang University, College of Medicine, Chung-Ang University, Seoul, 06974, Republic of Korea.
| | - Hyoung-Chun Kim
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon, 24341, Republic of Korea.
| |
Collapse
|
16
|
Oxidation-reduction mechanisms in psychiatric disorders: A novel target for pharmacological intervention. Pharmacol Ther 2020; 210:107520. [PMID: 32165136 DOI: 10.1016/j.pharmthera.2020.107520] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2019] [Accepted: 03/02/2020] [Indexed: 12/16/2022]
Abstract
While neurotransmitter dysfunction represents a key component in mental illnesses, there is now a wide agreement for a central pathophysiological hub that includes hormones, neuroinflammation, redox mechanisms as well as oxidative stress. With respect to oxidation-reduction (redox) mechanisms, preclinical and clinical evidence suggests that an imbalance in the pro/anti-oxidative homeostasis toward the increased production of substances with oxidizing potential may contribute to the etiology and manifestation of different psychiatric disorders. The substantial and continous demand for energy renders the brain highly susceptible to disturbances in its energy supply, especially following exposure to stressful events, which may lead to overproduction of reactive oxygen and nitrogen species under conditions of perturbed antioxidant defenses. This will eventually induce different molecular alterations, including extensive protein and lipid peroxidation, increased blood-brain barrier permeability and neuroinflammation, which may contribute to the changes in brain function and morphology observed in mental illnesses. This view may also reconcile different key concepts for psychiatric disorders, such as the neurodevelopmental origin of these diseases, as well as the vulnerability of selective cellular populations that are critical for specific functional abnormalities. The possibility to pharmacologically modulate the redox system is receiving increasing interest as a novel therapeutic strategy to counteract the detrimental effects of the unbalance in brain oxidative mechanisms. This review will describe the main mechanisms and mediators of the redox system and will examine the alterations of oxidative stress found in animal models of psychiatric disorders as well as in patients suffering from mental illnesses, such as schizophrenia and major depressive disorder. In addition, it will discuss studies that examined the effects of psychotropic drugs, including antipsychotics and antidepressants, on the oxidative balance as well as studies that investigated the effectiveness of a direct modulation of oxidative mechanisms in counteracting the behavioral and functional alterations associated with psychiatric disorders, which supports the promising role of the redox system as a novel therapeutic target for the improved treatment of brain disorders.
Collapse
|
17
|
Sharma N, Shin EJ, Kim NH, Cho EH, Nguyen BT, Jeong JH, Jang CG, Nah SY, Kim HC. Far-infrared Ray-mediated Antioxidant Potentials are Important for Attenuating Psychotoxic Disorders. Curr Neuropharmacol 2020; 17:990-1002. [PMID: 30819085 PMCID: PMC7052827 DOI: 10.2174/1570159x17666190228114318] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Revised: 02/02/2019] [Accepted: 02/14/2019] [Indexed: 12/14/2022] Open
Abstract
Far-infrared ray (FIR) is an electromagnetic wave that produces various health benefits against pathophysiological conditions, such as diabetes mellitus, renocardiovascular disorders, stress, and depression etc. However, the therapeutic ap-plication on the FIR-mediated protective potentials remains to be further extended. To achieve better understanding on FIR-mediated therapeutic potentials, we summarized additional findings in the present study that exposure to FIR ameliorates stressful condition, memory impairments, drug dependence, and mitochondrial dysfunction in the central nervous system. In this review, we underlined that FIR requires modulations of janus kinase 2 / signal transducer and activator of transcription 3 (JAK2/STAT3), nuclear factor E2-related factor 2 (Nrf-2), muscarinic M1 acetylcholine receptor (M1 mAChR), dopamine D1 receptor, protein kinase C δ gene, and glutathione peroxidase-1 gene for exerting the protective potentials in response to neuropsychotoxic conditions
Collapse
Affiliation(s)
- Naveen Sharma
- Neuropsychopharmacology and Toxicology Program, BK21 PLUS Project, College of Pharmacy, Kangwon National University, Chunchon 24341, Korea
| | - Eun-Joo Shin
- Neuropsychopharmacology and Toxicology Program, BK21 PLUS Project, College of Pharmacy, Kangwon National University, Chunchon 24341, Korea
| | - Nam Hun Kim
- College of Forest and Environmental Sciences, Kangwon National University, Chunchon 24341, Korea
| | - Eun-Hee Cho
- Department of Internal Medicine, Medical School, Kangwon National University, Chunchon 24341, Korea
| | - Bao Trong Nguyen
- Neuropsychopharmacology and Toxicology Program, BK21 PLUS Project, College of Pharmacy, Kangwon National University, Chunchon 24341, Korea
| | - Ji Hoon Jeong
- Department of Pharmacology, College of Medicine, Chung-Ang University, Seoul 06974, Korea
| | - Choon Gon Jang
- Department of Pharmacology, School of Pharmacy, Sungkyunkwan University Suwon 16419, Korea
| | - Seung-Yeol Nah
- Ginsentology Research Laboratory and Department of Physiology, College of Veterinary Medicine and Bio/Molecular Informatics Center, Konkuk University, Seoul, Korea
| | - Hyoung-Chun Kim
- Neuropsychopharmacology and Toxicology Program, BK21 PLUS Project, College of Pharmacy, Kangwon National University, Chunchon 24341, Korea
| |
Collapse
|
18
|
