101
|
Pentoxifylline Enhances Antioxidative Capability and Promotes Mitochondrial Biogenesis in D-Galactose-Induced Aging Mice by Increasing Nrf2 and PGC-1 α through the cAMP-CREB Pathway. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:6695613. [PMID: 34257818 PMCID: PMC8245236 DOI: 10.1155/2021/6695613] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 04/21/2021] [Accepted: 05/13/2021] [Indexed: 02/07/2023]
Abstract
Aging is a complex phenomenon associated with oxidative stress and mitochondrial dysfunction. The objective of this study was to investigate the potential ameliorative effects of the phosphodiesterase inhibitor pentoxifylline (PTX) on the aging process and its underlying mechanisms. We treated D-galactose- (D-gal-) induced aging mice with PTX and measured the changes in behavior, degree of oxidative damage, and mitochondrial ultrastructure and content as well as the expression of nuclear factor erythroid 2-related factor 2- (Nrf2-) mediated antioxidant genes and peroxisome proliferator-activated receptor-gamma coactivator 1-alpha- (PGC-1α-) dependent mitochondrial biogenesis genes. The results demonstrated that PTX improved cognitive deficits, reduced oxidative damage, ameliorated abnormal mitochondrial ultrastructure, increased mitochondrial content and Nrf2 activation, and upregulated antioxidant and mitochondrial biogenesis gene expression in the hippocampus of wild-type aging mice. However, the above antiaging effects of PTX were obviously decreased in the brains of Nrf2-deficient D-gal-induced aging mice. Moreover, in hydrogen peroxide-treated SH-SY5Y cells, we found that cyclic adenosine monophosphate (cAMP) response element-binding protein (CREB) and Nrf2/PGC-1α act in a linear way by CREB siRNA transfection. Thus, PTX administration improved the aging-related decline in brain function by enhancing antioxidative capability and promoting mitochondrial biogenesis, which might depend on increasing Nrf2 and PGC-1α by activating the cAMP-CREB pathway.
Collapse
|
102
|
Liu T, Lv YF, Zhao JL, You QD, Jiang ZY. Regulation of Nrf2 by phosphorylation: Consequences for biological function and therapeutic implications. Free Radic Biol Med 2021; 168:129-141. [PMID: 33794311 DOI: 10.1016/j.freeradbiomed.2021.03.034] [Citation(s) in RCA: 94] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 03/22/2021] [Accepted: 03/25/2021] [Indexed: 12/18/2022]
Abstract
The transcription factor nuclear factor erythroid-derived 2-like 2 (NRF2) participates in the activation of the antioxidant cytoprotective pathway and other important physiological processes to maintain cellular homeostasis. The dysregulation of NRF2 activity plays a role in various diseases, such as cardiovascular diseases, neurodegenerative diseases, and cancer. Thus, NRF2 activity is tightly regulated through multiple mechanisms, among which phosphorylation by kinases is critical in the posttranslational regulation of NRF2. For instance, PKC, casein kinase 2, and AMP-activated kinase positively, while GSK-3 negatively regulates NRF2 activity through phosphorylation of different sites. Here, we provide an overview of the phosphorylation regulation pattern of NRF2 and discuss the therapeutic potential of interventions targeting NRF2 phosphorylation.
Collapse
Affiliation(s)
- Tian Liu
- State Key Laboratory of Natural Medicines, And Jiang Su Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, 210009, China
| | - Yi-Fei Lv
- State Key Laboratory of Natural Medicines, And Jiang Su Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, 210009, China
| | - Jing-Long Zhao
- State Key Laboratory of Natural Medicines, And Jiang Su Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, 210009, China
| | - Qi-Dong You
- State Key Laboratory of Natural Medicines, And Jiang Su Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China.
| | - Zheng-Yu Jiang
- State Key Laboratory of Natural Medicines, And Jiang Su Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China.
| |
Collapse
|
103
|
Sousa A, Ribeiro D, Fernandes E, Freitas M. The Effect of Chalcones on the Main Sources of Reactive Species Production: Possible Therapeutic Implications in Diabetes Mellitus. Curr Med Chem 2021; 28:1625-1669. [PMID: 32448100 DOI: 10.2174/0929867327666200525010007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 03/24/2020] [Accepted: 03/25/2020] [Indexed: 11/22/2022]
Abstract
Diabetes mellitus (DM) is characterized by hyperglycaemia, resulting from defects in insulin secretion, insulin action or both. There are several factors such as hyperlipidemia and oxidative stress (OS), namely the production of reactive oxygen/nitrogen species (ROS/RNS), that actively contribute to the development and worsening of DM. Chalcones, also termed as benzalacetophenone or benzylidene acetophenone, present a 1,3-diaryl-2-propen-1-one scaffold that has been shown to be highly promising in the development of new antioxidant compounds. Considering the potential interest of antioxidant therapy, the present review scrutinizes the role of the main sources of ROS/RNS production during DM. The modulatory effect of chalcones against nicotinamide adenine dinucleotide phosphate (NADPH) oxidase, xanthine oxidase, mitochondrial respiratory chain and nitric oxide synthase, is also thoroughly discussed, establishing, whenever possible, a structure-activity relationship (SAR). From the SAR analysis, it can be stated that the presence of catechol groups, hydroxyl and methoxyl substituents in the chalcones scaffold improves their modulatory activity against the main sources of ROS/RNS production in DM.
Collapse
Affiliation(s)
- Adelaide Sousa
- LAQV, REQUIMTE, Laboratory of Applied Chemistry, Department of Chemical, Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Daniela Ribeiro
- LAQV, REQUIMTE, Laboratory of Applied Chemistry, Department of Chemical, Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Eduarda Fernandes
- LAQV, REQUIMTE, Laboratory of Applied Chemistry, Department of Chemical, Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Marisa Freitas
- LAQV, REQUIMTE, Laboratory of Applied Chemistry, Department of Chemical, Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| |
Collapse
|
104
|
Hushpulian DM, Ammal Kaidery N, Ahuja M, Poloznikov AA, Sharma SM, Gazaryan IG, Thomas B. Challenges and Limitations of Targeting the Keap1-Nrf2 Pathway for Neurotherapeutics: Bach1 De-Repression to the Rescue. Front Aging Neurosci 2021; 13:673205. [PMID: 33897412 PMCID: PMC8060438 DOI: 10.3389/fnagi.2021.673205] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Accepted: 03/15/2021] [Indexed: 12/30/2022] Open
Abstract
The Keap1-Nrf2 signaling axis is a validated and promising target for cellular defense and survival pathways. This minireview discusses the potential off-target effects and their impact on future drug development originating from Keap1-targeting small molecules that function as displacement activators of the redox-sensitive transcription factor Nrf2. We argue that small-molecule displacement activators, similarly to electrophiles, will release both Nrf2 and other Keap1 client proteins from the ubiquitin ligase complex. This non-specificity is likely unavoidable and may result in off-target effects during Nrf2 activation by targeting Keap1. The small molecule displacement activators may also target Kelch domains in proteins other than Keap1, causing additional off-target effects unless designed to ensure specificity for the Kelch domain only in Keap1. A potentially promising and alternative therapeutic approach to overcome this non-specificity emerging from targeting Keap1 is to inhibit the Nrf2 repressor Bach1 for constitutive activation of the Nrf2 pathway and bypass the Keap1-Nrf2 complex.
Collapse
Affiliation(s)
- Dmitry M. Hushpulian
- P. A. Hertsen Moscow Oncology Research Center, Branch of the National Medical Research Radiological Center, Ministry of Health of the Russian Federation, Moscow, Russia
- Faculty of Biology and Biotechnologies, Higher School of Economics, Moscow, Russia
| | - Navneet Ammal Kaidery
- Darby Children’s Research Institute, Medical University of South Carolina, Charleston, SC, United States
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC, United States
| | - Manuj Ahuja
- Darby Children’s Research Institute, Medical University of South Carolina, Charleston, SC, United States
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC, United States
- Department of Pharmaceutical Sciences, University at Buffalo, Buffalo, NY, United States
| | - Andrey A. Poloznikov
- Faculty of Biology and Biotechnologies, Higher School of Economics, Moscow, Russia
| | - Sudarshana M. Sharma
- Hollings Cancer Center, Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC, United States
| | - Irina G. Gazaryan
- P. A. Hertsen Moscow Oncology Research Center, Branch of the National Medical Research Radiological Center, Ministry of Health of the Russian Federation, Moscow, Russia
- Faculty of Biology and Biotechnologies, Higher School of Economics, Moscow, Russia
- Department of Chemical Enzymology, M.V. Lomonosov Moscow State University, Moscow, Russia
- Department of Chemistry and Physical Sciences, Dyson College of Arts and Sciences, Pace University, Pleasantville, NY, United States
| | - Bobby Thomas
- Darby Children’s Research Institute, Medical University of South Carolina, Charleston, SC, United States
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC, United States
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, United States
- Department of Drug Discovery, Medical University of South Carolina, Charleston, SC, United States
| |
Collapse
|
105
|
Naeem K, Tariq Al Kury L, Nasar F, Alattar A, Alshaman R, Shah FA, Khan AU, Li S. Natural Dietary Supplement, Carvacrol, Alleviates LPS-Induced Oxidative Stress, Neurodegeneration, and Depressive-Like Behaviors via the Nrf2/HO-1 Pathway. J Inflamm Res 2021; 14:1313-1329. [PMID: 33854358 PMCID: PMC8041651 DOI: 10.2147/jir.s294413] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 03/01/2021] [Indexed: 12/20/2022] Open
Abstract
PURPOSE Major depressive disorder (MDD) is a debilitating human health condition characterized by mood swings and is associated with a high probability of suicide attempts. Several studies have reported a role of neuroinflammation in MMD, yet the efficacy of natural drug substances on neuroinflammation-associated depression has not been well-investigated. The present study examined the neuroprotective effects of carvacrol on lipopolysaccharide (LPS)-induced neuroinflammation, depression, and anxiety-like behavior. METHODS Male Sprague Dawley rats were divided into two experimental cohorts to determine the effects and the effective dose of carvacrol (whether 20 mg/kg or 50 mg/kg), and further demonstrate the mechanism of action of nuclear factor E2-related factor (Nrf2) in depression. RESULTS We found marked neuronal alterations in the cortex and hippocampus of LPS-intoxicated animals that were associated with higher inflammatory cytokine expression such as cyclooxygenase (COX2), tumor necrosis factor-alpha (TNF-α), and c-Jun N-terminal kinase (p-JNK). These detrimental effects exacerbated oxidative stress, as documented by a compromised antioxidant system due to high lipid peroxidase (LPO). Carvacrol (20 mg/kg) significantly reverted these changes by positively modulating the antioxidant gene Nrf2, a master regulator of the downstream antioxidant pathway. To further investigate the role of Nrf2, an inhibitor of Nrf2 called all-trans retinoic acid (ATRA) was used, which further exacerbated LPS toxicity with a higher oxidative and inflammatory cytokine level. To further support our notion, we performed virtual docking of carvacrol with the Nrf2-Keap1 target and the resultant drug-protein interactions validated the in vivo findings. CONCLUSION Collectively, our findings suggest that carvacrol (20 mg/kg) could activate the endogenous master antioxidant Nrf2, which further regulates the expression of downstream antioxidants, eventually ameliorating LPS-induced neuroinflammation and neurodegeneration.
Collapse
Affiliation(s)
- Komal Naeem
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Riphah International University, Islamabad, 747424, Pakistan
- State Key Laboratory of Oncogenomics, School of Chemical Biology and Biotechnology, Shenzhen Graduate School, Peking University, Shenzhen, 518000, People’s Republic of China
| | - Lina Tariq Al Kury
- College of Natural and Health Sciences, Zayed University, Abu Dhabi, 49153, United Arab Emirates
| | - Faiza Nasar
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Riphah International University, Islamabad, 747424, Pakistan
| | - Abdullah Alattar
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, University of Tabuk, Tabuk, 71421, Saudi Arabia
| | - Reem Alshaman
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, University of Tabuk, Tabuk, 71421, Saudi Arabia
| | - Fawad Ali Shah
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Riphah International University, Islamabad, 747424, Pakistan
| | - Arif-ullah Khan
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Riphah International University, Islamabad, 747424, Pakistan
| | - Shupeng Li
- State Key Laboratory of Oncogenomics, School of Chemical Biology and Biotechnology, Shenzhen Graduate School, Peking University, Shenzhen, 518000, People’s Republic of China
| |
Collapse
|
106
|
Qu L, Ji L, Wang C, Luo H, Li S, Peng W, Yin F, Lu D, Liu X, Kong L, Wang X. Synthesis and evaluation of multi-target-directed ligands with BACE-1 inhibitory and Nrf2 agonist activities as potential agents against Alzheimer's disease. Eur J Med Chem 2021; 219:113441. [PMID: 33862517 DOI: 10.1016/j.ejmech.2021.113441] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 03/15/2021] [Accepted: 03/31/2021] [Indexed: 12/17/2022]
Abstract
Cumulative evidence suggests that β-amyloid and oxidative stress are closely related with each other and play key roles in the process of Alzheimer's disease (AD). Multitarget regulation of both pathways might represent a promising therapeutic strategy. Here, a series of selenium-containing compounds based on ebselen and verubecestat were designed and synthesized. Biological evaluation showed that 13f exhibited good BACE-1 inhibitory activity (IC50 = 1.06 μΜ) and potent GPx-like activity (ν0 = 183.0 μM min-1). Aβ production experiment indicated that 13f could reduce the secretion of Aβ1-40 in HEK APPswe 293T cells. Moreover, 13f exerted a cytoprotective effect against the H2O2 or 6-OHDA caused cell damage via alleviation of intracellular ROS, mitochondrial dysfunction, Ca2+ overload and cell apoptosis. The mechanism studies indicated that 13f exhibited cytoprotective effect by activating the Keap1-Nrf2-ARE pathway and stimulating downstream anti-oxidant protein including HO-1, NQO1, TrxR1, GCLC, and GCLM. In addition, 13f significantly reduced the production of NO and IL-6 induced by LPS in BV2 cells, which confirmed its anti-inflammatory activity as a Nrf2 activator. The BBB permeation assay predicted that 13f was able to cross the BBB. In summary, 13f might be a promising multi-target-directed ligand for the treatment of AD.
Collapse
Affiliation(s)
- Lailiang Qu
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Limei Ji
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Cheng Wang
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Heng Luo
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Shang Li
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Wan Peng
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Fucheng Yin
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Dehua Lu
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Xingchen Liu
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Lingyi Kong
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, China.
| | - Xiaobing Wang
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, China.
| |
Collapse
|
107
|
Singh S, Nagalakshmi D, Sharma KK, Ravichandiran V. Natural antioxidants for neuroinflammatory disorders and possible involvement of Nrf2 pathway: A review. Heliyon 2021; 7:e06216. [PMID: 33659743 PMCID: PMC7890213 DOI: 10.1016/j.heliyon.2021.e06216] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 05/21/2020] [Accepted: 02/04/2021] [Indexed: 12/12/2022] Open
Abstract
The transcription factor Nrf2 (nuclear factor-erythroid 2 p45-related factor 2) play a crucial role in cellular redox and metabolic system. Activation of Nrf2 may be an effective therapeutic approach for neuroinflammatory disorders, through activation of antioxidant defences system, lower the inflammation, line up the mitochondrial function, and balancing of protein homeostasis. Various recent studies revealed that many of active substance obtained from plants have been found to activate the Nrf2 and to exert neuroprotective effects in various experimental models, raising the possibility that activation of Nrf2 may be an effective therapeutic approaches for neuroinflammatory disorders. The objective of this review was to evaluate the neuroprotective property of natural substance against neuroinflammatory disorders by reviewing the studies done till today. The outcomes of various in vitro and in vivo examinations have shown that natural compounds producing neuroprotective effects in neuronal system via activation of Nrf2. Herein, we also reviewed the studies to understand the role of Nrf2 for curing CNS disorders. Here we can conclude, herbal/natural moieties having potency to fight and prevent from neuroinflammatory disorders due to their abilities to activate Nrf2 pathway.
