101
|
Shilpa O, Anupama KP, Antony A, Gurushankara HP. Lead (Pb) induced Oxidative Stress as a Mechanism to Cause Neurotoxicity in Drosophila melanogaster. Toxicology 2021; 462:152959. [PMID: 34560124 DOI: 10.1016/j.tox.2021.152959] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 09/14/2021] [Accepted: 09/16/2021] [Indexed: 12/20/2022]
Abstract
The widespread use of lead (Pb) has caused global contamination, inevitable human exposure, and public health problems. Pb neurotoxicity has been linked to various human diseases, but its associated mechanism causing neurotoxicity is unknown. Drosophila melanogaster as a model organism has been used to study the mechanism involved in Pb-caused neurotoxicity and the potential role of antioxidants in ameliorating its harmful effects. The larval feeding technique was adopted to administer different concentrations of Pb (0.2-0.8 mM) to Oregon-R (ORR), superoxide dismutase (Sod), or catalase (Cat) overexpressing, and Sod or Cat knockdown flies to analyse Pb load, oxidative stress components, DNA damage, apoptosis and vacuolation in the brain. The results revealed that Pb accumulation in the Drosophila brain induces oxidative stress by generating reactive oxygen species (ROS) and lipid peroxidation (LPO), depleting antioxidant enzymes. Molecular docking studies have evidenced it. Pb directly binds to antioxidants and major grooves of DNA, leading to DNA damage. Increased DNA damage, apoptosis, vacuolation in brains of Pb-treated ORR, Sod, or Cat knockdown flies; and on the contrary, reduced oxidative DNA damage, apoptosis, and vacuolation in brains of Pb treated Sod or Cat overexpressed flies put forward that oxidative stress is the mechanism in Pb caused neurotoxicity.
Collapse
Affiliation(s)
- Olakkaran Shilpa
- Department of Zoology, School of Biological Sciences, Central University of Kerala, Tejaswini Hills, Periya, 671320, Kasaragod, India
| | - Kizhakke Purayil Anupama
- Department of Zoology, School of Biological Sciences, Central University of Kerala, Tejaswini Hills, Periya, 671320, Kasaragod, India
| | - Anet Antony
- Department of Zoology, School of Biological Sciences, Central University of Kerala, Tejaswini Hills, Periya, 671320, Kasaragod, India
| | | |
Collapse
|
102
|
Garcia A, Wang K, Bedier F, Benavides M, Wan Z, Wang S, Wang Y. Plasmonic Imaging of Electrochemical Reactions at Individual Prussian Blue Nanoparticles. Front Chem 2021; 9:718666. [PMID: 34552911 PMCID: PMC8450507 DOI: 10.3389/fchem.2021.718666] [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: 06/01/2021] [Accepted: 08/23/2021] [Indexed: 11/25/2022] Open
Abstract
Prussian blue is an iron-cyanide-based pigment steadily becoming a widely used electrochemical sensor in detecting hydrogen peroxide at low concentration levels. Prussian blue nanoparticles (PBNPs) have been extensively studied using traditional ensemble methods, which only provide averaged information. Investigating PBNPs at a single entity level is paramount for correlating the electrochemical activities to particle structures and will shed light on the major factors governing the catalyst activity of these nanoparticles. Here we report on using plasmonic electrochemical microscopy (PEM) to study the electrochemistry of PBNPs at the individual nanoparticle level. First, two types of PBNPs were synthesized; type I synthesized with double precursors method and type II synthesized with polyvinylpyrrolidone (PVP) assisted single precursor method. Second, both PBNPs types were compared on their electrochemical reduction to form Prussian white, and the effect from the different particle structures was investigated. Type I PBNPs provided better PEM sensitivity and were used to study the catalytic reduction of hydrogen peroxide. Progressively decreasing plasmonic signals with respect to increasing hydrogen peroxide concentration were observed, demonstrating the capability of sensing hydrogen peroxide at a single nanoparticle level utilizing this optical imaging technique.
Collapse
Affiliation(s)
- Adaly Garcia
- Department of Chemistry and Biochemistry, California State University, Los Angeles, Los Angeles, CA, United States
| | - Kinsley Wang
- Department of Chemistry and Biochemistry, California State University, Los Angeles, Los Angeles, CA, United States
| | - Fatima Bedier
- Department of Chemistry and Biochemistry, California State University, Los Angeles, Los Angeles, CA, United States
| | - Miriam Benavides
- Department of Chemistry and Biochemistry, California State University, Los Angeles, Los Angeles, CA, United States
| | - Zijian Wan
- Biodesign Center for Biosensors and Bioelectronics, Arizona State University, Tempe, AZ, United States.,School of Electrical, Computer and Energy Engineering, Arizona State University, Tempe, AZ, United States
| | - Shaopeng Wang
- Biodesign Center for Biosensors and Bioelectronics, Arizona State University, Tempe, AZ, United States.,School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, United States
| | - Yixian Wang
- Department of Chemistry and Biochemistry, California State University, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
103
|
Klichkhanov NK, Dzhafarova AM. Effect of Mild Hypothermia on the Catalytic Characteristics of Synaptic Acetylcholinesterase during Subtotal Global Cerebral Ischemia in Rats. NEUROCHEM J+ 2021. [DOI: 10.1134/s1819712421030077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
104
|
Gadhave K, Kumar D, Uversky VN, Giri R. A multitude of signaling pathways associated with Alzheimer's disease and their roles in AD pathogenesis and therapy. Med Res Rev 2021; 41:2689-2745. [PMID: 32783388 PMCID: PMC7876169 DOI: 10.1002/med.21719] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 07/13/2020] [Accepted: 07/29/2020] [Indexed: 02/06/2023]
Abstract
The exact molecular mechanisms associated with Alzheimer's disease (AD) pathology continue to represent a mystery. In the past decades, comprehensive data were generated on the involvement of different signaling pathways in the AD pathogenesis. However, the utilization of signaling pathways as potential targets for the development of drugs against AD is rather limited due to the immense complexity of the brain and intricate molecular links between these pathways. Therefore, finding a correlation and cross-talk between these signaling pathways and establishing different therapeutic targets within and between those pathways are needed for better understanding of the biological events responsible for the AD-related neurodegeneration. For example, autophagy is a conservative cellular process that shows link with many other AD-related pathways and is crucial for maintenance of the correct cellular balance by degrading AD-associated pathogenic proteins. Considering the central role of autophagy in AD and its interplay with many other pathways, the finest therapeutic strategy to fight against AD is the use of autophagy as a target. As an essential step in this direction, this comprehensive review represents recent findings on the individual AD-related signaling pathways, describes key features of these pathways and their cross-talk with autophagy, represents current drug development, and introduces some of the multitarget beneficial approaches and strategies for the therapeutic intervention of AD.
Collapse
Affiliation(s)
- Kundlik Gadhave
- School of Basic Sciences, Indian Institute of Technology Mandi, Kamand, Himachal Pradesh, 175005, India
| | - Deepak Kumar
- School of Basic Sciences, Indian Institute of Technology Mandi, Kamand, Himachal Pradesh, 175005, India
| | - Vladimir N. Uversky
- Department of Molecular Medicine and Byrd Alzheimer’s Research Institute, Morsani College of Medicine, University of South Florida, Tampa, Florida, United States of America
- Laboratory of New Methods in Biology, Institute for Biological Instrumentation, Russian Academy of Sciences, Pushchino, Moscow Region, Russia
| | - Rajanish Giri
- School of Basic Sciences, Indian Institute of Technology Mandi, Kamand, Himachal Pradesh, 175005, India
| |
Collapse
|
105
|
Fernandez‐Perez EJ, Muñoz B, Bascuñan DA, Peters C, Riffo‐Lepe NO, Espinoza MP, Morgan PJ, Filippi C, Bourboulou R, Sengupta U, Kayed R, Epsztein J, Aguayo LG. Synaptic dysregulation and hyperexcitability induced by intracellular amyloid beta oligomers. Aging Cell 2021; 20:e13455. [PMID: 34409748 PMCID: PMC8441418 DOI: 10.1111/acel.13455] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 07/21/2021] [Accepted: 08/05/2021] [Indexed: 12/19/2022] Open
Abstract
Intracellular amyloid beta oligomer (iAβo) accumulation and neuronal hyperexcitability are two crucial events at early stages of Alzheimer's disease (AD). However, to date, no mechanism linking iAβo with an increase in neuronal excitability has been reported. Here, the effects of human AD brain-derived (h-iAβo) and synthetic (iAβo) peptides on synaptic currents and action potential firing were investigated in hippocampal neurons. Starting from 500 pM, iAβo rapidly increased the frequency of synaptic currents and higher concentrations potentiated the AMPA receptor-mediated current. Both effects were PKC-dependent. Parallel recordings of synaptic currents and nitric oxide (NO)-associated fluorescence showed that the increased frequency, related to pre-synaptic release, was dependent on a NO-mediated retrograde signaling. Moreover, increased synchronization in NO production was also observed in neurons neighboring those dialyzed with iAβo, indicating that iAβo can increase network excitability at a distance. Current-clamp recordings suggested that iAβo increased neuronal excitability via AMPA-driven synaptic activity without altering membrane intrinsic properties. These results strongly indicate that iAβo causes functional spreading of hyperexcitability through a synaptic-driven mechanism and offers an important neuropathological significance to intracellular species in the initial stages of AD, which include brain hyperexcitability and seizures.
Collapse
Affiliation(s)
| | - Braulio Muñoz
- Laboratory of NeurophysiologyDepartment of PhysiologyUniversidad de ConcepciónConcepciónChile
| | - Denisse A. Bascuñan
- Laboratory of NeurophysiologyDepartment of PhysiologyUniversidad de ConcepciónConcepciónChile
| | - Christian Peters
- Laboratory of NeurophysiologyDepartment of PhysiologyUniversidad de ConcepciónConcepciónChile
| | - Nicolas O. Riffo‐Lepe
- Laboratory of NeurophysiologyDepartment of PhysiologyUniversidad de ConcepciónConcepciónChile
| | - Maria P. Espinoza
- Laboratory of NeurophysiologyDepartment of PhysiologyUniversidad de ConcepciónConcepciónChile
| | - Peter J. Morgan
- Institute of Neurobiology of the Mediterranean Sea (INMED)Institut National de la Santé et de la Recherche Médicale (INSERM) U901, Aix-Marseille UniversitéMarseilleFrance
| | - Caroline Filippi
- Institute of Neurobiology of the Mediterranean Sea (INMED)Institut National de la Santé et de la Recherche Médicale (INSERM) U901, Aix-Marseille UniversitéMarseilleFrance
| | - Romain Bourboulou
- Institute of Neurobiology of the Mediterranean Sea (INMED)Institut National de la Santé et de la Recherche Médicale (INSERM) U901, Aix-Marseille UniversitéMarseilleFrance
| | - Urmi Sengupta
- Mitchell Center for Neurodegenerative DiseasesUniversity of Texas Medical BranchGalvestonTexasUSA
- Department of Neurology, Neuroscience and Cell BiologyUniversity of Texas Medical BranchGalvestonTexasUSA
| | - Rakez Kayed
- Mitchell Center for Neurodegenerative DiseasesUniversity of Texas Medical BranchGalvestonTexasUSA
- Department of Neurology, Neuroscience and Cell BiologyUniversity of Texas Medical BranchGalvestonTexasUSA
| | - Jérôme Epsztein
- Institute of Neurobiology of the Mediterranean Sea (INMED)Institut National de la Santé et de la Recherche Médicale (INSERM) U901, Aix-Marseille UniversitéMarseilleFrance
| | - Luis G. Aguayo
- Laboratory of NeurophysiologyDepartment of PhysiologyUniversidad de ConcepciónConcepciónChile
| |
Collapse
|
106
|
Srivastava R, Choudhury PK, Dev SK, Rathore V. Neuroprotective effect of α-pinene self-emulsifying nanoformulation against 6-OHDA induced neurotoxicity on human SH-SY5Y cells and its in vivo validation for anti-Parkinson's effect. J Biochem Mol Toxicol 2021; 35:e22902. [PMID: 34464010 DOI: 10.1002/jbt.22902] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 08/15/2021] [Accepted: 08/20/2021] [Indexed: 11/07/2022]
Abstract
Oxidative stress (OS) is involved in the multifaceted pathogenic paradigm of neurodegenerative diseases like Parkinson's disease (PD). Monoterpenes like α-pinene (ALP) is considered to be a therapeutically potent antioxidant agent able to attenuate and scavenge various reactive oxygen species and reactive nitrogen species. The present study aimed to evaluate the in vitro and in vivo neuroprotective effect of α-pinene self-emulsifying nanoformulation (ALP-SENF) for PD. 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide assay was done to evaluate the neurotoxic dose of the ALP-SENF; however, the neuroprotective effect was assessed by 6-hydroxydopamine (6-OHDA) induced neurotoxicity model on SH-SY5Y taking NAC (N-acetyl-l-cysteine) as standard. The in vivo anti-Parkinson's activity of the ALP-SENF was compared with that of the plain ALP suspension by using reserpine antagonism and haloperidol-induced Parkinsonism model in rats. Various behavioral tests and biochemical antioxidant enzymes were estimated. The in vitro results revealed that treatment with ALP-SENF at a concentration of 100 and 200 µM was found to show significant neuronal SH-SY5Y cell viability against 50 µM 6-OHDA. ALP-SENF treated animals have seen significant neurobehavioral improvement. Furthermore, the levels of antioxidative enzymes in biochemical test reveals a marked enhancement in the expression of antioxidant enzymes that significantly attenuated the OS induced neurodegeneration. Due to the mechanisms of their antioxidant action, it was probably due to the scavenging of free radicals and the expression of antioxidant enzymes. It also improved neurobehavioral changes induced by reserpine and haloperidol.
