1
|
Rajan RK, Engels M, Ramanathan M. Predicting phase-I metabolism of piceatannol: an in silico study. In Silico Pharmacol 2024; 12:52. [PMID: 38854674 PMCID: PMC11153392 DOI: 10.1007/s40203-024-00228-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 05/28/2024] [Indexed: 06/11/2024] Open
Abstract
Piceatannol is a natural compound found in plants and can be derived from resveratrol. While resveratrol has been extensively researched for its effects and how the body processes it, there are concerns about its use. These concerns include its limited absorption in the body, the need for specific dosages, potential interactions with other drugs, lack of standardization, and limited clinical evidence to support its benefits. Interestingly, Piceatannol, another compound derived from resveratrol, has received less attention from researchers but appears to offer advantages. It has better bioavailability and seems to have a more favorable therapeutic profile compared to resveratrol. Surprisingly, no previous attempts have been made to explore or predict the metabolites of piceatannol when it interacts with the enzyme cytochrome P450. This study aims to fill that gap by predicting how piceatannol is metabolized by cytochrome P450 and assessing any potential toxicity associated with its metabolites. This research is interesting because it's the first of its kind to investigate the metabolic fate of piceatannol, especially in the context of cytochrome P450. The findings have the potential to significantly contribute to the field of piceatannol research, particularly in the food industry where this compound has applications and implications. Graphical abstract
Collapse
Affiliation(s)
- Ravi Kumar Rajan
- Department of Pharmacology, School of Pharmaceutical Sciences, Girijananda Chowdhury University, Tezpur Campus, Tezpur, Assam India
- Present Address: Department of Pharmacology, Himalayan Pharmacy Institute, Majitar, East Sikkim 737136 India
| | - Maida Engels
- Department of Pharmaceutical Chemistry, PSG College of Pharmacy, Coimbatore, Tamil Nadu India
| | - Muthiah Ramanathan
- Department of Pharmacology, PSG College of Pharmacy, Coimbatore, Tamil Nadu India
| |
Collapse
|
2
|
Pala M, Meral I, Pala Acikgoz N, Mengi M, Erdim Gokce MB, Unsal R, Polat Y, Akbas F, Gorucu Yilmaz S. Thymoquinone ameliorates symptoms of Parkinson's disease in a 6-OHDA rat model by downregulation of miR-204-3p. Behav Pharmacol 2024; 35:201-210. [PMID: 38660812 DOI: 10.1097/fbp.0000000000000776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
microRNAs (miRNAs) play a significant role in the pathophysiology of Parkinson's disease. In this study, we evaluated the neuroprotective effect of thymoquinone on the expression profiles of miRNA and cognitive functions in the 6-hydroxydopamine (6-OHDA)-induced Parkinson's model. Male adult Wistar albino rats (200-230 g, n = 36) were randomly assigned to six groups: Sham, thymoquinone (10 mg/kg, p.o.), 6-OHDA, 6-OHDA + thymoquinone (10 mg/kg), 6-OHDA + thymoquinone (20 mg/kg), and 6-OHDA + thymoquinone (50 mg/kg). Behavioral changes were detected using the open field and the elevated plus maze tests. The mature 728 miRNA expressions were evaluated by miRNA microarray (GeneChip miRNA 4.0). Ten miRNAs were selected (rno-miR-212-5p, rno-miR-146b-5p, rno-miR-150-5p, rno-miR-29b-2-5p, rno-miR-126a-3p, rno-miR-187-3p, rno-miR-34a-5p, rno-miR-181d-5p, rno-miR-204-3p, and rno-miR-30c-2-3p) and confirmed by real-time PCR. Striatum samples were stained with hematoxylin-eosin to determine the effect of dopaminergic lesions. One-way ANOVA test and independent sample t -test were used for statistical analyses. rno-miR-204-3p was upregulated at 6-OHDA and downregulated at the 50 mg/kg dose of thymoquinone. In conclusion, thymoquinone at a dose of 50 mg/kg ameliorates symptoms of Parkinson's disease in a 6-OHDA rat model by downregulation of miR-204-3p. Also, the results showed that thymoquinone can improve locomotor activity and willing exploration and decreased anxiety. Therefore, thymoquinone can be used as a therapeutic agent.