Collapse
Affiliation(s)
- Sanjiv Singh
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Export Promotion Industrial Park (EPIP), Zandaha Road, Hajipur, Bihar, India
| | - Devarapati Nagalakshmi
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Export Promotion Industrial Park (EPIP), Zandaha Road, Hajipur, Bihar, India
| | - K K Sharma
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Export Promotion Industrial Park (EPIP), Zandaha Road, Hajipur, Bihar, India
| | - V Ravichandiran
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Export Promotion Industrial Park (EPIP), Zandaha Road, Hajipur, Bihar, India
| |
Collapse
|
108
|
Baxter PS, Márkus NM, Dando O, He X, Al-Mubarak BR, Qiu J, Hardingham GE. Targeted de-repression of neuronal Nrf2 inhibits α-synuclein accumulation. Cell Death Dis 2021; 12:218. [PMID: 33637689 PMCID: PMC7910424 DOI: 10.1038/s41419-021-03507-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 02/02/2021] [Accepted: 02/05/2021] [Indexed: 12/12/2022]
Abstract
Many neurodegenerative diseases are associated with neuronal misfolded protein accumulation, indicating a need for proteostasis-promoting strategies. Here we show that de-repressing the transcription factor Nrf2, epigenetically shut-off in early neuronal development, can prevent protein aggregate accumulation. Using a paradigm of α-synuclein accumulation and clearance, we find that the classical electrophilic Nrf2 activator tBHQ promotes endogenous Nrf2-dependent α-synuclein clearance in astrocytes, but not cortical neurons, which mount no Nrf2-dependent transcriptional response. Moreover, due to neuronal Nrf2 shut-off and consequent weak antioxidant defences, electrophilic tBHQ actually induces oxidative neurotoxicity, via Nrf2-independent Jun induction. However, we find that epigenetic de-repression of neuronal Nrf2 enables them to respond to Nrf2 activators to drive α-synuclein clearance. Moreover, activation of neuronal Nrf2 expression using gRNA-targeted dCas9-based transcriptional activation complexes is sufficient to trigger Nrf2-dependent α-synuclein clearance. Thus, targeting reversal of the developmental shut-off of Nrf2 in forebrain neurons may alter neurodegenerative disease trajectory by boosting proteostasis.
Collapse
Affiliation(s)
- Paul S Baxter
- UK Dementia Research Institute at the University of Edinburgh, Edinburgh Medical School, University of Edinburgh, Edinburgh, UK.
- Deanery of Biomedical Sciences, Edinburgh Medical School, University of Edinburgh, Edinburgh, UK.
| | - Nóra M Márkus
- UK Dementia Research Institute at the University of Edinburgh, Edinburgh Medical School, University of Edinburgh, Edinburgh, UK
- Deanery of Biomedical Sciences, Edinburgh Medical School, University of Edinburgh, Edinburgh, UK
| | - Owen Dando
- UK Dementia Research Institute at the University of Edinburgh, Edinburgh Medical School, University of Edinburgh, Edinburgh, UK
- Deanery of Biomedical Sciences, Edinburgh Medical School, University of Edinburgh, Edinburgh, UK
- Simons Initiative for the Developing Brain, Edinburgh Medical School, University of Edinburgh, Edinburgh, EH8 9XD, UK
| | - Xin He
- UK Dementia Research Institute at the University of Edinburgh, Edinburgh Medical School, University of Edinburgh, Edinburgh, UK
- Deanery of Biomedical Sciences, Edinburgh Medical School, University of Edinburgh, Edinburgh, UK
| | - Bashayer R Al-Mubarak
- UK Dementia Research Institute at the University of Edinburgh, Edinburgh Medical School, University of Edinburgh, Edinburgh, UK
- Deanery of Biomedical Sciences, Edinburgh Medical School, University of Edinburgh, Edinburgh, UK
| | - Jing Qiu
- UK Dementia Research Institute at the University of Edinburgh, Edinburgh Medical School, University of Edinburgh, Edinburgh, UK
- Deanery of Biomedical Sciences, Edinburgh Medical School, University of Edinburgh, Edinburgh, UK
| | - Giles E Hardingham
- UK Dementia Research Institute at the University of Edinburgh, Edinburgh Medical School, University of Edinburgh, Edinburgh, UK.
- Deanery of Biomedical Sciences, Edinburgh Medical School, University of Edinburgh, Edinburgh, UK.
| |
Collapse
|
109
|
Park JE, Park JS, Leem YH, Kim DY, Kim HS. NQO1 mediates the anti-inflammatory effects of nootkatone in lipopolysaccharide-induced neuroinflammation by modulating the AMPK signaling pathway. Free Radic Biol Med 2021; 164:354-368. [PMID: 33460769 DOI: 10.1016/j.freeradbiomed.2021.01.015] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Revised: 12/18/2020] [Accepted: 01/08/2021] [Indexed: 01/07/2023]
Abstract
Neuroinflammation and oxidative stress play key roles in the progression of neurodegenerative diseases. Thus, the use of potent anti-inflammatory/antioxidant agents has been suggested as a promising therapeutic strategy for neurodegenerative diseases. In the present study, we investigated the anti-inflammatory and antioxidant effects of nootkatone (NKT), a sesquiterpenoid compound isolated from grapefruit, in in vitro and in vivo models of neuroinflammation. In lipopolysaccharide (LPS)-stimulated BV2 microglial cells, NKT inhibited the expression of iNOS, COX-2, and pro-inflammatory cytokines, and increased the expression of the anti-inflammatory cytokine, IL-10. In addition, NKT inhibited reactive oxygen species (ROS) production and upregulated the expression of antioxidant enzymes, such as NQO1 and HO-1. Molecular mechanistic studies showed that NKT inhibited Akt, p38 MAPK, and NF-κB activities, while increasing AMPK, PKA/CREB, and Nrf2/ARE signaling in LPS-stimulated BV2 cells. Since NKT dramatically increased NQO1 expression, we investigated the role of this enzyme using pharmacological inhibition or knockdown experiments. Treatment of BV2 cells with the NQO1-specific inhibitor, dicoumarol, or with NQO1 siRNA significantly blocked NKT-mediated inhibition of NO, ROS, TNF-α, IL-1β, and upregulation of IL-10. Furthermore, NQO1 inhibition reversed the effects of NKT on pro- and anti-inflammatory signaling molecules. Intriguingly, we found that the AMPK inhibitor, compound C, mimicked the effects of dicoumarol, suggesting the presence of a crosstalk between NQO1 and AMPK. Finally, we demonstrated that NKT inhibited microglial activation, lipid peroxidation, and the expression of pro-inflammatory markers in the brains of LPS-injected mice, which was also reversed by dicoumarol. These data collectively suggest that NQO1 plays a critical role in mediating the anti-inflammatory and antioxidant effects of NKT in LPS-induced neuroinflammation by modulating AMPK and its downstream signaling pathways.
Collapse
Affiliation(s)
- Jung-Eun Park
- Department of Molecular Medicine and the Ewha Medical Research Institute, School of Medicine, Ewha Womans University, Seoul, South Korea
| | - Jin-Sun Park
- Department of Molecular Medicine and the Ewha Medical Research Institute, School of Medicine, Ewha Womans University, Seoul, South Korea
| | - Yea-Hyun Leem
- Department of Molecular Medicine and the Ewha Medical Research Institute, School of Medicine, Ewha Womans University, Seoul, South Korea
| | - Do-Yeon Kim
- Department of Molecular Medicine and the Ewha Medical Research Institute, School of Medicine, Ewha Womans University, Seoul, South Korea
| | - Hee-Sun Kim
- Department of Molecular Medicine and the Ewha Medical Research Institute, School of Medicine, Ewha Womans University, Seoul, South Korea; Department of Brain & Cognitive Sciences, Ewha Womans University, Seoul, South Korea.
| |
Collapse
|
110
|
Ji L, Qu L, Wang C, Peng W, Li S, Yang H, Luo H, Yin F, Lu D, Liu X, Kong L, Wang X. Identification and optimization of piperlongumine analogues as potential antioxidant and anti-inflammatory agents via activation of Nrf2. Eur J Med Chem 2021; 210:112965. [PMID: 33148493 DOI: 10.1016/j.ejmech.2020.112965] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 10/14/2020] [Accepted: 10/21/2020] [Indexed: 02/07/2023]
Abstract
Oxidative stress and inflammation are significant risk factors for neurodegenerative disease. The Keap1-Nrf2-ARE pathway is one of the most promising defensive systems against oxidative stress. Here, dozens of piperlongumine analogues were designed, synthesized, and tested on PC12 cells to examine neuroprotective effects against H2O2 and 6-OHDA induced damage. Among them, 6d was found to be able to alleviate the accumulation of ROS, inhibit the production of NO and downregulate the level of IL-6, which indicated its potential antioxidant and anti-inflammatory activity. Further studies proved that 6d could activate Nrf2 signaling pathway, induce the translocation of Nrf2 from cell cytosol to nucleus and upregulate the related phase II antioxidant enzymes including NQO1, HO-1, GCLC, GCLM and TrxR1. These results confirmed that 6d exerted antioxidant and anti-inflammatory activities by activating Nrf2 signaling pathway. Moreover, the parallel artificial membrane permeability assay indicated that 6d can cross the blood-brain barrier. In general, 6d is promising for further development as a therapeutic drug against oxidative stress and inflammation related neurodegenerative disorders.
Collapse
Affiliation(s)
- Limei Ji
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Lailiang Qu
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Cheng Wang
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Wan Peng
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Shang Li
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Huali Yang
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Heng Luo
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Fucheng Yin
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Dehua Lu
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Xingchen Liu
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Lingyi Kong
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China.
| | - Xiaobing Wang
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
111
|
Currenti W, Godos J, Castellano S, Caruso G, Ferri R, Caraci F, Grosso G, Galvano F. Association between Time Restricted Feeding and Cognitive Status in Older Italian Adults. Nutrients 2021; 13:nu13010191. [PMID: 33435416 PMCID: PMC7827225 DOI: 10.3390/nu13010191] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 01/05/2021] [Accepted: 01/07/2021] [Indexed: 12/26/2022] Open
Abstract
Background: Due to the increased life expectancy, the prevalence of aging-related health conditions, such as cognitive impairment, dementia and Alzheimer’s disease is increasing. Among the modifiable risk factors, dietary factors have proved to be of primary importance in preserving and improving mental health and cognitive status in older adults, possibly through the modulation of adult neurogenesis, neuronal plasticity and brain signaling. Feeding/fasting timing manipulation has emerged as an innovative strategy to counteract and treat cognitive decline. The aim of this study was to investigate the association between the timing of the feeding period and cognitive status in a cross-sectional cohort of adults living in the Mediterranean area. Methods: Demographic and dietary characteristics of 883 adults living in Southern Italy (Sicily) were analyzed. Food frequency questionnaires were used to calculate the time window between the first and the last meal of an average day. Participants with an eating time window duration of more than 10 h were then identified, as well as those with eating time restricted to less than 10 h (TRF). Results: After adjusting for potential confounding factors, individuals adherent to TRF were less likely to have cognitive impairment, compared to those with no eating time restrictions [odds ratio (OR) = 0.28; 95% confidence intervals (CI): 0.07–0.90]; a similar association was found for individuals having breakfast (OR = 0.37, 95% CI: 0.16–0.89), but not for those having dinner. Conclusions: The results of this study reveal that time restricted eating may be positively associated with cognitive status, and thus exert plausible effects on brain health.
Collapse
Affiliation(s)
- Walter Currenti
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (W.C.); (F.G.)
| | - Justyna Godos
- Oasi Research Institute—IRCCS, 94018 Troina, Italy; (J.G.); (R.F.); (F.C.)
| | - Sabrina Castellano
- Department of Educational Sciences, University of Catania, 95124 Catania, Italy;
| | - Giuseppe Caruso
- Department of Drug Sciences, University of Catania, 95125 Catania, Italy;
| | - Raffaele Ferri
- Oasi Research Institute—IRCCS, 94018 Troina, Italy; (J.G.); (R.F.); (F.C.)
| | - Filippo Caraci
- Oasi Research Institute—IRCCS, 94018 Troina, Italy; (J.G.); (R.F.); (F.C.)
- Department of Drug Sciences, University of Catania, 95125 Catania, Italy;
| | - Giuseppe Grosso
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (W.C.); (F.G.)
- Correspondence: ; Tel.: +39-0954-781-187
| | - Fabio Galvano
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (W.C.); (F.G.)
| |
Collapse
|
112
|
Murru E, Carta G, Manca C, Sogos V, Pistis M, Melis M, Banni S. Conjugated Linoleic Acid and Brain Metabolism: A Possible Anti-Neuroinflammatory Role Mediated by PPARα Activation. Front Pharmacol 2021; 11:587140. [PMID: 33505308 PMCID: PMC7832089 DOI: 10.3389/fphar.2020.587140] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 11/17/2020] [Indexed: 12/24/2022] Open
Abstract
Fatty acids play a crucial role in the brain as specific receptor ligands and as precursors of bioactive metabolites. Conjugated linoleic acid (CLA), a group of positional and geometric isomers of linoleic acid (LA, 18:2 n-6) present in meat and dairy products of ruminants and synthesized endogenously in non-ruminants and humans, has been shown to possess different nutritional properties associated with health benefits. Its ability to bind to peroxisome proliferator-activated receptor (PPAR) α, a nuclear receptor key regulator of fatty acid metabolism and inflammatory responses, partly mediates these beneficial effects. CLA is incorporated and metabolized into brain tissue where induces the biosynthesis of endogenous PPARα ligands palmitoylethanolamide (PEA) and oleoylethanolamide (OEA), likely through a positive feedback mechanism where PPARα activation sustains its own cellular effects through ligand biosynthesis. In addition to PPARα, PEA and OEA may as well bind to other receptors such as TRPV1, further extending CLA own anti-neuroinflammatory actions. Future studies are needed to investigate whether dietary CLA may exert anti-inflammatory activity, particularly in the setting of neurodegenerative diseases and neuropsychiatric disorders with a neuroinflammatory basis.
Collapse
Affiliation(s)
- Elisabetta Murru
- Department of Biomedical Sciences, University of Cagliari, Monserrato, Italy
| | - Gianfranca Carta
- Department of Biomedical Sciences, University of Cagliari, Monserrato, Italy
| | - Claudia Manca
- Department of Biomedical Sciences, University of Cagliari, Monserrato, Italy
| | - Valeria Sogos
- Department of Biomedical Sciences, University of Cagliari, Monserrato, Italy
| | - Marco Pistis
- Department of Biomedical Sciences, University of Cagliari, Monserrato, Italy.,Neuroscience Institute, National Research Council of Italy (CNR), Cagliari, Italy
| | - Miriam Melis
- Department of Biomedical Sciences, University of Cagliari, Monserrato, Italy
| | - Sebastiano Banni
- Department of Biomedical Sciences, University of Cagliari, Monserrato, Italy
| |
Collapse
|
113
|
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
|
114
|
Khan H, Tundis R, Ullah H, Aschner M, Belwal T, Mirzaei H, Akkol EK. Flavonoids targeting NRF2 in neurodegenerative disorders. Food Chem Toxicol 2020; 146:111817. [PMID: 33069760 DOI: 10.1016/j.fct.2020.111817] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 10/11/2020] [Accepted: 10/12/2020] [Indexed: 02/05/2023]
Abstract
Neurodegenerative disorders are characterized by progressive loss of neurons. To date, no efficacious therapies exist for these disorders, and current therapies provide only symptomatic relief. The neuroprotective effects of natural compounds have been reported in several neurological disorders, such as Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD) amyotrophic lateral sclerosis (ALS), cerebral ischemia and brain tumors. Flavonoids are the most widely studied natural products for the prevention and treatment of neurodegenerative disorders. The nuclear factor (erythroid-derived 2)-like 2 (Nrf2) represents a complex gene regulated cytoprotective pathway. Several natural compounds have been identified as Nrf2 regulators in various chronic disorders, including carcinogenic, liver ailments, inflammatory conditions, neurodegeneration, diabetes and cardiotoxicities. The current review focuses on Nrf2 targeting by flavonoids in the prevention and treatment of neurodegenerative disorders, addressing the most contemporary information available on this timely subject.