Collapse
Affiliation(s)
- Rajnish Srivastava
- Department of Pharmaceutical Sciences, Mohanlal Sukhadia University, Udaipur, India
| | - Pratim K Choudhury
- Department of Pharmaceutical Sciences, Mohanlal Sukhadia University, Udaipur, India
| | - Suresh K Dev
- Department of Pharmaceutical Sciences, Mohanlal Sukhadia University, Udaipur, India
| | - Vaibhav Rathore
- Department of Pharmaceutical Sciences, Mohanlal Sukhadia University, Udaipur, India
| |
Collapse
|
107
|
Fasae KD, Abolaji AO, Faloye TR, Odunsi AY, Oyetayo BO, Enya JI, Rotimi JA, Akinyemi RO, Whitworth AJ, Aschner M. Metallobiology and therapeutic chelation of biometals (copper, zinc and iron) in Alzheimer's disease: Limitations, and current and future perspectives. J Trace Elem Med Biol 2021; 67:126779. [PMID: 34034029 DOI: 10.1016/j.jtemb.2021.126779] [Citation(s) in RCA: 65] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Revised: 04/03/2021] [Accepted: 05/10/2021] [Indexed: 02/08/2023]
Abstract
BACKGROUND Alzheimer's disease (AD) is the most prevalent cause of cognitive impairment and dementia worldwide. The pathobiology of the disease has been studied in the form of several hypotheses, ranging from oxidative stress, amyloid-beta (Aβ) aggregation, accumulation of tau forming neurofibrillary tangles (NFT) through metal dysregulation and homeostasis, dysfunction of the cholinergic system, and to inflammatory and autophagic mechanism. However, none of these hypotheses has led to confirmed diagnostics or approved cure for the disease. OBJECTIVE This review is aimed as a basic and an encyclopedic short course into metals in AD and discusses the advances in chelation strategies and developments adopted in the treatment of the disease. Since there is accumulating evidence of the role of both biometal dyshomeostasis (iron (Fe), copper (Cu), and zinc (Zn)) and metal-amyloid interactions that lead to the pathogenesis of AD, this review focuses on unraveling therapeutic chelation strategies that have been considered in the treatment of the disease, aiming to sequester free and protein-bound metal ions and reducing cerebral metal burden. Promising compounds possessing chemically modified moieties evolving as multi-target ligands used as anti-AD drug candidates are also covered. RESULTS AND CONCLUSION Several multidirectional and multifaceted studies on metal chelation therapeutics show the need for improved synthesis, screening, and analysis of compounds to be able to effectively present chelating anti-AD drugs. Most drug candidates studied have limitations in their physicochemical properties; some enhance redistribution of metal ions, while others indirectly activate signaling pathways in AD. The metal chelation process in vivo still needs to be established and the design of potential anti-AD compounds that bi-functionally sequester metal ions as well as inhibit the Aβ aggregation by competing with the metal ions and reducing metal-induced oxidative damage and neurotoxicity may signal a bright end in chelation-based therapeutics of AD.
Collapse
Affiliation(s)
- Kehinde D Fasae
- Department of Biochemistry, Molecular Drug Metabolism and Toxicology Unit, College of Medicine, University of Ibadan, Nigeria
| | - Amos O Abolaji
- Department of Biochemistry, Molecular Drug Metabolism and Toxicology Unit, College of Medicine, University of Ibadan, Nigeria.
| | - Tolulope R Faloye
- Department of Biochemistry, Molecular Drug Metabolism and Toxicology Unit, College of Medicine, University of Ibadan, Nigeria
| | - Atinuke Y Odunsi
- Department of Biochemistry, Molecular Drug Metabolism and Toxicology Unit, College of Medicine, University of Ibadan, Nigeria
| | - Bolaji O Oyetayo
- Department of Pharmacology and Therapeutics, Neuropharmacology Unit, College of Medicine, University of Ibadan, Nigeria
| | - Joseph I Enya
- Department of Anatomy, University of Ilorin, Kwara State, Nigeria
| | - Joshua A Rotimi
- Department of Biochemistry and Molecular Biology, Obafemi Awolowo University, Ile-Ife, Nigeria
| | - Rufus O Akinyemi
- Neuroscience and Ageing Research Unit, Institute for Advanced Medical Research and Training, College of Medicine, University of Ibadan, Ibadan, Oyo State, Nigeria
| | | | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
108
|
Garrick JM, Cole TB, Bammler TK, MacDonald JW, Marsillach J, Furlong CE, Costa LG. Paraoxonase 2 deficiency in mice alters motor behavior and causes region-specific transcript changes in the brain. Neurotoxicol Teratol 2021; 87:107010. [PMID: 34216730 PMCID: PMC8440460 DOI: 10.1016/j.ntt.2021.107010] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 06/07/2021] [Accepted: 06/28/2021] [Indexed: 01/01/2023]
Abstract
Paraoxonase 2 (PON2) is an intracellular antioxidant enzyme shown to play an important role in mitigating oxidative stress in the brain. Oxidative stress is a common mechanism of toxicity for neurotoxicants and is increasingly implicated in the etiology of multiple neurological diseases. While PON2 deficiency increases oxidative stress in the brain in-vitro, little is known about its effects on behavior in-vivo and what global transcript changes occur from PON2 deficiency. We sought to characterize the effects of PON2 deficiency on behavior in mice, with an emphasis on locomotion, and evaluate transcriptional changes with RNA-Seq. Behavioral endpoints included home-cage behavior (Noldus PhenoTyper), motor coordination (Rotarod) and various gait metrics (Noldus CatWalk). Home-cage behavior analysis showed PON2 deficient mice had increased activity at night compared to wildtype controls and spent more time in the center of the cage, displaying a possible anxiolytic phenotype. PON2 deficient mice had significantly shorter latency to fall when tested on the rotarod, suggesting impaired motor coordination. Minimal gait alterations were observed, with decreased girdle support posture noted as the only significant change in gait with PON2 deficiency. Beyond one home-cage metric, no significant sex-based behavioral differences were found in this study. Finally, A subset of samples were utilized for RNA-Seq analysis, looking at three discrete brain regions: cerebral cortex, striatum, and cerebellum. Highly regional- and sex-specific changes in RNA expression were found when comparing PON2 deficient and wildtype mice, suggesting PON2 may play distinct regional roles in the brain in a sex-specific manner. Taken together, these findings demonstrates that PON2 deficiency significantly alters the brain on both a biochemical and phenotypic level, with a specific impact on motor function. These data have implications for future gene-environment toxicological studies and warrants further investigation of the role of PON2 in the brain.
Collapse
Affiliation(s)
- Jacqueline M Garrick
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA, United States..
| | - Toby B Cole
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA, United States.; Center on Human Development and Disabilities, University of Washington, United States
| | - Theo K Bammler
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA, United States
| | - James W MacDonald
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA, United States
| | - Judit Marsillach
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA, United States
| | - Clement E Furlong
- Depts. of Medicine (Div. Medical Genetics) and of Genome Sciences, University of Washington, United States
| | - Lucio G Costa
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA, United States.; Dept. of Medicine and Surgery, University of Parma, Italy
| |
Collapse
|
109
|
Coulibaly AP, Pezuk P, Varghese P, Gartman W, Triebwasser D, Kulas JA, Liu L, Syed M, Tvrdik P, Ferris H, Provencio JJ. Neutrophil Enzyme Myeloperoxidase Modulates Neuronal Response in a Model of Subarachnoid Hemorrhage by Venous Injury. Stroke 2021; 52:3374-3384. [PMID: 34404234 PMCID: PMC8478903 DOI: 10.1161/strokeaha.120.033513] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Mariam Syed
- Department of Neurology (M.S., J.J.P.), University of Virginia, Charlottesville
| | - Petr Tvrdik
- Department of Neuroscience (P.T., H.F., J.J.P.), University of Virginia, Charlottesville.,Department of Neurosurgery (P.T.), University of Virginia, Charlottesville
| | - Heather Ferris
- Department of Neuroscience (P.T., H.F., J.J.P.), University of Virginia, Charlottesville.,Division of Endocrinology, Department of Internal Medicine (H.F.), University of Virginia, Charlottesville
| | - J Javier Provencio
- Department of Neurology (M.S., J.J.P.), University of Virginia, Charlottesville.,Department of Neuroscience (P.T., H.F., J.J.P.), University of Virginia, Charlottesville
| |
Collapse
|
110
|
Han Y, Wang T, Li C, Wang Z, Zhao Y, He J, Fu L, Han B. Ginsenoside Rg3 exerts a neuroprotective effect in rotenone-induced Parkinson's disease mice via its anti-oxidative properties. Eur J Pharmacol 2021; 909:174413. [PMID: 34391769 DOI: 10.1016/j.ejphar.2021.174413] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 07/24/2021] [Accepted: 08/06/2021] [Indexed: 01/05/2023]
Abstract
Ginsenoside Rg3, extracted from Panax ginseng C.A. Meyer, has been shown to possess neuroprotective properties. The present study aims to investigate the neuroprotective effects of ginsenoside Rg3 on rotenone-induced Parkinson's disease mice. Rotenone, a mitochondrial complex I inhibitor, leads to the augmentation of reactive oxygen species production in cells. Male C57/BL6 mice were intragastrically administered rotenone (30 mg/kg) and then treated with ginsenoside Rg3 (5, 10, or 20 mg/kg). Pole, rotarod, and open field tests were performed to evaluate motor function. Ginsenoside Rg3 decreased the climbing time in the pole test (p < 0.01), whereas it increased the latency in the rotarod test (p < 0.01) and the total distance (p < 0.01) and mean speed in the open field test (p < 0.01). Ginsenoside Rg3 treatment augmented the number of tyrosine hydroxylase-positive neurons in the substantia nigra (p < 0.01), mean density of tyrosine hydroxylase-positive nerve fibers (p < 0.01), and dopamine content (p < 0.01) in the striatum and reduced the reactive oxygen species level in the substantia nigra (p < 0.01). Glutathione cysteine ligase regulatory subunit and glutathione cysteine ligase modulatory subunit expression levels were elevated in the ginsenoside Rg3 groups. Ginsenoside Rg3 also improved motor function in rotenone-induced Parkinson's disease mice. The neuroprotective effects of ginsenoside Rg3 are at least partly associated with its anti-oxidative properties via regulation of glutathione cysteine ligase modulatory subunit and glutathione cysteine ligase regulatory subunit expression.
Collapse
Affiliation(s)
- Yingjie Han
- Center for Mitochondria and Healthy Aging, College of Life Science, Yantai University, Yantai, 264005, PR China
| | - Tian Wang
- School of Pharmacy, Yantai University, Yantai, Shandong, 264005, PR China
| | - Chunyan Li
- 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
| | - Yue Zhao
- 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
| | - Li Fu
- Dalian Fusheng Natural Medicine Development Co., Ltd, Dalian, 116600, PR China.
| | - Bing Han
- Center for Mitochondria and Healthy Aging, College of Life Science, Yantai University, Yantai, 264005, PR China.
| |
Collapse
|
111
|
Kimura AM, Tsuji M, Yasumoto T, Mori Y, Oguchi T, Tsuji Y, Umino M, Umino A, Nishikawa T, Nakamura S, Inoue T, Kiuchi Y, Yamada M, Teplow DB, Ono K. Myricetin prevents high molecular weight Aβ 1-42 oligomer-induced neurotoxicity through antioxidant effects in cell membranes and mitochondria. Free Radic Biol Med 2021; 171:232-244. [PMID: 34015458 DOI: 10.1016/j.freeradbiomed.2021.05.019] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 04/27/2021] [Accepted: 05/10/2021] [Indexed: 12/14/2022]
Abstract
Excessive accumulation of amyloid β-protein (Aβ) is one of the primary mechanisms that leads to neuronal death with phosphorylated tau in the pathogenesis of Alzheimer's disease (AD). Protofibrils, one of the high-molecular-weight Aβ oligomers (HMW-Aβo), are implicated to be important targets of disease modifying therapy of AD. We previously reported that phenolic compounds such as myricetin inhibit Aβ1-40, Aβ1-42, and α-synuclein aggregations, including their oligomerizations, which may exert protective effects against AD and Parkinson's disease. The purpose of this study was to clarify the detailed mechanism of the protective effect of myricetin against the neurotoxicity of HMW-Aβo in SH-SY5Y cells. To assess the effect of myricetin on HMW-Aβo-induced oxidative stress, we systematically examined the level of membrane oxidative damage by measuring cell membrane lipid peroxidation, membrane fluidity, and cell membrane potential, and the mitochondrial oxidative damage was evaluated by mitochondrial permeability transition (MPT), mitochondrial reactive oxygen species (ROS), and manganese-superoxide dismutase (Mn-SOD), and adenosine triphosphate (ATP) assay in SH-SY5Y cells. Myricetin has been found to increased cell viability by suppression of HMW-Aβo-induced membrane disruption in SH-SY5Y cells, as shown in reducing membrane phospholipid peroxidation and increasing membrane fluidity and membrane resistance. Myricetin has also been found to suppress HMW-Aβo-induced mitochondria dysfunction, as demonstrated in decreasing MPT, Mn-SOD, and ATP generation, raising mitochondrial membrane potential, and increasing mitochondrial-ROS generation. These results suggest that myricetin preventing HMW-Aβo-induced neurotoxicity through multiple antioxidant functions may be developed as a disease-modifying agent against AD.