Collapse
Affiliation(s)
- Mukaddes Pala
- Department of Physiology, Faculty of Medicine, Malatya Turgut Ozal University, Malatya
| | | | - Nilgün Pala Acikgoz
- Department of Neurology, Faculty of Medicine, Bezmialem Vakif University, Istanbul
| | - Murat Mengi
- Department of Physiology, Faculty of Medicine, Namik Kemal University, Tekirdag
| | | | - Rumeysa Unsal
- Bakirkoy Prof. Dr. Mazhar Osman Mental Health and Nervous Diseases Training and Research Hospital
| | - Yalcin Polat
- Department of Pathology, Faculty of Medicine, Biruni University
| | - Fahri Akbas
- Department of Medical Biology, Faculty of Medicine, Bezmialem Vakif University, Istanbul
| | - Senay Gorucu Yilmaz
- Department of Nutrition and Dietetics, Faculty of Health Science, Gaziantep University, Gaziantep, Turkey
| |
Collapse
|
3
|
Athari SZ, Farajdokht F, Sadigh-Eteghad S, Mohajeri D, Nourazar MA, Mohaddes G. Hydroxychloroquine attenuated motor impairment and oxidative stress in a rat 6-hydroxydopamine model of Parkinson's disease. Int J Neurosci 2023; 133:1252-1261. [PMID: 35522252 DOI: 10.1080/00207454.2022.2074848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 04/27/2022] [Indexed: 10/18/2022]
Abstract
PURPOSE Parkinson's disease (PD) is associated with the destruction of dopaminergic neurons in the substantia nigra (SN). Hydroxychloroquine (HCQ) has the capability to cross the blood-brain barrier and promote a neuroprotective potential. This study evaluated the effects of HCQ on the 6-hydroxydopamine (6-OHDA)-induced PD model in rats. METHODS Wistar rats were randomly divided into sham, PD, PD + levodopa and PD + HCQ groups. The PD model was induced by a stereotactic administration of 6-OHDA into the left SN pars compacta (SNpc) and confirmed by rotation and the Murprogo's tests. HCQ (100 mg/kg, p.o.) and levodopa (12 mg/kg, p.o.) were administered once a day for 21 days. Three weeks after surgery, the behavioral tests were performed. Brain lipid peroxidation index (MDA), glutathione peroxidase activity (GPx), total antioxidant capacity (TAC) levels and α-synuclein protein expression in the SN were also measured. RESULTS The behavioral tests demonstrated that induction of PD increased the muscle rigidity and the number of rotations, which were reversed by HCQ treatment. Also, induction of PD was associated with an increase in α-synuclein protein levels and MDA and decreased TAC levels and GPx activity. However, HCQ decreased α-synuclein and MDA levels while increased TAC levels and GPx activity. In addition, histopathological data showed that HCQ protects dopaminergic neurons against 6-OHDA-induced toxicity. CONCLUSION According to the results, HCQ has a beneficial effect in improving PD-related pathophysiology, in part, by mitigating oxidative stress and protecting the dopaminergic neurons in the SN.
Collapse
Affiliation(s)
- Seyed Zanyar Athari
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
- Neurosciences Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Fereshteh Farajdokht
- Neurosciences Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Saeed Sadigh-Eteghad
- Neurosciences Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Daryoush Mohajeri
- Department of Pathobiology, Faculty of Veterinary Medicine, Tabriz Medical Sciences, Islamic Azad University, Tabriz, Iran
| | - Mir Alireza Nourazar
- Department of Pathobiology, Faculty of Veterinary Medicine, Tabriz Medical Sciences, Islamic Azad University, Tabriz, Iran
- Department of Basic Sciences, Faculty of Veterinary Medicine, Tabriz Medical Sciences, Islamic Azad University, Tabriz, Iran
| | - Gisou Mohaddes
- Neurosciences Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
4
|
Nakamori H, Hashitani H. Neural targets of the enteric dopaminergic system in regulating motility of rat proximal colon. Pflugers Arch 2023; 475:1315-1327. [PMID: 37589734 DOI: 10.1007/s00424-023-02849-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 08/02/2023] [Accepted: 08/08/2023] [Indexed: 08/18/2023]
Abstract
In isolated segments of the rat proximal colon, the dopamine reuptake inhibitor GBR 12909 (GBR) causes a dilatation, while the D1-like receptor antagonist SCH 23390 (SCH) induces a tonic constriction, suggesting that neurally released dopamine tonically stimulates enteric inhibitory efferent neurons. Here, the targets of the enteric dopaminergic neurons were investigated. Cannulated segments of rat proximal colon were bathed in physiological salt solution and luminally perfused with 0.9% saline, while all drugs were applied to the bath. Spatio-temporal maps of colonic motility were constructed from video recordings of peristaltic contractions, and the maximum diameter was measured as an index of colonic contractility. GBR (1 μM)-induced dilatations of colonic segments were prevented by SCH (5 μM), L-nitro arginine (L-NA; 100 μM), a nitric oxide synthase inhibitor, or tetrodotoxin (0.6 μM). In contrast, constrictions induced by a higher concentration of SCH (20 μM) were unaffected by either L-NA or tetrodotoxin. The vasoactive intestinal peptide (VIP) receptor antagonist VIP10-28 (3 μM) or P2Y1 receptor antagonist MRS 2500 (1 μM) had no effect on either the GBR-induced dilatation or the SCH-induced constriction. In colonic segments that had been pretreated with 6-hydroxydopamine (100 μM, 3 h) to deplete enteric dopamine, GBR failed to increase the colonic diameter, while SCH was still capable of constricting colonic segments. Enteric dopaminergic neurons appear to project to nitrergic neurons to dilate the proximal colon by activating neuronal D1-like receptors. In addition, constitutively activated D1-like receptors expressed in cells yet to be determined may provide a tonic inhibition on colonic constrictions.