Collapse
Affiliation(s)
- Haroon Khan
- Department of Pharmacy, Abdul Wali Khan University Mardan, 23200, Pakistan.
| | - Rosa Tundis
- Department of Pharmacy, Health and Nutritional Sciences University of Calabria, Via P. Bucci 87036 Rende (CS), Italy.
| | - Hammad Ullah
- Department of Pharmacy, Abdul Wali Khan University Mardan, 23200, Pakistan
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA.
| | - Tarun Belwal
- Centre for Biodiversity Conservation and Management, G.B. Plant National Institute of Himalayan Environment and Sustainable Development, Kosi-Katarmal, Almora, Uttarakhand, India.
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, I.R, Iran.
| | - Esra Kupeli Akkol
- Department of Pharmacognosy, Faculty of Pharmacy Gazi University 06330, Etiler/Ankara Turkey.
| |
Collapse
|
115
|
Binvignat O, Olloquequi J. Excitotoxicity as a Target Against Neurodegenerative Processes. Curr Pharm Des 2020; 26:1251-1262. [PMID: 31931694 DOI: 10.2174/1381612826666200113162641] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 11/27/2019] [Indexed: 12/20/2022]
Abstract
The global burden of neurodegenerative diseases is alarmingly increasing in parallel to the aging of population. Although the molecular mechanisms leading to neurodegeneration are not completely understood, excitotoxicity, defined as the injury and death of neurons due to excessive or prolonged exposure to excitatory amino acids, has been shown to play a pivotal role. The increased release and/or decreased uptake of glutamate results in dysregulation of neuronal calcium homeostasis, leading to oxidative stress, mitochondrial dysfunctions, disturbances in protein turn-over and neuroinflammation. Despite the anti-excitotoxic drug memantine has shown modest beneficial effects in some patients with dementia, to date, there is no effective treatment capable of halting or curing neurodegenerative diseases such as Alzheimer's disease, Parkinson disease, Huntington's disease or amyotrophic lateral sclerosis. This has led to a growing body of research focusing on understanding the mechanisms associated with the excitotoxic insult and on uncovering potential therapeutic strategies targeting these mechanisms. In the present review, we examine the molecular mechanisms related to excitotoxic cell death. Moreover, we provide a comprehensive and updated state of the art of preclinical and clinical investigations targeting excitotoxic- related mechanisms in order to provide an effective treatment against neurodegeneration.
Collapse
Affiliation(s)
| | - Jordi Olloquequi
- Laboratory of Cellular and Molecular Pathology, Instituto de Ciencias Biomedicas, Facultad de Ciencias de la Salud, Universidad Autonoma de Chile, Talca, Chile
| |
Collapse
|
116
|
Apigenin-7- O- β-D-(-6"- p-coumaroyl)-glucopyranoside treatment elicits a neuroprotective effect through GSK-3β phosphorylation-mediated Nrf2 activation. Aging (Albany NY) 2020; 12:23872-23888. [PMID: 33263567 PMCID: PMC7762462 DOI: 10.18632/aging.104050] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 07/14/2020] [Indexed: 02/06/2023]
Abstract
The current study was designed to seek the role of the glycogen synthase kinase-3β (GSK-β)-regulated NF-E2-related factor 2 (Nrf2) pathway in the antioxidant effect induced by Apigenin-7-O-β-D-(-6”-p-coumaroyl)-glucopyranoside (APG). Rat primary cultured cortical neurons were challenged by oxygen and glucose deprivation/reoxygenation (OGD/R) and then treated with APG. Cell viability, phosphorylation of GSK-β at Ser9 and nuclear expression of Nrf2 were measured. Male Sprague Dawley rats challenged by 2-h middle cerebral artery occlusion were treated with 50 mg/kg APG, and the neurological score, infarct volume, phosphorylation of GSK-3β and nuclear expression of Nrf2 were analyzed. The neuroprotective effect of APG and the expression levels of antioxidant enzymes and oxidative products were also examined in the presence and absence of Nrf2-siRNA and PI3K inhibitors. APG reduced the apoptotic proportion, attenuated LDH release and increased cell viability, and in vivo, APG improved neurological scores and reduced infarct volume. APG increased GSK-3β phosphorylation and Nrf2 nuclear translocation, while these effects were prevented by PI3K inhibitors or Nrf2-siRNA treatment in both OGD/R cell cultures and ischemic/reperfusion rats. These findings reveal that GSK-3β phosphorylation-mediated Nrf2 activation is involved in the neuroprotective effect of APG.
Collapse
|
117
|
Chartoumpekis DV, Fu CY, Ziros PG, Sykiotis GP. Patent Review (2017-2020) of the Keap1/Nrf2 Pathway Using PatSeer Pro: Focus on Autoimmune Diseases. Antioxidants (Basel) 2020; 9:antiox9111138. [PMID: 33212784 PMCID: PMC7697445 DOI: 10.3390/antiox9111138] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 11/04/2020] [Accepted: 11/14/2020] [Indexed: 12/18/2022] Open
Abstract
Research on the antioxidant pathway comprising the transcription factor nuclear factor erythroid 2-related factor 2 (Nrf2) and its cytoplasmic inhibitor Kelch-like ECH-associated protein 1 (Keap1) is ever increasing. As modulators of this pathway have started to be used in clinical trials and clinical practice, Nrf2 has become the subject of several patents. To assess the patent landscape of the last three years on Nrf2 and evaluate the main fields they refer to, we used the web-based tool PatSeer Pro to identify patents mentioning the Nrf2 pathway between January 2017 and May 2020. This search resulted in 509 unique patents that focus on topics such as autoimmune, neurodegenerative, liver, kidney, and lung diseases and refer to modulators (mainly activators) of the Nrf2 pathway as potential treatments. Autoimmunity emerged as the main theme among the topics of Nrf2 patents, including a broad range of diseases, such as systemic sclerosis, systemic lupus erythematosus, multiple sclerosis, inflammatory bowel diseases, Hashimoto's thyroiditis, etc.; however, there was a dearth of experimental support for the respective patents' claims. Given that chronic inflammation is the main element of the pathophysiology of most autoimmune diseases, the majority of patents referring to activation of Nrf2 as a method to treat autoimmune diseases base their claims on the well-established anti-inflammatory role of Nrf2. In conclusion, there is strong interest in securing intellectual property rights relating to the potential use of Nrf2 pathway activators in a variety of diseases, and this trend parallels the rise in related research publications. However, in the case of autoimmunity, more research is warranted to support the potential beneficial effects of Nrf2 modulation in each disease.
Collapse
Affiliation(s)
- Dionysios V. Chartoumpekis
- Service of Endocrinology and Diabetology, Lausanne University Hospital, and Faculty of Biology and Medicine, University of Lausanne, 1011 Lausanne, Switzerland; (D.V.C.); (P.G.Z.)
- Division of Endocrinology, Department of Internal Medicine, University of Patras, 26504 Patras, Greece
| | - Chun-Yan Fu
- Department of Pathology and Pathophysiology, Zhejiang University School of Medicine, Hangzhou 310058, China;
| | - Panos G. Ziros
- Service of Endocrinology and Diabetology, Lausanne University Hospital, and Faculty of Biology and Medicine, University of Lausanne, 1011 Lausanne, Switzerland; (D.V.C.); (P.G.Z.)
| | - Gerasimos P. Sykiotis
- Service of Endocrinology and Diabetology, Lausanne University Hospital, and Faculty of Biology and Medicine, University of Lausanne, 1011 Lausanne, Switzerland; (D.V.C.); (P.G.Z.)
- Correspondence: ; Tel.: +41-21-314-0606
| |
Collapse
|
118
|
Sesaminol prevents Parkinson's disease by activating the Nrf2-ARE signaling pathway. Heliyon 2020; 6:e05342. [PMID: 33163674 PMCID: PMC7609457 DOI: 10.1016/j.heliyon.2020.e05342] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 09/11/2020] [Accepted: 10/21/2020] [Indexed: 12/17/2022] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative disease caused by the degeneration of substantia nigra neurons due to oxidative stress. Sesaminol has strong antioxidant and anti-cancer effects. We investigated the preventive effect on PD as a new physiological action of sesaminol produced from sesaminol glycoside using in vitro and in vivo PD models. To prepare an in vitro PD model, 6-hydroxydopamine (6-OHDA) was added to human neuroblastoma (SH-SY5Y cells). The viability of SH-SY5Y cells decreased dose-dependently following 6-OHDA treatment, but the addition of sesaminol restored viability to the control level. 6-OHDA increased intracellular reactive oxygen species production, and the addition of sesaminol significantly suppressed this increase. No Nrf2 expression in the nucleus was observed in the control group, but a slight increase was observed in the 6-OHDA group. The sesaminol group showed strong expression of Nrf2 in the cytoplasm and nucleus. NAD(P)H: quinone oxidoreductase (NQO1) activity was enhanced in the 6-OHDA group and further enhanced in the sesaminol group. Furthermore, the neurotoxine rotenone was orally administrated to mice to prepare an in vivo PD model. The motor function of rotenone-treated mice was shorter than that of the control group, but a small amount of sesaminol restored it to the control level. The intestinal motility in the rotenone group was significantly lower than that in the control group, but it remained at the control level in the sesaminol group. The expression of α-synuclein in the substantia nigra increased in the rotenone group but decreased in the sesaminol group. The rotenone group exhibited shortening and damage to the colonic mucosa, but these abnormalities of the colonic mucosa were scarcely observed in the sesaminol group. These results suggest that sesaminol has a preventative effect on PD.
Collapse
|
119
|
Qu Z, Sun J, Zhang W, Yu J, Zhuang C. Transcription factor NRF2 as a promising therapeutic target for Alzheimer's disease. Free Radic Biol Med 2020; 159:87-102. [PMID: 32730855 DOI: 10.1016/j.freeradbiomed.2020.06.028] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 06/15/2020] [Accepted: 06/17/2020] [Indexed: 12/12/2022]
Abstract
Oxidative stress is considered as one of the pathogenesis of Alzheimer's disease (AD) and plays an important role in the occurrence and development of AD. Nuclear factor erythroid 2 related factor 2 (NRF2) is a key regulatory of oxidative stress defence. There is growing evidence indicating the relationship between NRF2 and AD. NRF2 activation mitigates multiple pathogenic processes involved in AD by upregulating antioxidative defense, inhibiting neuroinflammation, improving mitochondrial function, maintaining proteostasis, and inhibiting ferroptosis. In addition, several NRF2 activators are currently being evaluated as AD therapeutic agents in clinical trials. Thus, targeting NRF2 has been the focus of a new strategy for prevention and treatment of AD. In this review, the role of NRF2 in AD and the NRF2 activators advanced into clinical and preclinical studies will be summarized.
Collapse
Affiliation(s)
- Zhuo Qu
- School of Pharmacy, Key Laboratory of Hui Ethnic Medicine Modernization, Ministry of Education, Ningxia Medical University, 1160 Shengli Street, Yinchuan, 750004, China
| | - Jiachen Sun
- School of Biotechnology and Food Science, Tianjin University of Commerce, 409 Guangrong Road, Tianjin, 300134, China
| | - Wannian Zhang
- School of Pharmacy, Key Laboratory of Hui Ethnic Medicine Modernization, Ministry of Education, Ningxia Medical University, 1160 Shengli Street, Yinchuan, 750004, China; School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai, 200433, China
| | - Jianqiang Yu
- School of Pharmacy, Key Laboratory of Hui Ethnic Medicine Modernization, Ministry of Education, Ningxia Medical University, 1160 Shengli Street, Yinchuan, 750004, China.
| | - Chunlin Zhuang
- School of Pharmacy, Key Laboratory of Hui Ethnic Medicine Modernization, Ministry of Education, Ningxia Medical University, 1160 Shengli Street, Yinchuan, 750004, China; School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai, 200433, China.
| |
Collapse
|
120
|
Hoffman TE, Hanneman WH, Moreno JA. Network Simulations Reveal Molecular Signatures of Vulnerability to Age-Dependent Stress and Tau Accumulation. Front Mol Biosci 2020; 7:590045. [PMID: 33195439 PMCID: PMC7606936 DOI: 10.3389/fmolb.2020.590045] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 09/30/2020] [Indexed: 01/02/2023] Open
Abstract
Alzheimer’s disease (AD) is the leading cause of dementia and one of the most common causes of death worldwide. As an age-dependent multifactorial disease, the causative triggers of AD are rooted in spontaneous declines in cellular function and metabolic capacity with increases in protein stressors such as the tau protein. This multitude of age-related processes that cause neurons to change from healthy states to ones vulnerable to the damage seen in AD are difficult to simultaneously investigate and even more difficult to quantify. Here we aimed to diminish these gaps in our understanding of neuronal vulnerability in AD development by using simulation methods to theoretically quantify an array of cellular stress responses and signaling molecules. This temporally-descriptive molecular signature was produced using a novel multimethod simulation approach pioneered by our laboratory for biological research; this methodology combines hierarchical agent-based processes and continuous equation-based modeling in the same interface, all while maintaining intrinsic distributions that emulate natural biological stochasticity. The molecular signature was validated for a normal organismal aging trajectory using experimental longitudinal data from Caenorhabditis elegans and rodent studies. In addition, we have further predicted this aging molecular signature for cells impacted by the pathogenic tau protein, giving rise to distinct stress response conditions needed for cytoprotective aging. Interestingly, our simulation experiments showed that oxidative stress signaling (via daf-16 and skn-1 activities) does not substantially protect cells from all the early stressors of aging, but that it is essential in preventing a late-life degenerative cellular phenotype. Together, our simulation experiments aid in elucidating neurodegenerative triggers in the onset of AD for different genetic conditions. The long-term goal of this work is to provide more detailed diagnostic and prognostic tools for AD development and progression, and to provide more comprehensive preventative measures for this disease.
Collapse
Affiliation(s)
- Timothy E Hoffman
- Department of Environmental and Radiological Health Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
| | - William H Hanneman
- Department of Environmental and Radiological Health Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
| | - Julie A Moreno
- Department of Environmental and Radiological Health Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
| |
Collapse
|
121
|
Motor Coordination Disorders Evaluated through the Grid Test and Changes in the Nigral Nrf2 mRNA Expression in Rats with Pedunculopontine Lesion. Behav Sci (Basel) 2020; 10:bs10100156. [PMID: 33066049 PMCID: PMC7600924 DOI: 10.3390/bs10100156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 09/22/2020] [Accepted: 09/30/2020] [Indexed: 11/16/2022] Open
Abstract
Neurotoxic lesion of the pedunculopontine nucleus (PPN) is known to cause subtle motor dysfunctions. However, motor coordination during advance on a discontinuous and elevated surface has not been studied. It is also not known whether there are changes in the mRNA expression of nuclear factor (erythroid-derived 2)-like 2 (Nrf2) in nigral tissue. Methods: The effects of the unilateral neurotoxic lesion of the PPN in motor coordination evaluated through grid test and Nrf2 mRNA expression in nigral tissue were evaluated. Two experimental designs (ED) were organized: ED#1 behavioral study (7 and 30 days after PPN lesion) and ED#2 molecular biology study (24 h, 48 h and 7 days) after PPN lesion. Results: ED#1—The number of faults made with left limbs, were significant higher in the lesioned groups (p < 0.01) both 7 and 30 days post-lesion. The number of failures made by the right limbs, was also significantly higher (p < 0.05) vs. control groups. ED#2—Nrf2 mRNA expression showed an increase 24 h after PPN injury (p < 0.01), followed by a peak of expression 48 h post injury (p < 0.001). Conclusions: Disorders of motor coordination associated with PPN injury are bilateral. The increased Nrf2 mRNA expression could represent an adaptive response to oxidative stress in the nigral tissue following pontine injury.