Collapse
Affiliation(s)
- Atsushi Michael Kimura
- Department of Pharmacology, Division of Medical Pharmacology, School of Medicine, Showa University, Tokyo, 142-8555, Japan; Pharmacological Research Center, Showa University, Tokyo, 142-8555, Japan; Department of Internal Medicine, Division of Neurology, School of Medicine, Showa University, Tokyo, 142-8666, Japan
| | - Mayumi Tsuji
- Pharmacological Research Center, Showa University, Tokyo, 142-8555, Japan.
| | - Taro Yasumoto
- Department of Internal Medicine, Division of Neurology, School of Medicine, Showa University, Tokyo, 142-8666, Japan
| | - Yukiko Mori
- Department of Internal Medicine, Division of Neurology, School of Medicine, Showa University, Tokyo, 142-8666, Japan
| | - Tatsunori Oguchi
- Department of Pharmacology, Division of Medical Pharmacology, School of Medicine, Showa University, Tokyo, 142-8555, Japan; Pharmacological Research Center, Showa University, Tokyo, 142-8555, Japan
| | - Yuya Tsuji
- Department of Pharmacology, Division of Medical Pharmacology, School of Medicine, Showa University, Tokyo, 142-8555, Japan; Pharmacological Research Center, Showa University, Tokyo, 142-8555, Japan
| | - Masakazu Umino
- Department of Pharmacology, Division of Medical Pharmacology, School of Medicine, Showa University, Tokyo, 142-8555, Japan; Pharmacological Research Center, Showa University, Tokyo, 142-8555, Japan
| | - Asami Umino
- Department of Pharmacology, Division of Medical Pharmacology, School of Medicine, Showa University, Tokyo, 142-8555, Japan; Pharmacological Research Center, Showa University, Tokyo, 142-8555, Japan
| | - Toru Nishikawa
- Department of Pharmacology, Division of Medical Pharmacology, School of Medicine, Showa University, Tokyo, 142-8555, Japan; Pharmacological Research Center, Showa University, Tokyo, 142-8555, Japan
| | - Shiro Nakamura
- Department of Oral Physiology, School of Dentistry, Showa University, Tokyo, 142-8555, Japan
| | - Tomio Inoue
- Department of Oral Physiology, School of Dentistry, Showa University, Tokyo, 142-8555, Japan
| | - Yuji Kiuchi
- Department of Pharmacology, Division of Medical Pharmacology, School of Medicine, Showa University, Tokyo, 142-8555, Japan; Pharmacological Research Center, Showa University, Tokyo, 142-8555, Japan
| | - Masahito Yamada
- Department of Neurology and Neurobiology of Aging, Kanazawa University Graduate School of Medical Sciences, Kanazawa University, Kanazawa, 920-8640, Japan
| | - David B Teplow
- Department of Neurology, David Geffen School of Medicine at UCLA, 635 Charles E. Young Drive South, Room 445, Los Angeles, CA, 90095, USA
| | - Kenjiro Ono
- Department of Internal Medicine, Division of Neurology, School of Medicine, Showa University, Tokyo, 142-8666, Japan.
| |
Collapse
|
112
|
Crawford RA, Bowman KR, Cagle BS, Doorn JA. In vitro inhibition of glutathione-S-transferase by dopamine and its metabolites, 3,4-dihydroxyphenylacetaldehyde and 3,4-dihydroxyphenylacetic acid. Neurotoxicology 2021; 86:85-93. [PMID: 34314733 DOI: 10.1016/j.neuro.2021.07.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 07/22/2021] [Accepted: 07/23/2021] [Indexed: 02/06/2023]
Abstract
Parkinson's disease is characterized by dopamine dyshomeostasis and oxidative stress. The aldehyde metabolite of dopamine, 3,4-dihydroxyphenylacetaldehyde (DOPAL), has been reported to be cytotoxic and capable of protein modification. Protein modification by DOPAL has been implicated in the pathogenesis of Parkinson's disease, but the complete pathology is unknown. Our findings show that DOPAL modifies glutathione S-transferase (GST), an important enzyme in the antioxidant defense system. DOPAL, dopamine, and the metabolite 3,4-dihydroxyphenylacetic acid (DOPAC), inhibited the activity of GST isolated from N27 dopaminergic cells at an IC50 of 31.46 μM, 82.32 μM, and 260.0 μM, respectively. DOPAL, dopamine, and DOPAC inhibited commercially available equine liver GST at an IC50 of 23.72 μM, 32.17 μM, and 73.70 μM, respectively. This inhibition was time dependent and irreversible. 1 mM ʟ-cysteine or glutathione fully protected GST activity from DOPAL, DA, and DOPAC inhibition. 1 mM carnosine partially protected GST activity from DA inhibition. Furthermore, ʟ-cysteine was found to protect GST by forming a putative thiazolidine conjugate with DOPAL. We conclude that GST inactivation may be a part of the broader etiopathology of Parkinson's disease.
Collapse
Affiliation(s)
- Rachel A Crawford
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA, United States
| | - Kate R Bowman
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA, United States
| | - Brianna S Cagle
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA, United States
| | - Jonathan A Doorn
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA, United States.
| |
Collapse
|
113
|
Ibrahim KA, Abdelgaid HA, Eleyan M, Khwanes SA, Abdel-Daim MM. Ethoprophos induces rats' brain injury and neurobehavioral impairment via transcriptional activation of glial fibrillary acidic protein and tubulin-associated unit even at the threshold inhibition of acetylcholinesterase: A 90-days study. THE SCIENCE OF THE TOTAL ENVIRONMENT 2021. [DOI: 10.1016/j.scitotenv.2021.146216] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
114
|
Unsal V, Cicek M, Sabancilar İ. Toxicity of carbon tetrachloride, free radicals and role of antioxidants. REVIEWS ON ENVIRONMENTAL HEALTH 2021; 36:279-295. [PMID: 32970608 DOI: 10.1515/reveh-2020-0048] [Citation(s) in RCA: 86] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 09/02/2020] [Indexed: 06/11/2023]
Abstract
Several chemicals, including environmental toxicants and clinically useful drugs, cause severe cellular damage to different organs of our body through metabolic activation to highly reactive substances such as free radicals. Carbon tetrachloride is an organic compound of which chemical formula is CCl₄. CCl4 is strong toxic in the kidney, testicle, brain, heart, lung, other tissues, and particularly in the liver. CCl4 is a powerful hepatoxic, nephrotoxic and prooxidant agent which is widely used to induce hepatotoxicity in experimental animals and to create hepatocellular carcinoma, hepatic fibrosis/cirrhosis and liver injury, chemical hepatitis model, renal failure model, and nephrotoxicity model in recent years. The damage-causing mechanism of CCl4 in tissues can be explained as oxidative damage caused by lipid peroxidation which starts after the conversion of CCl4 to free radicals of highly toxic trichloromethyl radicals (•CCl₃) and trichloromethyl peroxyl radical (•CCl₃O2) via cytochrome P450 enzyme. Complete disruption of lipids (i.e., peroxidation) is the hallmark of oxidative damage. Free radicals are structures that contain one or more unpaired electrons in atomic or molecular orbitals. These toxic free radicals induce a chain reaction and lipid peroxidation in membrane-like structures rich in phospholipids, such as mitochondria and endoplasmic reticulum. CCl4-induced lipid peroxidation is the cause of oxidative stress, mitochondrial stress, endoplasmic reticulum stress. Free radicals trigger many biological processes, such as apoptosis, necrosis, ferroptosis and autophagy. Recent researches state that the way to reduce or eliminate these CCl4-induced negative effects is the antioxidants originated from natural sources. For normal physiological function, there must be a balance between free radicals and antioxidants. If this balance is in favor of free radicals, various pathological conditions occur. Free radicals play a role in various pathological conditions including Pulmonary disease, ischemia / reperfusion rheumatological diseases, autoimmune disorders, cardiovascular diseases, cancer, kidney diseases, hypertension, eye diseases, neurological disorders, diabetes and aging. Free radicals are antagonized by antioxidants and quenched. Antioxidants do not only remove free radicals, but they also have anti-inflammatory, anti-allergic, antithrombotic, antiviral, and anti-carcinogenic activities. Antioxidants contain high phenol compounds and antioxidants have relatively low side effects compared to synthetic drugs. The antioxidants investigated in CCI4 toxicity are usually antioxidants from plants and are promising because of their rich resources and low side effects. Data were investigated using PubMed, EBSCO, Embase, Web of Science, DOAJ, Scopus and Google Scholar, Carbon tetrachloride, carbon tetrachloride-induced toxicity, oxidative stress, and free radical keywords. This study aims to enlighten the damage-causing mechanism created by free radicals which are produced by CCl4 on tissues/cells and to discuss the role of antioxidants in the prevention of tissue/cell damage. In the future, Antioxidants can be used as a therapeutic strategy to strengthen effective treatment against substances with high toxicity such as CCl4 and increase the antioxidant capacity of cells.
Collapse
Affiliation(s)
- Velid Unsal
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Mardin Artuklu University, Mardin, Turkey
| | - Mustafa Cicek
- Department of Anatomy, Faculty of Medicine, Kahramanmaraş Sütçü imam University, Kahramanmaraş, Turkey
| | - İlhan Sabancilar
- Department of Biochemistry, Health Sciences Institute, Dicle University, Diyarbakır, Turkey
| |
Collapse
|
115
|
Zhang C, Lv Y, Bai R, Xie Y. Structural exploration of multifunctional monoamine oxidase B inhibitors as potential drug candidates against Alzheimer's disease. Bioorg Chem 2021; 114:105070. [PMID: 34126574 DOI: 10.1016/j.bioorg.2021.105070] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 05/25/2021] [Accepted: 06/05/2021] [Indexed: 10/21/2022]
Abstract
AD is one of the most typical neurodegenerative disorders that suffer many seniors worldwide. Recently, MAO inhibitors have received increasing attention not only for their roles involved in monoamine neurotransmitters metabolism and oxidative stress but also for their additional neuroprotective and neurorescue effects against AD. The curiosity in MAO inhibitors is reviving, and novel MAO-B inhibitors recently developed with ancillary activities (e.g., Aβ aggregation and AChE inhibition, anti-ROS and chelating activities) have been proposed as multitarget drugs foreshadowing a positive outlook for the treatment of AD. The current review describes the recent development of the design, synthesis, and screening of multifunctional ligands based on MAO-B inhibition for AD therapy. Structure-activity relationships and rational design strategies of the synthetic or natural product derivatives (chalcones, coumarins, chromones, and homoisoflavonoids) are discussed.
Collapse
Affiliation(s)
- Changjun Zhang
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceutical, Zhejiang University of Technology, Hangzhou, PR China
| | - Yangjing Lv
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, PR China
| | - Renren Bai
- College of Pharmacy, School of Medicine, Hangzhou Normal University, Hangzhou, PR China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, PR China.
| | - Yuanyuan Xie
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceutical, Zhejiang University of Technology, Hangzhou, PR China; College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, PR China.
| |
Collapse
|
116
|
Perluigi M, Di Domenico F, Barone E, Butterfield DA. mTOR in Alzheimer disease and its earlier stages: Links to oxidative damage in the progression of this dementing disorder. Free Radic Biol Med 2021; 169:382-396. [PMID: 33933601 PMCID: PMC8145782 DOI: 10.1016/j.freeradbiomed.2021.04.025] [Citation(s) in RCA: 60] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 04/15/2021] [Indexed: 12/11/2022]
Abstract
Alzheimer's disease (AD) is the most prevalent form of dementia in the elderly population and has worldwide impact. The etiology of the disease is complex and results from the confluence of multiple mechanisms ultimately leading to neuronal loss and cognitive decline. Among risk factors, aging is the most relevant and accounts for several pathogenic events that contribute to disease-specific toxic mechanisms. Accumulating evidence linked the alterations of the mammalian target of rapamycin (mTOR), a serine/threonine protein kinase playing a key role in the regulation of protein synthesis and degradation, to age-dependent cognitive decline and pathogenesis of AD. To date, growing studies demonstrated that aberrant mTOR signaling in the brain affects several pathways involved in energy metabolism, cell growth, mitochondrial function and proteostasis. Recent advances associated alterations of the mTOR pathway with the increased oxidative stress. Disruption of all these events strongly contribute to age-related cognitive decline including AD. The current review discusses the main regulatory roles of mTOR signaling network in the brain, focusing on its role in autophagy, oxidative stress and energy metabolism. Collectively, experimental data suggest that targeting mTOR in the CNS can be a valuable strategy to prevent/slow the progression of AD.
Collapse
Affiliation(s)
- M Perluigi
- Department of Biochemical Sciences "A. Rossi-Fanelli", Sapienza University of Rome, Piazzale A. Moro 5, 00185, Roma, Italy
| | - F Di Domenico
- Department of Biochemical Sciences "A. Rossi-Fanelli", Sapienza University of Rome, Piazzale A. Moro 5, 00185, Roma, Italy
| | - E Barone
- Department of Biochemical Sciences "A. Rossi-Fanelli", Sapienza University of Rome, Piazzale A. Moro 5, 00185, Roma, Italy
| | - D A Butterfield
- Department of Chemistry, Sapienza University of Rome, Piazzale A. Moro 5, 00185, Roma, Italy; Department of Chemistry and Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, 40506-0055, USA.
| |
Collapse
|
117
|
Responsive optical probes for deep-tissue imaging: Photoacoustics and second near-infrared fluorescence. Adv Drug Deliv Rev 2021; 173:141-163. [PMID: 33774116 DOI: 10.1016/j.addr.2021.03.008] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Revised: 01/23/2021] [Accepted: 03/11/2021] [Indexed: 02/08/2023]
Abstract
Optical imaging has played a vital role in development of biomedicine and image-guided theragnostic. Nevertheless, the clinical translation of optical molecular imaging for deep-tissue visualization is still limited by poor signal-to-background ratio and low penetration depth owing to light scattering and tissue autofluorescence. Hence, to facilitate precise diagnosis and accurate surgery excision in clinical practices, the responsive optical probes (ROPs) are broadly designed for specific reaction with biological analytes or disease biomarkers via chemical/physical interactions for photoacoustic and second near-infrared fluorescence (NIR-II, 900-1700 nm) fluorescence imaging. Herein, the recent advances in the development of ROPs including molecular design principles, activated mechanisms and treatment responses for photoacoustic and NIR-II fluorescence imaging are reviewed. Furthermore, the present challenges and future perspectives of ROPs for deep-tissue imaging are also discussed.
Collapse
|
118
|
Ma L, Gholam Azad M, Dharmasivam M, Richardson V, Quinn RJ, Feng Y, Pountney DL, Tonissen KF, Mellick GD, Yanatori I, Richardson DR. Parkinson's disease: Alterations in iron and redox biology as a key to unlock therapeutic strategies. Redox Biol 2021; 41:101896. [PMID: 33799121 PMCID: PMC8044696 DOI: 10.1016/j.redox.2021.101896] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Revised: 02/05/2021] [Accepted: 02/09/2021] [Indexed: 12/13/2022] Open
Abstract
A plethora of studies indicate that iron metabolism is dysregulated in Parkinson's disease (PD). The literature reveals well-documented alterations consistent with established dogma, but also intriguing paradoxical observations requiring mechanistic dissection. An important fact is the iron loading in dopaminergic neurons of the substantia nigra pars compacta (SNpc), which are the cells primarily affected in PD. Assessment of these changes reveal increased expression of proteins critical for iron uptake, namely transferrin receptor 1 and the divalent metal transporter 1 (DMT1), and decreased expression of the iron exporter, ferroportin-1 (FPN1). Consistent with this is the activation of iron regulator protein (IRP) RNA-binding activity, which is an important regulator of iron homeostasis, with its activation indicating cytosolic iron deficiency. In fact, IRPs bind to iron-responsive elements (IREs) in the 3ꞌ untranslated region (UTR) of certain mRNAs to stabilize their half-life, while binding to the 5ꞌ UTR prevents translation. Iron loading of dopaminergic neurons in PD may occur through these mechanisms, leading to increased neuronal iron and iron-mediated reactive oxygen species (ROS) generation. The "gold standard" histological marker of PD, Lewy bodies, are mainly composed of α-synuclein, the expression of which is markedly increased in PD. Of note, an atypical IRE exists in the α-synuclein 5ꞌ UTR that may explain its up-regulation by increased iron. This dysregulation could be impacted by the unique autonomous pacemaking of dopaminergic neurons of the SNpc that engages L-type Ca+2 channels, which imparts a bioenergetic energy deficit and mitochondrial redox stress. This dysfunction could then drive alterations in iron trafficking that attempt to rescue energy deficits such as the increased iron uptake to provide iron for key electron transport proteins. Considering the increased iron-loading in PD brains, therapies utilizing limited iron chelation have shown success. Greater therapeutic advancements should be possible once the exact molecular pathways of iron processing are dissected.
