1
|
Rizk SK, Ali EA, Sheref AAM, Tayel SG, El Derbaly SA. Vitamin D and canagliflozin combination alleviates Parkinson's disease in rats through modulation of RAC1/NF-κB/Nrf2 interaction. Immunopharmacol Immunotoxicol 2025; 47:328-344. [PMID: 40134204 DOI: 10.1080/08923973.2025.2481849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Accepted: 03/16/2025] [Indexed: 03/27/2025]
Abstract
OBJECTIVE Oxidative stress and neuroinflammation are crucial factors in the pathogenesis of Parkinson's disease (PD). Vitamin D (Vit D) and canagliflozin (CAN) are known to have anti-inflammatory and antioxidant properties. Together, they target key molecular pathways involved in PD, including oxidative stress and neuroinflammation, specifically, the small GTPase protein (RAC1)/nuclear factor-kappa B (NF-κB)/nuclear factor erythroid 2-related factor 2 (Nrf2) signaling, which regulates brain's oxidative stress and inflammation. This study investigates the effects of Vit D and CAN alone and in combination in a rat model of PD. MATERIALS AND METHODS Fifty male Wistar rats were assigned to five groups (n = 10), including control, rotenone (ROT), Vit D + ROT, CAN + ROT, and Vit D + CAN + ROT. We assessed weight changes, brain weight, neurobehavioral functions, biochemical markers, and immunohistopathology of brain tissues. RESULTS The results showed that Vit D treatment was more effective than CAN in alleviating PD symptoms, with the combination of Vit D and CAN offering the best therapeutic outcome. This combination therapy significantly improved serum Vit D, striatal dopamine (DA) levels, antioxidant status (reduced glutathione (GSH) and catalase (CAT), reduced oxidative stress (malondialdehyde (MDA)), and ameliorated inflammation (tumor necrosis factor-α (TNF-α), interleukin 6 (IL-6), and interleukin 10 (IL-10)). Additionally, the combination therapy modulated the expression of RAC1, NF-κB, Nrf2, vitamin D receptors (VDR), and vitamin D-binding protein (DBP) and immunoexpression of tyrosine hydroxylase (TH), and α-synuclein (α-SYN). CONCLUSION These findings suggest that Vit D and CAN synergistically modulate the RAC1/NF-κB/Nrf2 pathway, leading to improved neuroprotection in PD.
Collapse
Affiliation(s)
- Sara Kamal Rizk
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Menoufia University, Menoufia, Egypt
| | - Eman A Ali
- Clinical Pharmacology Department, Faculty of Medicine, Menoufia University, Menoufia, Egypt
- Clinical Pharmacology Department, Faculty of Medicine, Menoufia National University, Menoufia, Egypt
| | - AlZahraa A M Sheref
- Medical Physiology Department, Faculty of Medicine, Menoufia University, Menoufia, Egypt
- Medical Physiology Department, Faculty of Medicine, Menoufia National University, Menoufia, Egypt
| | - Sara G Tayel
- Anatomy and Embryology Department, Faculty of Medicine, Menoufia University, Menoufia, Egypt
- Anatomy and Embryology Department, Faculty of Medicine, Menoufia National University, Menoufia, Egypt
| | - Sara A El Derbaly
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Menoufia University, Menoufia, Egypt
- Department of Basic Medical Sciences, Faculty of Medicine, Ibn Sina University for Medical Sciences, Amman, Jordan
| |
Collapse
|
2
|
Guo YE, Lv J, Shu P, Li X, Li Y, Guo J, Chen G, Li Y, Lu B, Zhang W, Liu Y. Drug screening identifies pyrrolidinedithiocarbamate ammonium ameliorating DSS-induced mouse ulcerative colitis via suppressing Th17 differentiation. Cell Immunol 2024; 405-406:104887. [PMID: 39503083 DOI: 10.1016/j.cellimm.2024.104887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 10/09/2024] [Accepted: 10/20/2024] [Indexed: 12/02/2024]
Abstract
T helper 17 (Th17) cells play crucial roles in various autoimmune diseases, including ulcerative colitis (UC), which is characterized by widespread inflammation in the mucosa of the colon and rectum. To identify small-molecule compounds capable of inhibiting CD4+ T cell differentiation into Th17 cells, we established a screening system. Through drug screening, we found that pyrrolidinedithiocarbamate ammonium (PDTC) effectively inhibits Th17 differentiation. In a dextran sulfate sodium (DSS)-induced UC mouse model, administration of PDTC significantly ameliorated colitis. PDTC treatment decreased the production of proinflammatory mediators and inhibited the proportion of Th17 cells in colitis-afflicted mice by suppressing NF-κB activation. These findings showed that PDTC can alleviate colitis by inhibiting NF-κB activation. The therapeutic effects of PDTC observed in a mouse model of UC provided a rationale for its application in clinical settings.
Collapse
Affiliation(s)
- Yu-E Guo
- Department of Pharmacy, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jie Lv
- Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Ping Shu
- Department of Pharmacy, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xi Li
- Department of Pharmacy, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Ying Li
- Department of Pharmacy, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Junhong Guo
- Department of Pathology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Guofang Chen
- Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yuping Li
- Department of Pharmacy, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Bo Lu
- Department of Pharmacy, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China.
| | - Wei Zhang
- Department of Pharmacy, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China.
| | - Yin Liu
- Department of Laboratory Medicine, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China.
| |
Collapse
|
3
|
Wang F, Xu J, Hu C, Lai J, Shen P, Lu Y, Jiang F. β-glucan regulates the intestinal immunity of pearl gentian grouper via the nuclear factor kappa B signaling pathway. FISH & SHELLFISH IMMUNOLOGY 2024; 154:109941. [PMID: 39368523 DOI: 10.1016/j.fsi.2024.109941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 09/21/2024] [Accepted: 10/03/2024] [Indexed: 10/07/2024]
Abstract
The preceding study observed that yeast β-glucan supplementation enhanced intestinal health and augmented disease resistance in pearl gentian grouper (Epinephelus lanceolatus♂ × Epinephelus fuscoguttatus♀), which occurred concurrently with the activation of the nuclear factor kappa B (NFκB) signaling pathway. Thus, we hypothesized that β-glucan improves intestinal health in grouper by modulating the NFκB pathway. Accordingly, the present study examined the effects of NFκB pathway disruption using a specific inhibitor on the intestinal health of pearl gentian grouper that had been injected with β-glucan. The experimental groups were as follows, (1) CD group: PBS injected; (2) βG group: β-glucan injected at a dose of 80 mg/kg; (3) PDTC group: NFκB inhibitor PDTC injected at a dose of 30 mg/kg; (4) βG + PDTC group: a combination of β-glucan (80 mg/kg) and PDTC (30 mg/kg) injected together. The results demonstrated that β-glucan-induced increases in mRNA expression levels of NFκB inhibitor α (iκbα) and p65, the degradation and phosphorylation of IκBα, and the phosphorylation of NFκB p65 were significantly inhibited following NFκB inhibition using PDTC in the intestine of grouper. The PDTC injection resulted in a significant reduction in the β-glucan-induced increase in mucin levels. The β-glucan-induced elevation of alkaline phosphatase (AKP) activity, component 3 (C3) content, and inflammatory factors were significantly suppressed following NFκB inhibition. The βG + PDTC treatment resulted in a restoration of catalase (CAT) enzyme activity to the level observed in the CD treatment, while total antioxidant capacity (T-AOC) was decreased to the level of the βG treatment. The β-glucan-induced downregulation of caspase8 (casp8) was reversed following NFκB inhibition, as well as the mRNA levels of casp3 and casp9 being elevated to a greater extent. In conclusion, the β-glucan-regulated intestinal immunity in grouper may be mediated by the NFκB pathway. Furthermore, the inhibitory effect of β-glucan on apoptosis and oxidative stress may not be related to the NFκB signaling pathway.
Collapse
Affiliation(s)
- Fan Wang
- Guangxi Key Laboratory of Marine Environmental Science, Guangxi Academy of Marine Sciences, Guangxi Academy of Sciences, Nanning, China; College of Life Science and Technology of Guangxi University, Nanning, 530005, Guangxi, China
| | - Jia Xu
- Guangxi Key Laboratory of Marine Environmental Science, Guangxi Academy of Marine Sciences, Guangxi Academy of Sciences, Nanning, China
| | - Chaoqun Hu
- Guangxi Key Laboratory of Marine Environmental Science, Guangxi Academy of Marine Sciences, Guangxi Academy of Sciences, Nanning, China
| | - Junxiang Lai
- Guangxi Key Laboratory of Marine Environmental Science, Guangxi Academy of Marine Sciences, Guangxi Academy of Sciences, Nanning, China
| | - Peihong Shen
- College of Life Science and Technology of Guangxi University, Nanning, 530005, Guangxi, China
| | - Yishan Lu
- College of Fishery, Guangdong Ocean University, Zhanjiang, 524088, China.
| | - Fajun Jiang
- Guangxi Key Laboratory of Marine Environmental Science, Guangxi Academy of Marine Sciences, Guangxi Academy of Sciences, Nanning, China.
| |
Collapse
|
4
|
Ulloa M, Macías F, Clapp C, Martínez de la Escalera G, Arnold E. Prolactin is an Endogenous Antioxidant Factor in Astrocytes That Limits Oxidative Stress-Induced Astrocytic Cell Death via the STAT3/NRF2 Signaling Pathway. Neurochem Res 2024; 49:1879-1901. [PMID: 38755517 PMCID: PMC11144156 DOI: 10.1007/s11064-024-04147-3] [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: 02/05/2024] [Revised: 03/29/2024] [Accepted: 05/02/2024] [Indexed: 05/18/2024]
Abstract
Oxidative stress-induced death of neurons and astrocytes contributes to the pathogenesis of numerous neurodegenerative diseases. While significant progress has been made in identifying neuroprotective molecules against neuronal oxidative damage, little is known about their counterparts for astrocytes. Prolactin (PRL), a hormone known to stimulate astroglial proliferation, viability, and cytokine expression, exhibits antioxidant effects in neurons. However, its role in protecting astrocytes from oxidative stress remains unexplored. Here, we investigated the effect of PRL against hydrogen peroxide (H2O2)-induced oxidative insult in primary cortical astrocyte cultures. Incubation of astrocytes with PRL led to increased enzymatic activity of superoxide dismutase (SOD) and glutathione peroxidase (GPX), resulting in higher total antioxidant capacity. Concomitantly, PRL prevented H2O2-induced cell death, reactive oxygen species accumulation, and protein and lipid oxidation. The protective effect of PRL upon H2O2-induced cell death can be explained by the activation of both signal transducer and activator of transcription 3 (STAT3) and NFE2 like bZIP transcription factor 2 (NRF2) transduction cascades. We demonstrated that PRL induced nuclear translocation and transcriptional upregulation of Nrf2, concurrently with the transcriptional upregulation of the NRF2-dependent genes heme oxygenase 1, Sod1, Sod2, and Gpx1. Pharmacological blockade of STAT3 suppressed PRL-induced transcriptional upregulation of Nrf2, Sod1 and Gpx1 mRNA, and SOD and GPX activities. Furthermore, genetic ablation of the PRL receptor increased astroglial susceptibility to H2O2-induced cell death and superoxide accumulation, while diminishing their intrinsic antioxidant capacity. Overall, these findings unveil PRL as a potent antioxidant hormone that protects astrocytes from oxidative insult, which may contribute to brain neuroprotection.
Collapse
Affiliation(s)
- Miriam Ulloa
- Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus UNAM-Juriquilla, 76230, Querétaro, México
- Posgrado en Ciencias Biológicas, Universidad Nacional Autónoma de México, Ciudad Universitaria, 04510, Mexico City, México
| | - Fernando Macías
- Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus UNAM-Juriquilla, 76230, Querétaro, México
| | - Carmen Clapp
- Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus UNAM-Juriquilla, 76230, Querétaro, México
| | | | - Edith Arnold
- Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus UNAM-Juriquilla, 76230, Querétaro, México.