Collapse
|
122
|
Direct inhibition of Keap1-Nrf2 Protein-Protein interaction as a potential therapeutic strategy for Alzheimer's disease. Bioorg Chem 2020; 103:104172. [DOI: 10.1016/j.bioorg.2020.104172] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 07/24/2020] [Accepted: 08/02/2020] [Indexed: 12/11/2022]
|
123
|
Farkhondeh T, Pourbagher-Shahri AM, Ashrafizadeh M, Folgado SL, Rajabpour-Sanati A, Khazdair MR, Samarghandian S. Green tea catechins inhibit microglial activation which prevents the development of neurological disorders. Neural Regen Res 2020; 15:1792-1798. [PMID: 32246619 PMCID: PMC7513986 DOI: 10.4103/1673-5374.280300] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Revised: 11/09/2019] [Accepted: 12/31/2019] [Indexed: 12/18/2022] Open
Abstract
The over-activated microglial cells induce neuroinflammation which has the main role in neurological disorders. The over-activated microglia can disturb neuronal function by releasing inflammatory mediators leading to neuronal dysfunctions and death. Thus, inhibition of over-activated microglia may be an effective therapeutic approach for modulating neuroinflammation. Experimental studies have indicated anti-neuroinflammatory effects of flavonoids such as green tea catechins. The current research was aimed to review the effect of green tea catechins in inhibiting microglial cells, inflammatory cascades, and subsequent neurological diseases.
Collapse
Affiliation(s)
- Tahereh Farkhondeh
- Cardiovascular Diseases Research Center, Birjand University of Medical Sciences, Birjand, Iran
| | | | - Milad Ashrafizadeh
- Department of Basic Science, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran
| | - Silvia Llorens Folgado
- Department of Medical Sciences, Faculty of Medicine of Albacete, Centro Regional de Investigaciones Biomédicas (CRIB), University of Castilla-La Mancha, Albacete, Spain
| | | | - Mohammad Reza Khazdair
- Cardiovascular Diseases Research Center, Birjand University of Medical Sciences, Birjand, Iran
| | - Saeed Samarghandian
- Healthy Ageing Research Center, Neyshabur University of Medical Sciences, Neyshabur, Iran
- Department of Basic Medical Sciences, Neyshabur University of Medical Sciences, Neyshabur, Iran
| |
Collapse
|
124
|
Mhillaj E, Papi M, Paciello F, Silvestrini A, Rolesi R, Palmieri V, Perini G, Fetoni AR, Trabace L, Mancuso C. Celecoxib Exerts Neuroprotective Effects in β-Amyloid-Treated SH-SY5Y Cells Through the Regulation of Heme Oxygenase-1: Novel Insights for an Old Drug. Front Cell Dev Biol 2020; 8:561179. [PMID: 33134292 PMCID: PMC7550645 DOI: 10.3389/fcell.2020.561179] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 08/31/2020] [Indexed: 12/25/2022] Open
Abstract
The formation and aggregation of amyloid-β-peptide (Aβ) into soluble and insoluble species represent the pathological hallmarks of Alzheimer’s disease (AD). Over the last few years, however, soluble Aβ (sAβ) prevailed over fibrillar Aβ (fAβ) as determinant of neurotoxicity. One of the main therapeutic strategies for challenging neurodegeneration is to fight against neuroinflammation and prevent free radical-induced damage: in this light, the heme oxygenase/biliverdin reductase (HO/BVR) system is considered a promising drug target. The aim of this work was to investigate whether or not celecoxib (CXB), a selective inhibitor of the pro-inflammatory cyclooxygenase-2, modulates the HO/BVR system and prevents lipid peroxidation in SH-SY5Y neuroblastoma cells. Both sAβ (6.25–50 nM) and fAβ (1.25–50 nM) dose-dependently over-expressed inducible HO (HO-1) after 24 h of incubation, reaching statistical significance at 25 and 6.25 nM, respectively. Interestingly, CXB (1–10 μM, for 1 h) further enhanced Aβ-induced HO-1 expression through the nuclear translocation of the transcriptional factor Nrf2. Furthermore, 10 μM CXB counteracted the Aβ-induced ROS production with a mechanism fully dependent on HO-1 up-regulation; nevertheless, 10 μM CXB significantly counteracted only 25 nM sAβ-induced lipid peroxidation damage in SH-SY5Y neurons by modulating HO-1. Both carbon monoxide (CORM-2, 50 nM) and bilirubin (50 nM) significantly prevented ROS production in Aβ-treated neurons and favored both the slowdown of the growth rate of Aβ oligomers and the decrease in oligomer/fibril final size. In conclusion, these results suggest a novel mechanism through which CXB is neuroprotective in subjects with early AD or mild cognitive impairment.
Collapse
Affiliation(s)
- Emanuela Mhillaj
- Department of Healthcare Surveillance and Bioethics, Section of Pharmacology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Massimiliano Papi
- Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy.,Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Fabiola Paciello
- Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy.,Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Andrea Silvestrini
- Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy.,Department of Basic Biotechnological Sciences, Intensivological and Perioperative Clinics, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Rolando Rolesi
- Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy.,Department of Head and Neck Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Valentina Palmieri
- Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy.,Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Giordano Perini
- Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy.,Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Anna Rita Fetoni
- Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy.,Department of Head and Neck Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Luigia Trabace
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Cesare Mancuso
- Department of Healthcare Surveillance and Bioethics, Section of Pharmacology, Università Cattolica del Sacro Cuore, Rome, Italy.,Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| |
Collapse
|
125
|
Marchetti B. Nrf2/Wnt resilience orchestrates rejuvenation of glia-neuron dialogue in Parkinson's disease. Redox Biol 2020; 36:101664. [PMID: 32863224 PMCID: PMC7395594 DOI: 10.1016/j.redox.2020.101664] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 07/15/2020] [Accepted: 07/27/2020] [Indexed: 12/11/2022] Open
Abstract
Oxidative stress and inflammation have long been recognized to contribute to Parkinson's disease (PD), a common movement disorder characterized by the selective loss of midbrain dopaminergic neurons (mDAn) of the substantia nigra pars compacta (SNpc). The causes and mechanisms still remain elusive, but a complex interplay between several genes and a number of interconnected environmental factors, are chiefly involved in mDAn demise, as they intersect the key cellular functions affected in PD, such as the inflammatory response, mitochondrial, lysosomal, proteosomal and autophagic functions. Nuclear factor erythroid 2 -like 2 (NFE2L2/Nrf2), the master regulator of cellular defense against oxidative stress and inflammation, and Wingless (Wnt)/β-catenin signaling cascade, a vital pathway for mDAn neurogenesis and neuroprotection, emerge as critical intertwinned actors in mDAn physiopathology, as a decline of an Nrf2/Wnt/β-catenin prosurvival axis with age underlying PD mutations and a variety of noxious environmental exposures drive PD neurodegeneration. Unexpectedly, astrocytes, the so-called "star-shaped" cells, harbouring an arsenal of "beneficial" and "harmful" molecules represent the turning point in the physiopathological and therapeutical scenario of PD. Fascinatingly, "astrocyte's fil rouge" brings back to Nrf2/Wnt resilience, as boosting the Nrf2/Wnt resilience program rejuvenates astrocytes, in turn (i) mitigating nigrostriatal degeneration of aged mice, (ii) reactivating neural stem progenitor cell proliferation and neuron differentiation in the brain and (iii) promoting a beneficial immunomodulation via bidirectional communication with mDAns. Then, through resilience of Nrf2/Wnt/β-catenin anti-ageing, prosurvival and proregenerative molecular programs, it seems possible to boost the inherent endogenous self-repair mechanisms. Here, the cellular and molecular aspects as well as the therapeutical options for rejuvenating glia-neuron dialogue will be discussed together with major glial-derived mechanisms and therapies that will be fundamental to the identification of novel diagnostic tools and treatments for neurodegenerative diseases (NDs), to fight ageing and nigrostriatal DAergic degeneration and promote functional recovery.
Collapse
Affiliation(s)
- Bianca Marchetti
- Department of Biomedical and Biotechnological Sciences (BIOMETEC), Pharmacology Section, Medical School, University of Catania, Via S. Sofia 65, 95125, Catania, Italy; Oasi Research Institute-IRCCS, Neuropharmacology Section, Via Conte Ruggero 73, 94018, Troina, EN, Italy.
| |
Collapse
|
126
|
Colarusso S, De Simone D, Frattarelli T, Andreini M, Cerretani M, Missineo A, Moretti D, Tambone S, Kempf G, Augustin M, Steinbacher S, Munoz-Sanjuan I, Park L, Summa V, Tomei L, Bresciani A, Dominguez C, Toledo-Sherman L, Bianchi E. Optimization of linear and cyclic peptide inhibitors of KEAP1-NRF2 protein-protein interaction. Bioorg Med Chem 2020; 28:115738. [PMID: 33065433 DOI: 10.1016/j.bmc.2020.115738] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 08/20/2020] [Accepted: 08/21/2020] [Indexed: 01/16/2023]
Abstract
Inhibition of KEAP1-NRF2 protein-protein interaction is considered a promising strategy to selectively and effectively activate NRF2, a transcription factor which is involved in several pathologies such as Huntington's disease (HD). A library of linear peptides based on the NRF2-binding motifs was generated on the nonapeptide lead Ac-LDEETGEFL-NH2 spanning residues 76-84 of the Neh2 domain of NRF2 with the aim to replace E78, E79 and E82 with non-acidic amino acids. A deeper understanding of the features and accessibility of the T80 subpocket was also targeted by structure-based design. Approaches to improve cell permeability were investigated using both different classes of cyclic peptides and conjugation to cell-penetrating peptides. This insight will guide future design of macrocycles, peptido-mimetics and, most importantly, small neutral brain-penetrating molecules to evaluate whether NRF2 activators have utility in HD.
Collapse
Affiliation(s)
- Stefania Colarusso
- Department of Drug Discovery, IRBM Spa, Via Pontina km 30.600, 00071 Pomezia, Rome, Italy.
| | - Daniele De Simone
- Department of Drug Discovery, IRBM Spa, Via Pontina km 30.600, 00071 Pomezia, Rome, Italy
| | - Tommaso Frattarelli
- Department of Drug Discovery, IRBM Spa, Via Pontina km 30.600, 00071 Pomezia, Rome, Italy
| | - Matteo Andreini
- Department of Drug Discovery, IRBM Spa, Via Pontina km 30.600, 00071 Pomezia, Rome, Italy
| | - Mauro Cerretani
- Translational & Discovery Research, IRBM Spa, Via Pontina km 30.600, 00071 Pomezia, Rome, Italy
| | - Antonino Missineo
- Translational & Discovery Research, IRBM Spa, Via Pontina km 30.600, 00071 Pomezia, Rome, Italy
| | - Daniele Moretti
- Translational & Discovery Research, IRBM Spa, Via Pontina km 30.600, 00071 Pomezia, Rome, Italy
| | - Sara Tambone
- Translational & Discovery Research, IRBM Spa, Via Pontina km 30.600, 00071 Pomezia, Rome, Italy
| | - Georg Kempf
- Proteros Biostructures GmbH, Bunsenstraße 7 a, 82152 Planegg, Germany
| | - Martin Augustin
- Proteros Biostructures GmbH, Bunsenstraße 7 a, 82152 Planegg, Germany
| | | | | | - Larry Park
- CHDI Management/CHDI Foundation, Los Angeles, CA, United States
| | - Vincenzo Summa
- Department of Drug Discovery, IRBM Spa, Via Pontina km 30.600, 00071 Pomezia, Rome, Italy
| | - Licia Tomei
- Translational & Discovery Research, IRBM Spa, Via Pontina km 30.600, 00071 Pomezia, Rome, Italy
| | - Alberto Bresciani
- Translational & Discovery Research, IRBM Spa, Via Pontina km 30.600, 00071 Pomezia, Rome, Italy
| | - Celia Dominguez
- CHDI Management/CHDI Foundation, Los Angeles, CA, United States.
| | - Leticia Toledo-Sherman
- Translational & Discovery Research, IRBM Spa, Via Pontina km 30.600, 00071 Pomezia, Rome, Italy
| | - Elisabetta Bianchi
- Department of Drug Discovery, IRBM Spa, Via Pontina km 30.600, 00071 Pomezia, Rome, Italy
| |
Collapse
|
127
|
dos Santos Maia M, Rodrigues GCS, de Sousa NF, Scotti MT, Scotti L, Mendonça-Junior FJB. Identification of New Targets and the Virtual Screening of Lignans against Alzheimer's Disease. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:3098673. [PMID: 32879651 PMCID: PMC7448245 DOI: 10.1155/2020/3098673] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 06/22/2020] [Accepted: 07/17/2020] [Indexed: 02/08/2023]
Abstract
Alzheimer's disease (AD) is characterized by the progressive disturbance in cognition and affects approximately 36 million people, worldwide. However, the drugs used to treat this disease are only moderately effective and do not alter the course of the neurodegenerative process. This is because the pathogenesis of AD is mainly associated with oxidative stress, and current drugs only target two enzymes involved in neurotransmission. Therefore, the present study sought to identify potential multitarget compounds for enzymes that are directly or indirectly involved in the oxidative pathway, with minimal side effects, for AD treatment. A set of 159 lignans were submitted to studies of QSAR and molecular docking. A combined analysis was performed, based on ligand and structure, followed by the prediction of absorption, distribution, metabolism, excretion, and toxicity (ADMET) properties. The results showed that the combined analysis was able to select 139 potentially active and multitarget lignans targeting two or more enzymes, among them are c-Jun N-terminal kinase 3 (JNK-3), protein tyrosine phosphatase 1B (PTP1B), nicotinamide adenine dinucleotide phosphate oxidase 1 (NOX1), NADPH quinone oxidoreductase 1 (NQO1), phosphodiesterase 5 (PDE5), nuclear factor erythroid 2-related factor 2 (Nrf2), cycloxygenase 2 (COX-2), and inducible nitric oxide synthase (iNOS). The authors conclude that compounds (06) austrobailignan 6, (11) anolignan c, (19) 7-epi-virolin, (64) 6-[(2R,3R,4R,5R)-3,4-dimethyl-5-(3,4,5-trimethoxyphenyl)oxolan-2-yl]-4-methoxy-1,3-benzodioxole, (116) ococymosin, and (135) mappiodoinin b have probabilities that confer neuroprotection and antioxidant activity and represent potential alternative AD treatment drugs or prototypes for the development of new drugs with anti-AD properties.