Tran HQ, Shin EJ, Saito K, Tran TV, Phan DH, Sharma N, Kim DW, Choi SY, Jeong JH, Jang CG, Cheong JH, Nabeshima T, Kim HC. Indoleamine-2,3-dioxygenase-1 is a molecular target for the protective activity of mood stabilizers against mania-like behavior induced by d-amphetamine. Food Chem Toxicol 2019; 136:110986. [PMID: 31760073 DOI: 10.1016/j.fct.2019.110986] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 11/17/2019] [Accepted: 11/18/2019] [Indexed: 01/07/2023]
Abstract
It is recognized that d-amphetamine (AMPH)-induced hyperactivity is thought to be a valid animal model of mania. In the present study, we investigated whether a proinflammatory oxidative gene indoleamine-2,3-dioxygenase (IDO) is involved in AMPH-induced mitochondrial burden, and whether mood stabilizers (i.e., lithium and valproate) modulate IDO to protect against AMPH-induced mania-like behaviors. AMPH-induced IDO-1 expression was significantly greater than IDO-2 expression in the prefrontal cortex of wild type mice. IDO-1 expression was more pronounced in the mitochondria than in the cytosol. AMPH treatment activated intra-mitochondrial Ca2+ accumulation and mitochondrial oxidative burden, while inhibited mitochondrial membrane potential and activity of the mitochondrial complex (I > II), mitochondrial glutathione peroxidase, and superoxide dismutase, indicating that mitochondrial burden might be contributable to mania-like behaviors induced by AMPH. The behaviors were significantly attenuated by lithium, valproate, or IDO-1 knockout, but not in IDO-2 knockout mice. Lithium, valproate administration, or IDO-1 knockout significantly attenuated mitochondrial burden. Neither lithium nor valproate produced additive effects above the protective effects observed in IDO-1 KO in mice. Collectively, our results suggest that mood stabilizers attenuate AMPH-induced mania-like behaviors via attenuation of IDO-1-dependent mitochondrial stress, highlighting IDO-1 as a novel molecular target for the protective potential of mood stabilizers.
Collapse
Affiliation(s)
- Hai-Quyen Tran
- Neuropsychopharmacology and Toxicology Program, BK21 PLUS Project, College of Pharmacy, Kangwon National University, Chunchon, 24341, Republic of Korea
| | - Eun-Joo Shin
- Neuropsychopharmacology and Toxicology Program, BK21 PLUS Project, College of Pharmacy, Kangwon National University, Chunchon, 24341, Republic of Korea
| | - Kuniaki Saito
- Advanced Diagnostic System Research Laboratory, Fujita Health University Graduate School of Health Sciences, Toyoake, Japan.
| | - The-Vinh Tran
- Neuropsychopharmacology and Toxicology Program, BK21 PLUS Project, College of Pharmacy, Kangwon National University, Chunchon, 24341, Republic of Korea
| | - Dieu-Hien Phan
- Neuropsychopharmacology and Toxicology Program, BK21 PLUS Project, College of Pharmacy, Kangwon National University, Chunchon, 24341, Republic of Korea
| | - Naveen Sharma
- Neuropsychopharmacology and Toxicology Program, BK21 PLUS Project, College of Pharmacy, Kangwon National University, Chunchon, 24341, Republic of Korea
| | - Dae-Won Kim
- Department of Biochemistry and Molecular Biology, Research Institute of Oral Sciences, College of Dentistry, Gangneung-Wonju National University, Gangneung, 25457, South Korea
| | - Soo Young Choi
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chunchon, 24252, Republic of Korea
| | - Ji Hoon Jeong
- Department of Pharmacology, College of Medicine, Chung-Ang University, Seoul, Republic of Korea
| | - Choon-Gon Jang
- Department of Pharmacology, School of Pharmacy, Sungkyunkwan University, Suwon, South Korea
| | - Jae Hoon Cheong
- Department of Pharmacy, Sahmyook University, 815 Hwarangro, Nowon-gu, Seoul, 01795, Republic of Korea
| | - Toshitaka Nabeshima
- Advanced Diagnostic System Research Laboratory, Fujita Health University Graduate School of Health Sciences, Toyoake, Japan; Japanese Drug Organization of Appropriate Use and Research, Nagoya, Japan
| | - Hyoung-Chun Kim
- Neuropsychopharmacology and Toxicology Program, BK21 PLUS Project, College of Pharmacy, Kangwon National University, Chunchon, 24341, Republic of Korea.
| |
Collapse
|
19
|
Shin MG, Cha HN, Park S, Kim YW, Kim JY, Park SY. Selenoprotein W deficiency does not affect oxidative stress and insulin sensitivity in the skeletal muscle of high-fat diet-fed obese mice. Am J Physiol Cell Physiol 2019; 317:C1172-C1182. [PMID: 31509445 DOI: 10.1152/ajpcell.00064.2019] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Selenoprotein W (SelW) is a selenium-containing protein with a redox motif found abundantly in the skeletal muscle of rodents. Previous in vitro studies suggest that SelW plays an antioxidant role; however, relatively few in vivo studies have addressed the antioxidant role of SelW. Since oxidative stress is a causative factor for the development of insulin resistance in obese subjects, we hypothesized that if SelW plays a role as an antioxidant, SelW deficiency could aggravate the oxidative stress and insulin resistance caused by a high-fat diet. SelW deficiency did not affect insulin sensitivity and H2O2 levels in the skeletal muscle of control diet-fed mice. SelW levels in the skeletal muscle were decreased by high-fat diet feeding for 12 wk. High-fat diet induced obesity and insulin resistance and increased the levels of H2O2 and oxidative stress makers, which were not affected by SelW deficiency. High-fat diet feeding increased the expression of antioxidant enzymes; however, SelW deficiency did not affect the expression levels of antioxidants. These results suggest that SelW does not play a protective role against oxidative stress and insulin resistance in the skeletal muscle of high-fat diet-fed obese mice.