- CONAHCYT-Universidad Nacional Autónoma de México, Campus UNAM-Juriquilla, Querétaro, México.
| |
Collapse
|
5
|
Liddell JR, Hilton JBW, Kysenius K, Billings JL, Nikseresht S, McInnes LE, Hare DJ, Paul B, Mercer SW, Belaidi AA, Ayton S, Roberts BR, Beckman JS, McLean CA, White AR, Donnelly PS, Bush AI, Crouch PJ. Microglial ferroptotic stress causes non-cell autonomous neuronal death. Mol Neurodegener 2024; 19:14. [PMID: 38317225 PMCID: PMC10840184 DOI: 10.1186/s13024-023-00691-8] [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: 10/20/2023] [Accepted: 11/28/2023] [Indexed: 02/07/2024] Open
Abstract
BACKGROUND Ferroptosis is a form of regulated cell death characterised by lipid peroxidation as the terminal endpoint and a requirement for iron. Although it protects against cancer and infection, ferroptosis is also implicated in causing neuronal death in degenerative diseases of the central nervous system (CNS). The precise role for ferroptosis in causing neuronal death is yet to be fully resolved. METHODS To elucidate the role of ferroptosis in neuronal death we utilised co-culture and conditioned medium transfer experiments involving microglia, astrocytes and neurones. We ratified clinical significance of our cell culture findings via assessment of human CNS tissue from cases of the fatal, paralysing neurodegenerative condition of amyotrophic lateral sclerosis (ALS). We utilised the SOD1G37R mouse model of ALS and a CNS-permeant ferroptosis inhibitor to verify pharmacological significance in vivo. RESULTS We found that sublethal ferroptotic stress selectively affecting microglia triggers an inflammatory cascade that results in non-cell autonomous neuronal death. Central to this cascade is the conversion of astrocytes to a neurotoxic state. We show that spinal cord tissue from human cases of ALS exhibits a signature of ferroptosis that encompasses atomic, molecular and biochemical features. Further, we show the molecular correlation between ferroptosis and neurotoxic astrocytes evident in human ALS-affected spinal cord is recapitulated in the SOD1G37R mouse model where treatment with a CNS-permeant ferroptosis inhibitor, CuII(atsm), ameliorated these markers and was neuroprotective. CONCLUSIONS By showing that microglia responding to sublethal ferroptotic stress culminates in non-cell autonomous neuronal death, our results implicate microglial ferroptotic stress as a rectifiable cause of neuronal death in neurodegenerative disease. As ferroptosis is currently primarily regarded as an intrinsic cell death phenomenon, these results introduce an entirely new pathophysiological role for ferroptosis in disease.
Collapse
Affiliation(s)
- Jeffrey R Liddell
- Department of Anatomy and Physiology, The University of Melbourne, Parkville, VIC, 3010, Australia.
| | - James B W Hilton
- Department of Anatomy and Physiology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Kai Kysenius
- Department of Anatomy and Physiology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Jessica L Billings
- Department of Anatomy and Physiology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Sara Nikseresht
- Department of Anatomy and Physiology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Lachlan E McInnes
- School of Chemistry and Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Dominic J Hare
- Atomic Medicine Initiative, University of Technology Sydney, Ultimo, NSW, 2007, Australia
| | - Bence Paul
- School of Earth Science, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Stephen W Mercer
- Department of Anatomy and Physiology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Abdel A Belaidi
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Scott Ayton
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Blaine R Roberts
- Department of Biochemistry, Emory University, Atlanta, GA, 30322, USA
| | - Joseph S Beckman
- Linus Pauling Institute, Oregon State University, Corvallis, OR, 97331, USA
| | - Catriona A McLean
- Anatomical Pathology, Alfred Hospital, Melbourne, VIC, 3005, Australia
| | - Anthony R White
- QIMR Berghofer Medical Research Institute, Herston, QLD, 4006, Australia
| | - Paul S Donnelly
- School of Chemistry and Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Ashley I Bush
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Peter J Crouch
- Department of Anatomy and Physiology, The University of Melbourne, Parkville, VIC, 3010, Australia.
| |
Collapse
|
6
|
Chen J, Akhtar M, Hardej D. Exposure to dithiocarbamate fungicide Ziram results in hepatic and renal toxicity in Long Evan rats. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2023; 99:104116. [PMID: 37011740 DOI: 10.1016/j.etap.2023.104116] [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: 12/05/2022] [Revised: 03/28/2023] [Accepted: 03/30/2023] [Indexed: 06/19/2023]
Abstract
Ziram is a dimethyldithiocarbamate fungicide that is complexed to the metal zinc. The focus of this study is to examine the effects of dimethyldithiocarbamate exposure on metal homeostasis, glutathione levels, and the physiological parameters of the kidney and liver in Long-Evan rats. Our results indicate significant accumulation of copper or zinc, and changes in total GSH or GSH/GSSG ratio in the liver and kidneys of animals treated with Ziram only. Histopathological examination of liver and kidney sections indicate the presence of infiltrates in the liver of animals treated with Ziram only, whereas protein aggregates, sloughing of cells and increased KIM-1 positive cells, an indicator of tubule deterioration, are seen in the kidneys of animals treated with Ziram and sodium-dimethyldithiocarbamate, the salt form of the dimethyldithiocarbmate backbone. These findings suggest that the overall toxicological effect of Ziram is mediated by an intrinsic property rather than to dimethyldithiocarbamate backbone or metal moiety.
Collapse
Affiliation(s)
- Jeffrey Chen
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Jamaica, NY 11439, USA
| | - Mumtaz Akhtar
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Jamaica, NY 11439, USA
| | - Diane Hardej
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Jamaica, NY 11439, USA.
| |
Collapse
|
7
|
Egbujor MC, Tucci P, Onyeije UC, Emeruwa CN, Saso L. NRF2 Activation by Nitrogen Heterocycles: A Review. Molecules 2023; 28:2751. [PMID: 36985723 PMCID: PMC10058096 DOI: 10.3390/molecules28062751] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 03/14/2023] [Accepted: 03/16/2023] [Indexed: 03/30/2023] Open
Abstract
Several nitrogen heterocyclic analogues have been applied to clinical practice, and about 75% of drugs approved by the FDA contain at least a heterocyclic moiety. Thus, nitrogen heterocycles are beneficial scaffolds that occupy a central position in the development of new drugs. The fact that certain nitrogen heterocyclic compounds significantly activate the NRF2/ARE signaling pathway and upregulate the expression of NRF2-dependent genes, especially HO-1 and NQO1, underscores the need to study the roles and pharmacological effects of N-based heterocyclic moieties in NRF2 activation. Furthermore, nitrogen heterocycles exhibit significant antioxidant and anti-inflammatory activities. NRF2-activating molecules have been of tremendous research interest in recent times due to their therapeutic roles in neuroinflammation and oxidative stress-mediated diseases. A comprehensive review of the NRF2-inducing activities of N-based heterocycles and their derivatives will broaden their therapeutic prospects in a wide range of diseases. Thus, the present review, as the first of its kind, provides an overview of the roles and effects of nitrogen heterocyclic moieties in the activation of the NRF2 signaling pathway underpinning their antioxidant and anti-inflammatory actions in several diseases, their pharmacological properties and structural-activity relationship are also discussed with the aim of making new discoveries that will stimulate innovative research in this area.
Collapse
Affiliation(s)
- Melford C. Egbujor
- Department of Chemical Sciences, Rhema University Nigeria, Aba 453115, Nigeria
| | - Paolo Tucci
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy
| | - Ugomma C. Onyeije
- Department of Pure and Industrial Chemistry, Nnamdi Azikiwe University, Awka 420007, Nigeria
| | | | - Luciano Saso
- Department of Physiology and Pharmacology, Vittorio Erspamer, Sapienza University of Rome, 00161 Rome, Italy
| |
Collapse
|
8
|
Akintoye OO, Ajibare AJ, Folawiyo MA, Jimoh-Abdulghaffaar HO, Asuku A, Owolabi GA, Babalola KT. Zinc supplement reverses short-term memory deficit in sodium benzoate-induced neurotoxicity in male Wistar rats by enhancing anti-oxidative capacity via Nrf 2 up-regulation. Behav Brain Res 2023; 437:114163. [PMID: 36265761 DOI: 10.1016/j.bbr.2022.114163] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Revised: 10/10/2022] [Accepted: 10/14/2022] [Indexed: 11/06/2022]
Abstract
Sodium benzoate (SB) is a commonly-used food preservative, with a controversial report to its neurological benefit and toxicity. Zinc (Zn) is a trace element that plays a crucial role in memory, inflammation and oxidative stress. This study was to investigate the effect of SB on rat cognition and memory and the possible modulatory effect of Zn supplement. Twenty four male Wistar rats were divided into four groups of six animals each. Animals in groups 1-4 were treated with normal saline 1 ml/kg, SB 200 mg/kg, zinc sulphate 10 ml/kg and SB 200 mg/kg + zinc sulphate 10 ml/kg/day daily respectively for three weeks. After treatment, the animals were subjected to different behavioural tests, and then sacrificed. Their blood samples were collected for catalase(CAT), superoxide dismutase(SOD) and interleukin-1B(IL-1B) assay. Brain samples were also collected for nuclear factor-erythroid-related factor 2(Nrf2), and acetylcholinesterase (AchE) mRNA gene expression. The serum levels of CAT and SOD were (p < 0.0001; p < 0.0001) reduced in the SB only-treated group compared to the other groups. Nrf2 gene expression was totally shut down in the SB only-treated group but, up-regulated in the Zn-treated groups (p < 0.0001). The serum level of IL-1B was higher in the SB only-treated group compared to the other groups. SB-treated group spent longer time in the close arm (p = <0.0001), shorter time in the open arm (p = <0.0001) and had higher anxiety index (p = 0.0045) than the Zn-treated groups. Conclusively, Zinc improves memory deficit, has anxiolytic, anti-oxidant and anti-inflammatory properties.
Collapse
Affiliation(s)
- O O Akintoye
- Physiology Department, Faculty of Basic Medical Sciences, College of Medicine, Ekiti State University, Nigeria.
| | - A J Ajibare
- Physiology Department, Faculty of Basic Medical Sciences, College of Medicine, Ekiti State University, Nigeria
| | - M A Folawiyo
- Physiology Department, Faculty of Basic Medical Sciences, College of Medicine, Ekiti State University, Nigeria
| | - H O Jimoh-Abdulghaffaar
- Physiology Department, Faculty of Basic Medical Sciences, College of Health Science, University of Ilorin, Nigeria
| | - A Asuku
- Department of Medical Biotechnology, Bioresources Development Centre, National Biotechnology Development Agrncy, Ogbomoso, Nigeria
| | - G A Owolabi
- Physiology Department, Faculty of Basic Medical Sciences, College of Medicine, Ekiti State University, Nigeria
| | - K T Babalola
- Physiology Department, Faculty of Basic Medical Sciences, College of Medicine, Leadcity University, Ibadan, Nigeria
| |
Collapse
|
9
|
Sandouka S, Saadi A, Singh PK, Olowe R, Shekh-Ahmad T. Nrf2 is predominantly expressed in hippocampal neurons in a rat model of temporal lobe epilepsy. Cell Biosci 2023; 13:3. [PMID: 36600279 DOI: 10.1186/s13578-022-00951-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 12/28/2022] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Drug resistance is a particular problem in patients with temporal lobe epilepsy, where seizures originate mainly from the hippocampus. Many of these epilepsies are acquired conditions following an insult to the brain such as a prolonged seizure. Such conditions are characterized by pathophysiological mechanisms including massive oxidative stress that synergistically mediate the secondary brain damage, contributing to the development of epilepsy. The transcription factor nuclear factor (erythroid-derived 2)-like 2 (Nrf2) has emerged in recent years as an attractive therapeutic approach targeting to upregulate the antioxidative defenses in the cell, to ameliorate the oxidative stress-induced damage. Thus, it is important to understand the characteristics of Nrf2 activation during epileptogenesis and epilepsy. Here, we studied the temporal, regional, and cell-type specific expression of Nrf2 in the brain, in a rat model of temporal lobe epilepsy. RESULTS Early after status-epilepticus, Nrf2 is mainly activated in the hippocampus and maintained during the whole period of epileptogenesis. Only transient expression of Nrf2 was observed in the cortex. Nevertheless, the expression of several Nrf2 antioxidant target genes was increased within 24 h after status-epilepticus in both the cortex and the hippocampus. We demonstrated that after status-epilepticus in rats, Nrf2 is predominantly expressed in neurons in the CA1 and CA3 regions of the hippocampus, and only astrocytes in the CA1 increase their Nrf2 expression. CONCLUSIONS In conclusion, our data identify previously unrecognized spatial and cell-type dependent activation of Nrf2 during epilepsy development, highlighting the need for a time-controlled, and cell-type specific activation of the Nrf2 pathway for mediating anti-oxidant response after brain insult, to modify the development of epilepsy.