Collapse
Affiliation(s)
- Hiroyuki Nakamori
- Department of Cell Physiology, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-Cho, Mizuho-Ku, Nagoya, 467-8601, Japan.
| | - Hikaru Hashitani
- Department of Cell Physiology, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-Cho, Mizuho-Ku, Nagoya, 467-8601, Japan
| |
Collapse
|
5
|
Newly Synthesized Creatine Derivatives as Potential Neuroprotective and Antioxidant Agents on In Vitro Models of Parkinson's Disease. LIFE (BASEL, SWITZERLAND) 2023; 13:life13010139. [PMID: 36676090 PMCID: PMC9864416 DOI: 10.3390/life13010139] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 12/23/2022] [Accepted: 12/28/2022] [Indexed: 01/06/2023]
Abstract
Oxidative stress is one of the key factors responsible for many diseases-neurodegenerative (Parkinson and Alzheimer) diseases, diabetes, atherosclerosis, etc. Creatine, a natural amino acid derivative, is capable of exerting mild, direct antioxidant activity in cultured mammalian cells acutely injured with an array of different reactive oxygen species (ROS) generating compounds. The aim of the study was in vitro (on isolated rat brain sub-cellular fractions-synaptosomes, mitochondria and microsomes) evaluation of newly synthetized creatine derivatives for possible antioxidant and neuroprotective activity. The synaptosomes and mitochondria were obtained by multiple centrifugations with Percoll, while microsomes-only by multiple centrifugations. Varying models of oxidative stress were used to study the possible antioxidant and neuroprotective effects of the respective compounds: on synaptosomes-6-hydroxydopamine; on mitochondria-tert-butyl hydroperoxide; and on microsomes-iron/ascorbate (non-enzyme-induced lipid peroxidation). Administered alone, creatine derivatives and creatine (at concentration 38 µM) revealed neurotoxic and pro-oxidant effects on isolated rat brain subcellular fractions (synaptosomes, mitochondria and microsomes). In models of 6-hydroxydopamine (on synaptosomes), tert-butyl hydroperoxide (on mitochondria) and iron/ascorbate (on microsomes)-induced oxidative stress, the derivatives showed neuroprotective and antioxidant effects. These effects may be due to the preservation of the reduced glutathione level, ROS scavenging and membranes' stabilizers against free radicals. Thus, they play a role in the antioxidative defense system and have a promising potential as therapeutic neuroprotective agents for the treatment of neurodegenerative disorders, connected with oxidative stress, such as Parkinson's disease.
Collapse
|
6
|
Gonçalves DF, Duarte T, Foletto JVP, Senger LR, Vargas Brabosa N, Soares FAA, Dalla Corte CL. Mitochondrial function and cellular energy maintenance during aging in a Drosophila melanogaster model of Parkinson Disease. Mitochondrion 2022; 65:166-175. [PMID: 35787469 DOI: 10.1016/j.mito.2022.06.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 06/04/2022] [Accepted: 06/26/2022] [Indexed: 10/17/2022]
Abstract
Parkinson's disease (PD) is a common neurodegenerative disease characterized by movement disorders as well as loss of dopaminergic neurons. Moreover, genes affecting mitochondrial function, such as SNCA, Parkin, PINK1, DJ-1 and LRRK2, were demonstrated to be associated with PD and other neurodegenerative disease. Additionally, mitochondrial dysfunction and cellular energy imbalance are common markers found in PD. In this study, we used the pink1 null mutants of Drosophila melanogaster as a Parkinson's disease model to investigate how the energetic pathways and mitochondrial functions change during aging in a PD model. In our study, the loss of the pink1 gene decreased the survival percent and the decreased climbing index during aging in pink1-/- flies. Furthermore, there was an impairment in mitochondrial function demonstrated by a decrease in OXPHOS CI&CII-Linked and ETS CI&CII-Linked in pink1-/- flies at 3, 15 and 30 days of life. Interestingly, OXPHOS CII-Linked and ETS CII-Linked presented decreases only at 15 days of life in pink1-/- flies. Moreover, there was an increase in peroxide (H2O2) levels in pink1-/- flies at 15 and 30 days of life. Loss of the pink1 gene also decreased the activity of citrate synthase (CS) and increased the activity of lactate dehydrogenase (LDH) in pink1-/- flies head. Our results demonstrate a metabolic shift in ATP production in pink1-/- flies, which changed from oxidative to glycolytic pathways from 15 days of age, and is apparently more pronounced in the central nervous system.