Collapse
Affiliation(s)
- Min-Gyeong Shin
- Department of Physiology, College of Medicine, Yeungnam University, Daegu, Korea.,Smart-aging Convergence Research Center, College of Medicine, Yeungnam University, Daegu, Korea
| | - Hye-Na Cha
- Department of Physiology, College of Medicine, Yeungnam University, Daegu, Korea.,Smart-aging Convergence Research Center, College of Medicine, Yeungnam University, Daegu, Korea
| | - Soyoung Park
- Department of Physiology, College of Medicine, Yeungnam University, Daegu, Korea.,Smart-aging Convergence Research Center, College of Medicine, Yeungnam University, Daegu, Korea
| | - Yong-Woon Kim
- Department of Physiology, College of Medicine, Yeungnam University, Daegu, Korea
| | - Jong-Yeon Kim
- Department of Physiology, College of Medicine, Yeungnam University, Daegu, Korea
| | - So-Young Park
- Department of Physiology, College of Medicine, Yeungnam University, Daegu, Korea.,Smart-aging Convergence Research Center, College of Medicine, Yeungnam University, Daegu, Korea
| |
Collapse
|
20
|
Pham DT, Chung YH, Mai HN, Sharma N, Yun J, Kim HJ, Cheong JH, Jeong JH, Kim DJ, Shin EJ, Kim HC. Glutathione peroxidase-1 gene rescues cocaine-induced conditioned place preference in mice by inhibiting σ-1 receptor expression. Clin Exp Pharmacol Physiol 2019; 46:791-797. [PMID: 31332816 DOI: 10.1111/1440-1681.13140] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 06/26/2019] [Accepted: 07/15/2019] [Indexed: 12/29/2022]
Abstract
The aim of this study was to investigate whether the glutathione peroxidase-1 gene (GPx-1) affects cocaine-induced conditioned place preference (CPP) using a mouse model. Cocaine-induced CPP was accompanied by an increase in the level of σ-1 receptor in the nucleus accumbens (NAc). This phenomenon was more pronounced in the GPx-1 gene knockout (GPx-1 KO) than in wild type (WT) mice. In contrast, the CPP and expression of σ-1 receptor were much less pronounced in GPx-1-overexpressing transgenic (GPx-1 TG) mice than non-transgenic (non-TG) mice. Treatment of the mice with BD1047, a σ-1 receptor antagonist, significantly attenuated both cocaine-induced CPP and c-Fos-immunoreactivity (c-Fos-IR) in WT and GPx-1 KO mice, although the effects were more evident in the latter group. Despite the protective effects of BD1047 on cocaine-induced CPP and c-Fos in non-TG mice, there were no additional protective effects in cocaine-treated GPx-1 TG mice, indicating that the σ-1 receptor is a critical target for GPx-1-mediated psychoprotective activity. Overall, our results suggest that GPx-1 attenuates cocaine-induced CPP via inhibition of σ-1 receptor expression.
Collapse
Affiliation(s)
- Duc Toan Pham
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon, Korea
| | - Yoon Hee Chung
- Department of Anatomy, College of Medicine, Chung-Ang University, Seoul, Korea
| | - Huynh Nhu Mai
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon, Korea
| | - Naveen Sharma
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon, Korea
| | - Jaesuk Yun
- College of Pharmacy, Chungbuk National University, Chungbuk, Korea
| | - Hee Jin Kim
- Department of Pharmacy, Sahmyook University, Seoul, Korea
| | | | - Ji Hoon Jeong
- Department of Pharmacology, College of Medicine, Chung-Ang University, Seoul, Korea
| | - Dae-Joong Kim
- Department of Anatomy and Cell Biology, Medical School, Kangwon National University, Chunchon, Korea
| | - Eun-Joo Shin
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon, Korea
| | - Hyoung-Chun Kim
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon, Korea
| |
Collapse
|
21
|
Mai HN, Chung YH, Shin EJ, Kim DJ, Sharma N, Lee YJ, Jeong JH, Nah SY, Jang CG, Kim HC. Glutathione peroxidase-1 overexpressing transgenic mice are protected from cocaine-induced drug dependence. Neurochem Int 2019; 124:264-273. [DOI: 10.1016/j.neuint.2019.01.018] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2018] [Revised: 01/13/2019] [Accepted: 01/22/2019] [Indexed: 11/29/2022]
|
22
|
Sharma N, Shin EJ, Kim NH, Cho EH, Jeong JH, Jang CG, Nah SY, Nabeshima T, Yoneda Y, Cadet JL, Kim HC. Protective potentials of far-infrared ray against neuropsychotoxic conditions. Neurochem Int 2019; 122:144-148. [DOI: 10.1016/j.neuint.2018.11.019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2018] [Revised: 11/19/2018] [Accepted: 11/27/2018] [Indexed: 12/30/2022]
|
23
|
Tran HQ, Park SJ, Shin EJ, Tran TV, Sharma N, Lee YJ, Jeong JH, Jang CG, Kim DJ, Nabeshima T, Kim HC. Clozapine attenuates mitochondrial burdens and abnormal behaviors elicited by phencyclidine in mice via inhibition of p47 phox; Possible involvements of phosphoinositide 3-kinase/Akt signaling. J Psychopharmacol 2018; 32:1233-1251. [PMID: 30207504 DOI: 10.1177/0269881118795244] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
BACKGROUND Oxidative stress and mitochondrial dysfunction have been implicated in the pathophysiology of schizophrenia. AIMS We investigated whether antipsychotic clozapine modulates nicotinamide adenine dinucleotide phosphate oxidase and mitochondrial burdens induced by phencyclidine in mice. METHODS We examined the effect of clozapine on nicotinamide adenine dinucleotide phosphate oxidase activation, mitochondrial burdens (i.e. oxidative stress and mitochondrial dysfunction), and activities of enzymatic antioxidant in the prefrontal cortex, and subsequent abnormal behaviors induced by repeated treatment with phencyclidine. p47 phox Knockout mice and LY294002, a phosphoinositide 3-kinase inhibitor, were employed to elucidate the pharmacological mechanism of clozapine. RESULTS Phencyclidine treatment resulted in an early increase nicotinamide adenine dinucleotide phosphate oxidase activity, membrane translocation of p47 phox, interaction between p-Akt and p47 phox, and mitochondrial burdens in wild-type mice. Although these increases returned to near control level four days post-phencyclidine, mitochondrial superoxide dismutase and glutathione peroxidase activities were decreased at that time. Clozapine, LY294002, or p47 phox knockout significantly ameliorated social withdrawal and recognition memory deficits produced by phencyclidine. Importantly, LY294002 did not significantly alter the effects of clozapine against abnormal behaviors and the interaction between p-Akt and p47 phox induced by phencyclidine. Furthermore, neither LY294002 nor clozapine exhibited any additive effects to the protection afforded by p47 phox knockout against phencyclidine insult. CONCLUSION Our results suggest that p47 phox gene mediates phencyclidine-induced mitochondrial burdens and abnormal behaviors, and that the interactive modulation between p47 phox and phosphoinositide 3-kinase/Akt is important for the understanding on the pharmacological mechanism of clozapine.