Collapse
Affiliation(s)
- Sereen Sandouka
- The Institute for Drug Research, The School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, 91120, Jerusalem, Israel
| | - Aseel Saadi
- The Institute for Drug Research, The School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, 91120, Jerusalem, Israel
| | - Prince Kumar Singh
- The Institute for Drug Research, The School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, 91120, Jerusalem, Israel
| | - Rhoda Olowe
- The Institute for Drug Research, The School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, 91120, Jerusalem, Israel
| | - Tawfeeq Shekh-Ahmad
- The Institute for Drug Research, The School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, 91120, Jerusalem, Israel.
| |
Collapse
|
10
|
Role of Nrf2 in aging, Alzheimer's and other neurodegenerative diseases. Ageing Res Rev 2022; 82:101756. [PMID: 36243357 DOI: 10.1016/j.arr.2022.101756] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 09/14/2022] [Accepted: 10/09/2022] [Indexed: 01/31/2023]
Abstract
Nuclear Factor-Erythroid Factor 2 (Nrf2) is an important transcription factor that regulates the expression of large number of genes in healthy and disease states. Nrf2 is made up of 605 amino acids and contains 7 conserved regions known as Nrf2-ECH homology domains. Nrf2 regulates the expression of several key components of oxidative stress, mitochondrial biogenesis, mitophagy, autophagy and mitochondrial function in all organs of the human body, in the peripheral and central nervous systems. Mounting evidence also suggests that altered expression of Nrf2 is largely involved in aging, neurodegenerative diseases, including Alzheimer's disease, Parkinson's disease, Huntington's diseases, Amyotrophic lateral sclerosis, Stroke, Multiple sclerosis and others. The purpose of this article is to detail the essential role of Nrf2 in oxidative stress, antioxidative defense, detoxification, inflammatory responses, transcription factors, proteasomal and autophagic/mitophagic degradation, and metabolism in aging and neurodegenerative diseases. This article also highlights the Nrf2 structural and functional activities in healthy and disease states, and also discusses the current status of Nrf2 research and therapeutic strategies to treat aging and neurodegenerative diseases.
Collapse
|
11
|
Lu Y, Li B, Xu A, Liang X, Xu T, Jin H, Xie Y, Wang R, Liu X, Gao X, Han Y, Zeng J. NF-κB and AP-1 are required for the lipopolysaccharide-induced expression of MCP-1, CXCL1, and Cx43 in cultured rat dorsal spinal cord astrocytes. Front Mol Neurosci 2022; 15:859558. [PMID: 35966011 PMCID: PMC9368326 DOI: 10.3389/fnmol.2022.859558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 06/28/2022] [Indexed: 11/13/2022] Open
Abstract
TLR4 and Cx43 signaling in dorsal spinal cord has been shown to be involved in the development of neuropathic pain. However, it is not clear whether TLR4 signaling is associated with the expression of MCP-1, CXCL1, and Cx43 in LPS (lipopolysaccharide)-treated rat dorsal spinal cord astrocytes under in vitro condition. In the present study, we found that TLR4 antagonist TAK-242 significantly inhibited LPS-induced MCP-1, CXCL1, and Cx43 expression, suggesting the role of TLR4 in response to LPS in cultured dorsal spinal cord astrocytes. Application of TAK-242 significantly blocked LPS-induced NF-κB and AP-1 activity and the expression of MCP-1, CXCL1 and Cx43. Furthermore, NF-κB inhibitor PDTC and AP-1 inhibitor SR11302 significantly blocked LPS-induced MCP-1, CXCL1, and Cx43 expression. DNA-binding activity of NF-κB, its effect on MCP-1 expression was suppressed by PDTC and SR11302. On the other hand, DNA-binding activity of AP-1, its effect on CXCL1 or Cx43 expression was also suppressed by PDTC and SR11302. In addition, PDTC was found to inhibit the nuclear translocation of AP-1 and the expression of c-Jun induced by LPS, which suggested that NF-κBp65 is essential for the AP-1 activity. Similarly, SR11302 significantly blocked LPS-induced the nuclear translocation of NF-κBp65 and the expression of NF-κBp65 induced by LPS. Pretreatment with CBX, Gap26, or Gap19 (Cx43 blockers) significantly inhibited abnormal astrocytic hemichannel opening and chemokines (MCP-1 and CXCL1) release in LPS-stimulated astrocytes. In summary, cell culture experiments revealed that LPS stimulation could evoke TLR4 signaling with the subsequent activation of NF-κB and AP-1, resulting in the expression of MCP-1, CXCL1, and Cx43. TLR4 activation increased Cx43 hemichannel, but not gap-junction activities and induced the release of the MCP-1 and CXCL1 from astrocytes via Cx43 hemichannel. These findings may help us to understand the role of astrocytic signaling in inflammatory response within dorsal spinal cord tissue.
Collapse
|
12
|
Wang T, Sun Q, Yang J, Wang G, Zhao F, Chen Y, Jin Y. Reactive astrocytes induced by 2-chloroethanol modulate microglia polarization through IL-1β, TNF-α, and iNOS upregulation. Food Chem Toxicol 2021; 157:112550. [PMID: 34517076 DOI: 10.1016/j.fct.2021.112550] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Revised: 09/02/2021] [Accepted: 09/08/2021] [Indexed: 01/06/2023]
Abstract
The synthetic organic chemical, 1,2-dichloroethane (1,2-DCE), can cause brain edemas under subacute poisoning. Our previous studies indicated that neuroinflammation could be induced due to astrocytes and microglia activation during brain edemas in 1,2-DCE-intoxicated mice. However, the crosstalk between these two glial cells in 1,2-DCE-induced neuroinflammation remained unclear. In this study, primary cultured rat astrocytes and microglia, as well as an immortalized microglia cell line were employed to study the effects of 2-chloroethanol (2-CE, a 1,2-DCE intermediate metabolite in vivo) treated astrocytes on microglia polarization. Our current results revealed that 2-CE treated rat astrocytes were activated through p38 mitogen-activated protein kinase (p38 MAPK)/nuclear factor-κB (NF-κB), and activator protein-1 (AP-1) signaling pathways. Theses pathways were triggered by reactive oxygen species (ROS) produced during 2-CE metabolism. Also, astrocytes were more sensitive to 2-CE effects than microglia. Interleukin-1β (IL-1β), tumor necrosis factor-α (TNF-α), and inducible nitric oxide synthase (iNOS) expressions were upregulated in 2-CE-induced reactive astrocytes, enhancing IL-1β, TNF-α, and nitric oxide (NO) excretions, which stimulated microglia polarization. Therefore, the neuroinflammation induced by 1,2-DCE in mice's brains is probably triggered by reactive astrocytes.
Collapse
Affiliation(s)
- Tong Wang
- Department of Occupational and Environmental Health, School of Public Health, China Medical University, Shenyang, China
| | - Qi Sun
- Department of Child and Adolescent Health, China Medical University, Shenyang, China
| | - Jinhan Yang
- Department of Occupational and Environmental Health, School of Public Health, China Medical University, Shenyang, China
| | - Gaoyang Wang
- Department of Occupational and Environmental Health, School of Public Health, China Medical University, Shenyang, China
| | - Fenghong Zhao
- Department of Occupational and Environmental Health, School of Public Health, China Medical University, Shenyang, China
| | - Yuhua Chen
- Department of Developmental Biology, Key Laboratory of Cell Biology, Ministry of Public Health and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China
| | - Yaping Jin
- Department of Occupational and Environmental Health, School of Public Health, China Medical University, Shenyang, China.
| |
Collapse
|
13
|
Cui SB, Wang TX, Liu ZW, Yan JY, Zhang K. Zinc finger protein A20 regulates the development and progression of osteoarthritis by affecting the activity of NF-κB p65. Immunopharmacol Immunotoxicol 2021; 43:713-723. [PMID: 34463587 DOI: 10.1080/08923973.2021.1970764] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
OBJECTIVE To investigate the role of Zinc finger protein A20 in osteoarthritis (OA) by regulating NF-κB p65. METHODS A20, MMP1, MMP13 and IL-1β expressions in human OA cartilage samples were detected by qRT-PCR. IL-1β-induced chondrocyte was treated with A20 lentivirus activation particle, pyrrolidine dithiocarbamate (PDTC, a NF-κB inhibitor) with/without A20 siRNA. IL-6, TNF-α, and PGE2 levels were measured by ELISA, and NO production by Greiss reaction. Destabilization of the medial meniscus (DMM) surgery was used to construct the OA models, followed by injection of A20 adenovirus. MMP1 and MMP13 expression was measured by immunohistochemistry. The mRNA and protein expression were performed by qRT-PCR and western blotting, respectively. RESULTS A20 was down-regulated in human OA cartilage samples, and negatively correlated with the expressions of MMP1, MMP13 and IL-1β. The IL-1β-induced chondrocyte manifested decreased A20 with increased NF-κB p65 activity. A20 overexpression suppressed the NF-κB p65 activity in IL-1β-induced chondrocyte. Furthermore, PDTC decreased IL-1β-induced chondrocyte apoptosis with the upregulated COL1A1, COL2A1, COL10A1 and ACAN, as well as the down-regulated MMP1, MMP13, COX2, iNOS, IL-6, TNF-α, NO and PGE2, which was reversed by A20 siRNA. In vivo, OA mice gained higher OARSI score and Mankin's score, exhibited up-regulations of MMP1 and MMP13, and decreased NF-κB p65 activity, which was improved after injection of A20 adenovirus. CONCLUSION A20 was reduced in OA cartilage samples, and its overexpression, by suppressing the activity of NF-κB p65, could improve IL-1β-induced chondrocyte degradation and apoptosis in vitro, as well as mitigate the inflammation in OA mice.
Collapse
Affiliation(s)
- Shu-Bei Cui
- The First Department of Orthopedics, Handan Central Hospital, Handan, China
| | - Tao-Xia Wang
- Department of Nephrology, Affiliated Hospital of Hebei University of Technology, Handan, China
| | - Zhen-Wu Liu
- The First Department of Orthopedics, Handan Central Hospital, Handan, China
| | - Ji-Ying Yan
- The First Department of Orthopedics, Handan Central Hospital, Handan, China
| | - Kai Zhang
- The First Department of Orthopedics, Handan Central Hospital, Handan, China
| |
Collapse
|
14
|
Tampio J, Löffler S, Guillon M, Hugele A, Huttunen J, Huttunen KM. Improved l-Type amino acid transporter 1 (LAT1)-mediated delivery of anti-inflammatory drugs into astrocytes and microglia with reduced prostaglandin production. Int J Pharm 2021; 601:120565. [PMID: 33812973 DOI: 10.1016/j.ijpharm.2021.120565] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 03/26/2021] [Accepted: 03/29/2021] [Indexed: 12/14/2022]
Abstract
Non-steroidal anti-inflammatory drugs (NSAIDs) can have protective effects in the brain by inhibition of cyclooxygenases (COX). However, the delivery into the brain across the blood-brain barrier (BBB) and particularly into the brain parenchymal cells is hindered. Therefore, in the present study, we developed four l-type amino acid transporter 1 (LAT1)-utilizing prodrugs of flurbiprofen, ibuprofen, naproxen, and ketoprofen, since LAT1 is expressed on both, the BBB endothelial cells as well as parenchymal cells. The cellular uptake and utilization of LAT1 by novel prodrugs were studied in mouse cortical primary astrocytes and immortalized microglia (BV2), and the release of the parent NSAID in several tissue and cell homogenates. Finally, the effects of the studied prodrugs on prostaglandin E2 (PGE2) production and cell viability were explored. The gained results showed that all four prodrugs were carried into their target cells via LAT1. They also released their parent NSAIDs via carboxylesterases (CES) and most likely also other un-identified enzymes, which need to be carefully considered when administrating these compounds orally or intravenously. Most importantly, all the studied prodrugs reduced the PGE2 production in astrocytes and microglia after lipopolysaccharide (LPS)-induced inflammation by 29-94% and without affecting the cell viability with the studied concentration (20 µM).