Collapse
Affiliation(s)
- Débora F Gonçalves
- Universidade Federal de Santa Maria, Centro de Ciências Naturais e Exatas, Departamento de Bioquímica e Biologia Molecular, Programa de Pós-graduação em Ciências Biológicas: Bioquímica Toxicológica, Camobi, Zip code 97105-900, Santa Maria, RS, Brazil
| | - Tâmie Duarte
- Universidade Federal de Santa Maria, Centro de Ciências Naturais e Exatas, Departamento de Bioquímica e Biologia Molecular, Programa de Pós-graduação em Ciências Biológicas: Bioquímica Toxicológica, Camobi, Zip code 97105-900, Santa Maria, RS, Brazil
| | - João V P Foletto
- Universidade Federal de Santa Maria, Centro de Ciências Naturais e Exatas, Departamento de Bioquímica e Biologia Molecular, Programa de Pós-graduação em Ciências Biológicas: Bioquímica Toxicológica, Camobi, Zip code 97105-900, Santa Maria, RS, Brazil
| | - Leahn R Senger
- Universidade Federal de Santa Maria, Centro de Ciências Naturais e Exatas, Departamento de Bioquímica e Biologia Molecular, Programa de Pós-graduação em Ciências Biológicas: Bioquímica Toxicológica, Camobi, Zip code 97105-900, Santa Maria, RS, Brazil
| | - Nilda Vargas Brabosa
- Universidade Federal de Santa Maria, Centro de Ciências Naturais e Exatas, Departamento de Bioquímica e Biologia Molecular, Programa de Pós-graduação em Ciências Biológicas: Bioquímica Toxicológica, Camobi, Zip code 97105-900, Santa Maria, RS, Brazil
| | - Félix A A Soares
- Universidade Federal de Santa Maria, Centro de Ciências Naturais e Exatas, Departamento de Bioquímica e Biologia Molecular, Programa de Pós-graduação em Ciências Biológicas: Bioquímica Toxicológica, Camobi, Zip code 97105-900, Santa Maria, RS, Brazil
| | - Cristiane L Dalla Corte
- Universidade Federal de Santa Maria, Centro de Ciências Naturais e Exatas, Departamento de Bioquímica e Biologia Molecular, Programa de Pós-graduação em Ciências Biológicas: Bioquímica Toxicológica, Camobi, Zip code 97105-900, Santa Maria, RS, Brazil.
| |
Collapse
|
7
|
Duarte P, Michalska P, Crisman E, Cuadrado A, León R. Novel Series of Dual NRF2 Inducers and Selective MAO-B Inhibitors for the Treatment of Parkinson’s Disease. Antioxidants (Basel) 2022; 11:antiox11020247. [PMID: 35204129 PMCID: PMC8868346 DOI: 10.3390/antiox11020247] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 01/21/2022] [Accepted: 01/25/2022] [Indexed: 12/04/2022] Open
Abstract
Parkinson’s disease (PD) is the second most prevalent neurodegenerative disease. It is characterized by a complex network of physiopathological events where oxidative stress plays a central role among other factors such as neuroinflammation and protein homeostasis. Nuclear factor-erythroid 2 p45-related factor 2 (NRF2) has a multitarget profile itself as it controls a plethora of cellular processes involved in the progression of the disease. In this line, we designed a novel family of 2-(1H-indol-3-yl)ethan-1-amine derivatives as NRF2 inducers with complementary activities. Novel compounds are based on melatonin scaffold and include, among other properties, selective monoamine oxidase B (MAO-B) inhibition activity. Novel multitarget compounds exhibited NRF2 induction activity and MAO-B selective inhibition, combined with anti-inflammatory, antioxidant, and blood–brain barrier permeation properties. Furthermore, they exert neuroprotective properties against oxidative stress toxicity in PD-related in vitro. Hit compound 14 reduced oxidative stress markers and exerted neuroprotection in rat striatal slices exposed to 6-hydroxydopamine or rotenone. In conclusion, we developed a promising family of dual NRF2 inducers and selective MAO-B inhibitors that could serve as a novel therapeutic strategy for PD treatment.
Collapse
Affiliation(s)
- Pablo Duarte
- Instituto de Química Médica, Consejo Superior de Investigaciones Científicas (IQM-CSIC), 28006 Madrid, Spain; (P.D.); (E.C.)
- Instituto Teófilo Hernando y Departamento de Farmacología y Terapéutica, Facultad de Medicina, Universidad Autónoma de Madrid, 28029 Madrid, Spain
| | | | - Enrique Crisman
- Instituto de Química Médica, Consejo Superior de Investigaciones Científicas (IQM-CSIC), 28006 Madrid, Spain; (P.D.); (E.C.)
- Instituto Teófilo Hernando y Departamento de Farmacología y Terapéutica, Facultad de Medicina, Universidad Autónoma de Madrid, 28029 Madrid, Spain
- Instituto de Investigación Sanitaria La Princesa (IIS-IS), Hospital Universitario de la Princesa, 28006 Madrid, Spain
| | - Antonio Cuadrado
- Departamento de Bioquímica, Facultad de Medicina, Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Investigación Sanitaria La Paz (IdiPaz), Instituto de Investigaciones Biomédicas ‘Alberto Sols’ UAM-CSIC, Universidad Autónoma de Madrid, 28029 Madrid, Spain;
| | - Rafael León
- Instituto de Química Médica, Consejo Superior de Investigaciones Científicas (IQM-CSIC), 28006 Madrid, Spain; (P.D.); (E.C.)