Collapse
Affiliation(s)
- Hai-Quyen Tran
- 1 Neuropsychopharmacology and Toxicology Program, Kangwon National University, Chunchon, Republic of Korea
| | - Se J Park
- 2 School of Natural Resources and Environmental Sciences, Kangwon National University, Chunchon, Republic of Korea
| | - Eun-Joo Shin
- 1 Neuropsychopharmacology and Toxicology Program, Kangwon National University, Chunchon, Republic of Korea
| | - The-Vinh Tran
- 1 Neuropsychopharmacology and Toxicology Program, Kangwon National University, Chunchon, Republic of Korea
| | - Naveen Sharma
- 1 Neuropsychopharmacology and Toxicology Program, Kangwon National University, Chunchon, Republic of Korea
| | - Yu J Lee
- 3 Clinical Pharmacy, Kangwon National University, Chunchon, Republic of Korea
| | - Ji H Jeong
- 4 Department of Pharmacology, Chung-Ang University, Seoul, Republic of Korea
| | - Choon-Gon Jang
- 5 Department of Pharmacology, Sungkyunkwan University, Suwon, Korea
| | - Dae-Joong Kim
- 6 Department of Anatomy and Cell Biology, Kangwon National University, Chunchon, Korea
| | - Toshitaka Nabeshima
- 7 Advanced Diagnostic System Research Laboratory, Fujita Health University Graduate School of Health Sciences, Toyoake, Japan.,9 Japanese Drug Organization of Appropriate Use and Research, Nagoya, Japan
| | - Hyoung-Chun Kim
- 1 Neuropsychopharmacology and Toxicology Program, Kangwon National University, Chunchon, Republic of Korea
| |
Collapse
|
24
|
Tran HQ, Shin EJ, Hoai Nguyen BC, Phan DH, Kang MJ, Jang CG, Jeong JH, Nah SY, Mouri A, Saito K, Nabeshima T, Kim HC. 5-HT 1A receptor agonist 8-OH-DPAT induces serotonergic behaviors in mice via interaction between PKCδ and p47phox. Food Chem Toxicol 2018; 123:125-141. [PMID: 30366073 DOI: 10.1016/j.fct.2018.10.049] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 10/18/2018] [Accepted: 10/22/2018] [Indexed: 12/22/2022]
Abstract
Serotonin syndrome is an adverse reaction due to increased serotonin (5-hydroxytryptophan: 5-HT) concentrations in the central nervous system (CNS). The full 5-HT1A receptor (5-HT1AR) agonist (±)-8-hydroxy-dipropylaminotetralin (8-OH-DPAT) has been recognized to elicit traditional serotonergic behaviors. Treatment with 8-OH-DPAT selectively increased PKCδ expression out of PKC isoforms and 5-HT turnover rate in the hypothalamus of wild-type mice. Treatment with 8-OH-DPAT resulted in oxidative burdens, co-immunoprecipitation of 5-HT1AR and PKCδ, and phosphorylation and membrane translocation of p47phox. Importantly, p47phox also interacted with 5-HT1AR or PKCδ in the presence of 8-OH-DPAT. Consistently, the interaction and oxidative burdens were attenuated by 5-HT1AR antagonism (i.e., WAY100635), PKCδ inhibition (i.e., rottlerin and genetic depletion of PKCδ), or NADPH oxidase/p47phox inhibition (i.e., apocynin and genetic depletion of p47phox). However, WAY100635, apocynin, or rottlerin did not exhibit any additive effects against the protective effect by inhibition of PKCδ or p47phox. Furthermore, apocynin, rottlerin, or WAY100635 also significantly protected from pro-inflammatory/pro-apoptotic changes induced by 8-OH-DPAT. Therefore, we suggest that 8-OH-DPAT-induced serotonergic behaviors requires oxidative stress, pro-inflammatory, and pro-apoptotic changes, that PKCδ or p47phox mediates the serotonergic behaviors induced by 8-OH-DPAT, and that the inhibition of PKCδ-dependent p47phox activation is critical for protecting against serotonergic behaviors.