Collapse
Affiliation(s)
- Janne Tampio
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland
| | - Susanne Löffler
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland
| | - Melina Guillon
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland
| | - Agathe Hugele
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland
| | - Johanna Huttunen
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland
| | - Kristiina M Huttunen
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland.
| |
Collapse
|
15
|
Ehrke E, Steinmeier J, Stapelfeldt K, Dringen R. The Menadione-Mediated WST1 Reduction by Cultured Astrocytes Depends on NQO1 Activity and Cytosolic Glucose Metabolism. Neurochem Res 2021; 46:88-99. [PMID: 31902045 DOI: 10.1007/s11064-019-02930-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 12/03/2019] [Accepted: 12/05/2019] [Indexed: 01/07/2023]
Abstract
The reduction of water-soluble tetrazolium salts (WSTs) is frequently used to determine the metabolic integrity and the viability of cultured cells. Recently, we have reported that the electron cycler menadione can efficiently connect intracellular oxidation reactions in cultured astrocytes with the extracellular reduction of WST1 and that this menadione cycling reaction involves an enzyme. The enzymatic reaction involved in the menadione-dependent WST1 reduction was found strongly enriched in the cytosolic fraction of cultured astrocytes and is able to efficiently use both NADH and NADPH as electron donors. In addition, the reaction was highly sensitive towards dicoumarol with Kic values in the low nanomolar range, suggesting that the NAD(P)H:quinone oxidoreductase 1 (NQO1) catalyzes the menadione-dependent WST1 reduction in astrocytes. Also, in intact astrocytes, dicoumarol inhibited the menadione-dependent WST1 reduction in a concentration-dependent manner with half-maximal inhibition observed at around 50 nM. Moreover, the menadione-dependent WST1 reduction by viable astrocytes was strongly affected by the availability of glucose. In the absence of glucose only residual WST1 reduction was observed, while a concentration-dependent increase in WST1 reduction was found during a 30 min incubation with maximal WST1 reduction already determined in the presence of 0.5 mM glucose. Mannose could fully replace glucose as substrate for astrocytic WST1 reduction, while other hexoses, lactate and the mitochondrial substrate β-hydroxybutyrate failed to provide electrons for the cell-dependent WST1 reduction. These results demonstrate that the menadione-mediated WST1 reduction involves cytosolic NQO1 activity and that this process is strongly affected by the availability of glucose as metabolic substrate.
Collapse
Affiliation(s)
- Eric Ehrke
- Center for Biomolecular Interactions Bremen (CBIB), Faculty 2 (Biology/Chemistry), University of Bremen, P.O. Box 330440, 28334, Bremen, Germany
- Center for Environmental Research and Sustainable Technology (UFT), University of Bremen, Bremen, Germany
| | - Johann Steinmeier
- Center for Biomolecular Interactions Bremen (CBIB), Faculty 2 (Biology/Chemistry), University of Bremen, P.O. Box 330440, 28334, Bremen, Germany
- Center for Environmental Research and Sustainable Technology (UFT), University of Bremen, Bremen, Germany
| | - Karsten Stapelfeldt
- Center for Biomolecular Interactions Bremen (CBIB), Faculty 2 (Biology/Chemistry), University of Bremen, P.O. Box 330440, 28334, Bremen, Germany
- Institute for Biophysics, University of Bremen, Bremen, Germany
| | - Ralf Dringen
- Center for Biomolecular Interactions Bremen (CBIB), Faculty 2 (Biology/Chemistry), University of Bremen, P.O. Box 330440, 28334, Bremen, Germany.
- Center for Environmental Research and Sustainable Technology (UFT), University of Bremen, Bremen, Germany.
| |
Collapse
|
16
|
Wang G, Yang Q, Zheng C, Li D, Li J, Zhang F. Physiological Concentration of H 2O 2 Supports Dopamine Neuronal Survival via Activation of Nrf2 Signaling in Glial Cells. Cell Mol Neurobiol 2021; 41:163-171. [PMID: 32318898 PMCID: PMC11448570 DOI: 10.1007/s10571-020-00844-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 04/07/2020] [Indexed: 10/24/2022]
Abstract
Traditionally, hydrogen peroxide (H2O2) was formed from cellular oxidative metabolism and often viewed as toxic waste. In fact, H2O2 was a benefit messenger for neuron-glia signaling and synaptic transmission. Thus, H2O2 was a double-edged sword and neuroprotection vs. neurotoxicity produced by H2O2 was difficult to define. Nuclear factor erythroid 2-related factor 2 (Nrf2) has been implicated as an intracellular regulator of neuronal growth. Inactivation of Nrf2 participated in the development of Parkinson's disease (PD). Thus, suitable activation of Nrf2 was essential for the prevention and treatment of PD. This study aimed to explore whether H2O2-conferred neuroprotective effects to support neuronal survival. H2O2 were added into primary neuron-glia, neuron-astroglia and neuron-microglia co-cultures in concentration- and time-dependent manners. H2O2 increased dopamine (DA) neuronal survival in concentration- and time-dependent manners. In addition, glial cells Nrf2 activation involved in H2O2-supported DA neuronal survival with the following phenomenons. First, H2O2 activated Nrf2 signaling pathway. Second, H2O2 generated beneficial neuroprotection in neuron-glia, neuron-astroglia and neuron-microglia co-cultures but not in neuron-enriched cultures. Third, silence of Nrf2 in glial cells abolished H2O2-conferred DA neuronal survival. This study demonstrated that physiological concentration of H2O2-supported DA neuronal survival via activation of Nrf2 signaling in glial cells. Our data permit to re-evaluate the role of H2O2 in the pathogenesis and therapeutic strategies for PD.
Collapse
Affiliation(s)
- Guoqing Wang
- Joint International Research Laboratory of Ethnomedicine of Ministry of Education and Key Laboratory of Basic Pharmacology of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou, China
| | - Qiuyu Yang
- Joint International Research Laboratory of Ethnomedicine of Ministry of Education and Key Laboratory of Basic Pharmacology of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou, China
| | - Changqing Zheng
- Joint International Research Laboratory of Ethnomedicine of Ministry of Education and Key Laboratory of Basic Pharmacology of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou, China
| | - Daidi Li
- Joint International Research Laboratory of Ethnomedicine of Ministry of Education and Key Laboratory of Basic Pharmacology of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou, China
| | - Jingjie Li
- Joint International Research Laboratory of Ethnomedicine of Ministry of Education and Key Laboratory of Basic Pharmacology of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou, China
| | - Feng Zhang
- Joint International Research Laboratory of Ethnomedicine of Ministry of Education and Key Laboratory of Basic Pharmacology of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou, China.
| |
Collapse
|
17
|
Tampio J, Huttunen J, Montaser A, Huttunen KM. Targeting of Perforin Inhibitor into the Brain Parenchyma Via a Prodrug Approach Can Decrease Oxidative Stress and Neuroinflammation and Improve Cell Survival. Mol Neurobiol 2020; 57:4563-4577. [PMID: 32754897 PMCID: PMC7515946 DOI: 10.1007/s12035-020-02045-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 07/28/2020] [Indexed: 12/31/2022]
Abstract
The cytolytic protein perforin has a crucial role in infections and tumor surveillance. Recently, it has also been associated with many brain diseases, such as neurodegenerative diseases and stroke. Therefore, inhibitors of perforin have attracted interest as novel drug candidates. We have previously reported that converting a perforin inhibitor into an L-type amino acid transporter 1 (LAT1)-utilizing prodrug can improve the compound's brain drug delivery not only across the blood-brain barrier (BBB) but also into the brain parenchymal cells: neurons, astrocytes, and microglia. The present study evaluated whether the increased uptake into mouse primary cortical astrocytes and subsequently improvements in the cellular bioavailability of this brain-targeted perforin inhibitor prodrug could enhance its pharmacological effects, such as inhibition of production of caspase-3/-7, lipid peroxidation products and prostaglandin E2 (PGE2) in the lipopolysaccharide (LPS)-induced neuroinflammation mouse model. It was demonstrated that increased brain and cellular drug delivery could improve the ability of perforin inhibitors to elicit their pharmacological effects in the brain at nano- to picomolar levels. Furthermore, the prodrug displayed multifunctional properties since it also inhibited the activity of several key enzymes related to Alzheimer's disease (AD), such as the β-site amyloid precursor protein (APP) cleaving enzyme 1 (BACE1), acetylcholinesterase (AChE), and most probably also cyclooxygenases (COX) at micromolar concentrations. Therefore, this prodrug is a potential drug candidate for preventing Aβ-accumulation and ACh-depletion in addition to combatting neuroinflammation, oxidative stress, and neural apoptosis within the brain. Graphical abstract.
Collapse
Affiliation(s)
- Janne Tampio
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211, Kuopio, Finland
| | - Johanna Huttunen
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211, Kuopio, Finland
| | - Ahmed Montaser
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211, Kuopio, Finland
| | - Kristiina M Huttunen
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211, Kuopio, Finland.
| |
Collapse
|
18
|
Ajibade PA, Fatokun AA, Andrew FP. Synthesis, characterization and anti-cancer studies of Mn(II), Cu(II), Zn(II) and Pt(II) dithiocarbamate complexes - crystal structures of the Cu(II) and Pt(II) complexes. Inorganica Chim Acta 2020. [DOI: 10.1016/j.ica.2020.119431] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
19
|
Astrocyte-Targeted Transporter-Utilizing Derivatives of Ferulic Acid Can Have Multifunctional Effects Ameliorating Inflammation and Oxidative Stress in the Brain. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:3528148. [PMID: 31814871 PMCID: PMC6877910 DOI: 10.1155/2019/3528148] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/19/2019] [Revised: 09/19/2019] [Accepted: 10/03/2019] [Indexed: 12/27/2022]
Abstract
Ferulic acid (FA) is a natural phenolic antioxidant, which can exert also several other beneficial effects to combat neuroinflammation and neurodegenerative diseases, such as Alzheimer's disease. One of these properties is the inhibition of several enzymes and factors, such as β-site amyloid precursor protein (APP) cleaving enzyme 1 (BACE1), cyclooxygenases (COXs), lipoxygenases (LOXs), mammalian (or mechanistic) target for rapamycin (mTOR), and transcription factor NF-κB. We have previously synthesized three L-type amino acid transporter 1- (LAT1-) utilizing FA-derivatives with the aim to develop brain-targeted prodrugs of FA. In the present study, the cellular uptake and bioavailability of these FA-derivatives were evaluated in mouse primary astrocytic cell cultures together with their inhibitory effects towards BACE1, COX/LOX, mTOR, NF-κB, acetylcholinesterase (AChE), and oxidative stress. According to the results, all three FA-derivatives were taken up 200–600 times more effectively at 10 μM concentration into the astrocytes than FA, with one derivative having a high intracellular bioavailability (Kp,uu), particularly at low concentrations. Moreover, all of the derivatives were able to inhibit BACE1, COX/LOX, AChE, and oxidative stress measured as decreased cellular lipid peroxidation. Furthermore, one of the derivatives modified the total mTOR amount. Therefore, these derivatives have the potential to act as multifunctional compounds preventing β-amyloid accumulation as well as combating inflammation and reducing oxidative stress in the brain. Thus, this study shows that converting a parent drug into a transporter-utilizing derivative not only may increase its brain and cellular uptake, and bioavailability but can also broaden the spectrum of pharmacological effects elicited by the derivative.
Collapse
|
20
|
Li Q, Xing S, Chen Y, Liao Q, Li Q, Liu Y, He S, Feng F, Chen Y, Zhang J, Liu W, Guo Q, Sun Y, Sun H. Reasonably activating Nrf2: A long-term, effective and controllable strategy for neurodegenerative diseases. Eur J Med Chem 2019; 185:111862. [PMID: 31735576 DOI: 10.1016/j.ejmech.2019.111862] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2019] [Revised: 11/06/2019] [Accepted: 11/06/2019] [Indexed: 02/06/2023]
Abstract
Neurodegenerative diseases are a variety of debilitating and fatal disorder in central nervous system (CNS). Besides targeting neuronal activity by influencing neurotransmitters or their corresponding receptors, modulating the underlying processes that lead to cell death, such as oxidative stress and mitochondrial dysfunction, should also be emphasized as an assistant strategy for neurodegeneration therapy. Nuclear factor (erythroid-derived 2)-like 2 (Nrf2) has been closely verified to be related to anti-inflammation and oxidative stress, rationally regulating its belonging pathway and activating Nrf2 is emphasized to be a potential treatment approach. There have existed multiple Nrf2 activators with different mechanisms and diverse structures, but those applied for neuro-disorders are still limited. On the basis of research arrangement and compound summary, we put forward the limitations of existing Nrf2 activators for neurodegenerative diseases and their future developing directions in enhancing the blood-brain barrier permeability to make Nrf2 activators function in CNS and designing Nrf2-based multi-target-directed ligands to affect multiple nodes in pathology of neurodegenerative diseases.