- Correspondence:
| |
Collapse
|
8
|
Wąsik A, Białoń M, Jantas D, Żarnowska M. The Impact of the Combined Administration of 1MeTIQ and MK-801 on Cell Viability, Oxidative Stress Markers, and Glutamate Release in the Rat Hippocampus. Neurotox Res 2021; 39:1747-1761. [PMID: 34665405 PMCID: PMC8639582 DOI: 10.1007/s12640-021-00428-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 10/01/2021] [Accepted: 10/04/2021] [Indexed: 12/18/2022]
Abstract
MK-801, as an N-methyl-D-aspartate (NMDA) receptor inhibitor, causes elevation in glutamate release, which may lead to an increase in excitotoxicity, oxidative stress and, consequently, cell death. 1-Methyl-1,2,3,4-tetrahydroisoquinoline (1MeTIQ) shows antioxidant activity. The aim of the present study was to evaluate the effect of combined treatment with 1MeTIQ and MK-801 on cell viability, antioxidant enzyme activity, and glutamate release in the rat hippocampus. Cytotoxicity was measured using lactate dehydrogenase leakage assay (LDH) and the methyl tetrazolium (MTT) assay; antioxidant enzyme activity (glutathione peroxidase (GPx), glutathione reductase (GR), superoxide dismutase (SOD), and catalase (CAT)) were measured by ELISA kits. The release of glutamate in the rat hippocampus was measured using in vivo microdialysis methodology. An in vitro study showed that MK-801 induced cell death in a concentration-dependent manner and that 1MeTIQ partially reduced this adverse effect of MK-801. An ex vivo study indicated that MK-801 produced an increase in antioxidant enzyme activity (GPx, GR, and SOD), whereas coadministration of MK-801 and 1MeTIQ restored the activity of these enzymes to the control level. An in vivo microdialysis study demonstrated that combined treatment with both drugs decreased the release of glutamate in the rat hippocampus. The above results revealed that 1MeTIQ shows limited neuroprotective activity under conditions of glutamate-induced neurotoxicity.
Collapse
Affiliation(s)
- Agnieszka Wąsik
- Department of Neurochemistry, Maj Institute of Pharmacology PAS, Krakow, Poland.
| | - Magdalena Białoń
- Department of Neurochemistry, Maj Institute of Pharmacology PAS, Krakow, Poland
| | - Danuta Jantas
- Department of Experimental Neuroendocrinology, Maj Institute of Pharmacology PAS, Krakow, Poland
| | - Marcelina Żarnowska
- Department of Neurochemistry, Maj Institute of Pharmacology PAS, Krakow, Poland
| |
Collapse
|
9
|
Neuroprotective effect of bromelain in 6-hydroxydopamine induced in vitro model of Parkinson's disease. Mol Biol Rep 2021; 48:7711-7717. [PMID: 34643923 DOI: 10.1007/s11033-021-06779-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 09/20/2021] [Indexed: 10/20/2022]
Abstract
BACKGROUND This study was designed to investigate the neuroprotective effects of bromelain, which is known to have anti-oxidant and anti-inflammatory properties, against the neurotoxicity (induced by 6-OHDA) in SH-SY5Y cells. METHODS AND RESULTS To establish Parkinson's Disease (PD) model in cell culture conditions, SH-SY5Y cells were exposed to 200 µM 6-OHDA for 1 day. Prior to 6-OHDA treatment, SH-SY5Y cells had been pre-treated with bromelain (25 µg/mL, 50 µg/mL, 75 µg/mL and 100 µg/mL). After 1 day, cell viability was determined with the 3-(4,5-Dimethylthiazol-2-yl)-2,5-Diphenyltetrazolium Bromide (MTT) and lactate dehydrogenase (LDH) assays. Oxidative stress was assessed with total antioxidant capacity (TAC), total oxidant status (TOS), glutathione reductase (GR) and malondialdehyde (MDA) analyses. The effect of the bromelain in SH-SY5Ycells was also examined by 4',6-diamidino-2-phenylindole (DAPI) staining. We found that 6-OHDA increased LDH leakage, and cellular apoptosis in SH-SY5Y cells. 6-OHDA aggravated oxidative stress by increasing TOS, MDA and GR and eventually promoted apoptosis in SH-SY5Y cells, while pretreatment with bromelain attenuated these toxic effects of 6-OHDA. CONCLUSIONS These findings indicated that bromelain, with its neuroprotective features can be useful for neuroprotection in PD.
Collapse
|
10
|
Massari CM, Constantino LC, Tasca CI. Adenosine A 1 and A 2A receptors are involved on guanosine protective effects against oxidative burst and mitochondrial dysfunction induced by 6-OHDA in striatal slices. Purinergic Signal 2021; 17:247-254. [PMID: 33548045 PMCID: PMC8155135 DOI: 10.1007/s11302-021-09765-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 01/17/2021] [Indexed: 12/13/2022] Open
Abstract
6-Hydroxydopamine (6-OHDA) is the most used toxin in experimental Parkinson's disease (PD) models. 6-OHDA shows high affinity for the dopamine transporter and once inside the neuron, it accumulates and undergoes non-enzymatic auto-oxidation, promoting reactive oxygen species (ROS) formation and selective damage of catecholaminergic neurons. In this way, our group has established a 6-OHDA in vitro protocol with rat striatal slices as a rapid and effective model for screening of new drugs with protective effects against PD. We have shown that co-incubation with guanosine (GUO, 100 μM) prevented the 6-OHDA-induced damage in striatal slices. As the exact GUO mechanism of action remains unknown, the aim of this study was to investigate if adenosine A1 (A1R) and/or A2A receptors (A2AR) are involved on GUO protective effects on striatal slices. Pre-incubation with DPCPX, an A1R antagonist prevented guanosine effects on 6-OHDA-induced ROS formation and mitochondrial membrane potential depolarization, while CCPA, an A1R agonist, did not alter GUO effects. Regarding A2AR, the antagonist SCH58261 had similar protective effect as GUO in ROS formation and mitochondrial membrane potential. Additionally, SCH58261 did not affect GUO protective effects. The A2AR agonist CGS21680, although, completely blocked GUO effects. Finally, the A1R antagonist DPCPX, and the A2AR agonist CGS21680 also abolished the preventive guanosine effect on 6-OHDA-induced ATP levels decrease. These results reinforce previous evidence for a putative interaction of GUO with A1R-A2AR heteromer as its molecular target and clearly indicate a dependence on adenosine receptors modulation to GUO protective effect.