Collapse
Affiliation(s)
- Hai-Quyen Tran
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon, 24341, Republic of Korea
| | - Eun-Joo Shin
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon, 24341, Republic of Korea
| | - Bao-Chau Hoai Nguyen
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon, 24341, Republic of Korea
| | - Dieu-Hien Phan
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon, 24341, Republic of Korea
| | - Min-Ji Kang
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon, 24341, Republic of Korea
| | - Choon-Gon Jang
- Department of Pharmacology, School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Ji Hoon Jeong
- Department of Pharmacology, College of Medicine, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Seung-Yeol Nah
- Ginsentology Research Laboratory and Department of Physiology, College of Veterinary Medicine, Konkuk University, Seoul, 05029, Republic of Korea
| | - Akihiro Mouri
- Advanced Diagnostic System Research Laboratory, Fujita Health University Graduate School of Health Science, Aichi, 470-1192, Japan
| | - Kuniaki Saito
- Advanced Diagnostic System Research Laboratory, Fujita Health University Graduate School of Health Science, Aichi, 470-1192, Japan
| | - Toshitaka Nabeshima
- Advanced Diagnostic System Research Laboratory, Fujita Health University Graduate School of Health Science, Aichi, 470-1192, Japan; Aino University, Ibaraki, 576-0012, Japan; Japanese Drug Organization of Appropriate and Research, Nagoya, 468-0069, Japan
| | - Hyoung-Chun Kim
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon, 24341, Republic of Korea.
| |
Collapse
|
25
|
Shi HY, Yan SM, Guo YM, Zhang BQ, Guo XY, Shi BL. Vitamin A pretreatment protects NO-induced bovine mammary epithelial cells from oxidative stress by modulating Nrf2 and NF-κB signaling pathways. J Anim Sci 2018; 96:1305-1316. [PMID: 29669072 DOI: 10.1093/jas/sky037] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Accepted: 03/09/2018] [Indexed: 12/16/2022] Open
Abstract
It is known that physiological overproduction of nitric oxide (NO) contributes to oxidative stress and inflammation. Our published studies indicated that vitamin A (VA) reduces NO-induced oxidative stress in bovine mammary epithelial cells (BMECs) by increasing antioxidant enzyme activities. However, the precise mechanism is unclear. The present study was conducted to examine the protective effects of VA on NO-induced damage to BMECs in vitro using diethylenetriamine nitric oxide (DETA-NO) as the NO donor and to explore the intracellular signaling mechanisms of VA that involve nuclear factor erythroid 2-related factor (Nrf2) and nuclear factor kappa-B (NF-κB). Subconfluent BMECs were divided into 10 treatment groups with 6 replicates per treatment and were cultured with dimethyl sulfoxide (DMSO, vehicle negative control) or 0, 0.05, 0.1, 0.2, 0.5, 1, 2, 3, or 4 μg/mL of VA for 24 h and then incubated in the absence or presence of DETA-NO (1,000 μmol/liter) and VA for an additional 6 h. The results showed that exposure to DETA alone decreased cell proliferation compared with the negative control. Pretreatment with VA promoted the proliferation of BMECs, increased the activities of antioxidative enzymes including selenoprotein glutathione peroxidase (GPx) and thioredoxin reductase (TrxR) and their gene and protein expression but decreased NO and interleukin 1 (IL-1) contents in a quadratic manner (P < 0.05). In addition, the expression of mRNA and protein of factors that are related to NF-κB or Nrf2 signaling pathways in BMECs were regulated by VA in a quadratic dose-dependent manner; VA at a concentration of 1 μg/mL exhibited the strongest effect. Together, these results suggest that VA promotes antioxidant functions of BMECs by regulating the synthesis of selenoproteins including GPx and TrxR and by reducing concentrations of IL-1 and NO in vitro by modulating Nrf2 and NF-κB signaling pathways.
Collapse
Affiliation(s)
- H Y Shi
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot, P.R. China
| | - S M Yan
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot, P.R. China
| | - Y M Guo
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot, P.R. China
| | - B Q Zhang
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot, P.R. China
| | - X Y Guo
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot, P.R. China
| | - B L Shi
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot, P.R. China
| |
Collapse
|
26
|
Shin EJ, Hwang YG, Pham DT, Lee JW, Lee YJ, Pyo D, Jeong JH, Lei XG, Kim HC. Glutathione peroxidase-1 overexpressing transgenic mice are protected from neurotoxicity induced by microcystin-leucine-arginine. ENVIRONMENTAL TOXICOLOGY 2018; 33:1019-1028. [PMID: 30076769 DOI: 10.1002/tox.22580] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Revised: 05/15/2018] [Accepted: 05/20/2018] [Indexed: 06/08/2023]
Abstract
Although it has been well-recognized that microcystin-leucine-arginine (MCLR), the most common form of microcystins, induces neurotoxicity, little is currently known about the underlying mechanism for this neurotoxicity. Here, we found that MCLR (10 ng/μL/mouse, i.c.v.) induces significant neuronal loss in the hippocampus of mice. MCLR-induced neurotoxicity was accompanied by oxidative stress, as shown by a significant increase in the level of 4-hydroxynonenal, protein carbonyl, and reactive oxygen species (ROS). Superoxide dismutase-1 (SOD-1) activity was significantly increased, but glutathione peroxidase (GPx) level was significantly decreased following MCLR insult. In addition, MCLR significantly inhibited GSH/GSSG ratio, and significantly induced NFκB DNA binding activity. Because reduced activity of GPx appeared to be critical for the imbalance between activities of SODs and GPx, we utilized GPx-1 overexpressing transgenic mice to ascertain the role of GPx-1 in this neurotoxicity. Genetic overexpression of GPx-1 or NFκB inhibitor pyrrolidine dithiocarbamate (PDTC) significantly attenuated MCLR-induced hippocampal neuronal loss in mice. However, PDTC did not exert any additive effect on neuroprotection mediated by GPx-1 overexpression, indicating that NFκB is a neurotoxic target of MCLR. Combined, these results suggest that MCLR-induced neurotoxicity requires oxidative stress associated with failure in compensatory induction of GPx, possibly through activation of the transcription factor NFκB.