Collapse
Affiliation(s)
- L Ma
- School of Environment and Science, Griffith University Nathan, Brisbane, Queensland, Australia; Griffith Institute for Drug Discovery, Griffith University, Nathan, Brisbane, Queensland, Australia
| | - M Gholam Azad
- School of Environment and Science, Griffith University Nathan, Brisbane, Queensland, Australia; Griffith Institute for Drug Discovery, Griffith University, Nathan, Brisbane, Queensland, Australia; Centre for Cancer Cell Biology and Drug Discovery, Griffith Institute for Drug Discovery, Griffith University, Nathan, Brisbane, Queensland, Australia
| | - M Dharmasivam
- School of Environment and Science, Griffith University Nathan, Brisbane, Queensland, Australia; Griffith Institute for Drug Discovery, Griffith University, Nathan, Brisbane, Queensland, Australia; Centre for Cancer Cell Biology and Drug Discovery, Griffith Institute for Drug Discovery, Griffith University, Nathan, Brisbane, Queensland, Australia
| | - V Richardson
- School of Environment and Science, Griffith University Nathan, Brisbane, Queensland, Australia; Griffith Institute for Drug Discovery, Griffith University, Nathan, Brisbane, Queensland, Australia; Centre for Cancer Cell Biology and Drug Discovery, Griffith Institute for Drug Discovery, Griffith University, Nathan, Brisbane, Queensland, Australia
| | - R J Quinn
- Griffith Institute for Drug Discovery, Griffith University, Nathan, Brisbane, Queensland, Australia
| | - Y Feng
- School of Environment and Science, Griffith University Nathan, Brisbane, Queensland, Australia; Griffith Institute for Drug Discovery, Griffith University, Nathan, Brisbane, Queensland, Australia
| | - D L Pountney
- School of Medical Science, Griffith University, Gold Coast, Queensland, Australia
| | - K F Tonissen
- School of Environment and Science, Griffith University Nathan, Brisbane, Queensland, Australia; Griffith Institute for Drug Discovery, Griffith University, Nathan, Brisbane, Queensland, Australia
| | - G D Mellick
- School of Environment and Science, Griffith University Nathan, Brisbane, Queensland, Australia; Griffith Institute for Drug Discovery, Griffith University, Nathan, Brisbane, Queensland, Australia
| | - I Yanatori
- Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan
| | - D R Richardson
- School of Environment and Science, Griffith University Nathan, Brisbane, Queensland, Australia; Griffith Institute for Drug Discovery, Griffith University, Nathan, Brisbane, Queensland, Australia; Centre for Cancer Cell Biology and Drug Discovery, Griffith Institute for Drug Discovery, Griffith University, Nathan, Brisbane, Queensland, Australia; Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan.
| |
Collapse
|
119
|
Wang J, Wang S, Guo H, Li Y, Jiang Z, Gu T, Su B, Hou W, Zhong H, Cheng D, Zhang X, Fang Z. Rosmarinic acid protects rats against post-stroke depression after transient focal cerebral ischemic injury through enhancing antioxidant response. Brain Res 2021; 1757:147336. [PMID: 33548269 DOI: 10.1016/j.brainres.2021.147336] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 01/22/2021] [Accepted: 01/26/2021] [Indexed: 12/26/2022]
Abstract
Rosmarinic acid (RA), a natural polyphenol, possesses potent antioxidant and anti-inflammatory activities. To evaluate the ability of RA to cure ischemic stroke and post-stroke depression (PSD), rats were treated with various doses of RA after cerebral ischemia. Neurological deficits and infarct volume of the brain were measured. The activities of superoxide dismutase (SOD), catalase (CAT), and glutathione (GSH) were examined at different time points. In addition, a forced swimming test and sucrose preference test were performed to detect the anti-depressive effects of RA. Our results revealed RA administration significantly alleviated neurological deficits and reduced infarct volumes. RA attenuated the decrease of SOD, CAT activities and GSH levels in the ischemic penumbra of the brain. Most importantly, RA treatment alleviated the depression behaviors. Increased expression of Nrf2 was also induced by RA, while down regulation Nrf2 by Nrf2-short-hairpin RNA sequences reversed the increasing activity of SOD and CAT induced by RA, as well as the protection against PSD. The present study indicates that RA exerts a potent neuroprotective effect against stroke and PSD, which could be a promising therapeutic intervention for stroke.
Collapse
Affiliation(s)
- Jiajia Wang
- Department of Anesthesiology, Xijing Hospital, the Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Shiquan Wang
- Department of Anesthesiology, Xijing Hospital, the Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Haiyun Guo
- Department of Anesthesiology, Xijing Hospital, the Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Yi Li
- Department of Anesthesiology, Xijing Hospital, the Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Zhenhua Jiang
- Department of Anesthesiology, Xijing Hospital, the Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Ting Gu
- Department of Anesthesiology, Xijing Hospital, the Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Binxiao Su
- Department of Anesthesiology, Xijing Hospital, the Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Wugang Hou
- Department of Anesthesiology, Xijing Hospital, the Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Haixing Zhong
- Department of Anesthesiology, Xijing Hospital, the Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Dandan Cheng
- Department of Anesthesiology, Xijing Hospital, the Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Xijing Zhang
- Department of Anesthesiology, Xijing Hospital, the Fourth Military Medical University, Xi'an, Shaanxi 710032, China.
| | - Zongping Fang
- Department of Anesthesiology, Xijing Hospital, the Fourth Military Medical University, Xi'an, Shaanxi 710032, China.
| |
Collapse
|
120
|
Nagu P, Parashar A, Behl T, Mehta V. Gut Microbiota Composition and Epigenetic Molecular Changes Connected to the Pathogenesis of Alzheimer's Disease. J Mol Neurosci 2021; 71:1436-1455. [PMID: 33829390 DOI: 10.1007/s12031-021-01829-3] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 03/11/2021] [Indexed: 12/12/2022]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder, and its pathogenesis is not fully known. Although there are several hypotheses, such as neuroinflammation, tau hyperphosphorylation, amyloid-β plaques, neurofibrillary tangles, and oxidative stress, none of them completely explain the origin and progression of AD. Emerging evidence suggests that gut microbiota and epigenetics can directly influence the pathogenesis of AD via their effects on multiple pathways, including neuroinflammation, oxidative stress, and amyloid protein. Various gut microbes such as Actinobacteria, Bacteroidetes, E. coli, Firmicutes, Proteobacteria, Tenericutes, and Verrucomicrobia are known to play a crucial role in the pathogenesis of AD. These microbes and their metabolites modulate various physiological processes that contribute to AD pathogenesis, such as neuroinflammation and other inflammatory processes, amyloid deposition, cytokine storm syndrome, altered BDNF and NMDA signaling, impairing neurodevelopmental processes. Likewise, epigenetic markers associated with AD mainly include histone modifications and DNA methylation, which are under the direct control of a variety of enzymes, such as acetylases and methylases. The activity of these enzymes is dependent upon the metabolites generated by the host's gut microbiome, suggesting the significance of epigenetics in AD pathogenesis. It is interesting to know that both gut microbiota and epigenetics are dynamic processes and show a high degree of variation according to diet, stressors, and environmental factors. The bidirectional relation between the gut microbiota and epigenetics suggests that they might work in synchrony to modulate AD representation, its pathogenesis, and progression. They both also provide numerous targets for early diagnostic biomarkers and for the development of AD therapeutics. This review discusses the gut microbiota and epigenetics connection in the pathogenesis of AD and aims to highlight vast opportunities for diagnosis and therapeutics of AD.
Collapse
Affiliation(s)
- Priyanka Nagu
- Department of Pharmaceutics, Govt. College of Pharmacy, Rohru, Himachal Pradesh, India.,Department of Pharmacy, Shri Jagdishprasad Jhabarmal Tibrewala University, Jhunjhunu, Rajasthan, India
| | - Arun Parashar
- Faculty of Pharmaceutical Sciences, Shoolini University of Biotechnology and Management Sciences, Solan, Himachal Pradesh, India
| | - Tapan Behl
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Vineet Mehta
- Department of Pharmacology, Govt. College of Pharmacy, Rohru, Himachal Pradesh, India.
| |
Collapse
|
121
|
Hofmann J, Ginex T, Espargaró A, Scheiner M, Gunesch S, Aragó M, Stigloher C, Sabaté R, Luque FJ, Decker M. Azobioisosteres of Curcumin with Pronounced Activity against Amyloid Aggregation, Intracellular Oxidative Stress, and Neuroinflammation. Chemistry 2021; 27:6015-6027. [PMID: 33666306 PMCID: PMC8048673 DOI: 10.1002/chem.202005263] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 01/14/2021] [Indexed: 01/01/2023]
Abstract
Many (poly-)phenolic natural products, for example, curcumin and taxifolin, have been studied for their activity against specific hallmarks of neurodegeneration, such as amyloid-β 42 (Aβ42) aggregation and neuroinflammation. Due to their drawbacks, arising from poor pharmacokinetics, rapid metabolism, and even instability in aqueous medium, the biological activity of azobenzene compounds carrying a pharmacophoric catechol group, which have been designed as bioisoteres of curcumin has been examined. Molecular simulations reveal the ability of these compounds to form a hydrophobic cluster with Aβ42, which adopts different folds, affecting the propensity to populate fibril-like conformations. Furthermore, the curcumin bioisosteres exceeded the parent compound in activity against Aβ42 aggregation inhibition, glutamate-induced intracellular oxidative stress in HT22 cells, and neuroinflammation in microglial BV-2 cells. The most active compound prevented apoptosis of HT22 cells at a concentration of 2.5 μm (83 % cell survival), whereas curcumin only showed very low protection at 10 μm (21 % cell survival).
Collapse
Affiliation(s)
- Julian Hofmann
- Pharmaceutical and Medicinal ChemistryInstitute of, Pharmacy and Food ChemistryUniversity of WürzburgAm Hubland97074WürzburgGermany
| | - Tiziana Ginex
- Department of Nutrition Food Science and GastronomyFaculty of Pharmacy, Institute of Theoretical and Computational, Chemistry and Institute of Biomedicine, Campus TorriberaUniversity of BarcelonaSanta Coloma de Gramenet08921Spain
| | - Alba Espargaró
- Pharmacy and Pharmaceutical Technology and Physical-ChemistrySchool of Pharmacy Institute of Nanoscience and Nanotechnology, (IN2UB)University of Barcelona08028BarcelonaSpain
| | - Matthias Scheiner
- Pharmaceutical and Medicinal ChemistryInstitute of, Pharmacy and Food ChemistryUniversity of WürzburgAm Hubland97074WürzburgGermany
| | - Sandra Gunesch
- Pharmaceutical and Medicinal ChemistryInstitute of, Pharmacy and Food ChemistryUniversity of WürzburgAm Hubland97074WürzburgGermany
| | - Marc Aragó
- Department of Nutrition Food Science and GastronomyFaculty of Pharmacy, Institute of Theoretical and Computational, Chemistry and Institute of Biomedicine, Campus TorriberaUniversity of BarcelonaSanta Coloma de Gramenet08921Spain
| | - Christian Stigloher
- Imaging Core FacilityBiocenter/Theodor-Boveri-InstituteUniversity of WürzburgAm Hubland97074WürzburgGermany
| | - Raimon Sabaté
- Pharmacy and Pharmaceutical Technology and Physical-ChemistrySchool of Pharmacy Institute of Nanoscience and Nanotechnology, (IN2UB)University of Barcelona08028BarcelonaSpain
| | - F. Javier Luque
- Department of Nutrition Food Science and GastronomyFaculty of Pharmacy, Institute of Theoretical and Computational, Chemistry and Institute of Biomedicine, Campus TorriberaUniversity of BarcelonaSanta Coloma de Gramenet08921Spain
| | - Michael Decker
- Pharmaceutical and Medicinal ChemistryInstitute of, Pharmacy and Food ChemistryUniversity of WürzburgAm Hubland97074WürzburgGermany
| |
Collapse
|
122
|
Shen YC, Chang CE, Lin MN, Lin CL. Vegetarian Diet Is Associated with Lower Risk of Depression in Taiwan. Nutrients 2021; 13:nu13041059. [PMID: 33805124 PMCID: PMC8064096 DOI: 10.3390/nu13041059] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 03/15/2021] [Accepted: 03/21/2021] [Indexed: 12/22/2022] Open
Abstract
In order to determine whether Taiwanese vegetarian diets reduce the risks of depression, we analyzed data from the Tzu Chi Vegetarian Study (TCVS), which is a prospective cohort study following 12,062 participants from the Buddhist Tzu Chi Foundation of Taiwan since 2005. The cohort was prospectively followed by linking to the National Health Institute Research Database (NHIRD) of Taiwan and hazard ratios of depression between vegetarian and non-vegetarian groups were calculated by Cox proportional hazards regression. We assessed dietary intake using a detailed food frequency questionnaire (FFQ). Incident depression was ascertained through linkage to NHIRD which had claim records with the International Classification of Diseases, and a total of 3571 vegetarians and 7006 non-vegetarians were included in this analysis. Compared with non-vegetarians, the vegetarian group had a lower incidence of depressive disorders (2.37 vs. 3.21 per 10,000 person-years; adjusted hazard ratio (aHR): 0.70; 95% confidence interval (95% CI): 0.52–0.93). Thus, Taiwanese vegetarians had a lower risk of developing subsequent depressive disorders compared with non-vegetarians. This indicated that diet may be an important measure for the prevention of depression. However, to generalize to the global population requires further study.