Collapse
Affiliation(s)
- C M Massari
- Programa de Pós-graduação em Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, Brazil
| | - L C Constantino
- Programa de Pós-graduação em Neurociências, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, Brazil
| | - C I Tasca
- Programa de Pós-graduação em Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, Brazil.
- Programa de Pós-graduação em Neurociências, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, Brazil.
- Laboratório de Neuroquímica-4, Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, Brazil.
| |
Collapse
|
11
|
Hadi F, Akrami H, Totonchi M, Barzegar A, Nabavi SM, Shahpasand K. α-synuclein abnormalities trigger focal tau pathology, spreading to various brain areas in Parkinson disease. J Neurochem 2021; 157:727-751. [PMID: 33264426 DOI: 10.1111/jnc.15257] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Revised: 07/28/2020] [Accepted: 11/30/2020] [Indexed: 12/21/2022]
Abstract
Parkinson disease (PD) is the second most common neurodegenerative disorder, whose prevalence is 2~3% in the population over 65. α-Synuclein aggregation is the major pathological hallmark of PD. However, recent studies have demonstrated enhancing evidence of tau pathology in PD. Despite extensive considerations, thus far, the actual spreading mechanism of neurodegeneration has remained elusive in a PD brain. This study aimed to further investigate the development of α-synuclein and tau pathology. We employed various PD models, including cultured neurons treated with either 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) or with recombinant α-synuclein. Also, we studied dopaminergic neurons of cytokine Interferon-β knock-out. Moreover, we examined rats treated with 6-hydroxydopamine, Rhesus monkeys administrated with MPTP neurotoxin, and finally, human post-mortem brains. We found the α-synuclein phosphorylation triggers tau pathogenicity. Also, we observed more widespread phosphorylated tau than α-synuclein with prion-like nature in various brain areas. We optionally removed P-tau or P-α-synuclein from cytokine interferon-β knock out with respective monoclonal antibodies. We found that tau immunotherapy suppressed neurodegeneration more than α-synuclein elimination. Our findings indicate that the pathogenic tau could be one of the leading causes of comprehensive neurodegeneration triggered by PD. Thus, we can propose an efficient therapeutic target to fight the devastating disorder.
Collapse
Affiliation(s)
- Fatemeh Hadi
- Department of Biology, Faculty of Science, Razi University, Kermanshah, Iran
| | - Hassan Akrami
- Department of Biology, Faculty of Science, Razi University, Kermanshah, Iran
| | - Mehdi Totonchi
- Department of Brain and Cognitive Sciences, Cell Science Research Center, Royan Institute, ACECR, Tehran, Iran
| | | | - Seyed Massood Nabavi
- Department of Brain and Cognitive Sciences, Cell Science Research Center, Royan Institute, ACECR, Tehran, Iran
| | - Koorosh Shahpasand
- Department of Brain and Cognitive Sciences, Cell Science Research Center, Royan Institute, ACECR, Tehran, Iran
| |
Collapse
|
12
|
Evaluation of the neuroprotective potential of caffeic acid phenethyl ester in a cellular model of Parkinson's disease. Eur J Pharmacol 2020; 883:173342. [PMID: 32634439 DOI: 10.1016/j.ejphar.2020.173342] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 06/26/2020] [Accepted: 06/29/2020] [Indexed: 01/23/2023]
Abstract
Parkinson's disease (PD) is the second most prevalent neurodegenerative disease, and oxidative stress and mitochondrial dysfunction play a major role in the pathogenesis of PD. Since conventional therapeutics are not sufficient for the treatment of PD, the development of new agents with anti-oxidant potential is crucial. Caffeic Acid Phenethyl Ester (CAPE), a biologically active flavonoid of propolis, possesses several biological properties such as immunomodulatory, anti-inflammatory and anti-oxidative. In the present study, we investigated the neuroprotective effects of CAPE against 6-hydroxydopamine (6-OHDA)-induced SH-SY5Y cells. The neuroprotective effects were detected by using cell viability, Annexin V, Hoechst staining, total caspase activity, cell cycle, as well as western blotting. Besides, the anti-oxidative activity was measured by the production of reactive oxygen species and mitochondrial function was determined by measurement of mitochondrial membrane potential (ΔΨm). We found that CAPE significantly increased cell viability and decreased apoptotic cell death (~20%) after 150 μM 6-OHDA exposure following 24 h. 1.25 μM CAPE also prevented 6-OHDA-induced changes in condensed nuclear morphology. Furthermore, treatment with 1.25 μM CAPE increased mitochondrial membrane potential in 6-OHDA-exposed cells. CAPE inhibited 6-OHDA-induced caspase activity (~2 fold) and production of reactive oxygen species. In addition, 150 μM 6-OHDA-induced down-regulation of Bcl-2 and Akt levels and up-regulation of Bax and cleaved caspase-9/caspase-9 levels were partially restored by 1.25 μM CAPE treatment. These results revealed a neuroprotective potential of CAPE against 6-OHDA-induced apoptosis in an in vitro PD model and may be a potential therapeutic candidate for the prevention of neurodegeneration in Parkinson's Disease.