Collapse
Affiliation(s)
- Eun-Joo Shin
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Republic of Korea
| | - Yeong Gwang Hwang
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Republic of Korea
| | - Duc Toan Pham
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Republic of Korea
| | - Ji Won Lee
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Republic of Korea
| | - Yu Jeung Lee
- Clinical Pharmacy, College of Pharmacy, Kangwon National University, Republic of Korea
| | - Dongjin Pyo
- Department of Chemistry, College of Natural Sciences, Kangwon National University, Republic of Korea
| | - Ji Hoon Jeong
- Department of Pharmacology, College of Medicine, Chung-Ang University, Seoul, Republic of Korea
| | - Xin Gen Lei
- Department of Animal Science, Cornell University, New York
| | - Hyoung-Chun Kim
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Republic of Korea
| |
Collapse
|
27
|
Mai HN, Jung TW, Kim DJ, Sharma G, Sharma N, Shin EJ, Jang CG, Nah SY, Lee SH, Chung YH, Lei XG, Jeong JH, Kim HC. Protective potential of glutathione peroxidase-1 gene against cocaine-induced acute hepatotoxic consequences in mice. J Appl Toxicol 2018; 38:1502-1520. [DOI: 10.1002/jat.3666] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Revised: 05/29/2018] [Accepted: 06/10/2018] [Indexed: 01/22/2023]
Affiliation(s)
- Huynh Nhu Mai
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy; Kangwon National University; Chunchon 24341 Republic of Korea
| | - Tae Woo Jung
- Research Administration Team; Seoul National University Bundang Hospital; Seongnam 13620 Republic of Korea
| | - Dae-Joong Kim
- Department of Anatomy and Cell Biology, Medical School; Kangwon National University; Chunchon 24341 Republic of Korea
| | - Garima Sharma
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy; Kangwon National University; Chunchon 24341 Republic of Korea
| | - Naveen Sharma
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy; Kangwon National University; Chunchon 24341 Republic of Korea
| | - Eun-Joo Shin
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy; Kangwon National University; Chunchon 24341 Republic of Korea
| | - Choon-Gon Jang
- Department of Pharmacology, School of Pharmacy; Sungkyunkwan University; Suwon 16419 Republic of Korea
| | - Seung-Yeol Nah
- Ginsentology Research Laboratory and Department of Physiology, College of Veterinary Medicine; Konkuk University; Seoul 05029 Republic of Korea
| | - Sung Hoon Lee
- Department of Pharmacology, College of Pharmacy; Chung-Ang University; Seoul 06974 Republic of Korea
| | - Yoon Hee Chung
- Department of Anatomy, College of Medicine; Chung-Ang University; Seoul 06974 Republic of Korea
| | - Xin Gen Lei
- Department of Animal Science; Cornell University; Ithaca New York 14853 USA
| | - Ji Hoon Jeong
- Department of Pharmacology, College of Medicine; Chung-Ang University; Seoul 06974 Republic of Korea
| | - Hyoung-Chun Kim
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy; Kangwon National University; Chunchon 24341 Republic of Korea
| |
Collapse
|
28
|
Tian H, Sun W, Zhang Q, Li X, Sang Y, Li J, Niu Y, Ding H. Procyanidin B2 mitigates behavioral impairment and protects myelin integrity in cuprizone-induced schizophrenia in mice. RSC Adv 2018; 8:23835-23846. [PMID: 35540280 PMCID: PMC9081829 DOI: 10.1039/c8ra03854f] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2018] [Accepted: 06/11/2018] [Indexed: 01/18/2023] Open
Abstract
Numerous studies have suggested that neuropathological changes in schizophrenia may be related to damage to white matter or demyelination. Procyanidin B2, which is a constituent of many fruits such as grapes and strawberries, has various biological activities such as anti-inflammatory and anti-tumor activity, as has been reported. This study aimed to estimate the effects of procyanidin B2 on behavioral impairment and the protection of myelin integrity in a cuprizone-induced schizophrenia model. Mice were exposed to cuprizone (0.2% w/w in chow) for five weeks to induce schizophrenia-like behavioral changes and demyelination. Procyanidin B2 (20 or 100 mg kg−1 day−1) or vehicle was administered orally to mice after withdrawal from cuprizone. Behavioral impairment was detected with an open-field test, a rotarod test and a Morris water maze. Myelin integrity was assessed using LFB staining and MBP expression, including immunofluorescence and western blotting. In addition, enhancements in the expression of HO-1 and NQO1 suggested that procyanidin B2 may regulate oxidative homeostasis via promoting the translation of Nrf2 to the nucleus. Data indicated that procyanidin B2 could mitigate behavioral impairment and protect myelin integrity in the cuprizone-induced model via regulating oxidative stress by activating Nrf2 signaling. Numerous studies have suggested that neuropathological changes in schizophrenia may be related to damage to white matter or demyelination.![]()
Collapse
Affiliation(s)
- Hui Tian
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education, Wuhan University School of Pharmaceutical Sciences, Wuhan University Wuhan Hubei P. R. China +8613007162084
| | - Wanchun Sun
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education, Wuhan University School of Pharmaceutical Sciences, Wuhan University Wuhan Hubei P. R. China +8613007162084
| | - Qianying Zhang
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education, Wuhan University School of Pharmaceutical Sciences, Wuhan University Wuhan Hubei P. R. China +8613007162084
| | - Xiaofei Li
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education, Wuhan University School of Pharmaceutical Sciences, Wuhan University Wuhan Hubei P. R. China +8613007162084
| | - Ying Sang
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education, Wuhan University School of Pharmaceutical Sciences, Wuhan University Wuhan Hubei P. R. China +8613007162084