Collapse
Affiliation(s)
- Yu-Chih Shen
- Department of Psychiatric, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 970, Taiwan;
| | - Chiao-Erh Chang
- Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University, Taipei 100, Taiwan;
| | - Ming-Nan Lin
- Department of Family Medicine, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Chiayi County 622, Taiwan;
- Department of Family Medicine, College of Medicine, Tzu Chi University, Hualien 970, Taiwan
| | - Chin-Lon Lin
- Department of Cardiology, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Chiayi County 622, Taiwan
- Department of Internal Medicine, College of Medicine, Tzu Chi University, Hualien 970, Taiwan
- Correspondence: ; Tel.: +886-3-856-1825 (ext. 5305)
| |
Collapse
|
123
|
Behavior and oxidative stress parameters in rats subjected to the animal's models induced by chronic mild stress and 6-hydroxydopamine. Behav Brain Res 2021; 406:113226. [PMID: 33684423 DOI: 10.1016/j.bbr.2021.113226] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 03/01/2021] [Accepted: 03/02/2021] [Indexed: 12/16/2022]
Abstract
Major depressive disorder (MDD) is one of the most prevalent forms of mental illness also affecting older adults. Recent evidence suggests a relationship between MDD and neurodegenerative diseases, including Parkinson's disease (PD). Individuals with PD have a predisposition to developing MDD, and both neurobiological conditions are associated with oxidative stress. Thus, we conducted this study to investigate depressive-like behavior and oxidative stress parameters using both animal models of PD and stress. Adult Wistar rats were subjected to chronic mild stress (CMS) protocol by 40 days and then it was used 6-hydroxydopamine (6-OHDA) as a model of PD, into the striatum. The experimental groups were: Control + Sham, Stress + Sham, Control+6-OHDA, and Stress+6-OHDA. Depressive like-behavior was evaluated by the forced swimming test (FST) and spontaneous locomotor activity by open-field test. Oxidative stress parameters were measured in the striatum, hippocampus, and prefrontal cortex (PFC). The results showed effects to increase immobility and decrease climbing times in the FST in Stress + Sham, Control+6-OHDA, and Stress+6-OHDA groups. The number of crossings and rearings were decreased in the Stress+6-OHDA group. The lipid peroxidation was increased in the PFC of Stress + Sham, and the hippocampus and striatum of Stress + Sham and Control+6-OHDA groups. Carbonyl protein levels increased in the PFC of Stress + Sham and striatum in Control+6-OHDA. Nitrite/Nitrate concentration was elevated in the PFC of Stress + Sham, in the hippocampus of Control+6-OHDA, the striatum of Stress + Sham, and Control+6-OHDA groups. Myeloperoxidase (MPO) activity was increased in the PFC and hippocampus of Stress + Sham and Control+6-OHDA groups. The activity of catalase decreased in the PFC of the Stress + Sham group. The activity of the superoxide dismutase (SOD) was decreased in the PFC of the Stress + Sham group, in the hippocampus of Stress + Sham and Control+6-OHDA groups, and the striatum of Control+6-OHDA group. These findings suggest that both stress and 6-OHDA induce depressive-like behavior and oxidative stress in the brain. The joining models have little evidence of the effects. Thus these findings suggest that other pathways are involved in the common point of the pathophysiology of PD and MDD.
Collapse
|
124
|
Malankar GS, Sakunthala A, Navalkar A, Maji SK, Raju S, Manjare ST. Organoselenium-based BOPHY as a sensor for detection of hypochlorous acid in mammalian cells. Anal Chim Acta 2021; 1150:338205. [DOI: 10.1016/j.aca.2021.338205] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 01/03/2021] [Accepted: 01/04/2021] [Indexed: 02/08/2023]
|
125
|
Current Updates On the In vivo Assessment of Zinc Oxide Nanoparticles Toxicity Using Animal Models. BIONANOSCIENCE 2021. [DOI: 10.1007/s12668-021-00845-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
126
|
Akawa OB, Subair TI, Soremekun OS, Olotu FA, Soliman MES. Structural alterations in the catalytic core of hSIRT2 enzyme predict therapeutic benefits of Garcinia mangostana derivatives in Alzheimer's disease: molecular dynamics simulation study. RSC Adv 2021; 11:8003-8018. [PMID: 35423339 PMCID: PMC8695224 DOI: 10.1039/d0ra10459k] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Accepted: 02/03/2021] [Indexed: 12/15/2022] Open
Abstract
Recent studies have shown that inhibition of the hSIRT2 enzyme provides favorable effects in neurodegenerative diseases such as Alzheimer's disease. Prenylated xanthone phytochemicals including α-mangostin, β-mangostin and γ-mangostin obtained from Garcinia mangostana, a well-established tropical plant, have been shown experimentally to inhibit sirtuin enzymatic activity. However, the molecular mechanism of this sirtuin inhibition has not been reported. Using comprehensive integrated computational techniques, we provide molecular and timewise dynamical insights into the structural alterations capable of facilitating therapeutically beneficial effects of these phytochemicals at the catalytic core of the hSIRT2 enzyme. Findings revealed the enhanced conformational stability and compactness of the hSIRT2 catalytic core upon binding of γ-mangostin relative to the apoenzyme and better than α-mangostin and β-mangostin. Although thermodynamic calculations revealed favorable binding of all the phytochemicals to the hSIRT2 enzyme, the presence of only hydroxy functional groups on γ-mangostin facilitated the occurrence of additional hydrogen bonds involving Pro115, Phe119, Asn168 and His187 which are absent in α-mangostin- and β-mangostin-bound systems. Per-residue energy contributions showed that van der Waals and more importantly electrostatic interactions are involved in catalytic core stability with Phe96, Tyr104 and Phe235 notably contributing π-π stacking, π-π T shaped and π-sigma interactions. Cumulatively, our study revealed the structural alterations leading to inhibition of hSIRT2 catalysis and findings from this study could be significantly important for the future design and development of sirtuin inhibitors in the management of Alzheimer's disease.
Collapse
Affiliation(s)
- Oluwole B Akawa
- Molecular Bio-computation and Drug Design Laboratory, School of Health Sciences, University of KwaZulu-Natal Westville Campus Durban 4001 South Africa http://soliman.ukzn.ac.za +27 31 260 7872 +27 31 260 8048
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, Afe Babalola University Ado Ekiti Nigeria
| | - Temitayo I Subair
- Molecular Bio-computation and Drug Design Laboratory, School of Health Sciences, University of KwaZulu-Natal Westville Campus Durban 4001 South Africa http://soliman.ukzn.ac.za +27 31 260 7872 +27 31 260 8048
| | - Opeyemi S Soremekun
- Molecular Bio-computation and Drug Design Laboratory, School of Health Sciences, University of KwaZulu-Natal Westville Campus Durban 4001 South Africa http://soliman.ukzn.ac.za +27 31 260 7872 +27 31 260 8048
| | - Fisayo A Olotu
- Molecular Bio-computation and Drug Design Laboratory, School of Health Sciences, University of KwaZulu-Natal Westville Campus Durban 4001 South Africa http://soliman.ukzn.ac.za +27 31 260 7872 +27 31 260 8048
| | - Mahmoud E S Soliman
- Molecular Bio-computation and Drug Design Laboratory, School of Health Sciences, University of KwaZulu-Natal Westville Campus Durban 4001 South Africa http://soliman.ukzn.ac.za +27 31 260 7872 +27 31 260 8048
| |
Collapse
|
127
|
Diez V, Traikov S, Schmeisser K, Adhikari AKD, Kurzchalia TV. Glycolate combats massive oxidative stress by restoring redox potential in Caenorhabditis elegans. Commun Biol 2021; 4:151. [PMID: 33526793 PMCID: PMC7851149 DOI: 10.1038/s42003-021-01669-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 01/04/2021] [Indexed: 01/30/2023] Open
Abstract
Upon exposure to excessive reactive oxygen species (ROS), organismal survival depends on the strength of the endogenous antioxidant defense barriers that prevent mitochondrial and cellular deterioration. Previously, we showed that glycolic acid can restore the mitochondrial membrane potential of C. elegans treated with paraquat, an oxidant that produces superoxide and other ROS species, including hydrogen peroxide. Here, we demonstrate that glycolate fully suppresses the deleterious effects of peroxide on mitochondrial activity and growth in worms. This endogenous compound acts by entering serine/glycine metabolism. In this way, conversion of glycolate into glycine and serine ameliorates the drastically decreased NADPH/NADP+ and GSH/GSSG ratios induced by H2O2 treatment. Our results reveal the central role of serine/glycine metabolism as a major provider of reducing equivalents to maintain cellular antioxidant systems and the fundamental function of glycolate as a natural antioxidant that improves cell fitness and survival.
Collapse
Affiliation(s)
- Veronica Diez
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Sofia Traikov
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Kathrin Schmeisser
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | | | | |
Collapse
|
128
|
Pajarillo E, Nyarko-Danquah I, Adinew G, Rizor A, Aschner M, Lee E. Neurotoxicity mechanisms of manganese in the central nervous system. ADVANCES IN NEUROTOXICOLOGY 2021; 5:215-238. [PMID: 34263091 DOI: 10.1016/bs.ant.2020.11.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Edward Pajarillo
- Department of Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL, United States
| | - Ivan Nyarko-Danquah
- Department of Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL, United States
| | - Getinet Adinew
- Department of Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL, United States
| | - Asha Rizor
- Department of Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL, United States
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Eunsook Lee
- Department of Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL, United States
| |
Collapse
|
129
|
Kumar A, Datta LP, Samanta S, Arora H, Govindaraju T. Benzothiazole‐Phenothiazine Conjugate Based Molecular Probe for the Differential Detection of Glycated Albumin. Isr J Chem 2021. [DOI: 10.1002/ijch.202000098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Ashish Kumar
- Bioorganic Chemistry Laboratory New Chemistry Unit and The School of Advanced Materials (SAMat) Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR), Jakkur P. O. Bengaluru 560064 Karnataka India
| | - Lakshmi Priya Datta
- Bioorganic Chemistry Laboratory New Chemistry Unit and The School of Advanced Materials (SAMat) Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR), Jakkur P. O. Bengaluru 560064 Karnataka India
| | - Sourav Samanta
- Bioorganic Chemistry Laboratory New Chemistry Unit and The School of Advanced Materials (SAMat) Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR), Jakkur P. O. Bengaluru 560064 Karnataka India
| | - Harshit Arora
- Bioorganic Chemistry Laboratory New Chemistry Unit and The School of Advanced Materials (SAMat) Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR), Jakkur P. O. Bengaluru 560064 Karnataka India
| | - Thimmaiah Govindaraju
- Bioorganic Chemistry Laboratory New Chemistry Unit and The School of Advanced Materials (SAMat) Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR), Jakkur P. O. Bengaluru 560064 Karnataka India
| |
Collapse
|
130
|
Protection of 6-OHDA neurotoxicity by PGF 2α through FP-ERK-Nrf2 signaling in SH-SY5Y cells. Toxicology 2021; 450:152686. [PMID: 33486071 DOI: 10.1016/j.tox.2021.152686] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 01/07/2021] [Accepted: 01/16/2021] [Indexed: 12/26/2022]
Abstract
6-Hydroxydopamine (6-OHDA) is a neurotoxin that destroy dopaminergic neurons and widely used to establish animal models of Parkinson's disease. Prostaglandins (PGs) are involved in various cellular processes, including the damage and repair of neuronal cells. However, the function of PGF2α in neuronal cells remains unclear. In this study, we investigated the effects of PGF2α against 6-OHDA-mediated toxicity in human neuroblastoma SH-SY5Y cells and elucidated its underlying molecular mechanism. When the cells were treated with 6-OHDA (50 μM) for 6 h, the expression levels of PGF2α synthetic enzymes; cyclooxygenase-2 and aldo-keto reductase 1C3 as PGF2α synthase were enhanced in an incubation-time-dependent manner. In addition, the production of PGF2α was increased in 6-OHDA-treated cells. Fluprostenol, a PGF2α receptor (FP) agonist (500 nM), suppressed 6-OHDA-induced cell death by decreasing the production of reactive oxygen species (ROS) and increasing the expression of the anti-oxidant genes. These fluprostenol-mediated effects were inhibited by co-treatment with AL8810, an FP receptor antagonist (1 μM) or transfection with FP siRNA (20 nM). Moreover, 6-OHDA-induced phosphorylation of extracellular signal-regulated kinase (ERK), a member of the mitogen-activated protein kinase family, was inhibited by co-incubation with AL8810. Furthermore, fluprostenol itself enhanced ERK phosphorylation and further elevated the 6-OHDA-induced phosphorylation of ERK. In addition, 6-OHDA induced nuclear translocation of nuclear factor erythroid 2-related factor 2 (Nrf2), activating anti-oxidant gene expression, was repressed by co-culturing with AL8810. These results indicate that PGF2α suppressed 6-OHDA-induced neuronal cell death by enhancing anti-oxidant gene expression via the FP receptor-ERK-Nrf2 signaling. Thus, FP receptor is a potential target for inhibition of ROS-mediated neuronal cell death.
Collapse
|
131
|
Fracassi A, Marcatti M, Zolochevska O, Tabor N, Woltjer R, Moreno S, Taglialatela G. Oxidative Damage and Antioxidant Response in Frontal Cortex of Demented and Nondemented Individuals with Alzheimer's Neuropathology. J Neurosci 2021; 41:538-554. [PMID: 33239403 PMCID: PMC7821866 DOI: 10.1523/jneurosci.0295-20.2020] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 10/05/2020] [Accepted: 10/07/2020] [Indexed: 11/21/2022] Open
Abstract
Alzheimer's disease (AD) is characterized by progressive neurodegeneration in the cerebral cortex, histopathologically hallmarked by amyloid β (Aβ) extracellular plaques and intracellular neurofibrillary tangles, constituted by hyperphosphorylated tau protein. Correlation between these pathologic features and dementia has been challenged by the emergence of "nondemented with Alzheimer's neuropathology" (NDAN) individuals, cognitively intact despite displaying pathologic features of AD. The existence of these subjects suggests that some unknown mechanisms are triggered to resist Aβ-mediated detrimental events. Aβ accumulation affects mitochondrial redox balance, increasing oxidative stress status, which in turn is proposed as a primary culprit in AD pathogenesis. To clarify the relationship linking Aβ, oxidative stress, and cognitive impairment, we performed a comparative study on AD, NDAN, and aged-matched human postmortem frontal cortices of either sex. We quantitatively analyzed immunofluorescence distribution of oxidative damage markers, and of SOD2 (superoxide dismutase 2), PGC1α [peroxisome proliferator-activated receptor (PPAR) γ-coactivator 1α], PPARα, and catalase as key factors in antioxidant response, as well as the expression of miRNA-485, as a PGC1α upstream regulator. Our results confirm dramatic redox imbalance, associated with impaired antioxidant defenses in AD brain. By contrast, NDAN individuals display low oxidative damage, which is associated with high levels of scavenging systems, possibly resulting from a lack of PGC1α miRNA-485-related inhibition. Comparative analyses in neurons and astrocytes further highlighted cell-specific mechanisms to counteract redox imbalance. Overall, our data emphasize the importance of transcriptional and post-transcriptional regulation of antioxidant response in AD. This suggests that an efficient PGC1α-dependent "safety mechanism" may prevent Aβ-mediated oxidative stress, supporting neuroprotective therapies aimed at ameliorating defects in antioxidant response pathways in AD patients.