Collapse
|
13
|
Iglesias González PA, Conde MA, González-Pardo V, Uranga RM, Salvador GA. In vitro 6-hydroxydopamine-induced neurotoxicity: New insights on NFκB modulation. Toxicol In Vitro 2019; 60:400-411. [DOI: 10.1016/j.tiv.2019.06.019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 06/21/2019] [Accepted: 06/23/2019] [Indexed: 10/26/2022]
|
14
|
Marques NF, Binder LB, Roversi K, Sampaio TB, Constantino LC, Prediger RD, Tasca CI. Guanosine prevents depressive-like behaviors in rats following bilateral dorsolateral striatum lesion induced by 6-hydroxydopamine. Behav Brain Res 2019; 372:112014. [DOI: 10.1016/j.bbr.2019.112014] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 05/28/2019] [Accepted: 06/04/2019] [Indexed: 10/26/2022]
|
15
|
N-acetyl-cysteine blunts 6-hydroxydopamine- and l-buthionine-sulfoximine-induced apoptosis in human mesenchymal stromal cells. Mol Biol Rep 2019; 46:4423-4435. [DOI: 10.1007/s11033-019-04897-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Accepted: 05/24/2019] [Indexed: 02/07/2023]
|
16
|
Yi NX, Zhou LY, Wang XY, Song YJ, Han HH, Zhang TS, Wang YJ, Shi Q, Xu H, Liang QQ, Zhang T. MK-801 attenuates lesion expansion following acute brain injury in rats: a meta-analysis. Neural Regen Res 2019; 14:1919-1931. [PMID: 31290450 PMCID: PMC6676887 DOI: 10.4103/1673-5374.259619] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
OBJECTIVE: To evaluate the efficacy and safety of MK-801 and its effect on lesion volume in rat models of acute brain injury. DATA SOURCES: Key terms were “stroke”, “brain diseases”, “brain injuries”, “brain hemorrhage, traumatic”, “acute brain injury”, “dizocilpine maleate”, “dizocilpine”, “MK-801”, “MK801”, “rat”, “rats”, “rattus” and “murine”. PubMed, Cochrane library, EMBASE, the China National Knowledge Infrastructure, WanFang database, the VIP Journal Integration Platform (VJIP) and SinoMed databases were searched from their inception dates to March 2018. DATA SELECTION: Studies were selected if they reported the effects of MK-801 in experimental acute brain injury. Two investigators independently conducted literature screening, data extraction, and methodological quality assessments. OUTCOME MEASURES: The primary outcomes included lesion volume and brain edema. The secondary outcomes included behavioral assessments with the Bederson neurological grading system and the water maze test 24 hours after brain injury. RESULTS: A total of 52 studies with 2530 samples were included in the systematic review. Seventeen of these studies had a high methodological quality. Overall, the lesion volume (34 studies, n = 966, MD = −58.31, 95% CI: −66.55 to −50.07; P < 0.00001) and degree of cerebral edema (5 studies, n = 75, MD = −1.21, 95% CI: −1.50 to −0.91; P < 0.00001) were significantly decreased in the MK-801 group compared with the control group. MK-801 improved spatial cognition assessed with the water maze test (2 studies, n = 60, MD = −10.88, 95% CI: −20.75 to −1.00; P = 0.03) and neurological function 24 hours after brain injury (11 studies, n = 335, MD = −1.04, 95% CI: −1.47 to −0.60; P < 0.00001). Subgroup analysis suggested an association of reduction in lesion volume with various injury models (34 studies, n = 966, MD = −58.31, 95% CI: −66.55 to −50.07; P = 0.004). Further network analysis showed that 0–1 mg/kg MK-801 may be the optimal dose for treatment in the middle cerebral artery occlusion animal model. CONCLUSION: MK-801 effectively reduces brain lesion volume and the degree of cerebral edema in rat models of experimental acute brain injury, providing a good neuroprotective effect. Additionally, MK-801 has a good safety profile, and its mechanism of action is well known. Thus, MK-801 may be suitable for future clinical trials and applications.