Collapse
Affiliation(s)
- Qi Li
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, PR China
| | - Shuaishuai Xing
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, PR China
| | - Ying Chen
- Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing, 211198, PR China
| | - Qinghong Liao
- Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing, 211198, PR China
| | - Qihang Li
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, PR China
| | - Yang Liu
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, PR China
| | - Siyu He
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, PR China
| | - Feng Feng
- Jiangsu Food and Pharmaceutical Science College, No.4 Meicheng Road, Huai'an, 223003, PR China
| | - Yao Chen
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China
| | - Jie Zhang
- Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing, 211198, PR China
| | - Wenyuan Liu
- Department of Analytical Chemistry, School of Pharmacy, Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, Nanjing, 210009, PR China
| | - Qinglong Guo
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, 210009, PR China
| | - Yuan Sun
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California Davis, Sacramento, 95817, USA
| | - Haopeng Sun
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, PR China; Jiangsu Food and Pharmaceutical Science College, No.4 Meicheng Road, Huai'an, 223003, PR China.
| |
Collapse
|
21
|
Huttunen J, Peltokangas S, Gynther M, Natunen T, Hiltunen M, Auriola S, Ruponen M, Vellonen KS, Huttunen KM. L-Type Amino Acid Transporter 1 (LAT1/Lat1)-Utilizing Prodrugs Can Improve the Delivery of Drugs into Neurons, Astrocytes and Microglia. Sci Rep 2019; 9:12860. [PMID: 31492955 PMCID: PMC6731241 DOI: 10.1038/s41598-019-49009-z] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Accepted: 08/18/2019] [Indexed: 12/27/2022] Open
Abstract
l-Type Amino Acid Transporter 1 (LAT1/Lat1) is responsible for carrying large, neutral l-amino acids as well as several drugs and prodrugs across the blood-brain barrier (BBB). However, the BBB is not the only barrier that hinders drugs acting effectively within the brain; the brain parenchymal cell membranes represent a secondary barrier for the drugs with intracellular target sites. In this study, expression and function of Lat1 was quantified in mouse primary neuron, astrocyte and immortalized microglia (BV2) cultures. Moreover, ability of Lat1 to carry prodrugs inside these brain cells was evaluated. The results showed that Lat1 was localized at the similar level in all studied cells (3.07 ± 0.92–3.77 ± 0.91 fmol/µg protein). The transporter was also functional in all three cell types, astrocytes having the highest transport capacity and affinity for the LAT1/Lat1-substrate, [14C]-l-leucine, followed by neurons and microglia. The designed prodrugs (1-6) were able to utilize Lat1 for their cellular uptake and it was mainly much higher than the one of their parent drugs. Interestingly, improved cellular uptake was also achieved in cells representing Alzheimer’s Disease phenotype. Therefore, improved delivery and intra-brain targeting of drugs can be attained by utilizing LAT1/Lat1 and prodrug approach.
Collapse
Affiliation(s)
- Johanna Huttunen
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211, Kuopio, Finland
| | - Soile Peltokangas
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211, Kuopio, Finland
| | - Mikko Gynther
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211, Kuopio, Finland
| | - Teemu Natunen
- Institute of Biomedicine, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211, Kuopio, Finland
| | - Mikko Hiltunen
- Institute of Biomedicine, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211, Kuopio, Finland
| | - Seppo Auriola
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211, Kuopio, Finland
| | - Marika Ruponen
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211, Kuopio, Finland
| | - Kati-Sisko Vellonen
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211, Kuopio, Finland
| | - Kristiina M Huttunen
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211, Kuopio, Finland.
| |
Collapse
|
22
|
Wang G, He X, Zhu G, Li D, Shi J, Zhang F. Ellagic acid supports neuron by regulating astroglia Nrf2. Biotechnol Appl Biochem 2019; 66:738-743. [DOI: 10.1002/bab.1791] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 06/08/2019] [Indexed: 12/23/2022]
Affiliation(s)
- Guo‐Qing Wang
- Key Laboratory of Basic Pharmacology of Guizhou Zunyi Medical University Zunyi Guizhou People's Republic of China
- Joint International Research Laboratory of Ethnomedicine of Ministry of Education Zunyi Medical University Zunyi Guizhou People's Republic of China
- Key Laboratory of Basic Pharmacology of Ministry of Education Zunyi Medical University Zunyi Guizhou People's Republic of China
| | - Xue‐Mei He
- Key Laboratory of Basic Pharmacology of Guizhou Zunyi Medical University Zunyi Guizhou People's Republic of China
- Joint International Research Laboratory of Ethnomedicine of Ministry of Education Zunyi Medical University Zunyi Guizhou People's Republic of China
- Key Laboratory of Basic Pharmacology of Ministry of Education Zunyi Medical University Zunyi Guizhou People's Republic of China
| | - Guo‐Fu Zhu
- Key Laboratory of Basic Pharmacology of Guizhou Zunyi Medical University Zunyi Guizhou People's Republic of China
- Joint International Research Laboratory of Ethnomedicine of Ministry of Education Zunyi Medical University Zunyi Guizhou People's Republic of China
- Key Laboratory of Basic Pharmacology of Ministry of Education Zunyi Medical University Zunyi Guizhou People's Republic of China
| | - Dai‐Di Li
- Key Laboratory of Basic Pharmacology of Guizhou Zunyi Medical University Zunyi Guizhou People's Republic of China
- Joint International Research Laboratory of Ethnomedicine of Ministry of Education Zunyi Medical University Zunyi Guizhou People's Republic of China
- Key Laboratory of Basic Pharmacology of Ministry of Education Zunyi Medical University Zunyi Guizhou People's Republic of China
| | - Jing‐Shan Shi
- Key Laboratory of Basic Pharmacology of Guizhou Zunyi Medical University Zunyi Guizhou People's Republic of China
- Joint International Research Laboratory of Ethnomedicine of Ministry of Education Zunyi Medical University Zunyi Guizhou People's Republic of China
- Key Laboratory of Basic Pharmacology of Ministry of Education Zunyi Medical University Zunyi Guizhou People's Republic of China
| | - Feng Zhang
- Key Laboratory of Basic Pharmacology of Guizhou Zunyi Medical University Zunyi Guizhou People's Republic of China
- Joint International Research Laboratory of Ethnomedicine of Ministry of Education Zunyi Medical University Zunyi Guizhou People's Republic of China
- Key Laboratory of Basic Pharmacology of Ministry of Education Zunyi Medical University Zunyi Guizhou People's Republic of China
| |
Collapse
|
23
|
Zhu X, Liu J, Huang S, Zhu W, Wang Y, Chen O, Xue J. Neuroprotective effects of isoliquiritigenin against cognitive impairment via suppression of synaptic dysfunction, neuronal injury, and neuroinflammation in rats with kainic acid-induced seizures. Int Immunopharmacol 2019; 72:358-366. [PMID: 31030091 DOI: 10.1016/j.intimp.2019.04.028] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 03/28/2019] [Accepted: 04/14/2019] [Indexed: 02/06/2023]
Abstract
Epileptogenesis is a dynamic process initiated by insults to brain and commonly accompanied by cognitive impairment. Isoliquiritigenin (ISL), a flavonoid in licorice, has a broad spectrum of biological effects including anti-inflammatory and antioxidant activities. However, the protective effects of ISL against cognitive impairment in epileptic processes and the underlying molecular mechanism are not well understood. To address these questions, we established an reproducible seizure model by intracerebroventricular injection of kainic acid (KA) in 21-day-old rats; ISL was intraperitoneally administered three times prior to KA injection, and changes in cognitive function; synaptic plasticity; neuronal injury; number of glial cells; and expression of pro-inflammatory cytokines and nuclear factor-like (NRF)2 signaling and NACHT, LRR, and PYD domains-containing protein (NLRP)3 inflammasome components in the hippocampus were examined. Rats with KA-induced seizures showed longer average escape latency and decreases in the number of platform crossings and average time spent in the target quadrant in the Morris water maze; ISL pretreatment reversed this decline in cognitive impairment and increased the protein levels of synaptophysin, postsynaptic density-95 and brain-derived neurotrophic factor while reducing the number of Fluoro Jade B-positive cells, microglia, and astrocytes; cleaved-Caspase-3 and -9 protein levels; and tumor necrosis factor-α, interleukin (IL)-1β, and IL-18 production. It also enhanced the nuclear localization of NRF2, hemeoxygenase-1, and NAD(P)H:quinone oxidoreductase (NQO) 1, and reversed the upregulation of NLRP3 inflammasome components NLRP3 and Caspase-1 induced by KA injection. Thus, ISL protects against cognitive impairment in KA-induced epileptic processes possibly through regulation of NRF2 signaling and the NLRP3 inflammasome pathway.
Collapse
Affiliation(s)
- Xiaobo Zhu
- Department of Pediatrics, the Second Hospital of Shandong University, Jinan 250012, China
| | - Jiankun Liu
- Department of Ophthalmology, the Second People's Hospital of Jinan City, Jinan 250000, China
| | - Shanying Huang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Shandong University, Jinan 250012, China
| | - Weiwei Zhu
- Department of Pediatrics, Jinan Central Hospital Affiliated to Shandong University, Jinan 250000, China
| | - Yibiao Wang
- Department of Pediatrics, the Second Hospital of Shandong University, Jinan 250012, China
| | - Ou Chen
- Department of Pediatrics, the Second Hospital of Shandong University, Jinan 250012, China; Nursing School, Shandong University, Jinan 250012, China.
| | - Jiang Xue
- Department of Pediatrics, the Second Hospital of Shandong University, Jinan 250012, China.
| |
Collapse
|
24
|
Dong B, Yang Y, Zhang Z, Xie K, Su L, Yu Y. Hemopexin alleviates cognitive dysfunction after focal cerebral ischemia-reperfusion injury in rats. BMC Anesthesiol 2019; 19:13. [PMID: 30646866 PMCID: PMC6334464 DOI: 10.1186/s12871-019-0681-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Accepted: 01/04/2019] [Indexed: 01/15/2023] Open
Abstract
Background Ischemia-reperfusion (I/R) is a critical pathophysiological basis of cognitive dysfunction caused by ischemia stroke. Heme-oxygenase-1 (HO-1) is the rate-limiting enzyme for the elimination of excessive free heme by combining with hemopexin (HPX), a plasma protein that contributes to eliminating excessive free heme during ischemia stroke. This study aimed to elucidate whether HPX could alleviate cognitive dysfunction in rats subjected to cerebral I/R. Methods Rats were randomly divided into five groups: sham, MCAO, Vehicle, HPX and HPX + protoporphyrin IX (ZnPPIX). Cerebral I/R was induced by MCAO. Saline, vehicle, HPX and HPX + ZnPPIX were injected intracerebroventricularly at the moment after reperfusion. Morris water maze (MWM) test was used to detect the learning and cognitive function. Western blot was used to detect the expression of HO-1 in ischemic penumbra. CD31/vWF double labeling immunofluorescence was used to detect the neovascularization in the penumbra hippocampus. The structure and function of blood-brain barrier (BBB) was detected by the permeability of Evans Blue (EB), water content of the brain tissue, the Ang1/Ang2 and VE-cadherin expression. Results Our study verified that HPX improved the learning and memory capacity. Hemopexin up-regulated HO-1 protein expression, the average vessel density in the penumbra hippocampus and the VE- cadherin expression but decreased the permeability of EB, the water content of brain tissue and the ratio of Ang1/Ang2. The effects were reversed by ZnPPIX, an inhibitor of HO-1. Conclusion HPX can maintain the integrity of the blood-brain barrier and alleviate cognitive dysfunction after cerebral I/R through the HO-1 pathway.