Collapse
Affiliation(s)
- Anna Fracassi
- Mitchell Center for Neurodegenerative Diseases, Department of Neurology, University of Texas Medical Branch (UTMB), Galveston, Texas 77550
| | - Michela Marcatti
- Mitchell Center for Neurodegenerative Diseases, Department of Neurology, University of Texas Medical Branch (UTMB), Galveston, Texas 77550
| | - Olga Zolochevska
- Mitchell Center for Neurodegenerative Diseases, Department of Neurology, University of Texas Medical Branch (UTMB), Galveston, Texas 77550
| | - Natalie Tabor
- Neuroscience Summer Undergraduate Program, University of Texas Medical Branch, Galveston, Texas 77555
| | - Randall Woltjer
- Department of Pathology, Oregon Health and Science University, Portland, Oregon 97239-3098
| | - Sandra Moreno
- Department of Science, LIME, University Roma Tre, 00146 Rome, Italy
| | - Giulio Taglialatela
- Mitchell Center for Neurodegenerative Diseases, Department of Neurology, University of Texas Medical Branch (UTMB), Galveston, Texas 77550
| |
Collapse
|
132
|
Govindarajulu M, Ramesh S, Neel L, Fabbrini M, Buabeid M, Fujihashi A, Dwyer D, Lynd T, Shah K, Mohanakumar KP, Smith F, Moore T, Dhanasekaran M. Nutraceutical based SIRT3 activators as therapeutic targets in Alzheimer's disease. Neurochem Int 2021; 144:104958. [PMID: 33444675 DOI: 10.1016/j.neuint.2021.104958] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Revised: 12/21/2020] [Accepted: 01/06/2021] [Indexed: 02/07/2023]
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disease, and its incidence is increasing worldwide with increased lifespan. Currently, there is no effective treatment to cure or prevent the progression of AD, which indicates the need to develop novel therapeutic targets and agents. Sirtuins, especially SIRT3, a mitochondrial deacetylase, are NAD-dependent histone deacetylases involved in aging and longevity. Accumulating evidence indicates that SIRT3 dysfunction is strongly associated with pathologies of AD, hence, therapeutic modulation of SIRT3 activity may be a novel application to ameliorate the pathologies of AD. Natural products commonly used in traditional medicine have wide utility and appear to have therapeutic benefits for the treatment of neurodegenerative diseases such as AD. The present review summarizes the currently available natural SIRT3 activators and their potentially neuroprotective molecular mechanisms of action that make them a promising agent in the treatment and management of neurodegenerative diseases such as AD.
Collapse
Affiliation(s)
- Manoj Govindarajulu
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, AL 36849, USA
| | - Sindhu Ramesh
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, AL 36849, USA
| | - Logan Neel
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, AL 36849, USA
| | - Mary Fabbrini
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, AL 36849, USA
| | - Manal Buabeid
- Clinical Pharmacy Department, College of Pharmacy and Health Sciences, Ajman University, United Arab Emirates
| | - Ayaka Fujihashi
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, AL 36849, USA
| | - Darby Dwyer
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, AL 36849, USA
| | - Tyler Lynd
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, AL 36849, USA
| | - Karishma Shah
- Department of Ophthalmology, D.Y. Patil Medical College and Research Hospital, Mumbai, India
| | | | - Forrest Smith
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, AL 36849, USA
| | - Timothy Moore
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, AL 36849, USA
| | - Muralikrishnan Dhanasekaran
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, AL 36849, USA.
| |
Collapse
|
133
|
Zaiter J, Hibot A, Hafid A, Khouili M, Neves CMB, Simões MMQ, Neves MGPMS, Faustino MAF, Dagci T, Saso L, Armagan G. Evaluation of the cellular protection by novel spiropyrazole compounds in dopaminergic cell death. Eur J Med Chem 2021; 213:113140. [PMID: 33454549 DOI: 10.1016/j.ejmech.2020.113140] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 12/09/2020] [Accepted: 12/24/2020] [Indexed: 01/09/2023]
Abstract
The loss of neurons is strongly correlated with aging and aging-associated disorders. In this study, cell viability assays and mitochondrial function were performed to evaluate the effect of new spiro-pyrazole derivatives, prepared from aldehydes and 3-amino-1-phenyl-2-pyrazolin-5-one, on neuroprotection in an in vitro model of dopaminergic cell death induced by 1-methyl-4-phenylpyridinium (MPP+). The percentages of neuroprotection by derivatives were found between 21.26% and 52.67% at selected concentrations (10-50 μM) with compound 4d exerting the best neuroprotective effect. The results show that the studied spiropyrazolones perform important roles in dopaminergic neuroprotection and can be used for potential new therapies in the treatment of neurodegenerative disorders including Parkinson's disease.
Collapse
Affiliation(s)
- Jamila Zaiter
- Laboratoire de Chimie Organique et Analytique, Faculté des Sciences et Techniques, Université Sultan Moulay Slimane, BP 523, 23000, Beni-Mellal, Morocco
| | - Achraf Hibot
- Laboratoire de Chimie Organique et Analytique, Faculté des Sciences et Techniques, Université Sultan Moulay Slimane, BP 523, 23000, Beni-Mellal, Morocco
| | - Abderrafia Hafid
- Laboratoire de Chimie Organique et Analytique, Faculté des Sciences et Techniques, Université Sultan Moulay Slimane, BP 523, 23000, Beni-Mellal, Morocco
| | - Mostafa Khouili
- Laboratoire de Chimie Organique et Analytique, Faculté des Sciences et Techniques, Université Sultan Moulay Slimane, BP 523, 23000, Beni-Mellal, Morocco
| | - Claudia M B Neves
- LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, 3810-193, Aveiro, Portugal
| | - Mário M Q Simões
- LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, 3810-193, Aveiro, Portugal
| | - M Graça P M S Neves
- LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, 3810-193, Aveiro, Portugal
| | - M Amparo F Faustino
- LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, 3810-193, Aveiro, Portugal
| | - Taner Dagci
- Department of Physiology, Faculty of Medicine, Ege University, 35100, Bornova, Izmir, Turkey
| | - Luciano Saso
- Department of Physiology and Pharmacology "Vittorio Erspamer", Sapienza University of Rome, P. le Aldo Moro 5, 00185, Rome, Italy
| | - Güliz Armagan
- Department of Biochemistry, Faculty of Pharmacy, Ege University, 35100, Bornova, Izmir, Turkey.
| |
Collapse
|
134
|
Lovin LM, Kim S, Taylor RB, Scarlett KR, Langan LM, Chambliss CK, Chatterjee S, Scott JT, Brooks BW. Differential influences of (±) anatoxin-a on photolocomotor behavior and gene transcription in larval zebrafish and fathead minnows. ENVIRONMENTAL SCIENCES EUROPE 2021; 33:40. [PMID: 34367861 PMCID: PMC8345817 DOI: 10.1186/s12302-021-00479-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/07/2023]
Abstract
BACKGROUND Though anatoxin-a (antx-a) is a globally important cyanobacterial neurotoxin in inland waters, information on sublethal toxicological responses of aquatic organisms is limited. We examined influences of (±) antx-a (11-3490 μg/L) on photolocomotor behavioral responses and gene transcription associated with neurotoxicity, oxidative stress and hepatotoxicity, in two of the most common alternative vertebrate and fish models, Danio rerio (zebrafish) and Pimephales promelas (fathead minnow). We selected environmentally relevant treatment levels from probabilistic exposure distributions, employed standardized experimental designs, and analytically verified treatment levels using isotope-dilution liquid chromatography tandem mass spectrometry. Caffeine was examined as a positive control. RESULTS Caffeine influences on fish behavior responses were similar to previous studies. Following exposure to (±) antx-a, no significant photolocomotor effects were observed during light and dark transitions for either species. Though zebrafish behavioral responses profiles were not significantly affected by (±) antx-a at the environmentally relevant treatment levels examined, fathead minnow stimulatory behavior was significantly reduced in the 145-1960 μg/L treatment levels. In addition, no significant changes in transcription of target genes were observed in zebrafish; however, elavl3 and sod1 were upregulated and gst and cyp3a126 were significantly downregulated in fathead minnows. CONCLUSION We observed differential influences of (±) antx-a on swimming behavior and gene transcription in two of the most common larval fish models employed for prospective and retrospective assessment of environmental contaminants and water quality conditions. Sublethal responses of fathead minnows were consistently more sensitive than zebrafish to this neurotoxin at the environmentally relevant concentrations examined. Future studies are needed to understand such interspecies differences, the enantioselective toxicity of this compound, molecular initiation events within adverse outcome pathways, and subsequent individual and population risks for this emerging water quality threat.
Collapse
Affiliation(s)
- Lea M. Lovin
- Department of Environmental Science, Baylor University, Waco, TX 76798, USA
| | - Sujin Kim
- Department of Environmental Science, Baylor University, Waco, TX 76798, USA
| | | | | | - Laura M. Langan
- Department of Environmental Science, Baylor University, Waco, TX 76798, USA
| | | | - Saurabh Chatterjee
- Department of Environmental Health Sciences, University of South Carolina, Columbia, SC 29208, USA
| | - J. Thad Scott
- Department of Biology, Baylor University, Waco, TX 76798, USA
| | - Bryan W. Brooks
- Department of Environmental Science, Baylor University, Waco, TX 76798, USA
- Correspondence:
| |
Collapse
|
135
|
Almeida Â, Calisto V, Esteves VI, Schneider RJ, Figueira E, Soares AMVM, Freitas R. Can ocean warming alter sub-lethal effects of antiepileptic and antihistaminic pharmaceuticals in marine bivalves? AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2021; 230:105673. [PMID: 33221665 DOI: 10.1016/j.aquatox.2020.105673] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Revised: 10/23/2020] [Accepted: 10/25/2020] [Indexed: 06/11/2023]
Abstract
The negative effects induced in marine organisms by Climate Change related abiotic factors consequences, namely ocean warming, are well-known. However, few works studied the combined impacts of ocean warming and contaminants, as pharmaceutical drugs. Carbamazepine (CBZ) and cetirizine (CTZ) occur in the marine environment, showing negative effects in marine organisms. This study aimed to evaluate the impacts of ocean warming on the effects of CBZ and CTZ, when acting individually and combined (drug vs drug), in the edible clam Ruditapes philippinarum. For that, drugs concentration, bioconcentration factors and biochemical parameters, related with clam's metabolic capacity and oxidative stress, were evaluated after 28 days exposure to environmentally relevant scenarios of these stressors. The results showed limited impacts of the drugs (single and combined) at control and warming condition. Indeed, it appeared that warming improved the oxidative status of contaminated clams (higher reduced to oxidized glutathione ratio, lower lipid peroxidation and protein carbonylation levels), especially when both drugs were combined. This may result from clam's defence mechanisms activation and reduced metabolic capacity that, respectively, increased elimination and limited production of reactive oxygen species. At low stress levels, defence mechanisms were not activated which resulted into oxidative stress. The present findings highlighted that under higher stress levels clams may be able to activate defence strategies that were sufficient to avoid cellular damages and loss of redox homeostasis. Nevertheless, low concentrations were tested in the present study and the observed responses may greatly change under increased pollution levels or temperatures. Further research on this topic is needed since marine heat waves are increasing in frequency and intensity and pollution levels of some pharmaceuticals are also increasing in coastal systems.
Collapse
Affiliation(s)
- Ângela Almeida
- Biology Department & CESAM, University of Aveiro, 3810-193, Aveiro, Portugal
| | - Vânia Calisto
- Chemistry Department & CESAM, University of Aveiro, 3810-193, Aveiro, Portugal
| | - Valdemar I Esteves
- Chemistry Department & CESAM, University of Aveiro, 3810-193, Aveiro, Portugal
| | - Rudolf J Schneider
- BAM Federal Institute for Materials Research and Testing, Richard-Willstaetter -Str. 11, D-12489, Berlin, Germany
| | - Etelvina Figueira
- Biology Department & CESAM, University of Aveiro, 3810-193, Aveiro, Portugal
| | - Amadeu M V M Soares
- Biology Department & CESAM, University of Aveiro, 3810-193, Aveiro, Portugal
| | - Rosa Freitas
- Biology Department & CESAM, University of Aveiro, 3810-193, Aveiro, Portugal.
| |
Collapse
|
136
|
Almeida Â, Esteves VI, Soares AMVM, Freitas R. Effects of Carbamazepine in Bivalves: A Review. REVIEWS OF ENVIRONMENTAL CONTAMINATION AND TOXICOLOGY 2021; 254:163-181. [PMID: 32926215 DOI: 10.1007/398_2020_51] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Carbamazepine (CBZ) is among the ten most frequent pharmaceuticals that occur in the aquatic systems, with known effects on inhabiting organisms, including bivalves. Bivalves are important species in coastal ecosystems, often exhibiting a dominant biomass within invertebrate communities. These organisms play a major role in the functioning of the ecosystem and particularly in food webs (as suspension-feeders) and represent a significant fraction of the fisheries resource. They also have strong interactions with the environment, water and sediment and are considered good bioindicator species. The present paper reviews the known literature on the impacts of CBZ in biological endpoints of marine bivalves exposed to environmentally and non-environmentally relevant concentrations, highlighting differences in terms of biological responses, associated with exposure period, concentrations tested, and species used. Overall, the literature available showed that CBZ induces individual and sub-individual effects in marine bivalves (adults and life stages) and the most common effect reported was the induction of oxidative stress.
Collapse
Affiliation(s)
- Ângela Almeida
- Biology Department and CESAM, University of Aveiro, Aveiro, Portugal
| | | | | | - Rosa Freitas
- Biology Department and CESAM, University of Aveiro, Aveiro, Portugal.
| |
Collapse
|
137
|
Paraoxonase Role in Human Neurodegenerative Diseases. Antioxidants (Basel) 2020; 10:antiox10010011. [PMID: 33374313 PMCID: PMC7824310 DOI: 10.3390/antiox10010011] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 12/17/2020] [Accepted: 12/18/2020] [Indexed: 12/11/2022] Open
Abstract
The human body has biological redox systems capable of preventing or mitigating the damage caused by increased oxidative stress throughout life. One of them are the paraoxonase (PON) enzymes. The PONs genetic cluster is made up of three members (PON1, PON2, PON3) that share a structural homology, located adjacent to chromosome seven. The most studied enzyme is PON1, which is associated with high density lipoprotein (HDL), having paraoxonase, arylesterase and lactonase activities. Due to these characteristics, the enzyme PON1 has been associated with the development of neurodegenerative diseases. Here we update the knowledge about the association of PON enzymes and their polymorphisms and the development of multiple sclerosis (MS), amyotrophic lateral sclerosis (ALS), Alzheimer's disease (AD) and Parkinson's disease (PD).