Collapse
Affiliation(s)
- Mayara dos Santos Maia
- Laboratory of Cheminformatics, Program of Natural and Synthetic Bioactive Products (PgPNSB), Health Sciences Center, Federal University of Paraíba, João Pessoa, PB, Brazil
| | - Gabriela Cristina Soares Rodrigues
- Laboratory of Cheminformatics, Program of Natural and Synthetic Bioactive Products (PgPNSB), Health Sciences Center, Federal University of Paraíba, João Pessoa, PB, Brazil
| | - Natália Ferreira de Sousa
- Laboratory of Cheminformatics, Program of Natural and Synthetic Bioactive Products (PgPNSB), Health Sciences Center, Federal University of Paraíba, João Pessoa, PB, Brazil
| | - Marcus Tullius Scotti
- Laboratory of Cheminformatics, Program of Natural and Synthetic Bioactive Products (PgPNSB), Health Sciences Center, Federal University of Paraíba, João Pessoa, PB, Brazil
| | - Luciana Scotti
- Laboratory of Cheminformatics, Program of Natural and Synthetic Bioactive Products (PgPNSB), Health Sciences Center, Federal University of Paraíba, João Pessoa, PB, Brazil
| | | |
Collapse
|
128
|
Wu Y, Wang Y, Wu Y, Li T, Wang W. Salidroside shows anticonvulsant and neuroprotective effects by activating the Nrf2-ARE pathway in a pentylenetetrazol-kindling epileptic model. Brain Res Bull 2020; 164:14-20. [PMID: 32800786 DOI: 10.1016/j.brainresbull.2020.08.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 07/12/2020] [Accepted: 08/08/2020] [Indexed: 12/17/2022]
Abstract
Evidence points towards oxidative stress and neuroinflammation being major processes associated with brain dysfunction in epilepsy. Salidroside reportedly possesses anti-oxidative activity and neuroprotective potential, in addition to exerting an anti-neuroinflammatory response. This study was designed to evaluate the anticonvulsant and neuroprotective role of salidroside in rats with pentylenetetrazole (PTZ) kindling and to explore the underlying mechanism. Male Wistar rats were administered a sub-convulsive dose of PTZ (35 mg/kg) every other day for 15 injections, and salidroside (50 mg/kg) was injected intraperitoneally along with alternate-day PTZ. The seizure degree, cognitive function, and number of hippocampal neurons were investigated. The expression of nuclear factor erythroid 2-related factor- antioxidant response element (Nrf2-ARE) signaling pathways, oxidative stress parameters and inflammatory cytokines were also observed. Our study showed that salidroside treatment suppressed the kindling acquisition process, ameliorated cognitive impairment, and rescued the number of pyramidal neurons in the CA3 regions. Salidroside treatment could activate the Nrf2-ARE signal pathway, and suppressed oxidative stress and neuroinflammation. Our findings demonstrated that salidroside exerted anticonvulsant and neuroprotective effects in epileptic rats by activating the Nrf2-ARE signal pathway.
Collapse
Affiliation(s)
- Yanfen Wu
- Health management department, Aerospace Center Hospital, Peking University Aerospace Clinical College, Beijing, China
| | - Yong Wang
- Department of Anesthesiology, Pain Medicine and Critical Care Medicine, Aviation General Hospital of China Medical University & Beijing Institute of Translational Medicine, Chinese Academy of Sciences, Beijing, China
| | - Yarui Wu
- Health management department, Aerospace Center Hospital, Peking University Aerospace Clinical College, Beijing, China
| | - Tingting Li
- Inovation Center for Neurological Disorders, Department of Neurology, Xuan Wu Hospital, Capital Medical University, Beijing, China
| | - Wei Wang
- Inovation Center for Neurological Disorders, Department of Neurology, Xuan Wu Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
129
|
Li X, Zhang J, Zhang X, Dong M. Puerarin suppresses MPP +/MPTP-induced oxidative stress through an Nrf2-dependent mechanism. Food Chem Toxicol 2020; 144:111644. [PMID: 32763437 DOI: 10.1016/j.fct.2020.111644] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 07/18/2020] [Accepted: 07/21/2020] [Indexed: 12/24/2022]
Abstract
In this study, we hypothesized that anti-parkinsonian effect of puerarin is attributable to its antioxidant properties via Nrf2-dependent glutathione (GSH) biosynthesis mechanism. Experimentally, we found that puerarin attenuated 1-methyl-4-phenylpyridinium (MPP+)-induced oxidative stress through elevating biosynthetic capacity of GSH in PC12 cells. Mechanistically, puerarin suppressed Fyn phosphorylation by GSK-3β-dependent mechanism in MPP+-challenged PC12 cells. Furthermore, puerarin induced accumulation of Nrf2 in the nucleus via inhibiting its nuclear exclusion. In parallel, puerarin up-regulated antioxidant response element (ARE)-driven catalytic subunits from glutamate cysteine ligase (GCLc) expression at levels of transcription and translation. Most interestingly, pharmacological inhibitor of GSK-3β or Fyn shRNA blocked puerarin-induced Nrf2 activation in MPP+-challenged PC12 cells. Concomitantly, puerarin ameliorated motor deficits and inhibited oxidative stress in the ventral midbrain in MPTP-intoxicated wild-type (WT) mice, but failed to attenuate MPTP neurotoxicity and up-regulate GCLc gene in Nrf2-knockout (Nrf2-/-) mice, suggesting that anti-parkinsonian effect of puerarin was dependent on Nrf2. Additionally, puerarin regulated Fyn and GSK-3β phosphorylation in the ventral midbrain in MPTP-intoxicated WT mice. Collectively, the results of the study provide molecular insights into the potential therapeutic action of puerarin in Parkinson's disease, suggesting that puerarin may be a promising candidate for the treatment of Parkinson's disease.
Collapse
Affiliation(s)
- Xiaoming Li
- The Institute of Medicine, Qiqihar Medical University, Qiqihar, 161006, China
| | - Jing Zhang
- Department of Hematology, The First Affiliated Hospital, Harbin Medical University, Harbin, 150001, China
| | - Xiaojie Zhang
- The Institute of Medicine, Qiqihar Medical University, Qiqihar, 161006, China
| | - Miaoxian Dong
- The Institute of Medicine, Qiqihar Medical University, Qiqihar, 161006, China.
| |
Collapse
|
130
|
Scuderi SA, Ardizzone A, Paterniti I, Esposito E, Campolo M. Antioxidant and Anti-inflammatory Effect of Nrf2 Inducer Dimethyl Fumarate in Neurodegenerative Diseases. Antioxidants (Basel) 2020; 9:antiox9070630. [PMID: 32708926 PMCID: PMC7402174 DOI: 10.3390/antiox9070630] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 07/13/2020] [Accepted: 07/14/2020] [Indexed: 12/13/2022] Open
Abstract
Neurodegenerative diseases (NDs) represents debilitating conditions characterized by degeneration of neuronal cells in specific brain areas, causing disability and death in patients. In the pathophysiology of NDs, oxidative stress, apoptosis and neuroinflammation have a key role, as demonstrated by in vivo and in vitro models. Therefore, the use of molecules with antioxidant and anti-inflammatory activities represents a possible strategy for the treatment of NDs. Many studies demonstrated the beneficial effects of fumaric acid esters (FAEs) to counteract neuroinflammation and oxidative stress. Among these molecules, dimethyl fumarate (DMF) showed a valid therapeutic approach to slow down neurodegeneration and relieve symptoms in patients with NDs. DMF is a methyl ester of fumaric acid and acts as modulator of the nuclear factor erythroid 2-related factor 2 (Nrf2) pathway as well as nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) translocation. Therefore, this review aims to examine the potential beneficial effects of DMF to counteract oxidative stress and inflammation in patients with NDs.
Collapse
|
131
|
Mou Y, Wen S, Li YX, Gao XX, Zhang X, Jiang ZY. Recent progress in Keap1-Nrf2 protein-protein interaction inhibitors. Eur J Med Chem 2020; 202:112532. [PMID: 32668381 DOI: 10.1016/j.ejmech.2020.112532] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Revised: 05/28/2020] [Accepted: 05/31/2020] [Indexed: 12/16/2022]
Abstract
Therapeutic targeting the protein-protein interaction (PPI) of Nuclear factor (erythroid-derived 2)-like 2 (Nrf2) and its main regulator, Kelch-like ECH-Associating protein 1 (Keap1) has been emerged as a feasible way to combat oxidative stress related diseases, due to the key role of Nrf2 in oxidative stress regulation. In recent years, many efforts have been made to develop potent Keap1-Nrf2 inhibitors with new chemical structures. Various molecules with diverse chemical structures have been reported and some compounds exhibit high potency. This review summarizes peptide and small molecule Keap1-Nrf2 inhibitors reported recently. We also highlight the pharmacological effects and discuss the possible therapeutic application of Keap1-Nrf2 inhibitors.
Collapse
Affiliation(s)
- Yi Mou
- College of Pharmacy and Chemistry & Chemical Engineering, Taizhou University, Taizhou, 225300, China
| | - Shuai Wen
- College of Pharmacy and Chemistry & Chemical Engineering, Taizhou University, Taizhou, 225300, China
| | - Yu-Xiu Li
- College of Pharmacy and Chemistry & Chemical Engineering, Taizhou University, Taizhou, 225300, China
| | - Xin-Xing Gao
- College of Pharmacy and Chemistry & Chemical Engineering, Taizhou University, Taizhou, 225300, China
| | - Xin Zhang
- College of Pharmacy and Chemistry & Chemical Engineering, Taizhou University, Taizhou, 225300, China
| | - Zheng-Yu Jiang
- Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, 210009, China.
| |
Collapse
|
132
|
Al-Awadhi FH, Salvador-Reyes LA, Elsadek LA, Ratnayake R, Chen QY, Luesch H. Largazole is a Brain-Penetrant Class I HDAC Inhibitor with Extended Applicability to Glioblastoma and CNS Diseases. ACS Chem Neurosci 2020; 11:1937-1943. [PMID: 32559056 PMCID: PMC7390227 DOI: 10.1021/acschemneuro.0c00093] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Largazole is a potent class I selective histone deacetylase inhibitor prodrug with anticancer activity against solid tumors in preclinical models. Largazole possesses in vitro activity against glioblastoma multiforme (GBM) cells and sufficiently crosses the blood-brain barrier based on measurement of the active species, largazole thiol, to achieve therapeutically relevant concentrations in the mouse brain. The effective dose resulted in pronounced functional responses on the transcript level based on RNA sequencing and quantitative polymerase chain reaction after reverse transcription (RT-qPCR), revealing desirable expression changes of genes related to neuroprotection, including Bdnf and Pax6 upregulation, extending the applicability of largazole to the treatment of brain cancer and neurodegenerative disorders. The largazole-induced modulation of Pax6 unifies both activities, since Pax6 expression suppresses GBM proliferation and invasion and inversely correlates with GBM tumor grade, while it is also implicated in neurogenesis, neuronal plasticity, and cognitive ability. Our results suggest that largazole could be repurposed for diseases of the brain.
Collapse
Affiliation(s)
- Fatma H. Al-Awadhi
- Department of Medicinal Chemistry and Center for Natural Products, Drug Discovery and Development (CNPD3), University of Florida, 1345 Center Drive, Gainesville, Florida 32610, United States
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Kuwait University, P.O. Box 24923, Safat 13110, Kuwait
| | - Lilibeth A. Salvador-Reyes
- Department of Medicinal Chemistry and Center for Natural Products, Drug Discovery and Development (CNPD3), University of Florida, 1345 Center Drive, Gainesville, Florida 32610, United States
- Marine Science Institute, College of Science, University of the Philippines, Diliman, Quezon City, 1100 Philippines
| | - Lobna A. Elsadek
- Department of Medicinal Chemistry and Center for Natural Products, Drug Discovery and Development (CNPD3), University of Florida, 1345 Center Drive, Gainesville, Florida 32610, United States
| | - Ranjala Ratnayake
- Department of Medicinal Chemistry and Center for Natural Products, Drug Discovery and Development (CNPD3), University of Florida, 1345 Center Drive, Gainesville, Florida 32610, United States
| | - Qi-Yin Chen
- Department of Medicinal Chemistry and Center for Natural Products, Drug Discovery and Development (CNPD3), University of Florida, 1345 Center Drive, Gainesville, Florida 32610, United States
| | - Hendrik Luesch
- Department of Medicinal Chemistry and Center for Natural Products, Drug Discovery and Development (CNPD3), University of Florida, 1345 Center Drive, Gainesville, Florida 32610, United States
| |
Collapse
|
133
|
Zhao Y, Han Y, Wang Z, Chen T, Qian H, He J, Li J, Han B, Wang T. Rosmarinic acid protects against 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-induced dopaminergic neurotoxicity in zebrafish embryos. Toxicol In Vitro 2020; 65:104823. [PMID: 32147576 DOI: 10.1016/j.tiv.2020.104823] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2019] [Revised: 03/01/2020] [Accepted: 03/04/2020] [Indexed: 10/24/2022]
Abstract
Rosmarinic acid (RA) is an extract that can be obtained from Lamiaceae herbs and the Boraginaceae family. This study aimed to evaluate the effect of RA on 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced dopaminergic neurotoxicity in zebrafish embryos. Embryos were challenged with MPTP and then were treated with RA or brusatol (a Nrf2 inhibitor). Locomotor activity of zebrafish was recorded using a video camera. The swimming distance was analyzed with SMART 3.0 software. Tyrosine hydroxylase (TH) immunohistochemistry, reactive oxygen species (ROS), glutathione (GSH), and malondialdehyde (MDA) contents were evaluated. The expressions of proteins in the DJ-1/Akt/Nrf2 signaling pathway were measured. The results showed that RA not only prevented MPTP-induced dopaminergic neuron loss, but also attenuated the deficit in locomotor behavior. RA attenuated the increases of ROS and MDA induced by MPTP. Treatment with RA augmented expression of glutamate cysteine ligase catalytic subunit, glutamate cysteine ligase modifier subunit, and GSH. Furthermore, RA increased the expression of DJ-1, p-Akt, Nuclear-Nrf2, HO-1 and inhibited the expression of PTEN. Brusatol partially abolished the neuroprotective effect of RA in MPTP-induced Parkinson's disease (PD) model of zebrafish embryos. The results of this study indicate that RA exerts neuroprotective effects on MPTP-induced neurotoxicity in dopaminergic neurons of a zebrafish PD model. The mechanism underlying the effects of RA is associated with promotion of antioxidant gene expression via regulation of the DJ-1/Akt/Nrf2 signaling pathway.