| | - Jian Li
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education, Wuhan University School of Pharmaceutical Sciences, Wuhan University Wuhan Hubei P. R. China +8613007162084
| | - Yunhui Niu
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education, Wuhan University School of Pharmaceutical Sciences, Wuhan University Wuhan Hubei P. R. China +8613007162084
| | - Hong Ding
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education, Wuhan University School of Pharmaceutical Sciences, Wuhan University Wuhan Hubei P. R. China +8613007162084
| |
Collapse
|
29
|
Genetic overexpression of glutathione peroxidase-1 attenuates microcystin-leucine-arginine-induced memory impairment in mice. Neurochem Int 2018; 118:152-165. [PMID: 29908255 DOI: 10.1016/j.neuint.2018.06.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 05/14/2018] [Accepted: 06/12/2018] [Indexed: 11/21/2022]
Abstract
Microcystin-leucine-arginine (MCLR) is the most common form of microcystins, which are environmental toxins produced by cyanobacteria, and its hepatotoxicity has been well-documented. However, the neurotoxic potential of MCLR remains to be further elucidated. In the present study, we investigated whether intracerebroventricular (i.c.v.) infusion of MCLR induces mortality and neuronal loss in the hippocampus of mice. Because we found that MCLR impairs memory function in the hippocampus at a low dose (4 ng/μl/mouse, i.c.v.) without a significant neuronal loss, we focused on this dose for further analyses. Results showed that MCLR (4 ng/μl/mouse, i.c.v.) significantly increased oxidative stress (i.e., malondialdehyde, protein carbonyl, and synaptosomal ROS) in the hippocampus. In addition, MCLR significantly increased superoxide dismutase (SOD) activity without corresponding induction of glutathione peroxidase (GPx) activity, and thus led to significant decrease in the ratio of GPx/SODs activity. The GSH/GSSG ratio was also significantly reduced after MCLR treatment. GPx-1 overexpressing transgenic mice (GPx-1 Tg) were significantly protected from MCLR-induced memory impairment and oxidative stress. The DNA binding activity of nuclear factor erythroid-derived 2-related factor 2 (Nrf2) in these mice was significantly enhanced, and the ratios of GPx/SODs activity and GSH/GSSG returned to near control levels in the hippocampus. Importantly, memory function exhibited a significant positive correlation with the ratios of GPx/SODs activity and GSH/GSSG in the hippocampus of MCLR-treated non-transgenic (non-Tg)- and GPx-1 Tg-mice. Combined, our results suggest that MCLR induces oxidative stress and memory impairment without significant neuronal loss, and that GPx-1 gene constitutes an important protectant against MCLR-induced memory impairment and oxidative stress via maintaining antioxidant defense system homeostasis, possibly through the induction of Nrf2 transcription factor.
Collapse
|
30
|
Shi H, Yan S, Guo Y, Shi B, Guo X. The pre-protective effect of vitamin A on LPS-induced oxidative stress of bovine mammary epithelial cells. ITALIAN JOURNAL OF ANIMAL SCIENCE 2018. [DOI: 10.1080/1828051x.2018.1453757] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- Huiyu Shi
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot, China
| | - Sumei Yan
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot, China
| | - Yongmei Guo
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot, China
| | - Binlin Shi
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot, China
| | - Xiaoyu Guo
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot, China
| |
Collapse
|
31
|
Tran HQ, Lee Y, Shin EJ, Jang CG, Jeong JH, Mouri A, Saito K, Nabeshima T, Kim HC. PKCδ Knockout Mice Are Protected from Dextromethorphan-Induced Serotonergic Behaviors in Mice: Involvements of Downregulation of 5-HT 1A Receptor and Upregulation of Nrf2-Dependent GSH Synthesis. Mol Neurobiol 2018; 55:7802-7821. [PMID: 29468562 DOI: 10.1007/s12035-018-0938-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Accepted: 01/28/2018] [Indexed: 12/12/2022]
Abstract
We investigated whether a specific serotonin (5-HT) receptor-mediated mechanism was involved in dextromethorphan (DM)-induced serotonergic behaviors. We firstly observed that the activation of 5-HT1A receptor, but not 5-HT2A receptor, contributed to DM-induced serotonergic behaviors in mice. We aimed to determine whether the upregulation of 5-HT1A receptor induced by DM facilitates the specific induction of certain PKC isoform, because previous reports suggested that 5-HT1A receptor activates protein kinase C (PKC). A high dose of DM (80 mg/kg, i.p.) induced a selective induction of PKCδ out of PKCα, PKCβI, PKCβII, PKCξ, and PKCδ in the hypothalamus of wild-type (WT) mice. More importantly, 5-HT1A receptor co-immunoprecipitated PKCδ in the presence of DM. Consistently, rottlerin, a pharmacological inhibitor of PKCδ, or PKCδ knockout significantly protected against increases in 5-HT1A receptor gene expression, 5-HT turnover rate, and serotonergic behaviors induced by DM. Treatment with DM resulted in an initial increase in nuclear factor erythroid-2-related factor 2 (Nrf2) nuclear translocation and DNA-binding activity, γ-glutamylcysteine (GCL) mRNA expression, and glutathione (GSH) level. This compensative induction was further potentiated by rottlerin or PKCδ knockout. However, GCL mRNA and GSH/GSSG levels were decreased 6 and 12 h post-DM. These decreases were attenuated by PKCδ inhibition. Our results suggest that interaction between 5-HT1A receptor and PKCδ is critical for inducing DM-induced serotonergic behaviors and that inhibition of PKCδ attenuates the serotonergic behaviors via downregulation of 5-HT1A receptor and upregulation of Nrf2-dependent GSH synthesis.