Collapse
|
138
|
Volkert MR, Crowley DJ. Preventing Neurodegeneration by Controlling Oxidative Stress: The Role of OXR1. Front Neurosci 2020; 14:611904. [PMID: 33384581 PMCID: PMC7770112 DOI: 10.3389/fnins.2020.611904] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 11/23/2020] [Indexed: 12/31/2022] Open
Abstract
Parkinson’s disease, diabetic retinopathy, hyperoxia induced retinopathy, and neuronal damage resulting from ischemia are among the notable neurodegenerative diseases in which oxidative stress occurs shortly before the onset of neurodegeneration. A shared feature of these diseases is the depletion of OXR1 (oxidation resistance 1) gene products shortly before the onset of neurodegeneration. In animal models of these diseases, restoration of OXR1 has been shown to reduce or eliminate the deleterious effects of oxidative stress induced cell death, delay the onset of symptoms, and reduce overall severity. Moreover, increasing OXR1 expression in cells further increases oxidative stress resistance and delays onset of disease while showing no detectable side effects. Thus, restoring or increasing OXR1 function shows promise as a therapeutic for multiple neurodegenerative diseases. This review examines the role of OXR1 in oxidative stress resistance and its impact on neurodegenerative diseases. We describe the potential of OXR1 as a therapeutic in light of our current understanding of its function at the cellular and molecular level and propose a possible cascade of molecular events linked to OXR1’s regulatory functions.
Collapse
Affiliation(s)
- Michael R Volkert
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA, United States
| | - David J Crowley
- Department of Biological and Physical Sciences, Assumption University, Worcester, MA, United States
| |
Collapse
|
139
|
Aluko OM, Umukoro S. Methyl jasmonate reverses chronic stress-induced memory dysfunctions through modulation of monoaminergic neurotransmission, antioxidant defense system, and Nrf2 expressions. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2020; 393:2339-2353. [PMID: 32666287 DOI: 10.1007/s00210-020-01939-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Accepted: 07/03/2020] [Indexed: 12/12/2022]
Abstract
Unpredictable chronic mild stress (UCMS) has been shown to cause memory loss via increased oxidative stress and deregulation of monoaminergic and cholinergic neurotransmissions. Although the benefits of methyl jasmonate (MJ), a well-known anti-stress plant hormone against chronic stress-induced psychopathologies, have been earlier reported, its effects on antioxidant defense molecules, monoaminergic transmitters, and nuclear factor erythroid 2-related factor 2 (Nrf2) immunopositive cells have not been extensively studied. The present study was designed to examine its effect on memory functions, antioxidant biomarkers, monoaminergic transmitters, and Nrf2 immunopositive cell expression in rats exposed to UCMS. Rats received an intraperitoneal injection of MJ (10, 25, and 50 mg/kg) 30 min before exposure to UCMS daily for 28 days. Memory function was assessed on day 29 using a modified elevated plus maze and novel object recognition tests. The antioxidant biomarkers, level of monoamines (serotonin, noradrenaline, and dopamine), and Nrf2 immunopositive cell expression were determined in the rat brain tissues. The activity of cholinesterase and monoamine oxidase enzymes was also determined. MJ attenuated memory deficits and elevated the brain levels of monoamines in UCMS rats. UCMS-induced increase of brain cholinesterase and monoamine oxidase activities was inhibited by MJ. Also, MJ attenuated UCMS-induced decrease in antioxidant enzymes (CAT, GPx, GST, and SOD) and thiol contents in the brains of rats. UCMS-induced increase in NO level and Nrf2 immunopositive cell expression in the rat's brain was attenuated by MJ. Taken together, these findings suggest that increasing antioxidant defense molecules and monoaminergic/cholinergic neurotransmitters and decreasing the Nrf2 immunopositive cell expressions may contribute to the memory-promoting effects of MJ in rats exposed to UCMS.
Collapse
Affiliation(s)
- Oritoke M Aluko
- Department of Physiology, School of Health and Health Technology, Federal University of Technology, Akure, Nigeria.
- Neuropharmacology Unit, Department of Pharmacology and Therapeutics, Faculty of Basic Medical Sciences, University of Ibadan, Ibadan, Nigeria.
| | - Solomon Umukoro
- Neuropharmacology Unit, Department of Pharmacology and Therapeutics, Faculty of Basic Medical Sciences, University of Ibadan, Ibadan, Nigeria
| |
Collapse
|
140
|
Fassihi A, Hasanzadeh F, Attar AM, Saghaie L, Mohammadpour M. Synthesis and evaluation of antioxidant activity of some novel hydroxypyridinone derivatives: a DFT approach for explanation of their radical scavenging activity. Res Pharm Sci 2020; 15:515-528. [PMID: 33828595 PMCID: PMC8020857 DOI: 10.4103/1735-5362.301336] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Revised: 09/11/2020] [Accepted: 10/26/2020] [Indexed: 11/17/2022] Open
Abstract
Background and purpose: Reactive oxygen species (ROSs) are continuously produced as byproducts of cell metabolism. Free radicals are an unstable form of ROSs with the tendency to react readily with biomolecules such as amino acids, lipids and DNA. These reactions lead to oxidative damages to the cell. Oxidative stress occurs when the concentration of the ROSs exceeds the capacity of antioxidative protection systems of the body. 5-Hydroxypyridin-4-one derivatives can chelate Fe2+ and Fe3+ due to their α-hydroxyketone moiety. Also, tautomerism in hydroxypyridinone ring leads to enough level of aromaticity resulting in a catechol-like behavior that provides them with good chelating and radical scavenging properties. Experimental approach: Different compounds were synthesized with 5-hydroxypyridine-4-one moiety as the core. The antioxidant properties of molecules were evaluated experimentally by DPPH scavenging method and theoretically using DFT/B3LYP with a 6-31++G (d,p) basis set. Electronic properties were investigated using frontier molecular orbital theory calculations. Furthermore, global descriptive parameters were obtained to find the chemical reactivity of molecules. The natural bond orbital analysis was performed to investigate charge distribution and hydrogen bonding. Findings/Results: Structures of the synthesized compounds were confirmed using IR, 1H-NMR, and 13C-NMR spectral analyses. Among all the synthesized compounds, Va and Vb showed the best antioxidant effect experimentally and computationally. Conclusion and implications: Results of this study were valuable in terms of synthesis, in silico, and in vitro antioxidant evaluations and can be useful for future investigations about the design of novel 5-hydroxypyridin- 4-one derivatives possessing iron-chelating and radical scavenging abilities.
Collapse
Affiliation(s)
- Afshin Fassihi
- Department of Medicinal Chemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, I.R. Iran.,Bioinformatics Research Center, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, I.R. Iran
| | - Farshid Hasanzadeh
- Department of Medicinal Chemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, I.R. Iran.,Bioinformatics Research Center, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, I.R. Iran
| | - Ahmad Movahedian Attar
- Department of Biochemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, I.R. Iran
| | - Lotfalah Saghaie
- Department of Medicinal Chemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, I.R. Iran.,Bioinformatics Research Center, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, I.R. Iran
| | - Mehrdad Mohammadpour
- Department of Medicinal Chemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, I.R. Iran
| |
Collapse
|
141
|
Saeedi M, Rashidy-Pour A. Association between chronic stress and Alzheimer's disease: Therapeutic effects of Saffron. Biomed Pharmacother 2020; 133:110995. [PMID: 33232931 DOI: 10.1016/j.biopha.2020.110995] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 10/28/2020] [Accepted: 11/01/2020] [Indexed: 12/19/2022] Open
Abstract
Chronic stress and high levels of glucocorticoids produce functional and structural changes in brain and especially in the hippocampus, an important limbic system structure that plays a key role in cognitive functions including learning and memory. Alzheimer's disease (AD) is a chronic neurodegenerative disease that usually starts slowly and worsens over time. Indeed, cognitive dysfunction, neuronal atrophy, and synaptic loss are associated with both AD and chronic stress. Recent preclinical and clinical studies have highlighted a possible link between chronic stress, cognitive decline and the development of AD. It is suggested that Tau protein is an essential mediator of the neurodegenerative effects of stress and glucocorticoids towards the development of AD pathology. Recent findings from animal and humans studies demonstrated that saffron and its main constitutive crocin are effective against chronic stress-induced cognitive dysfunction and oxidative stress and slowed cognitive decline in AD. The inhibitory actions on acetylcholinesterase activity, aggregation of beta-amyloid protein into amyloid plaques and tau protein into neurofibrillary tangles, and also the antioxidant, anti-inflammatory, and the promotion of synaptic plasticity effects are among the possible mechanisms to explain the neuroprotective effects of saffron. New evidences demonstrate that saffron and its main component crocin might be a promising target for cognition improvement in AD and stress-related disorders.
Collapse
Affiliation(s)
- Mohammad Saeedi
- Student Research Committee, Semnan University of Medical Sciences, Semnan, Iran
| | - Ali Rashidy-Pour
- Research Center of Physiology, Semnan University of Medical Sciences, Semnan, Iran.
| |
Collapse
|
142
|
Ghasemzadeh Rahbardar M, Hemadeh B, Razavi BM, Eisvand F, Hosseinzadeh H. Effect of carnosic acid on acrylamide induced neurotoxicity: in vivo and in vitro experiments. Drug Chem Toxicol 2020; 45:1528-1535. [DOI: 10.1080/01480545.2020.1845715] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
| | - Batool Hemadeh
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Bibi Marjan Razavi
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
- Targeted Drug Delivery Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Farhad Eisvand
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hossein Hosseinzadeh
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
143
|
Hamezah HS, Durani LW, Yanagisawa D, Ibrahim NF, Aizat WM, Makpol S, Wan Ngah WZ, Damanhuri HA, Tooyama I. Modulation of Proteome Profile in AβPP/PS1 Mice Hippocampus, Medial Prefrontal Cortex, and Striatum by Palm Oil Derived Tocotrienol-Rich Fraction. J Alzheimers Dis 2020; 72:229-246. [PMID: 31594216 PMCID: PMC6839455 DOI: 10.3233/jad-181171] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Tocotrienol-rich fraction (TRF) is a mixture of vitamin E analogs derived from palm oil. We previously demonstrated that supplementation with TRF improved cognitive function and modulated amyloid pathology in AβPP/PS1 mice brains. The current study was designed to examine proteomic profiles underlying the therapeutic effect of TRF in the brain. Proteomic analyses were performed on samples of hippocampus, medial prefrontal cortex (mPFC), and striatum using liquid chromatography coupled to Q Exactive HF Orbitrap mass spectrometry. From these analyses, we profiled a total of 5,847 proteins of which 155 proteins were differentially expressed between AβPP/PS1 and wild-type mice. TRF supplementation of these mice altered the expression of 255 proteins in the hippocampus, mPFC, and striatum. TRF also negatively modulated the expression of amyloid beta A4 protein and receptor-type tyrosine-protein phosphatase alpha protein in the hippocampus. The expression of proteins in metabolic pathways, oxidative phosphorylation, and those involved in Alzheimer’s disease were altered in the brains of AβPP/PS1 mice that received TRF supplementation.
Collapse
Affiliation(s)
- Hamizah Shahirah Hamezah
- Molecular Neuroscience Research Center, Shiga University of Medical Science, Seta Tsukinowa-cho, Otsu, Japan
| | - Lina Wati Durani
- Molecular Neuroscience Research Center, Shiga University of Medical Science, Seta Tsukinowa-cho, Otsu, Japan
| | - Daijiro Yanagisawa
- Molecular Neuroscience Research Center, Shiga University of Medical Science, Seta Tsukinowa-cho, Otsu, Japan
| | - Nor Faeizah Ibrahim
- Department of Biochemistry, Faculty of Medicine, UKMMC, Universiti Kebangsaan Malaysia (UKM), Jalan Yaacob Latif, Cheras, Kuala Lumpur, Malaysia
| | - Wan Mohd Aizat
- Institute of Systems Biology, Universiti Kebangsaan Malaysia, Bangi, Selangor, Malaysia
| | - Suzana Makpol
- Department of Biochemistry, Faculty of Medicine, UKMMC, Universiti Kebangsaan Malaysia (UKM), Jalan Yaacob Latif, Cheras, Kuala Lumpur, Malaysia
| | - Wan Zurinah Wan Ngah
- Department of Biochemistry, Faculty of Medicine, UKMMC, Universiti Kebangsaan Malaysia (UKM), Jalan Yaacob Latif, Cheras, Kuala Lumpur, Malaysia
| | - Hanafi Ahmad Damanhuri
- Department of Biochemistry, Faculty of Medicine, UKMMC, Universiti Kebangsaan Malaysia (UKM), Jalan Yaacob Latif, Cheras, Kuala Lumpur, Malaysia
| | - Ikuo Tooyama
- Molecular Neuroscience Research Center, Shiga University of Medical Science, Seta Tsukinowa-cho, Otsu, Japan
| |
Collapse
|
144
|
Therapeutic Potential of Porcine Liver Decomposition Product: New Insights and Perspectives for Microglia-Mediated Neuroinflammation in Neurodegenerative Diseases. Biomedicines 2020; 8:biomedicines8110446. [PMID: 33105637 PMCID: PMC7690401 DOI: 10.3390/biomedicines8110446] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 10/16/2020] [Accepted: 10/21/2020] [Indexed: 12/23/2022] Open
Abstract
It is widely accepted that microglia-mediated inflammation contributes to the progression of neurodegenerative diseases; however, the precise mechanisms through which these cells contribute remain to be elucidated. Microglia, as the primary immune effector cells of the brain, play key roles in maintaining central nervous system (CNS) homeostasis. Microglia are located throughout the brain and spinal cord and may account for up to 15% of all cells in the brain. Activated microglia express pro-inflammatory cytokines that act on the surrounding brain and spinal cord. Microglia may also play a detrimental effect on nerve cells when they gain a chronic inflammatory function and promote neuropathologies. A key feature of microglia is its rapid morphological change upon activation, characterized by the retraction of numerous fine processes and the gradual acquisition of amoeba-like shapes. These morphological changes are also accompanied by the expression and secretion of inflammatory molecules, including cytokines, chemokines, and lipid mediators that promote systemic inflammation during neurodegeneration. This may be considered a protective response intended to limit further injury and initiate repair processes. We previously reported that porcine liver decomposition product (PLDP) induces a significant increase in the Hasegawa’s Dementia Scale-Revised (HDS-R) score and the Wechsler Memory Scale (WMS) in a randomized, double-blind, placebo-controlled study in healthy humans. In addition, the oral administration of porcine liver decomposition product enhanced visual memory and delayed recall in healthy adults. We believe that PLDP is a functional food that aids cognitive function. In this review, we provide a critical assessment of recent reports of lysophospholipids derived from PLDP, a rich source of phospholipids. We also highlight some recent findings regarding bidirectional interactions between lysophospholipids and microglia and age-related neurodegenerative diseases such as dementia and Alzheimer’s disease.