Collapse
Affiliation(s)
- Yue Zhao
- Center for Mitochondria and Healthy Aging, College of Life Science, Yantai University, Yantai 264005, PR China
| | - Yingjie Han
- Center for Mitochondria and Healthy Aging, College of Life Science, Yantai University, Yantai 264005, PR China
| | - Zhenhua Wang
- Center for Mitochondria and Healthy Aging, College of Life Science, Yantai University, Yantai 264005, PR China
| | - Tianrong Chen
- Center for Mitochondria and Healthy Aging, College of Life Science, Yantai University, Yantai 264005, PR China
| | - Haowen Qian
- Center for Mitochondria and Healthy Aging, College of Life Science, Yantai University, Yantai 264005, PR China
| | - Jie He
- Center for Mitochondria and Healthy Aging, College of Life Science, Yantai University, Yantai 264005, PR China
| | - Ji Li
- Center for Mitochondria and Healthy Aging, College of Life Science, Yantai University, Yantai 264005, PR China
| | - Bing Han
- Center for Mitochondria and Healthy Aging, College of Life Science, Yantai University, Yantai 264005, PR China.
| | - Tian Wang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, Shandong 264005, PR China.
| |
Collapse
|
134
|
Li X, Zhang J, Rong H, Zhang X, Dong M. Ferulic Acid Ameliorates MPP +/MPTP-Induced Oxidative Stress via ERK1/2-Dependent Nrf2 Activation: Translational Implications for Parkinson Disease Treatment. Mol Neurobiol 2020; 57:2981-2995. [PMID: 32445087 DOI: 10.1007/s12035-020-01934-1] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Accepted: 05/13/2020] [Indexed: 12/27/2022]
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder closely associated with oxidative stress. The biochemical and cellular alterations that occur after cell and mouse treatment with the parkinsonism-inducing neurotoxin MPP+/MPTP are remarkably similar to those observed in idiopathic PD. Previously, we showed that ferulic acid (FA) has antioxidant properties and the ability to activate nuclear factor E2-related factor 2 (Nrf2). The present study tested the hypothesis that FA attenuates MPP+/MPTP-induced oxidative stress by regulating crosstalk between sirtuin 2 (SIRT2) and Nrf2 pathways. To test this hypothesis, we performed in vitro and in vivo studies using MPP+/MPTP-challenged SH-SY5Y cells or mice treated with or not with FA. FA marginally inhibited SIRT2 in parallel with α-synuclein at levels of transcription and translation in SH-SY5Y cells challenged with MPP+. Moreover, FA attenuated MPP+-induced oxidative stress, as indicated by reactive oxygen species, lipid hydroperoxides, GSH/GSSG ratio, and NAD+/NADH ratio. Mechanistically, FA strongly upregulated the glutamate cysteine ligase catalytic subunit and heme oxygenase-1 expression at the levels of transcription and translation. Interestingly, FA-mediated extracellular signal-regulated kinases 1 and 2 (ERK1/2) activation contributed to nuclear accumulation of Nrf2 via de novo synthesis, which was validated by the use of dominant negative ERK2. Surprisingly, activation of the ERK1/2 and inhibition of SIRT2 by FA are mediated by independent mechanisms. Furthermore, FA ameliorated motor deficits and oxidative stress in the ventral midbrain in MPTP-treated (25 mg/kg, i.p., daily for 5 days) wild-type mice and α-synuclein knockout mice, but not in Nrf2 knockout mice. Collectively, FA exerts antioxidant effects through ERK1/2-mediated activation of the Nrf2 pathway, and these results may have important translational value for the treatment of PD.
Collapse
Affiliation(s)
- Xu Li
- The Institute of Medicine, Qiqihar Medical University, 333 BuKui Street, JianHua District, Qiqihar, 161006, China
| | - Jing Zhang
- Department of Hematology, the First Affiliated Hospital, Harbin Medical University, Harbin, 150001, China
| | - Hua Rong
- The Institute of Medicine, Qiqihar Medical University, 333 BuKui Street, JianHua District, Qiqihar, 161006, China
| | - Xiaojie Zhang
- The Institute of Medicine, Qiqihar Medical University, 333 BuKui Street, JianHua District, Qiqihar, 161006, China
| | - Miaoxian Dong
- The Institute of Medicine, Qiqihar Medical University, 333 BuKui Street, JianHua District, Qiqihar, 161006, China.
| |
Collapse
|
135
|
New Synthetic 3-Benzoyl-5-Hydroxy-2 H-Chromen-2-One (LM-031) Inhibits Polyglutamine Aggregation and Promotes Neurite Outgrowth through Enhancement of CREB, NRF2, and Reduction of AMPK α in SCA17 Cell Models. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:3129497. [PMID: 32377295 PMCID: PMC7195640 DOI: 10.1155/2020/3129497] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Revised: 03/25/2020] [Accepted: 04/02/2020] [Indexed: 12/31/2022]
Abstract
Spinocerebellar ataxia type 17 (SCA17) is caused by a CAG/CAA expansion mutation encoding an expanded polyglutamine (polyQ) tract in TATA-box binding protein (TBP), a general transcription initiation factor. Suppression of cAMP-responsive element binding protein- (CREB-) dependent transcription, impaired nuclear factor erythroid 2-related factor 2 (NRF2) signaling, and interaction of AMP-activated protein kinase (AMPK) with increased oxidative stress have been implicated to be involved in pathogenic mechanisms of polyQ-mediated diseases. In this study, we demonstrated decreased pCREB and NRF2 and activated AMPK contributing to neurotoxicity in SCA17 SH-SY5Y cells. We also showed that licochalcone A and the related in-house derivative compound 3-benzoyl-5-hydroxy-2H-chromen-2-one (LM-031) exhibited antiaggregation, antioxidative, antiapoptosis, and neuroprotective effects in TBP/Q79-GFP-expressing cell models. LM-031 and licochalcone A exerted neuroprotective effects by upregulating pCREB and its downstream genes, BCL2 and GADD45B, and enhancing NRF2. Furthermore, LM-031, but not licochalcone A, reduced activated AMPKα. Knockdown of CREB and NRF2 and treatment of AICAR (5-aminoimidazole-4-carboxamide 1-β-D-ribofuranoside), an AMPK activator, attenuated the aggregation-inhibiting and neurite outgrowth promoting effects of LM-031 on TBP/Q79 SH-SY5Y cells. The study results suggest the LM-031 as potential therapeutics for SCA17 and probable other polyQ diseases.
Collapse
|
136
|
Chen CM, Chen WL, Hung CT, Lin TH, Lee MC, Chen IC, Lin CH, Chao CY, Wu YR, Chang KH, Hsieh-Li HM, Lee-Chen GJ. Shaoyao Gancao Tang (SG-Tang), a formulated Chinese medicine, reduces aggregation and exerts neuroprotection in spinocerebellar ataxia type 17 (SCA17) cell and mouse models. Aging (Albany NY) 2020; 11:986-1007. [PMID: 30760647 PMCID: PMC6382417 DOI: 10.18632/aging.101804] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2018] [Accepted: 01/24/2019] [Indexed: 12/22/2022]
Abstract
Spinocerebellar ataxia (SCA) type 17 is an autosomal dominant ataxia caused by expanded polyglutamine (polyQ) tract in the TATA-box binding protein (TBP). Substantial studies have shown involvement of compromised mitochondria biogenesis regulator peroxisome proliferator-activated receptor gamma-coactivator 1 alpha (PGC-1α), nuclear factor erythroid 2-related factor 2 (NRF2), nuclear factor-Y subunit A (NFYA), and their downstream target genes in the pathogenesis of polyQ-expansion diseases. The extracts of Paeonia lactiflora (P. lactiflora) and Glycyrrhiza uralensis (G. uralensis) have long been used as a Chinese herbal medicine (CHM). Shaoyao Gancao Tang (SG-Tang) is a formulated CHM made of P. lactiflora and G. uralensis at a 1:1 ratio. In the present study, we demonstrated the aggregate-inhibitory and anti-oxidative effect of SG-Tang in 293 TBP/Q79 cells. We then showed that SG-Tang reduced the aggregates and ameliorated the neurite outgrowth deficits in TBP/Q79 SH-SY5Y cells. SG-Tang upregulated expression levels of NFYA, PGC-1α, NRF2, and their downstream target genes in TBP/Q79 SH-SY5Y cells. Knock down of NFYA, PGC-1α, and NRF2 attenuated the neurite outgrowth promoting effect of SG-Tang on TBP/Q79 SH-SY5Y cells. Furthermore, SG-Tang inhibited aggregation and rescued motor-deficits in SCA17 mouse model. The study results suggest the potential of SG-Tang in treating SCA17 and probable other polyQ diseases.
Collapse
Affiliation(s)
- Chiung-Mei Chen
- Department of Neurology, Chang-Gung Memorial Hospital, Chang-Gung University College of Medicine, Taoyuan 33305, Taiwan
| | - Wan-Ling Chen
- Department of Neurology, Chang-Gung Memorial Hospital, Chang-Gung University College of Medicine, Taoyuan 33305, Taiwan
| | - Chen-Ting Hung
- Department of Life Science, National Taiwan Normal University, Taipei 11677, Taiwan
| | - Te-Hsien Lin
- Department of Life Science, National Taiwan Normal University, Taipei 11677, Taiwan
| | | | - I-Cheng Chen
- Department of Neurology, Chang-Gung Memorial Hospital, Chang-Gung University College of Medicine, Taoyuan 33305, Taiwan
| | - Chih-Hsin Lin
- Department of Neurology, Chang-Gung Memorial Hospital, Chang-Gung University College of Medicine, Taoyuan 33305, Taiwan
| | - Chih-Ying Chao
- Department of Neurology, Chang-Gung Memorial Hospital, Chang-Gung University College of Medicine, Taoyuan 33305, Taiwan
| | - Yih-Ru Wu
- Department of Neurology, Chang-Gung Memorial Hospital, Chang-Gung University College of Medicine, Taoyuan 33305, Taiwan
| | - Kuo-Hsuan Chang
- Department of Neurology, Chang-Gung Memorial Hospital, Chang-Gung University College of Medicine, Taoyuan 33305, Taiwan
| | - Hsiu Mei Hsieh-Li
- Department of Life Science, National Taiwan Normal University, Taipei 11677, Taiwan
| | - Guey-Jen Lee-Chen
- Department of Life Science, National Taiwan Normal University, Taipei 11677, Taiwan
| |
Collapse
|
137
|
Saad El-Din S, Rashed L, Medhat E, Emad Aboulhoda B, Desoky Badawy A, Mohammed ShamsEldeen A, Abdelgwad M. Active form of vitamin D analogue mitigates neurodegenerative changes in Alzheimer's disease in rats by targeting Keap1/Nrf2 and MAPK-38p/ERK signaling pathways. Steroids 2020; 156:108586. [PMID: 31982424 DOI: 10.1016/j.steroids.2020.108586] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 12/06/2019] [Accepted: 01/20/2020] [Indexed: 12/19/2022]
Abstract
BACKGROUND The nuclear factor erythroid2-related factor2 (Nrf2), a chief transcriptional regulator of antioxidant response element (ARE), is considered a promising target for the prevention of Alzheimer's disease (AD). Vitamin D has been recognized to have a crucial role in improving AD cognitive functions. The present study was conducted to evaluate the effects of active vitamin D analogue, Maxacalcitol, on Keap1-Nrf2 signaling pathway in experimental Alzheimer's disease in rats. MATERIALS AND METHODS The study was conducted on thirty female white albino rats divided equally into 3 groups: control group, Alzheimer group induced by Lipopolysaccharide and Alzheimer group treated with active vitamin D3 analogue, Maxacalcitol. The following parameters were assessed in rat brain tissues: Gene expression of Nrf2, Keap1 and MAF by RT-PCR, protein levels of phosphorylated MAPK-38p and ERK1/2 by Western Blot Technique, estimation of HO-1, Amyloid β, p-Tau levels and serum levels of TNFα, IL-10 and total 25-hydroxyvitamin D, serum calcium levels, GSH and MDA levels were also estimated in addition to cognitive function tests and histo-pathological examination of rat brain tissues. RESULTS In Alzheimer group, there was a significant deficit in cognition along with down-regulation of gene expression of Nrf2 and the protein levels of its downstream antioxidant effectors (HO-1 and GSH) with increased levels of the lipid peroxidation biomarker MDA. Also, there was increased neuro-inflammation as evidenced by increased levels of TNFα and decreased levels of IL-10. Moreover, there were increased amyloid β load and enhanced levels of phosphorylation of MAPK-38 and ERK1/2 leading to hyperphosphorylation of Tau protein. In addition, there were decreased serum levels of both total 25-hydroxyvitamin D and calcium. Treatment with vitamin D3 analogue, Maxacalcitol significantly improved cognitive dysfunction and histopathological picture of the brains of Alzheimer rats. Also, Vitamin D analogue significantly increased expression of Nrf2 and its downstream effectors (HO-1 and GSH), improved serum levels of total 25-hydroxyvitamin D and calcium, decreased neuro-inflammation and Amyloid β load as well as hyperphosphorylation of MAPK-38, ERK1/2 and tau proteins were also observed. Therefore, these data suggest that vitamin D analogue, Maxacalcitol could be used as a therapeutic agent in treatment of Alzheimer disease.
Collapse
Affiliation(s)
- Shimaa Saad El-Din
- The Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Cairo University, Egypt
| | - Laila Rashed
- The Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Cairo University, Egypt
| | - Engy Medhat
- The Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Cairo University, Egypt
| | | | - Ahmed Desoky Badawy
- The Department of Physiology, Faculty of Medicine, October 6 University, Egypt
| | | | - Marwa Abdelgwad
- The Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Cairo University, Egypt
| |
Collapse
|
138
|
Jové M, Pradas I, Mota-Martorell N, Cabré R, Ayala V, Ferrer I, Pamplona R. Succination of Protein Thiols in Human Brain Aging. Front Aging Neurosci 2020; 12:52. [PMID: 32210786 PMCID: PMC7068737 DOI: 10.3389/fnagi.2020.00052] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Accepted: 02/17/2020] [Indexed: 01/07/2023] Open
Abstract
Human brain evolution toward complexity has been achieved with increasing energy supply as the main adaptation in brain metabolism. Energy metabolism, like other biochemical reactions in aerobic cells, is under enzymatic control and strictly regulated. Nevertheless, physiologically uncontrolled and deleterious reactions take place. It has been proposed that these reactions constitute the basic molecular mechanisms that underlie the maintenance or loss-of-function of neurons and, by extension, cerebral functions during brain aging. In this review article, we focus attention on the role of the nonenzymatic and irreversible adduction of fumarate to the protein thiols, which leads to the formation of S-(2-succino)cysteine (2SC; protein succination) in the human brain. In particular, we first offer a brief approach to the succination reaction, features related to the specificity of protein succination, methods for their detection and quantification, the bases for considering 2SC as a biomarker of mitochondrial stress, the succinated proteome, the cross-regional differences in 2SC content, and changes during brain aging, as well as the potential regulatory significance of fumarate and 2SC. We propose that 2SC defines cross-regional differences of metabolic mitochondrial stress in the human brain and that mitochondrial stress is sustained throughout the healthy adult lifespan in order to preserve neuronal function and survival.
Collapse
Affiliation(s)
- Mariona Jové
- Department of Experimental Medicine, University of Lleida-Lleida Biomedical Research Institute (UdL-IRBLleida), Lleida, Spain
| | - Irene Pradas
- Department of Experimental Medicine, University of Lleida-Lleida Biomedical Research Institute (UdL-IRBLleida), Lleida, Spain
| | - Natalia Mota-Martorell
- Department of Experimental Medicine, University of Lleida-Lleida Biomedical Research Institute (UdL-IRBLleida), Lleida, Spain
| | - Rosanna Cabré
- Department of Experimental Medicine, University of Lleida-Lleida Biomedical Research Institute (UdL-IRBLleida), Lleida, Spain
| | - Victoria Ayala
- Department of Experimental Medicine, University of Lleida-Lleida Biomedical Research Institute (UdL-IRBLleida), Lleida, Spain
| | - Isidre Ferrer
- Institute of Neuropathology, Bellvitge University Hospital, University of Barcelona, Biomedical Research Institute of Bellvitge (IDIBELL), L'Hospitalet de Llobregat (Barcelona), Barcelona, Spain.,Center for Biomedical Research on Neurodegenerative Diseases (CIBERNED), ISCIII, Madrid, Spain
| | - Reinald Pamplona
- Department of Experimental Medicine, University of Lleida-Lleida Biomedical Research Institute (UdL-IRBLleida), Lleida, Spain
| |
Collapse
|
139
|
Oksanen M, Hyötyläinen I, Trontti K, Rolova T, Wojciechowski S, Koskuvi M, Viitanen M, Levonen A, Hovatta I, Roybon L, Lehtonen Š, Kanninen KM, Hämäläinen RH, Koistinaho J. NF-E2-related factor 2 activation boosts antioxidant defenses and ameliorates inflammatory and amyloid properties in human Presenilin-1 mutated Alzheimer's disease astrocytes. Glia 2020; 68:589-599. [PMID: 31670864 PMCID: PMC7003860 DOI: 10.1002/glia.23741] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 10/11/2019] [Accepted: 10/11/2019] [Indexed: 12/31/2022]
Abstract
Alzheimer's disease (AD) is a common dementia affecting a vast number of individuals and significantly impairing quality of life. Despite extensive research in animal models and numerous promising treatment trials, there is still no curative treatment for AD. Astrocytes, the most common cell type of the central nervous system, have been shown to play a role in the major AD pathologies, including accumulation of amyloid plaques, neuroinflammation, and oxidative stress. Here, we show that inflammatory stimulation leads to metabolic activation of human astrocytes and reduces amyloid secretion. On the other hand, the activation of oxidative metabolism leads to increased reactive oxygen species production especially in AD astrocytes. While healthy astrocytes increase glutathione (GSH) release to protect the cells, Presenilin-1-mutated AD patient astrocytes do not. Thus, chronic inflammation is likely to induce oxidative damage in AD astrocytes. Activation of NRF2, the major regulator of cellular antioxidant defenses, encoded by the NFE2L2 gene, poses several beneficial effects on AD astrocytes. We report here that the activation of NRF2 pathway reduces amyloid secretion, normalizes cytokine release, and increases GSH secretion in AD astrocytes. NRF2 induction also activates the metabolism of astrocytes and increases the utilization of glycolysis. Taken together, targeting NRF2 in astrocytes could be a potent therapeutic strategy in AD.