Collapse
Affiliation(s)
- Beibei Dong
- Department of Anesthesiology, Tianjin Institute of Anesthesiology, General Hospital of Tianjin Medical University, No. 154 Anshan Road, Heping District, Tianjin, 300052, People's Republic of China
| | - Yongyan Yang
- Department of Anesthesiology, Tianjin Institute of Anesthesiology, General Hospital of Tianjin Medical University, No. 154 Anshan Road, Heping District, Tianjin, 300052, People's Republic of China
| | - Zhishen Zhang
- Department of Anesthesiology, Xiamen Medical University Zhongshan Hospital, Xiamen, 516211, China
| | - Keliang Xie
- Department of Anesthesiology, Tianjin Institute of Anesthesiology, General Hospital of Tianjin Medical University, No. 154 Anshan Road, Heping District, Tianjin, 300052, People's Republic of China.
| | - Lin Su
- Department of Anesthesiology, Tianjin Institute of Anesthesiology, General Hospital of Tianjin Medical University, No. 154 Anshan Road, Heping District, Tianjin, 300052, People's Republic of China
| | - Yonghao Yu
- Department of Anesthesiology, Tianjin Institute of Anesthesiology, General Hospital of Tianjin Medical University, No. 154 Anshan Road, Heping District, Tianjin, 300052, People's Republic of China.
| |
Collapse
|
25
|
Martinez RM, Ivan ALM, Vale DL, Campanini MZ, Ferreira VS, Steffen VS, Vicentini FTMC, Vilela FMP, Fonseca MJV, Baracat MM, Georgetti SR, Verri WA, Casagrande R. Topical emulsion containing pyrrolidine dithiocarbamate: effectiveness against ultraviolet B irradiation-induced injury of hairless mouse skin. J Pharm Pharmacol 2018; 70:1461-1473. [PMID: 30132896 DOI: 10.1111/jphp.12997] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Accepted: 07/21/2018] [Indexed: 01/08/2023]
Abstract
OBJECTIVES To evaluate the effects of a topical emulsion containing pyrrolidine dithiocarbamate (PDTC) (EcPDTC) in skin oxidative stress and inflammation triggered by ultraviolet B (UVB) irradiation (dose of 4.14 J/cm2 ). METHODS Hairless mouse received treatment with 0.5 g of EcPDTC or control emulsion (CTRLE) on the dorsal surface skin 12 h, 6 h and 5 min before and 6 h after the irradiation. Oxidative stress was evaluated by ferric reducing antioxidant power (FRAP), 2,2'-azino-bis (3-ethylbenzothiazoline-6-sulfonic acid) radical (ABTS) scavenging capacity, reduced glutathione quantitation, catalase activity, superoxide anion production and lipid peroxidation products. Inflammation parameters were as follows: skin oedema, myeloperoxidase activity (neutrophil marker), matrix metalloproteinase-9 activity, collagen fibre damage, mast cell and sunburn cell counts, and cytokine production. KEY FINDINGS Topical treatment with EcPDTC protected from UVB-induced skin injury by maintaining the antioxidant capacity levels similar to non-irradiated control group. Furthermore, EcPDTC inhibited UVB irradiation-induced superoxide anion production, lipid peroxidation and reduced skin inflammation by inhibiting skin oedema, neutrophil recruitment, metalloproteinase-9 activity, collagen fibre damage, mast cell and sunburn cell counts, and cytokine (TNF-α and IL-1β) production. CONCLUSIONS Topical treatment with EcPDTC improves antioxidant systems and inhibits inflammation, protecting the skin from the damaging effects of UVB irradiation.
Collapse
Affiliation(s)
- Renata M Martinez
- Department of Pharmaceutical Science, Health Science Center, State University of Londrina, Londrina, Brazil
| | - Ana L M Ivan
- Department of Pharmaceutical Science, Health Science Center, State University of Londrina, Londrina, Brazil
| | - David L Vale
- Department of Pharmaceutical Science, Health Science Center, State University of Londrina, Londrina, Brazil
| | - Marcela Z Campanini
- Department of Pharmaceutical Science, Health Science Center, State University of Londrina, Londrina, Brazil
| | - Vitor S Ferreira
- Department of Pharmaceutical Science, Health Science Center, State University of Londrina, Londrina, Brazil
| | - Vinicius S Steffen
- Department of Pharmaceutical Science, Health Science Center, State University of Londrina, Londrina, Brazil
| | - Fabiana T M C Vicentini
- Department of Pharmaceutical Science, Faculty of Pharmaceutical Sciences of Ribeirão Preto, University of Sao Paulo, Ribeirão Preto, Brazil
| | - Fernanda M P Vilela
- Department of Pharmaceutical Science, Faculty of Pharmaceutical Sciences of Ribeirão Preto, University of Sao Paulo, Ribeirão Preto, Brazil
| | - Maria J V Fonseca
- Department of Pharmaceutical Science, Faculty of Pharmaceutical Sciences of Ribeirão Preto, University of Sao Paulo, Ribeirão Preto, Brazil
| | - Marcela M Baracat
- Department of Pharmaceutical Science, Health Science Center, State University of Londrina, Londrina, Brazil
| | - Sandra R Georgetti
- Department of Pharmaceutical Science, Health Science Center, State University of Londrina, Londrina, Brazil
| | - Waldiceu A Verri
- Department of Pathology, Biological Science Center, State University of Londrina, Londrina, Brazil
| | - Rúbia Casagrande
- Department of Pharmaceutical Science, Health Science Center, State University of Londrina, Londrina, Brazil
| |
Collapse
|
26
|
Yang X, Chen S, Shao Z, Li Y, Wu H, Li X, Mao L, Zhou Z, Bai L, Mei X, Liu C. Apolipoprotein E Deficiency Exacerbates Spinal Cord Injury in Mice: Inflammatory Response and Oxidative Stress Mediated by NF-κB Signaling Pathway. Front Cell Neurosci 2018; 12:142. [PMID: 29875635 PMCID: PMC5974465 DOI: 10.3389/fncel.2018.00142] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Accepted: 05/09/2018] [Indexed: 12/30/2022] Open
Abstract
Spinal cord injury (SCI) is a severe neurological trauma that involves complex pathological processes. Inflammatory response and oxidative stress are prevalent during the second injury and can influence the functional recovery of SCI. Specially, Apolipoprotein E (APOE) induces neuronal repair and nerve regeneration, and the deficiency of Apoe impairs spinal cord-blood-barrier and reduces functional recovery after SCI. However, the mechanism by which Apoe mediates signaling pathways of inflammatory response and oxidative stress in SCI remains largely elusive. This study was designed to investigate the signaling pathways that regulate Apoe deficiency-dependent inflammatory response and oxidative stress in the acute stage of SCI. In the present study, Apoe-/- mice retarded functional recovery and had a larger lesion size when compared to wild-type mice after SCI. Moreover, deficiency of Apoe induced an exaggerated inflammatory response by increasing expression of interleukin-6 (IL-6) and interleukin-1β (IL-1β), and increased oxidative stress by reducing expression of Nrf2 and HO-1. Furthermore, lack of Apoe promoted neuronal apoptosis and decreased neuronal numbers in the anterior horn of the spinal cord after SCI. Mechanistically, we found that the absence of Apoe increased inflammation and oxidative stress through activation of NF-κB after SCI. In contrast, an inhibitor of nuclear factor-κB (NF-κB; Pyrrolidine dithiocarbamate) alleviates these changes. Collectively, these results indicate that a critical role for activation of NF-κB in regulating Apoe-deficiency dependent inflammation and oxidative stress is detrimental to recovery after SCI.
Collapse
Affiliation(s)
- Xuan Yang
- School of Nursing, Jinzhou Medical University, Jinzhou, China
| | - Shurui Chen
- Department of Orthopedics, First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Zhenya Shao
- Department of Orthopedics, First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Yuanlong Li
- Department of Orthopedics, First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - He Wu
- Department of Endocrinology, First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Xian Li
- Department of Orthopedics, Third Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Liang Mao
- Department of Oncology, First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Zipeng Zhou
- Department of Orthopedics, First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Liangjie Bai
- Department of Orthopedics, First Affiliated Hospital of China Medical University, Shenyang, China
| | - Xifan Mei
- Department of Orthopedics, First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Chang Liu
- Department of Endocrinology, First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| |
Collapse
|
27
|
Garaschuk O, Semchyshyn HM, Lushchak VI. Healthy brain aging: Interplay between reactive species, inflammation and energy supply. Ageing Res Rev 2018; 43:26-45. [PMID: 29452266 DOI: 10.1016/j.arr.2018.02.003] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Revised: 11/13/2017] [Accepted: 02/08/2018] [Indexed: 02/07/2023]
Abstract
Brains' high energy expenditure with preferable utilization of glucose and ketone bodies, defines the specific features of its energy homeostasis. The extensive oxidative metabolism is accompanied by a concomitant generation of high amounts of reactive oxygen, nitrogen, and carbonyl species, which will be here collectively referred to as RONCS. Such metabolism in combination with high content of polyunsaturated fatty acids creates specific problems in maintaining brains' redox homeostasis. While the levels of products of interaction between RONCS and cellular components increase slowly during the first two trimesters of individuals' life, their increase is substantially accelerated towards the end of life. Here we review the main mechanisms controlling the redox homeostasis of the mammalian brain, their age-dependencies as well as their adaptive potential, which might turn out to be much higher than initially assumed. According to recent data, the organism seems to respond to the enhancement of aging-related toxicity by forming a new homeostatic set point. Therefore, further research will focus on understanding the properties of the new set point(s), the general nature of this phenomenon and will explore the limits of brains' adaptivity.
Collapse
Affiliation(s)
- O Garaschuk
- Department of Neurophysiology, Institute of Physiology, University of Tübingen, 72074 Tübingen, Germany.
| | - H M Semchyshyn
- Department of Biochemistry and Biotechnology, Vasyl Stefanyk Precarpathian National University, 57 Shevchenko Str, Ivano-Frankivsk, 76018, Ukraine.
| | - V I Lushchak
- Department of Biochemistry and Biotechnology, Vasyl Stefanyk Precarpathian National University, 57 Shevchenko Str, Ivano-Frankivsk, 76018, Ukraine.
| |
Collapse
|
28
|
Ali T, Rehman SU, Shah FA, Kim MO. Acute dose of melatonin via Nrf2 dependently prevents acute ethanol-induced neurotoxicity in the developing rodent brain. J Neuroinflammation 2018; 15:119. [PMID: 29679979 PMCID: PMC5911370 DOI: 10.1186/s12974-018-1157-x] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 04/10/2018] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Melatonin is a well-known potent endogenous antioxidant pharmacological agent with significant neuroprotective actions. Here in the current study, we explored the nuclear factor erythroid 2-related factor 2 (Nrf2) gene-dependent antioxidant mechanism underlying the neuroprotective effects of the acute melatonin against acute ethanol-induced elevated reactive oxygen species (ROS)-mediated neuroinflammation and neurodegeneration in the developing rodent brain. METHODS In vivo rat pups were co-treated with a single dose of acute ethanol (5 g/kg, subcutaneous (S.C.)) and a single dose of acute melatonin (20 mg/kg, intraperitoneal (I.P.)). Four hours after a single S.C. and I.P. injections, all of the rat pups were sacrificed for further biochemical (Western blotting, ROS- assay, LPO-assay, and immunohistochemical) analyses. In order to corroborate the in vivo results, we used the in vitro murine-hippocampal HT22 and microglial BV2 cells, which were subjected to knockdown with small interfering RNA (siRNA) of Nrf2 genes and exposed with melatonin (100 μM) and ethanol (100 mM) and proceed for further biochemical analyses. RESULTS Our biochemical, immunohistochemical, and immunofluorescence results demonstrate that acute melatonin significantly upregulated the master endogenous antioxidant Nrf2 and heme oxygenase-1, consequently reversing the acute ethanol-induced elevated ROS and oxidative stress in the developing rodent brain, and in the murine-hippocampal HT22 and microglial BV2 cells. In addition, acute melatonin subsequently reduced the activated MAPK-p-P38-JNK pathways and attenuated neuroinflammation by decreasing the expression of activated gliosis and downregulated the p-NF-K-B/p-IKKβ pathway and decreased the expression levels of other inflammatory markers in the developing rodent brain and BV2 cells. Of note, melatonin acted through the Nrf2-dependent mechanism to attenuate neuronal apoptosis in the postnatal rodent brain and HT22 cells. Immunohistofluorescence results also showed that melatonin prevented ethanol-induced neurodegeneration in the developing rodent brain. The in vitro results indicated that melatonin induced neuroprotection via Nrf2-dependent manner and reduced ethanol-induced neurotoxicity. CONCLUSIONS The pleiotropic and potent neuroprotective antioxidant characteristics of melatonin, together with our in vivo and in vitro findings, suppose that acute melatonin could be beneficial to prevent and combat the acute ethanol-induced neurotoxic effects, such as elevated ROS, neuroinflammation, and neurodegeneration in the developing rodent brain.