Collapse
MESH Headings
- Acetophenones/pharmacology
- Animals
- Behavior, Animal
- Benzopyrans/pharmacology
- Cell Nucleus/drug effects
- Cell Nucleus/metabolism
- Dextromethorphan
- Down-Regulation
- Glutamate-Cysteine Ligase/genetics
- Glutamate-Cysteine Ligase/metabolism
- Glutathione/biosynthesis
- Glutathione Disulfide/metabolism
- Hypothalamus/metabolism
- Hypothermia, Induced
- Isoenzymes/metabolism
- Mice, Inbred C57BL
- Mice, Knockout
- NF-E2-Related Factor 2/metabolism
- Neuroprotective Agents/pharmacology
- Phosphorylation/drug effects
- Piperazines/pharmacology
- Protein Kinase C-delta/metabolism
- Pyridines/pharmacology
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Receptor, Serotonin, 5-HT1A/metabolism
- Receptor, Serotonin, 5-HT2A/metabolism
- Serotonin/metabolism
- Serotonin 5-HT1 Receptor Antagonists/pharmacology
- Time Factors
- Up-Regulation
Collapse
Affiliation(s)
- Hai-Quyen Tran
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon, 24341, Republic of Korea
| | - Youngho Lee
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon, 24341, Republic of Korea
| | - Eun-Joo Shin
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon, 24341, Republic of Korea.
| | - Choon-Gon Jang
- Department of Pharmacology, School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Ji Hoon Jeong
- Department of Pharmacology, College of Medicine, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Akihiro Mouri
- Advanced Diagnostic System Research Laboratory, Fujita Health University Graduate School of Health Science, Toyoake, Aichi, 470-1192, Japan
| | - Kuniaki Saito
- Advanced Diagnostic System Research Laboratory, Fujita Health University Graduate School of Health Science, Toyoake, Aichi, 470-1192, Japan
| | - Toshitaka Nabeshima
- Advanced Diagnostic System Research Laboratory, Fujita Health University Graduate School of Health Science, Toyoake, Aichi, 470-1192, Japan
- Aino University, Ibaraki, 576-0012, Japan
- Japanese Drug Organization of Appropriate and Research, Nagoya, 468-0069, Japan
| | - Hyoung-Chun Kim
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon, 24341, Republic of Korea.
| |
Collapse
|
32
|
Blockade of platelet-activating factor receptor attenuates abnormal behaviors induced by phencyclidine in mice through down-regulation of NF-κB. Brain Res Bull 2017; 137:71-78. [PMID: 29122692 DOI: 10.1016/j.brainresbull.2017.11.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Revised: 10/26/2017] [Accepted: 11/02/2017] [Indexed: 01/21/2023]
Abstract
Accumulating evidence suggests that neuroinflammation is one of the important etiologic factors of abusive and neuropsychiatric disorders. Platelet-activating factor (PAF) is potent proinflammatory lipid mediat1or and plays a pivotal role in neuroinflammatory disorders through the specific PAF receptor (PAF-R). Phencyclidine (PCP) induces a psychotomimetic state that closely resembles schizophrenia. Here, we investigated the role of PAF-R in the abnormal behaviors induced by PCP in mice. Repeated treatment with PCP resulted in a significant increase in PAF-R gene expression in the prefrontal cortex (PFC) and in the hippocampus. This increase was more pronounced in the PFC than hippocampus. Treatment with PCP resulted in a significant increase in nuclear translocation of the nuclear factor kappa beta (NF-κB) p65 and DNA binding activity, indicating that the proinflammatory molecule NF-κB was increased through up-regulation of PAF-R. Consistently, NF-κB activation was significantly protected by the PAF-R antagonist, ginkgolide B (Gink B), in PAF-R knockout mice and by the NF-κB inhibitor, pyrrolidine dithiocarbamate (PDTC). In addition, PCP-induced abnormal behaviors (i.e., reduced sociability, depression, cognitive impairment, and behavioral sensitization) were significantly attenuated by Gink B, in PAF-R knockout mice, and by PDTC. Importantly, PDTC did not significantly alter the attenuations observed in Gink B-treated mice or PAF-R knockout mice, indicating that NF-κB is a critical target for neuropsychotoxic modulation of PAF-R. Therefore, the results suggest that PAF-R mediates PCP-induced neuropsychotoxicity via a NF-κB-dependent mechanism, and that up-regulation of PAF-R may be associated with schizophrenia-like behavior in animal models.
Collapse
|
33
|
Tran TV, Shin EJ, Nguyen LTT, Lee Y, Kim DJ, Jeong JH, Jang CG, Nah SY, Toriumi K, Nabeshima T, Yamada K, Kim HC. Protein Kinase Cδ Gene Depletion Protects Against Methamphetamine-Induced Impairments in Recognition Memory and ERK1/2 Signaling via Upregulation of Glutathione Peroxidase-1 Gene. Mol Neurobiol 2017; 55:4136-4159. [DOI: 10.1007/s12035-017-0638-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2017] [Accepted: 05/24/2017] [Indexed: 11/28/2022]
|