Collapse
|
145
|
Kesh S, Kannan RR, Sivaji K, Balakrishnan A. Hesperidin downregulates kinases lrrk2 and gsk3β in a 6-OHDA induced Parkinson's disease model. Neurosci Lett 2020; 740:135426. [PMID: 33075420 DOI: 10.1016/j.neulet.2020.135426] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 09/03/2020] [Accepted: 10/04/2020] [Indexed: 12/11/2022]
Abstract
The depletion of dopamine in the striatum region and Lewy bodies are the hallmark characteristics of Parkinson's disease. The pathology also includes the upregulation of various Parkinson's disease (PARK) genes and kinases. Two such kinases, LRRK2 and GSK-3β have been directly implicated in the formation of tau and alpha-synuclein proteins, causing PD. Hesperidin (HES) is a flavanone glycoside that has multiple therapeutic benefits including neuroprotective effects. In this study, we examined the neuroprotective effects of HES against 6-hydroxydopamine (6-OHDA) induced-neurotoxicity in the in-vitro and in-vivo model. Hesperidin significantly protected the SH-SY5Y cells' stress against 6-OHDA induced toxicity by downregulating biomarkers of oxidative stress. Furthermore, HES downregulated the kinases lrrk2 and gsk3β along with casp3, casp9, and polg in the zebrafish model. The treatment with HES also improved the locomotor pattern of zebrafish that was affected by 6-OHDA. This study suggests that hesperidin could be a drug of choice in targeting kinases against a 6-OHDA model of PD.
Collapse
Affiliation(s)
- Swathi Kesh
- Neuroscience Lab, Centre for Molecular and Nanomedical Sciences, School of Bio and Chemical Engineering, Sathyabama Institute of Science and Technology, Chennai, 600119, Tamil Nadu, India.
| | - Rajaretinam Rajesh Kannan
- Neuroscience Lab, Centre for Molecular and Nanomedical Sciences, School of Bio and Chemical Engineering, Sathyabama Institute of Science and Technology, Chennai, 600119, Tamil Nadu, India.
| | - Kalaiarasi Sivaji
- Neuroscience Lab, Centre for Molecular and Nanomedical Sciences, School of Bio and Chemical Engineering, Sathyabama Institute of Science and Technology, Chennai, 600119, Tamil Nadu, India.
| | - Anandan Balakrishnan
- Department of Genetics, Dr. ALM PGIBMS Campus, University of Madras, Taramani, Chennai, Tamil Nadu, India.
| |
Collapse
|
146
|
Park SK, Hyun SH, In G, Park CK, Kwak YS, Jang YJ, Kim B, Kim JH, Han CK. The antioxidant activities of Korean Red Ginseng ( Panax ginseng) and ginsenosides: A systemic review through in vivo and clinical trials. J Ginseng Res 2020; 45:41-47. [PMID: 33437155 PMCID: PMC7790892 DOI: 10.1016/j.jgr.2020.09.006] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 09/24/2020] [Accepted: 09/29/2020] [Indexed: 12/19/2022] Open
Abstract
A wide range of studies have steadily pointed out the relation of oxidative stress to the primary and secondary causes of human disease and aging. As such, there have been multiple misconceptions about oxidative stress. Most of reactive oxygen species (ROS) generated from chronic diseases cause oxidative damage to cell membrane lipids and proteins. ROS production is increased by abnormal stimulation inside and outside in the body, and even though ROS are generated in cells in response to abnormal metabolic processes such as disease, it does not mean that they directly contribute to the pathogenesis of a disease. Therefore, the focus of treatment should not be on ROS production itself but on the prevention and treatment of diseases linked to ROS production, including types 1 and 2 diabetes, cancer, heart disease, schizophrenia, Parkinson's disease, and Alzheimer's disease. In this regard, Korean Red Ginseng (KRG) has been traditionally utilized to help prevent and treat diseases such as diabetes, cancer, inflammation, nervous system diseases, cardiovascular disease, and hyperlipidemia. Therefore, this review was intended to summarize in vivo animal and human clinical studies on the antioxidant activities of KRG and its components, ginsenosides.
Collapse
Affiliation(s)
- Soo Kyung Park
- Laboratory of Efficacy Research, Korea Ginseng Corporation, 30, Gajeong-ro, Shinseong-dong, Yuseong-gu, Daejeon, Republic of Korea
| | - Sun Hee Hyun
- Laboratory of Efficacy Research, Korea Ginseng Corporation, 30, Gajeong-ro, Shinseong-dong, Yuseong-gu, Daejeon, Republic of Korea
| | - Gyo In
- Laboratory of Efficacy Research, Korea Ginseng Corporation, 30, Gajeong-ro, Shinseong-dong, Yuseong-gu, Daejeon, Republic of Korea
| | - Chae-Kyu Park
- Laboratory of Efficacy Research, Korea Ginseng Corporation, 30, Gajeong-ro, Shinseong-dong, Yuseong-gu, Daejeon, Republic of Korea
| | - Yi-Seong Kwak
- Laboratory of Efficacy Research, Korea Ginseng Corporation, 30, Gajeong-ro, Shinseong-dong, Yuseong-gu, Daejeon, Republic of Korea
| | - Young-Jin Jang
- College of Veterinary Medicine, Biosafety Research Institute, Jeonbuk National University, Iksan-city, Republic of Korea
| | - Bumseok Kim
- College of Veterinary Medicine, Biosafety Research Institute, Jeonbuk National University, Iksan-city, Republic of Korea
| | - Jong-Hoon Kim
- College of Veterinary Medicine, Biosafety Research Institute, Jeonbuk National University, Iksan-city, Republic of Korea
| | - Chang-Kyun Han
- Laboratory of Efficacy Research, Korea Ginseng Corporation, 30, Gajeong-ro, Shinseong-dong, Yuseong-gu, Daejeon, Republic of Korea
| |
Collapse
|
147
|
Ghasemzadeh Rahbardar M, Hosseinzadeh H. Effects of rosmarinic acid on nervous system disorders: an updated review. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2020; 393:1779-1795. [PMID: 32725282 DOI: 10.1007/s00210-020-01935-w] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 06/29/2020] [Indexed: 02/07/2023]
Abstract
Nowadays, the worldwide interest is growing to use medicinal plants and their active constituents to develop new potent medicines with fewer side effects. Precise dietary compounds have prospective beneficial applications for various neurodegenerative ailments. Rosmarinic acid is a polyphenol and is detectable most primarily in many Lamiaceae families, for instance, Rosmarinus officinalis also called rosemary. This review prepared a broad and updated literature review on rosmarinic acid elucidating its biological activities on some nervous system disorders. Rosmarinic acid has significant antinociceptive, neuroprotective, and neuroregenerative effects. In this regard, we classified and discussed our findings in different nervous system disorders including Alzheimer's disease, epilepsy, depression, Huntington's disease, familial amyotrophic lateral sclerosis, Parkinson's disease, cerebral ischemia/reperfusion injury, spinal cord injury, stress, anxiety, and pain.
Collapse
Affiliation(s)
| | - Hossein Hosseinzadeh
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
148
|
Temitayo GI, Olawande B, Emmanuel YO, Timothy AT, Kehinde O, Susan LF, Ezra L, Joseph OO. Inhibitory potentials of Cymbopogon citratus oil against aluminium-induced behavioral deficits and neuropathology in rats. Anat Cell Biol 2020; 53:342-354. [PMID: 32839358 PMCID: PMC7527123 DOI: 10.5115/acb.20.099] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 05/19/2020] [Accepted: 05/25/2020] [Indexed: 01/04/2023] Open
Abstract
Cymbopogon citratus is a tropical phytomedicinal plant that is widely known for its hypoglycemic, hypolipidemic, anxiolytic, sedative, antioxidative and anti-inflammatory properties. In this study, we have examined the neuroprotective effects of the essential oil (ESO) of Cymbopogon citratus, following aluminum chloride (AlCl3)-induced neurotoxicity within the cerebellum of Wistar rats. A total of 40 adult male Wistar rats were assigned into five groups and treated orally as follows: A–phosphate-buffered saline (1 ml daily for 15 days); B–ESO (50 mg/kg daily for 15 days); C–AlCl3 (100 mg/kg daily for 15 days); D–AlCl3 then ESO (100 mg/kg AlCl3 daily for 15 days followed by 50 mg/kg ESO daily for subsequent 15 days); E–ESO then AlCl3 (50 mg/kg ESO daily for 15 days followed by 100 mg/kg AlCl3 daily for following 15 days). To address our questions, we observed the locomotion and exploratory behavior of the rats in the open field apparatus and subsequently evaluated cerebellar oxidative redox parameters, neural bioenergetics, acetylcholinesterase levels, transferrin receptor protein, and total protein profiles by biochemical assays. Furthermore, we investigated cerebellar histomorphology and Nissl profile by H&E and Cresyl violet Nissl staining procedures. ESO treatment markedly attenuated deficits in exploratory activities and rearing behavior following AlCl3 toxicity, indicating its anxiolytic potentials. Additionally, AlCl3 evokedincrease in malondialdehyde and nitric oxide levels, as well as repressed cerebellar catalase, glutathione peroxidase, and superoxide dismutase profiles were normalised to baseline levels by ESO treatment. Treatment with ESO, ergo, exhibits substantial neuroprotective and modulatory potentials in response to AlCl3 toxicity.
Collapse
Affiliation(s)
- Gbadamosi Ismail Temitayo
- Division of Neurobiology, Department of Anatomy, Faculty of Basic Medical Sciences, College of Health Sciences, University of Ilorin, Ilorin, Nigeria
| | - Bamisi Olawande
- Division of Neurobiology, Department of Anatomy, Faculty of Basic Medical Sciences, College of Health Sciences, University of Ilorin, Ilorin, Nigeria
| | - Yawson Olushola Emmanuel
- Division of Neurobiology, Department of Anatomy, Faculty of Basic Medical Sciences, College of Health Sciences, University of Ilorin, Ilorin, Nigeria.,Division of Neurobiology, Department of Anatomy, Faculty of Basic Medical Sciences, Adeleke University, Ede, Nigeria
| | - Arogundade Tolulope Timothy
- Division of Neurobiology, Department of Anatomy, Faculty of Basic Medical Sciences, College of Health Sciences, University of Ilorin, Ilorin, Nigeria.,Division of Neurobiology, Department of Anatomy, Faculty of Basic Medical Sciences, Adeleke University, Ede, Nigeria
| | - Ogunrinola Kehinde
- Division of Neurobiology, Department of Anatomy, Faculty of Basic Medical Sciences, College of Health Sciences, University of Ilorin, Ilorin, Nigeria
| | - Lewu Folashade Susan
- Division of Neurobiology, Department of Anatomy, Faculty of Basic Medical Sciences, College of Health Sciences, University of Ilorin, Ilorin, Nigeria
| | - Lambe Ezra
- Division of Neurobiology, Department of Anatomy, Faculty of Basic Medical Sciences, College of Health Sciences, University of Ilorin, Ilorin, Nigeria
| | - Olajide Olayemi Joseph
- Division of Neurobiology, Department of Anatomy, Faculty of Basic Medical Sciences, College of Health Sciences, University of Ilorin, Ilorin, Nigeria.,Center for Studies in Behavioral Neurobiology, Department of Psychology, Concordia University Montreal, Montreal, Quebec, Canada
| |
Collapse
|
149
|
Dantas LS, Viviani LG, Inague A, Piccirillo E, Rezende LD, Ronsein GE, Augusto O, Medeiros MHG, Amaral ATD, Miyamoto S. Lipid aldehyde hydrophobicity affects apo-SOD1 modification and aggregation. Free Radic Biol Med 2020; 156:157-167. [PMID: 32598986 DOI: 10.1016/j.freeradbiomed.2020.05.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 04/30/2020] [Accepted: 05/12/2020] [Indexed: 12/12/2022]
Abstract
Unsaturated lipids are oxidized by reactive oxygen species and enzymes, leading to the increased formation of lipid hydroperoxides and several electrophilic products. Lipid-derived electrophiles can modify macromolecules, such as proteins, resulting in the loss of function and/or aggregation. The accumulation of Cu,Zn-superoxide dismutase (SOD1) aggregates has been associated with familial cases of amyotrophic lateral sclerosis (ALS). The protein aggregation mechanisms in motor neurons remain unclear, although recent studies have shown that lipids and oxidized lipid derivatives may play roles in this process. Here, we aimed to compare the effects of different lipid aldehydes on the induction of SOD1 modifications and aggregation, in vitro. Human recombinant apo-SOD1 was incubated with 4-hydroxy-2-hexenal (HHE), 4-hydroxy-2-nonenal (HNE), 2-hexen-1-al (HEX), 2,4-nonadienal (NON), 2,4-decadienal (DEC), or secosterol aldehydes (SECO-A or SECO-B). High-molecular-weight apo-SOD1 aggregates dramatically increased in the presence of highly hydrophobic aldehydes (LogPcalc > 3). Notably, several Lys residues were modified by exposure to all aldehydes. The observed modifications were primarily observed on Lys residues located near the dimer interface (K3 and K9) and at the electrostatic loop (K122, K128, and K136). Moreover, HHE and HNE induced extensive apo-SOD1 modifications, by forming Schiff bases or Michael adducts with Lys, His, and Cys residues. However, these aldehydes were unable to induce large protein aggregates. Overall, our data shed light on the importance of lipid aldehyde hydrophobicity on the induction of apo-SOD1 aggregation and identified preferential sites of lipid aldehyde-induced modifications.
Collapse
Affiliation(s)
- Lucas S Dantas
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Lucas G Viviani
- Departamento de Química Fundamental, Instituto de Química, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Alex Inague
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Erika Piccirillo
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, SP, Brazil; Departamento de Química Fundamental, Instituto de Química, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Leandro de Rezende
- Departamento de Química Fundamental, Instituto de Química, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Graziella E Ronsein
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Ohara Augusto
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Marisa H G Medeiros
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Antonia T do Amaral
- Departamento de Química Fundamental, Instituto de Química, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Sayuri Miyamoto
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, SP, Brazil.
| |
Collapse
|
150
|
Chaturvedi S, Malik MY, Rashid M, Singh S, Tiwari V, Gupta P, Shukla S, Singh S, Wahajuddin M. Mechanistic exploration of quercetin against metronidazole induced neurotoxicity in rats: Possible role of nitric oxide isoforms and inflammatory cytokines. Neurotoxicology 2020; 79:1-10. [DOI: 10.1016/j.neuro.2020.03.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2019] [Revised: 03/02/2020] [Accepted: 03/05/2020] [Indexed: 02/06/2023]
|