Collapse
Affiliation(s)
- Tahir Ali
- Division of Applied Life Science (BK 21), College of Natural Sciences, Gyeongsang National University, Jinju, 660-701, Republic of Korea
| | - Shafiq Ur Rehman
- Division of Applied Life Science (BK 21), College of Natural Sciences, Gyeongsang National University, Jinju, 660-701, Republic of Korea
| | - Fawad Ali Shah
- Department of Pharmacology, Riphah Institute of Pharmaceutical Sciences, Riphah International University, Islamabad, Pakistan
| | - Myeong Ok Kim
- Division of Applied Life Science (BK 21), College of Natural Sciences, Gyeongsang National University, Jinju, 660-701, Republic of Korea.
| |
Collapse
|
29
|
Ren HL, Lv CN, Xing Y, Geng Y, Zhang F, Bu W, Wang MW. Downregulated Nuclear Factor E2-Related Factor 2 (Nrf2) Aggravates Cognitive Impairments via Neuroinflammation and Synaptic Plasticity in the Senescence-Accelerated Mouse Prone 8 (SAMP8) Mouse: A Model of Accelerated Senescence. Med Sci Monit 2018; 24:1132-1144. [PMID: 29474348 PMCID: PMC5833362 DOI: 10.12659/msm.908954] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Background We observed the effects of nuclear factor E2-related factor 2 (Nrf2) downregulation via intrahippocampal injection of a lentiviral vector on cognition in senescence-accelerated mouse prone 8 (SAMP8) to investigate the role of the (Nrf2)/antioxidant response element (ARE) pathway in age-related changes. Material/Methods Control lentivirus and Nrf2-shRNA-lentivirus were separately injected into the hippocampus of 4-month-old SAMR1 and SAMP8 mice and then successfully downregulated Nrf2 expression in this brain region. Five months later, cognitive function tests, including the novel object test, the Morris water maze test, and the passive avoidance task were conducted. Glial fibrillary acidic protein (GFAP) and ionized calcium-binding adapter molecule 1 (Iba1) immunohistochemistry was performed to observe an inflammatory response. Presynaptic synapsin (SYN) were observed by immunofluorescence. We then determined the Nrf2-regulated, heme oxygenase-1 (HO-1), P65, postsynaptic density protein (PSD), and SYN protein levels. The ultrastructure of neurons and synapses in the hippocampal CA1 region was observed by transmission electron microscopy. Results Aging led to a decline in cognitive function compared with SAMR1 mice and the Nrf2-shRNA-lentivirus further exacerbated the cognitive impairment in SAMP8 mice. Nrf2, HO-1, PSD, and SYN levels were significantly reduced (all P<0.05) but high levels of inflammation were detected in SAMP8 mice with low expression of Nrf2. Furthermore, neurons were vacuolated, the number of organelles decreased, and the number of synapses decreased. Conclusions Downregulation of Nrf2 suppressed the Nrf2/ARE pathway, activated oxidative stress and neuroinflammation, and accelerated cognitive impairment in SAMP8 mice. Downregulation of Nrf2 accelerates the aging process through neuroinflammation and synaptic plasticity.
Collapse
Affiliation(s)
- Hui Ling Ren
- Department of Neurology, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China (mainland)
| | - Chao Nan Lv
- Department of Neurology, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei, China (mainland).,Brain Aging and Cognitive Neuroscience Key Laboratory of Hebei, Shijiazhuang, Hebei, China (mainland)
| | - Ying Xing
- Department of Rehabilitation Medicine, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China (mainland)
| | - Yuan Geng
- Department of Neurology, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei, China (mainland).,Brain Aging and Cognitive Neuroscience Key Laboratory of Hebei, Shijiazhuang, Hebei, China (mainland)
| | - Feng Zhang
- Department of Rehabilitation Medicine, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China (mainland)
| | - Wei Bu
- Department of Neurosurgery, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China (mainland)
| | - Ming Wei Wang
- Department of Neurology, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei, China (mainland).,Brain Aging and Cognitive Neuroscience Key Laboratory of Hebei, Shijiazhuang, Hebei, China (mainland)
| |
Collapse
|
30
|
Zhang K, Dong R, Sun K, Wang X, Wang J, Yang CS, Zhang J. Synergistic toxicity of epigallocatechin-3-gallate and diethyldithiocarbamate, a lethal encounter involving redox-active copper. Free Radic Biol Med 2017; 113:143-156. [PMID: 28974447 DOI: 10.1016/j.freeradbiomed.2017.09.027] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Revised: 09/09/2017] [Accepted: 09/27/2017] [Indexed: 01/20/2023]
Abstract
Dithiocarbamates (DTC) are widely used in agricultural, industrial and therapeutic domains. There are ample opportunities for human exposure to DTC. Green tea extracts, with epigallocatechin-3-gallate (EGCG) being the most abundant constituent, have been used as dietary supplements for body weight reduction. Our hypothesis is that DTC can act as a copper ionophore to increase hepatic levels of redox-active copper which promotes EGCG auto-oxidation to produce oxidative stress and toxicity. The results of the present study in a mouse model is consistent with this hypothesis, showing that co-administration of EGCG and diethyldithiocarbamate - a metabolite of disulfiram (a drug for alcohol aversion therapy), both at tolerable levels, caused lethality. The liver was the major organ site of toxicity. The co-administration drastically increased lipid peroxidation, DNA damage and cell apoptosis as well as caused deleterious transcriptional responses including basal and Nrf2 antioxidant systems in the liver. The results suggest that exposure to DTC reduces toxic threshold of dietary polyphenols from green tea and possibly other plants, and vice versa. This novel hypothesis is important to human health, and the dose-response relationship of this synergistic toxicity needs to be further characterized.
Collapse
Affiliation(s)
- Ke Zhang
- State Key Laboratory of Tea Plant Biology and Utilization, School of Tea & Food Science, Anhui Agricultural University, Hefei, Anhui, China
| | - Ruixia Dong
- State Key Laboratory of Tea Plant Biology and Utilization, School of Tea & Food Science, Anhui Agricultural University, Hefei, Anhui, China
| | - Kang Sun
- State Key Laboratory of Tea Plant Biology and Utilization, School of Tea & Food Science, Anhui Agricultural University, Hefei, Anhui, China
| | - Xiaoxiao Wang
- State Key Laboratory of Tea Plant Biology and Utilization, School of Tea & Food Science, Anhui Agricultural University, Hefei, Anhui, China
| | - Jiajia Wang
- State Key Laboratory of Tea Plant Biology and Utilization, School of Tea & Food Science, Anhui Agricultural University, Hefei, Anhui, China
| | - Chung S Yang
- Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ, USA; International Joint Research Laboratory of Tea Chemistry and Health Effects, Anhui Agricultural University, Hefei, Anhui, China
| | - Jinsong Zhang
- State Key Laboratory of Tea Plant Biology and Utilization, School of Tea & Food Science, Anhui Agricultural University, Hefei, Anhui, China; International Joint Research Laboratory of Tea Chemistry and Health Effects, Anhui Agricultural University, Hefei, Anhui, China.
| |
Collapse
|
31
|
Are Astrocytes the Predominant Cell Type for Activation of Nrf2 in Aging and Neurodegeneration? Antioxidants (Basel) 2017; 6:antiox6030065. [PMID: 28820437 PMCID: PMC5618093 DOI: 10.3390/antiox6030065] [Citation(s) in RCA: 114] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Revised: 08/11/2017] [Accepted: 08/16/2017] [Indexed: 12/29/2022] Open
Abstract
Nuclear factor erythroid 2-related factor 2 (Nrf2) is a transcription factor that regulates hundreds of antioxidant genes, and is activated in response to oxidative stress. Given that many neurodegenerative diseases including Alzheimer’s disease, Parkinson’s disease, amyotrophic lateral sclerosis, Huntington’s disease and multiple sclerosis are characterised by oxidative stress, Nrf2 is commonly activated in these diseases. Evidence demonstrates that Nrf2 activity is repressed in neurons in vitro, and only cultured astrocytes respond strongly to Nrf2 inducers, leading to the interpretation that Nrf2 signalling is largely restricted to astrocytes. However, Nrf2 activity can be observed in neurons in post-mortem brain tissue and animal models of disease. Thus this interpretation may be false, and a detailed analysis of the cell type expression of Nrf2 in neurodegenerative diseases is required. This review describes the evidence for Nrf2 activation in each cell type in prominent neurodegenerative diseases and normal aging in human brain and animal models of neurodegeneration, the response to pharmacological and genetic modulation of Nrf2, and clinical trials involving Nrf2-modifying drugs.
Collapse
|
32
|
Zhang H, Zhou L, Yuen J, Birkner N, Leppert V, O'Day PA, Forman HJ. Delayed Nrf2-regulated antioxidant gene induction in response to silica nanoparticles. Free Radic Biol Med 2017; 108:311-319. [PMID: 28389405 PMCID: PMC5480609 DOI: 10.1016/j.freeradbiomed.2017.04.002] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Revised: 03/29/2017] [Accepted: 04/01/2017] [Indexed: 12/14/2022]
Abstract
Silica nanoparticles with iron on their surface cause the production of oxidants and stimulate an inflammatory response in macrophages. Nuclear factor erythroid-derived 2 - like factor 2 (Nrf2) signaling and its regulated antioxidant genes play critical roles in maintaining redox homeostasis. In this study we investigated the regulation of four representative Nrf2-regulated antioxidant genes; i.e., glutamate cysteine ligase (GCL) catalytic subunit (GCLC), GCL modifier subunit (GCLM), heme oxygenase 1 (HO-1), and NAD(P)H:quinone oxidoreductase-1 (NQO-1), by iron-coated silica nanoparticles (SiO2-Fe) in human THP-1 macrophages. We found that the expression of these four antioxidant genes was modified by SiO2-Fe in a time-dependent manner. At 6h, their expression was unchanged except for GCLC, which was reduced compared with controls. At 18h, the expression of these antioxidant genes was significantly increased compared with controls. In contrast, the Nrf2 activator sulforaphane induced all antioxidant genes at as early as 3h. The nuclear translocation of Nrf2 occurred later than that for NF-κB p65 protein and the induction of proinflammatory cytokines (TNFα and IL-1β). NF-κB inhibitor SN50 prevented the reduction of GCLC at 6h and abolished the induction of antioxidant genes at 18h by SiO2-Fe, but did not affect the basal and sulforaphane-induced expression of antioxidant genes, suggesting that NF-κB signaling plays a key role in the induction of Nrf2-mediated genes in response to SiO2-Fe. Consistently, SN50 inhibited the nuclear translocation of Nrf2 caused by SiO2-Fe. In addition, Nrf2 silencing decreased the basal and SiO2-induced expression of the four reprehensive antioxidant genes. Taken together, these data indicated that SiO2-Fe induced a delayed response of Nrf2-regulated antioxidant genes, likely through NF-κB-Nrf2 interactions.
Collapse
Affiliation(s)
- Hongqiao Zhang
- Leonard Davies School of Gerontology, University of Southern California, Los Angeles, CA 90089, United States.
| | - Lulu Zhou
- Leonard Davies School of Gerontology, University of Southern California, Los Angeles, CA 90089, United States
| | - Jenay Yuen
- Leonard Davies School of Gerontology, University of Southern California, Los Angeles, CA 90089, United States
| | - Nancy Birkner
- School of Natural Sciences and Sierra Nevada Research Institute, University of California at Merced, Merced, CA 95343, United States
| | - Valerie Leppert
- School of Engineering, University of California at Merced, Merced, CA 95343, United States
| | - Peggy A O'Day
- School of Natural Sciences and Sierra Nevada Research Institute, University of California at Merced, Merced, CA 95343, United States
| | - Henry Jay Forman
- Leonard Davies School of Gerontology, University of Southern California, Los Angeles, CA 90089, United States
| |
Collapse
|
33
|
Liddell JR, White AR. Nexus between mitochondrial function, iron, copper and glutathione in Parkinson's disease. Neurochem Int 2017; 117:126-138. [PMID: 28577988 DOI: 10.1016/j.neuint.2017.05.016] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Revised: 05/26/2017] [Accepted: 05/30/2017] [Indexed: 01/26/2023]
Abstract
Parkinson's disease is neuropathologically characterised by loss of catecholamine neurons in vulnerable brain regions including substantia nigra pars compacta and locus coeruleus. This review discusses how the susceptibility of these regions is defined by their shared biochemical characteristics that differentiate them from other neurons. Parkinson's disease is biochemically characterised by mitochondrial dysfunction, accumulation of iron, diminished copper content and depleted glutathione levels in these regions. This review also discusses this neuropathology, and provides evidence for how these pathological features are mechanistically linked to each other. This leads to the conclusion that disruption of mitochondrial function, or iron, copper or glutathione metabolism in isolation provokes the pathological impairment of them all. This creates a vicious cycle that drives pathology leading to mitochondrial failure and neuronal cell death in vulnerable brain regions.