Collapse
Affiliation(s)
- Minna Oksanen
- A. I. Virtanen Institute for Molecular SciencesUniversity of Eastern FinlandKuopioFinland
| | - Ida Hyötyläinen
- A. I. Virtanen Institute for Molecular SciencesUniversity of Eastern FinlandKuopioFinland
| | - Kalevi Trontti
- Neuroscience CenterUniversity of HelsinkiHelsinkiFinland
- SleepWell Research Program, Faculty of MedicineUniversity of HelsinkiHelsinkiFinland
- Department of Psychology and LogopedicsUniversity of HelsinkiHelsinkiFinland
| | - Taisia Rolova
- Neuroscience CenterUniversity of HelsinkiHelsinkiFinland
| | - Sara Wojciechowski
- A. I. Virtanen Institute for Molecular SciencesUniversity of Eastern FinlandKuopioFinland
| | - Marja Koskuvi
- A. I. Virtanen Institute for Molecular SciencesUniversity of Eastern FinlandKuopioFinland
- Neuroscience CenterUniversity of HelsinkiHelsinkiFinland
| | - Matti Viitanen
- Department of GeriatricsUniversity of Turku, Turku City HospitalTurkuFinland
- Department of GeriatricsKarolinska Institutet and Karolinska University HospitalStockholmSweden
| | - Anna‐Liisa Levonen
- A. I. Virtanen Institute for Molecular SciencesUniversity of Eastern FinlandKuopioFinland
| | - Iiris Hovatta
- Neuroscience CenterUniversity of HelsinkiHelsinkiFinland
- SleepWell Research Program, Faculty of MedicineUniversity of HelsinkiHelsinkiFinland
- Department of Psychology and LogopedicsUniversity of HelsinkiHelsinkiFinland
| | - Laurent Roybon
- Department of Experimental Medical Science and MultiPark and Lund Stem Cell CenterFaculty of Medicine, Lund UniversityLundSweden
| | - Šárka Lehtonen
- A. I. Virtanen Institute for Molecular SciencesUniversity of Eastern FinlandKuopioFinland
- Neuroscience CenterUniversity of HelsinkiHelsinkiFinland
| | - Katja M. Kanninen
- A. I. Virtanen Institute for Molecular SciencesUniversity of Eastern FinlandKuopioFinland
| | - Riikka H. Hämäläinen
- A. I. Virtanen Institute for Molecular SciencesUniversity of Eastern FinlandKuopioFinland
| | - Jari Koistinaho
- A. I. Virtanen Institute for Molecular SciencesUniversity of Eastern FinlandKuopioFinland
- Neuroscience CenterUniversity of HelsinkiHelsinkiFinland
| |
Collapse
|
140
|
Marchetti B, Tirolo C, L'Episcopo F, Caniglia S, Testa N, Smith JA, Pluchino S, Serapide MF. Parkinson's disease, aging and adult neurogenesis: Wnt/β-catenin signalling as the key to unlock the mystery of endogenous brain repair. Aging Cell 2020; 19:e13101. [PMID: 32050297 PMCID: PMC7059166 DOI: 10.1111/acel.13101] [Citation(s) in RCA: 123] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 11/27/2019] [Accepted: 12/25/2019] [Indexed: 12/14/2022] Open
Abstract
A common hallmark of age-dependent neurodegenerative diseases is an impairment of adult neurogenesis. Wingless-type mouse mammary tumor virus integration site (Wnt)/β-catenin (WβC) signalling is a vital pathway for dopaminergic (DAergic) neurogenesis and an essential signalling system during embryonic development and aging, the most critical risk factor for Parkinson's disease (PD). To date, there is no known cause or cure for PD. Here we focus on the potential to reawaken the impaired neurogenic niches to rejuvenate and repair the aged PD brain. Specifically, we highlight WβC-signalling in the plasticity of the subventricular zone (SVZ), the largest germinal region in the mature brain innervated by nigrostriatal DAergic terminals, and the mesencephalic aqueduct-periventricular region (Aq-PVR) Wnt-sensitive niche, which is in proximity to the SNpc and harbors neural stem progenitor cells (NSCs) with DAergic potential. The hallmark of the WβC pathway is the cytosolic accumulation of β-catenin, which enters the nucleus and associates with T cell factor/lymphoid enhancer binding factor (TCF/LEF) transcription factors, leading to the transcription of Wnt target genes. Here, we underscore the dynamic interplay between DAergic innervation and astroglial-derived factors regulating WβC-dependent transcription of key genes orchestrating NSC proliferation, survival, migration and differentiation. Aging, inflammation and oxidative stress synergize with neurotoxin exposure in "turning off" the WβC neurogenic switch via down-regulation of the nuclear factor erythroid-2-related factor 2/Wnt-regulated signalosome, a key player in the maintenance of antioxidant self-defense mechanisms and NSC homeostasis. Harnessing WβC-signalling in the aged PD brain can thus restore neurogenesis, rejuvenate the microenvironment, and promote neurorescue and regeneration.
Collapse
Affiliation(s)
- Bianca Marchetti
- Department of Biomedical and Biotechnological Sciences (BIOMETEC)Pharmacology and Physiology SectionsMedical SchoolUniversity of CataniaCataniaItaly
- Neuropharmacology SectionOASI Research Institute‐IRCCSTroinaItaly
| | - Cataldo Tirolo
- Neuropharmacology SectionOASI Research Institute‐IRCCSTroinaItaly
| | | | | | - Nunzio Testa
- Neuropharmacology SectionOASI Research Institute‐IRCCSTroinaItaly
| | - Jayden A. Smith
- Department of Clinical Neurosciences and NIHR Biomedical Research CentreUniversity of CambridgeCambridgeUK
| | - Stefano Pluchino
- Department of Clinical Neurosciences and NIHR Biomedical Research CentreUniversity of CambridgeCambridgeUK
| | - Maria F. Serapide
- Department of Biomedical and Biotechnological Sciences (BIOMETEC)Pharmacology and Physiology SectionsMedical SchoolUniversity of CataniaCataniaItaly
| |
Collapse
|
141
|
Wang T, Li C, Han B, Wang Z, Meng X, Zhang L, He J, Fu F. Neuroprotective effects of Danshensu on rotenone-induced Parkinson's disease models in vitro and in vivo. BMC Complement Med Ther 2020; 20:20. [PMID: 32020857 PMCID: PMC7076814 DOI: 10.1186/s12906-019-2738-7] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Accepted: 10/31/2019] [Indexed: 11/15/2022] Open
Abstract
Background Danshensu is an active constituent in the extracts of Danshen which is a traditional Chinese medical herb. Rotenone inhibits complex I of the mitochondrial electron transport chain in dopaminergic neurons leading to glutathione (GSH) level reduction and oxidative stress. The aim of this study is to investigate neuroprotective effects of Danshensu on rotenone-induced Parkinson’s disease (PD) in vitro and in vivo. Methods In vitro, SH-SY5Y human neuroblastoma cell line was pretreated with Danshensu and challenged with rotenone. Then the reactive oxygen species (ROS) production was assayed. In vivo, male C57BL/6 mice were intragastrically administered with Danshensu (15, 30, or 60 mg/kg), followed by oral administration with rotenone at a dose of 30 mg/kg. Pole and rotarod tests were carried out at 28 d to observe the effects of Danshensu on PD. Results Danshensu repressed ROS generation and therefore attenuated the rotenone-induced injury in SH-SY5Y cells. Danshensu improved motor dysfunction induced by rotenone, accompanied with reducing MDA content and increasing GSH level in striatum. Danshensu increased the number of TH positive neurons, the expression of TH and the dopamine contents. The expressions of p-PI3K, p-AKT, Nrf2, hemeoxygenase (HO-1), glutathione cysteine ligase regulatory subunit (GCLC), glutathione cysteine ligase modulatory subunit (GCLM) were significantly increased and the expression of Keap1 was decreased in Danshensu groups. Conclusions The neuroprotective effects of Danshensu on rotenone-induced PD are attributed to the anti-oxidative properties by activating PI3K/AKT/Nrf2 pathway and increasing Nrf2-induced expression of HO-1, GCLC, and GCLM, at least in part.
Collapse
Affiliation(s)
- Tian Wang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Qingquan Road 30#, Yantai, Shandong, 264005, People's Republic of China
| | - Cuiting Li
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Qingquan Road 30#, Yantai, Shandong, 264005, People's Republic of China
| | - Bing Han
- Center of Mitochondria and Healthy Aging, School of Life Science, Yantai University, Qingquan Road 30#, Yantai, Shandong, 264005, People's Republic of China
| | - Zhenhua Wang
- Center of Mitochondria and Healthy Aging, School of Life Science, Yantai University, Qingquan Road 30#, Yantai, Shandong, 264005, People's Republic of China
| | - Xiaoyu Meng
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Qingquan Road 30#, Yantai, Shandong, 264005, People's Republic of China
| | - Leiming Zhang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Qingquan Road 30#, Yantai, Shandong, 264005, People's Republic of China
| | - Jie He
- Center of Mitochondria and Healthy Aging, School of Life Science, Yantai University, Qingquan Road 30#, Yantai, Shandong, 264005, People's Republic of China
| | - Fenghua Fu
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Qingquan Road 30#, Yantai, Shandong, 264005, People's Republic of China.
| |
Collapse
|
142
|
Wu Y, Shi YG, Zheng XL, Dang YL, Zhu CM, Zhang RR, Fu YY, Zhou TY, Li JH. Lipophilic ferulic acid derivatives protect PC12 cells against oxidative damage via modulating β-amyloid aggregation and activating Nrf2 enzymes. Food Funct 2020; 11:4707-4718. [DOI: 10.1039/d0fo00800a] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Ferulic acid (FA) has been shown to have a neuroprotective effect on Alzheimer's disease induced by amyloid-beta (Aβ) neurotoxicity.
Collapse
Affiliation(s)
- Yu Wu
- School of Food Science and Biotechnology
- Zhejiang Gongshang University
- Hangzhou
- China
| | - Yu-gang Shi
- School of Food Science and Biotechnology
- Zhejiang Gongshang University
- Hangzhou
- China
| | - Xiao-liang Zheng
- Center for Molecular Medicine
- Hangzhou Medical College
- Hangzhou 310013
- China
| | - Ya-li Dang
- Key Laboratory of Animal Protein Food Processing Technology of Zhejiang Province
- Ningbo University
- Ningbo 315211
- China
| | - Chen-min Zhu
- School of Food Science and Biotechnology
- Zhejiang Gongshang University
- Hangzhou
- China
| | - Run-run Zhang
- School of Food Science and Biotechnology
- Zhejiang Gongshang University
- Hangzhou
- China
| | - Yu-ying Fu
- School of Food Science and Biotechnology
- Zhejiang Gongshang University
- Hangzhou
- China
| | - Tian-yi Zhou
- Center for Molecular Medicine
- Hangzhou Medical College
- Hangzhou 310013
- China
| | - Jun-hui Li
- Key Laboratory of Marine Food Quality and Hazard Controlling Technology of Zhejiang Province
- College of Life Sciences
- China Jiliang University
- Hangzhou, 310018
- China
| |
Collapse
|
143
|
Dong Y, Stewart T, Bai L, Li X, Xu T, Iliff J, Shi M, Zheng D, Yuan L, Wei T, Yang X, Zhang J. Coniferaldehyde attenuates Alzheimer's pathology via activation of Nrf2 and its targets. Am J Cancer Res 2020; 10:179-200. [PMID: 31903114 PMCID: PMC6929631 DOI: 10.7150/thno.36722] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 09/02/2019] [Indexed: 01/22/2023] Open
Abstract
Background: Alzheimer's disease (AD) currently lacks a cure. Because substantial neuronal damage usually occurs before AD is advanced enough for diagnosis, the best hope for disease-modifying AD therapies likely relies on early intervention or even prevention, and targeting multiple pathways implicated in early AD pathogenesis rather than focusing exclusively on excessive production of β-amyloid (Aβ) species. Methods: Coniferaldehyde (CFA), a food flavoring and agonist of NF-E2-related factor 2 (Nrf2), was selected by multimodal in vitro screening, followed by investigation of several downstream effects potentially involved. Furthermore, in the APP/PS1 AD mouse model, the therapeutic effects of CFA (0.2 mmol kg-1d-1) were tested beginning at 3 months of age. Behavioral phenotypes related to learning and memory capacity, brain pathology and biochemistry, including Aβ transport, were assessed at different time intervals. Results: CFA promoted neuron viability and showed potent neuroprotective effects, especially on mitochondrial structure and functions. In addition, CFA greatly enhanced the brain clearance of Aβ in both free and extracellular vesicle (EV)-contained Aβ forms. In the APP/PS1 mouse model, CFA effectively abolished brain Aβ deposits and reduced the level of toxic soluble Aβ peptides, thus eliminating AD-like pathological changes in the hippocampus and cerebral cortex and preserving learning and memory capacity of the mice. Conclusion: The experimental evidence overall indicated that Nrf2 activation may contribute to the potent anti-AD effects of CFA. With an excellent safety profile, further clinical investigation of coniferaldehyde might bring hope for AD prevention/therapy.
Collapse
|
144
|
Artemether Activation of AMPK/GSK3 β(ser9)/Nrf2 Signaling Confers Neuroprotection towards β-Amyloid-Induced Neurotoxicity in 3xTg Alzheimer's Mouse Model. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:1862437. [PMID: 31871541 PMCID: PMC6907052 DOI: 10.1155/2019/1862437] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/07/2019] [Revised: 09/06/2019] [Accepted: 09/14/2019] [Indexed: 12/15/2022]
Abstract
Alzheimer's disease is a severe neurodegenerative disease. Multiple factors involving neurofibrillary tangles and amyloid-β plaques lead to the progression of the AD, generated by aggregated hyperphosphorylated Tau protein. Inflammation, mitochondrial dysfunction, and oxidative stress play a significant role in the progression of AD. It has been therefore suggested that the multifactorial nature of AD pathogenesis requires the design of antioxidant drugs with a broad spectrum of neuroprotective activities. For this reason, the use of natural products, characterized by multiple pharmacological properties is advantageous as AD-modifying drugs over the single-targeted chemicals. Artemether, a peroxide sesquiterpenoid lipid-soluble compound, has been used in the clinic as an antimalarial drug. Also, it exhibits potent anti-inflammatory and antioxidant activities. Here, we report the neuroprotective effects of Artemether towards Aβ-induced neurotoxicity in neuronal cell cultures. A temporal correlation was found between Artemether neuroprotection towards Aβ-induced neurotoxicity and AMPK/GSK3β phosphorylation activity and increased expression of the activated Nrf2 signaling pathway. In 3xTg-AD mice, Artemether attenuated learning and memory deficits, inhibited cortical neuronal apoptosis and glial activation, inhibited oxidative stress through decrease of lipid peroxidation and increased expression of SOD, and reduced Aβ deposition and tau protein phosphorylation. Moreover, in 3xTg-AD mice, Artemether induced phosphorylation of the AMPK/GSK3β pathway which activated Nrf2, increasing the level of antioxidant protein HO-1. These activities probably produced the antioxidant and anti-inflammatory effects responsible for the neuroprotective effects of Artemether in the 3xTg-AD mouse model. These findings propose Artemether as a new drug for the treatment of AD disease.