Collapse
Affiliation(s)
- Jeffrey R Liddell
- Department of Pathology, The University of Melbourne, Victoria 3010, Australia.
| | - Anthony R White
- Cell and Molecular Biology, QIMR Berghofer Medical Research Institute, Herston, Queensland 4006, Australia
| |
Collapse
|
34
|
Moujalled D, Grubman A, Acevedo K, Yang S, Ke YD, Moujalled DM, Duncan C, Caragounis A, Perera ND, Turner BJ, Prudencio M, Petrucelli L, Blair I, Ittner LM, Crouch PJ, Liddell JR, White AR. TDP-43 mutations causing amyotrophic lateral sclerosis are associated with altered expression of RNA-binding protein hnRNP K and affect the Nrf2 antioxidant pathway. Hum Mol Genet 2017; 26:1732-1746. [PMID: 28334913 DOI: 10.1093/hmg/ddx093] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Accepted: 03/07/2017] [Indexed: 12/12/2022] Open
Abstract
TAR DNA binding protein 43 (TDP-43) is a major disease-associated protein involved in the pathogenesis of amyotrophic lateral sclerosis (ALS) and frontotemporal lobar degeneration with ubiquitin-positive inclusions (FTLD-U). Our previous studies found a direct association between TDP-43 and heterogeneous nuclear ribonucleoprotein K (hnRNP K). In this study, utilizing ALS patient fibroblasts harboring a TDP-43M337V mutation and NSC-34 motor neuronal cell line expressing TDP-43Q331K mutation, we show that hnRNP K expression is impaired in urea soluble extracts from mutant TDP-43 cell models. This was confirmed in vivo using TDP-43Q331K and inducible TDP-43A315T murine ALS models. We further investigated the potential pathological effects of mutant TDP-43-mediated changes to hnRNP K metabolism by RNA binding immunoprecipitation analysis. hnRNP K protein was bound to antioxidant NFE2L2 transcripts encoding Nrf2 antioxidant transcription factor, with greater enrichment in TDP-43M337V patient fibroblasts compared to healthy controls. Subsequent gene expression profiling revealed an increase in downstream antioxidant transcript expression of Nrf2 signaling in the spinal cord of TDP-43Q331K mice compared to control counterparts, yet the corresponding protein expression was not up-regulated in transgenic mice. Despite the elevated expression of antioxidant transcripts, we observed impaired levels of glutathione (downstream Nrf2 antioxidant) in TDP-43M337V patient fibroblasts and astrocyte cultures from TDP-43Q331K mice, indicative of elevated oxidative stress and failure of some upregulated antioxidant genes to be translated into protein. Our findings indicate that further exploration of the interplay between hnRNP K (or other hnRNPs) and Nrf2-mediated antioxidant signaling is warranted and may be an important driver for motor neuron degeneration in ALS.
Collapse
Affiliation(s)
- Diane Moujalled
- Department of Pathology, The University of Melbourne, Victoria 3010, Australia
| | - Alexandra Grubman
- Department of Pathology, The University of Melbourne, Victoria 3010, Australia
| | - Karla Acevedo
- Department of Pathology, The University of Melbourne, Victoria 3010, Australia
| | - Shu Yang
- The Australian School of Advanced Medicine, Macquarie University, NSW 2109, Australia
| | - Yazi D Ke
- Dementia Research Unit, Department of Anatomy, Faculty of Medicine, School of Medical Sciences, UNSW Australia, Sydney, NSW 2052, Australia
| | - Donia M Moujalled
- Australian Centre for Blood Diseases (ACBD), The Alfred Centre, Victoria 3004, Australia
| | - Clare Duncan
- Florey Institute of Neuroscience and Mental Health, Parkville, Victoria 3010, Australia
| | | | - Nirma D Perera
- Florey Institute of Neuroscience and Mental Health, Parkville, Victoria 3010, Australia
| | - Bradley J Turner
- Florey Institute of Neuroscience and Mental Health, Parkville, Victoria 3010, Australia
| | | | | | - Ian Blair
- The Australian School of Advanced Medicine, Macquarie University, NSW 2109, Australia
| | - Lars M Ittner
- Dementia Research Unit, Department of Anatomy, Faculty of Medicine, School of Medical Sciences, UNSW Australia, Sydney, NSW 2052, Australia
| | - Peter J Crouch
- Department of Pathology, The University of Melbourne, Victoria 3010, Australia
- Florey Institute of Neuroscience and Mental Health, Parkville, Victoria 3010, Australia
| | - Jeffrey R Liddell
- Department of Pathology, The University of Melbourne, Victoria 3010, Australia
| | - Anthony R White
- Department of Pathology, The University of Melbourne, Victoria 3010, Australia
- Florey Institute of Neuroscience and Mental Health, Parkville, Victoria 3010, Australia
- Berghofer Medical Research Institute, Herston, Queensland 4006, Australia
| |
Collapse
|
35
|
You W, Zuo G, Shen H, Tian X, Li H, Zhu H, Yin J, Zhang T, Wang Z. Potential dual role of nuclear factor-kappa B in experimental subarachnoid hemorrhage-induced early brain injury in rabbits. Inflamm Res 2016; 65:975-984. [PMID: 27554683 DOI: 10.1007/s00011-016-0980-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Revised: 07/18/2016] [Accepted: 08/11/2016] [Indexed: 12/17/2022] Open
Abstract
OBJECTIVE AND DESIGN Nuclear factor-kappa B (NF-κB) has multiple physiological and pathological functions. The role of NF-κB can be protective or destructive. We aim to investigate the biphasic activation of NF-κB in brain after subarachnoid hemorrhage (SAH). MATERIAL OR SUBJECTS Eighty male New Zealand rabbits are assigned to control, SAH, vehicle, and pyrrolidine dithiocarbamate (PDTC) groups. TREATMENT PDTC (3 mg/kg, dissolved in saline) was injected into cisterna magna. METHODS Immunofluorescence and electrophoretic mobility shift assay experiments were performed to assess the activation of NF-κB. The levels of inflammatory and apoptosis mediators were detected by ELISA and real-time polymerase chain reaction. Nissl and immunofluorescent stain was performed to evaluate neuron injury. RESULTS NF-κB activity in the brain cortex showed two peaks after SAH. Inflammatory mediators exhibited similar time course. PDTC could significantly inhibit the NF-κB activity and inflammatory mediators. Suppressing the early NF-κB activity significantly decreased neuron injury, while inhibiting the late one could statistically increase neuron injury. CONCLUSIONS The biphasic NF-κB activation in the brain cortex after SAH played a decisive role on neuronal fate through the inflammatory signaling pathway. The early NF-κB activity contributed to neuron damage after SAH. Nevertheless, the late activated NF-κB may serve as a protector.
Collapse
Affiliation(s)
- Wanchun You
- Department of Neurosurgery and Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, China
| | - Gang Zuo
- Department of Neurosurgery and Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, China.,Department of Neurosurgery, The First People's Hospital of Taicang City, Taicang, 215400, China
| | - Haitao Shen
- Department of Neurosurgery and Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, China
| | - Xiaodi Tian
- Department of Neurosurgery and Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, China
| | - Haiying Li
- Department of Neurosurgery and Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, China
| | - Haiping Zhu
- Department of Neurosurgery, Changshu No. 1 People's Hospital, Changshu, 215500, China.
| | - Jun Yin
- Department of Neurosurgery, Taixing Chinese Medicine Hospital, Taixing, 225400, China.
| | - Tiejun Zhang
- Department of Neurosurgery and Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, China
| | - Zhong Wang
- Department of Neurosurgery and Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, China
| |
Collapse
|
36
|
Pinho-Ribeiro FA, Fattori V, Zarpelon AC, Borghi SM, Staurengo-Ferrari L, Carvalho TT, Alves-Filho JC, Cunha FQ, Cunha TM, Casagrande R, Verri WA. Pyrrolidine dithiocarbamate inhibits superoxide anion-induced pain and inflammation in the paw skin and spinal cord by targeting NF-κB and oxidative stress. Inflammopharmacology 2016; 24:97-107. [PMID: 27160222 DOI: 10.1007/s10787-016-0266-3] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Accepted: 04/28/2016] [Indexed: 12/30/2022]
Abstract
We evaluated the effect of pyrrolidine dithiocarbamate (PDTC) in superoxide anion-induced inflammatory pain. Male Swiss mice were treated with PDTC and stimulated with an intraplantar or intraperitoneal injection of potassium superoxide, a superoxide anion donor. Subcutaneous PDTC treatment attenuated mechanical hyperalgesia, thermal hyperalgesia, paw oedema and leukocyte recruitment (neutrophils and macrophages). Intraplantar injection of superoxide anion activated NF-κB and increased cytokine production (IL-1β, TNF-α and IL-10) and oxidative stress (nitrite and lipid peroxidation levels) at the primary inflammatory foci and in the spinal cord (L4-L6). PDTC treatment inhibited superoxide anion-induced NF-κB activation, cytokine production and oxidative stress in the paw and spinal cord. Furthermore, intrathecal administration of PDTC successfully inhibited superoxide anion-induced mechanical hyperalgesia, thermal hyperalgesia and inflammatory response in peripheral foci (paw). These results suggest that peripheral stimulus with superoxide anion activates the local and spinal cord oxidative- and NF-κB-dependent inflammatory nociceptive mechanisms. PDTC targets these events, therefore, inhibiting superoxide anion-induced inflammatory pain in mice.
Collapse
Affiliation(s)
- Felipe A Pinho-Ribeiro
- Departamento de Ciências Patológicas, Centro de Ciências Biológicas, Universidade Estadual de Londrina, Rod. Celso Garcia Cid PR445 KM380, 86057-970, Londrina, Paraná, Brazil
| | - Victor Fattori
- Departamento de Ciências Patológicas, Centro de Ciências Biológicas, Universidade Estadual de Londrina, Rod. Celso Garcia Cid PR445 KM380, 86057-970, Londrina, Paraná, Brazil
| | - Ana C Zarpelon
- Departamento de Ciências Patológicas, Centro de Ciências Biológicas, Universidade Estadual de Londrina, Rod. Celso Garcia Cid PR445 KM380, 86057-970, Londrina, Paraná, Brazil
| | - Sergio M Borghi
- Departamento de Ciências Patológicas, Centro de Ciências Biológicas, Universidade Estadual de Londrina, Rod. Celso Garcia Cid PR445 KM380, 86057-970, Londrina, Paraná, Brazil
| | - Larissa Staurengo-Ferrari
- Departamento de Ciências Patológicas, Centro de Ciências Biológicas, Universidade Estadual de Londrina, Rod. Celso Garcia Cid PR445 KM380, 86057-970, Londrina, Paraná, Brazil
| | - Thacyana T Carvalho
- Departamento de Ciências Patológicas, Centro de Ciências Biológicas, Universidade Estadual de Londrina, Rod. Celso Garcia Cid PR445 KM380, 86057-970, Londrina, Paraná, Brazil
| | - Jose C Alves-Filho
- Departamento de Ciências Patológicas, Centro de Ciências Biológicas, Universidade Estadual de Londrina, Rod. Celso Garcia Cid PR445 KM380, 86057-970, Londrina, Paraná, Brazil
| | - Fernando Q Cunha
- Departamento de Ciências Patológicas, Centro de Ciências Biológicas, Universidade Estadual de Londrina, Rod. Celso Garcia Cid PR445 KM380, 86057-970, Londrina, Paraná, Brazil
| | - Thiago M Cunha
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Av. Bandeirantes, 3900, 14049-900, Ribeirão Preto, São Paulo, Brazil
| | - Rubia Casagrande
- Departamento de Ciências Farmacêuticas, Centro de Ciências da Saúde, Hospital Universitário, Universidade Estadual de Londrina, Av. Robert Koch, 60, 86038-350, Londrina, Paraná, Brazil
| | - Waldiceu A Verri
- Departamento de Ciências Patológicas, Centro de Ciências Biológicas, Universidade Estadual de Londrina, Rod. Celso Garcia Cid PR445 KM380, 86057-970, Londrina, Paraná, Brazil.
| |
Collapse
|