Collapse
|
145
|
Kwon SH, Lee SR, Park YJ, Ra M, Lee Y, Pang C, Kim KH. Suppression of 6-Hydroxydopamine-Induced Oxidative Stress by Hyperoside Via Activation of Nrf2/HO-1 Signaling in Dopaminergic Neurons. Int J Mol Sci 2019; 20:ijms20235832. [PMID: 31757050 PMCID: PMC6929192 DOI: 10.3390/ijms20235832] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2019] [Revised: 11/15/2019] [Accepted: 11/18/2019] [Indexed: 02/07/2023] Open
Abstract
In our ongoing research to discover natural products with neuroprotective effects, hyperoside (quercetin 3-O-galactoside) was isolated from Acer tegmentosum, which has been used in Korean traditional medicine to treat liver-related disorders. Here, we demonstrated that hyperoside protects cultured dopaminergic neurons from death via reactive oxygen species (ROS)-dependent mechanisms, although other relevant mechanisms of hyperoside activity remain largely uncharacterized. For the first time, we investigated the neuroprotective effects of hyperoside on 6-hydroxydopamine (6-OHDA)-induced neurotoxicity in neurons, and the possible underlying mechanisms. Hyperoside significantly ameliorated the loss of neuronal cell viability, lactate dehydrogenase release, excessive ROS accumulation and mitochondrial membrane potential dysfunction associated with 6-OHDA-induced neurotoxicity. Furthermore, hyperoside treatment activated the nuclear erythroid 2-related factor 2 (Nrf2), an upstream molecule of heme oxygenase-1 (HO-1). Hyperoside also induced the expression of HO-1, an antioxidant response gene. Remarkably, we found that the neuroprotective effects of hyperoside were weakened by an Nrf2 small interfering RNA, which blocked the ability of hyperoside to inhibit neuronal death, indicating the vital role of HO-1. Overall, we show that hyperoside, via the induction of Nrf2-dependent HO-1 activation, suppresses neuronal death caused by 6-OHDA-induced oxidative stress. Moreover, Nrf2-dependent HO-1 signaling activation represents a potential preventive and therapeutic target in Parkinson's disease management.
Collapse
Affiliation(s)
- Seung-Hwan Kwon
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.;
- Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Seoung Rak Lee
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Korea; (S.R.L.); (Y.J.P.)
| | - Yong Joo Park
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Korea; (S.R.L.); (Y.J.P.)
| | - Moonjin Ra
- Hongcheon Institute of Medicinal Herb, 101 Yeonbongri, Hongcheon 25142, Korea; (M.R.); (Y.L.)
| | - Yongjun Lee
- Hongcheon Institute of Medicinal Herb, 101 Yeonbongri, Hongcheon 25142, Korea; (M.R.); (Y.L.)
| | - Changhyun Pang
- School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Korea
- SKKU Advanced Institute of Nanotechnology Sungkyunkwan University, Suwon 16419, Korea
- Correspondence: (C.P.); (K.H.K.); Tel.: +82-31-290-7341 (C.P.); +82-31-290-7700 (K.H.K.)
| | - Ki Hyun Kim
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Korea; (S.R.L.); (Y.J.P.)
- Correspondence: (C.P.); (K.H.K.); Tel.: +82-31-290-7341 (C.P.); +82-31-290-7700 (K.H.K.)
| |
Collapse
|
146
|
Ghanekar SD, Miller WW, Meyer CJ, Fenelon KJ, Lacdao A, Zesiewicz TA. Orphan Drugs In Development For The Treatment Of Friedreich's Ataxia: Focus On Omaveloxolone. Degener Neurol Neuromuscul Dis 2019; 9:103-107. [PMID: 31686946 PMCID: PMC6800542 DOI: 10.2147/dnnd.s180027] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Accepted: 09/06/2019] [Indexed: 11/23/2022] Open
Abstract
Friedreich’s Ataxia (FRDA) is a devastating and progressive ataxia, marked by mitochondrial dysfunction and oxidative stress. Nrf2 activators such as omaveloxolone (Omav) modulate antioxidative mechanisms, and thus may be viable therapeutic agents in FRDA.
Collapse
Affiliation(s)
- Shaila D Ghanekar
- University of South Florida (USF), Department of Neurology, Tampa, FL, USA.,USF Ataxia Research Center, Department of Neurology, Tampa, FL, USA
| | - Wai Wai Miller
- University of South Florida (USF), Department of Neurology, Tampa, FL, USA.,USF Ataxia Research Center, Department of Neurology, Tampa, FL, USA.,USF Movement Disorders Neuromodulation Center, Department of Neurology, Tampa, FL, USA
| | - Colin J Meyer
- Reata Pharmaceuticals, Inc., Department of Product Development, Irving, TX, USA
| | - Kevin J Fenelon
- University of South Florida (USF), Department of Neurology, Tampa, FL, USA.,USF Ataxia Research Center, Department of Neurology, Tampa, FL, USA
| | - Alvin Lacdao
- University of South Florida (USF), Department of Neurology, Tampa, FL, USA.,USF Ataxia Research Center, Department of Neurology, Tampa, FL, USA
| | - Theresa A Zesiewicz
- University of South Florida (USF), Department of Neurology, Tampa, FL, USA.,USF Ataxia Research Center, Department of Neurology, Tampa, FL, USA.,USF Movement Disorders Neuromodulation Center, Department of Neurology, Tampa, FL, USA.,James A. Haley Veterans' Hospital, Department of Neurology, Tampa, FL, USA
| |
Collapse
|
147
|
Neuroprotection of Indole-Derivative Compound NC001-8 by the Regulation of the NRF2 Pathway in Parkinson's Disease Cell Models. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:5074367. [PMID: 31781339 PMCID: PMC6874971 DOI: 10.1155/2019/5074367] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 04/05/2019] [Accepted: 07/10/2019] [Indexed: 02/07/2023]
Abstract
Parkinson's disease (PD) is a common neurodegenerative disease accompanied by a loss of dopaminergic (DAergic) neurons. The development of therapies to prevent disease progression is the main goal of drug discovery. There is increasing evidence that oxidative stress and antioxidants may contribute to the pathogenesis and treatment of PD, respectively. In the present study, we investigated the antioxidative protective effects of the indole-derivative compound NC001-8 in DAergic neurons derived from SH-SY5Y cells and PD-specific induced pluripotent stem cells (PD-iPSCs) carrying a PARKIN ex5del mutation. In SH-SY5Y-differentiated DAergic neurons under 1-methyl-4-phenylpyridinium (MPP+) treatment, NC001-8 remarkably reduced the levels of reactive oxygen species (ROS) and cleaved caspase 3; upregulated nuclear factor erythroid 2-related factor 2 (NRF2) and NAD(P)H dehydrogenase, quinone 1 (NQO1); and promoted neuronal viability. In contrast, NRF2 knockdown abolished the effect of NC001-8 on the reduction of ROS and improvement of neuronal viability. In H2O2-treated DAergic neurons differentiated from PD-iPSCs, NC001-8 rescued the aberrant increase in ROS and cleaved caspase 3 by upregulating NRF2 and NQO1. Our results demonstrated the protective effect of NC001-8 in DAergic neurons via promoting the NRF2 antioxidative pathway and reducing ROS levels. We anticipate that our present in vitro assays may be a starting point for more sophisticated in vivo models or clinical trials that evaluate the potential of NC001-8 as a disease modifier for PD.
Collapse
|
148
|
Zhou Y, Jiang Z, Lu H, Xu Z, Tong R, Shi J, Jia G. Recent Advances of Natural Polyphenols Activators for Keap1-Nrf2 Signaling Pathway. Chem Biodivers 2019; 16:e1900400. [PMID: 31482617 DOI: 10.1002/cbdv.201900400] [Citation(s) in RCA: 110] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 09/03/2019] [Indexed: 01/01/2023]
Abstract
The Keap1-Nrf2/ARE signaling pathway is an important defense system against exogenous and endogenous oxidative stress injury. The dysregulation of the signaling pathway is associated with many diseases, such as cancer, diabetes, and respiratory diseases. Over the years, a wide range of natural products has provided sufficient resources for the discovery of potential therapeutic drugs. Among them, polyphenols possess Nrf2 activation, not only inhibit the production of ROS, inhibit Keap1-Nrf2 protein-protein interaction, but also degrade Keap1 and regulate the Nrf2 related pathway. In fact, with the continuous improvement of natural polyphenols separation and purification technology and further studies on the Keap1-Nrf2 molecular mechanism, more and more natural polyphenols monomer components of Nrf2 activators have been gradually discovered. In this view, we summarize the research status of natural polyphenols that have been found with apparent Nrf2 activation and their action modes. On the whole, this review may guide the design of novel Keap1-Nrf2 activator.
Collapse
Affiliation(s)
- Yanping Zhou
- Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine of University of Electronic Science and Technology of China, No. 32 West Second Section First Ring Road, Chengdu, 610072, P. R. China
| | - Zhongliang Jiang
- Department of Hematology, Miller School of Medicine, University of Miami, Miami, 33136, USA
| | - Haiying Lu
- Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine of University of Electronic Science and Technology of China, No. 32 West Second Section First Ring Road, Chengdu, 610072, P. R. China
| | - Zhuyu Xu
- Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine of University of Electronic Science and Technology of China, No. 32 West Second Section First Ring Road, Chengdu, 610072, P. R. China
| | - Rongsheng Tong
- Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine of University of Electronic Science and Technology of China, No. 32 West Second Section First Ring Road, Chengdu, 610072, P. R. China
| | - Jianyou Shi
- Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine of University of Electronic Science and Technology of China, No. 32 West Second Section First Ring Road, Chengdu, 610072, P. R. China
| | - Guiqing Jia
- Department of Gastrointestinal Surgery, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, 610072, P. R. China
| |
Collapse
|
149
|
Iqubal A, Sharma S, Najmi AK, Syed MA, Ali J, Alam MM, Haque SE. Nerolidol ameliorates cyclophosphamide-induced oxidative stress, neuroinflammation and cognitive dysfunction: Plausible role of Nrf2 and NF- κB. Life Sci 2019; 236:116867. [PMID: 31520598 DOI: 10.1016/j.lfs.2019.116867] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Revised: 08/31/2019] [Accepted: 09/10/2019] [Indexed: 12/27/2022]
Abstract
AIM Cyclophosphamide (CP) is a potent anticancer and immunosuppressant drug. Studies have shown significant oxidative stress and cognitive impairment but neuroinflammatory and histological aberrations with its administration is underexplored. Nerolidol (NER) is a lipophilic bioactive molecule with antioxidant and anti-inflammatory properties but it has not been explored for neuroprotective potential in CP-induced neurotoxic manifestations. Therefore, in the present study, we aimed to evaluate the neuroprotective potential of NER in CP-induced neuroinflammation and associated comorbid conditions like depression and cognitive dysfunctions. MATERIALS AND METHOD In-silico study using Schrödinger software was used to assess the binding affinity of NER with Nrf2. In the In vivo study, NER 200 and 400 mg/kg p.o. were given from 1st day to 14th day. CP 200 mg/kg, i.p., was administered on the 7th day. After 24 h of the last dosing, neurobehavioral tests like spontaneous body alternation, passive avoidance and forced swim test were performed. On completion of study, mice were sacrificed, hippocampus and cortex were removed for biochemical estimations, histopathology and immunohistochemistry of p65 NF- κB and Nrf2. KEY FINDINGS In-silico study showed significant binding of NER into the pocket domain of Nrf2. In-vivo study showed protective effect of NER against CP-induced neuroinflammation, oxidative stress, cognitive impairment and structural abnormalities in the hippocampus and cortex regions. SIGNIFICANCE Findings of the study suggested that NER is a potential therapeutic molecule which can mitigate CP-induced neurotoxic manifestations via Nrf2 and NF-κB pathway. However, more detailed studies are needed to explicate the mechanism underlying its neuroprotective effect.
Collapse
Affiliation(s)
- Ashif Iqubal
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Sumit Sharma
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Abul Kalam Najmi
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Mansoor Ali Syed
- Department of Biotechnology, Jamia Millia Islamia, New Delhi 110025, India
| | - Javed Ali
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - M Mumtaz Alam
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Syed Ehtaishamul Haque
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India.
| |
Collapse
|
150
|
Paunkov A, Chartoumpekis DV, Ziros PG, Sykiotis GP. A Bibliometric Review of the Keap1/Nrf2 Pathway and its Related Antioxidant Compounds. Antioxidants (Basel) 2019; 8:antiox8090353. [PMID: 31480567 PMCID: PMC6769514 DOI: 10.3390/antiox8090353] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 08/21/2019] [Accepted: 08/23/2019] [Indexed: 12/19/2022] Open
Abstract
Nrf2 is a master transcriptional regulator of antioxidant and cytoprotective pathways. Currently in its third decade, research on Nrf2 has expanded to encompass not only basic but also clinical studies. In the present bibliometric review, we employed the VOSviewer tool to describe the existing Nrf2 literature landscape. As of July 2019, 11,931 papers on Nrf2 were listed in the “Web of Science” database, with more than 1000 new papers published each year. As expected, terms related to oxidative stress and antioxidant molecules occur very often in the Nrf2 literature throughout the years. Interestingly, there is also a gradual increase in the occurrence of terms related to diseases or to natural compounds, the most prominent being sulforaphane, curcumin, and resveratrol that modulate the Nrf2 pathway. Going beyond molecular biology/biochemistry and related fields, Nrf2 research has begun to spread into more clinical areas like endocrinology/metabolism, cardiology, and nephrology, likely reflecting an increased interest in clinical applications of Nrf2 pathway activators. China has become the most prolific producer of Nrf2 papers the last five years followed by the USA and Japan, a reverse pattern compared to the past. In conclusion, Nrf2 is the subject of a globally active research field that keeps growing and extends from bench to bedside.
Collapse
Affiliation(s)
- Ana Paunkov
- Service of Endocrinology, Diabetology and Metabolism, Lausanne University Hospital and University of Lausanne, SA08/02/250, Ave de la Sallaz 8, CH-1011 Lausanne, Switzerland
| | - Dionysios V Chartoumpekis
- Service of Endocrinology, Diabetology and Metabolism, Lausanne University Hospital and University of Lausanne, SA08/02/250, Ave de la Sallaz 8, CH-1011 Lausanne, Switzerland
- Division of Endocrinology, Department of Internal Medicine, School of Medicine, University of Patras, 265 04 Patras, Greece
| | - Panos G Ziros
- Service of Endocrinology, Diabetology and Metabolism, Lausanne University Hospital and University of Lausanne, SA08/02/250, Ave de la Sallaz 8, CH-1011 Lausanne, Switzerland
| | - Gerasimos P Sykiotis
- Service of Endocrinology, Diabetology and Metabolism, Lausanne University Hospital and University of Lausanne, SA08/02/250, Ave de la Sallaz 8, CH-1011 Lausanne, Switzerland.
| |
Collapse
|