Collapse
Affiliation(s)
- Nan-Xing Yi
- Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine; Institute of Spine, Shanghai University of Traditional Chinese Medicine; Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education, Shanghai, China
| | - Long-Yun Zhou
- Institute of Spine, Shanghai University of Traditional Chinese Medicine; Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education; School of Rehabilitation Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiao-Yun Wang
- Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yong-Jia Song
- Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine; Institute of Spine, Shanghai University of Traditional Chinese Medicine; Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education, Shanghai, China
| | - Hai-Hui Han
- Institute of Spine; Guanghua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Tian-Song Zhang
- Jing'an District Center Hospital, Fudan University, Shanghai, China
| | - Yong-Jun Wang
- Institute of Spine, Shanghai University of Traditional Chinese Medicine; Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education, Shanghai, China
| | - Qi Shi
- Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine; Institute of Spine, Shanghai University of Traditional Chinese Medicine; Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education; Guanghua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hao Xu
- Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine; Institute of Spine, Shanghai University of Traditional Chinese Medicine; Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education, Shanghai, China
| | - Qian-Qian Liang
- Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine; Institute of Spine, Shanghai University of Traditional Chinese Medicine; Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education, Shanghai, China
| | - Ting Zhang
- Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine; Institute of Spine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
17
|
Marques NF, Massari CM, Tasca CI. Guanosine Protects Striatal Slices Against 6-OHDA-Induced Oxidative Damage, Mitochondrial Dysfunction, and ATP Depletion. Neurotox Res 2018; 35:475-483. [PMID: 30417317 DOI: 10.1007/s12640-018-9976-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 10/26/2018] [Accepted: 10/30/2018] [Indexed: 01/18/2023]
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disorder characterized by loss of dopaminergic neurons in substantia nigra pars compacta which induces severe motor symptoms. 6-OHDA is a neurotoxin widely used in PD animal models due to its high affinity by dopamine transporter, its rapid non-enzymatic auto-oxidation which generates reactive oxygen species (ROS), oxidative stress, and for induced mitochondrial dysfunction. We previously reported an in vitro protocol of 6-OHDA-induced toxicity in brain regions slices, as a simple and sensitive assay to screen for protective compounds related to PD. Guanosine (GUO), a guanine-based purine nucleoside, is a neuroprotective molecule that is showing promising effects as an antiparkinsonian agent. To investigate the mechanisms involved on GUO-induced neuroprotection, slices of cortex, striatum, and hippocampus were incubated with GUO in the presence of 6-OHDA (100 μM). 6-OHDA promoted a decrease in cellular viability and increased ROS generation in all brain regions. Disruption of mitochondrial potential, depletion in intracellular ATP levels, and increase in cell membrane permeabilization were evidenced in striatal slices. GUO prevented the increase in ROS generation, disruption in mitochondrial potential, and depletion of intracellular ATP induced by 6-OHDA in striatal slices. In conclusion, GUO was effective to prevent oxidative events before cell damage, such as mitochondrial disruption, intracellular ATP levels depletion, and ROS generation in striatal slices subjected to in vitro 6-OHDA-induced toxicity.
Collapse
Affiliation(s)
- Naiani Ferreira Marques
- Programa de Pós-Graduação em Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, Santa Catarina, Brazil
| | - Caio Marcos Massari
- Programa de Pós-Graduação em Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, Santa Catarina, Brazil
| | - Carla Inês Tasca
- Programa de Pós-Graduação em Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, Santa Catarina, Brazil. .,Departamento de Bioquímica, CCB, UFSC, Trindade, Florianópolis, Santa Catarina, 88040-900, Brazil.
| |
Collapse
|
18
|
Gonçalves DF, Courtes AA, Hartmann DD, da Rosa PC, Oliveira DM, Soares FAA, Dalla Corte CL. 6-Hydroxydopamine induces different mitochondrial bioenergetics response in brain regions of rat. Neurotoxicology 2018; 70:1-11. [PMID: 30359634 DOI: 10.1016/j.neuro.2018.10.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Revised: 10/07/2018] [Accepted: 10/10/2018] [Indexed: 11/30/2022]
Abstract
Mitochondrial dysfunction has been demonstrated to have a central role in Parkinson Disease (PD) pathophysiology. Some studies have indicated that PD causes an impairment in mitochondrial bioenergetics; however, the effects of PD on brain-region specific bioenergetics was never investigated before. This study aimed to evaluate mitochondrial bioenergetics in different rat brain structures in an in vitro model of PD using 6-OHDA. Rat brain slices of hippocampus, striatum, and cortex were exposed to 6-OHDA (100 μM) for 1 h and mitochondrial bioenergetic parameters, peroxide production, lactate dehydrogenase (LDH) and citrate synthase (CS) activities were analyzed. Hippocampus slices exposed to 6-OHDA presented increased peroxide production but, no mitochondrial adaptive response against 6-OHDA damage. Cortex slices exposed to 6-OHDA presented increased oxygen flux related to oxidative phosphorylation and energetic pathways exchange demonstrated by the increase in LDH activity, suggesting a mitochondrial compensatory response. Striatum slices exposed to 6-OHDA presented a decrease of oxidative phosphorylation and decrease of oxygen flux related to ATP-synthase indicating an impairment in the respiratory chain. The co-incubation of 6-OHDA with n-acetylcysteine (NAC) abolished the effects of 6-OHDA on mitochondrial function in all brain regions tested, indicating that the increased reactive oxygen species (ROS) production is responsible for the alterations observed in mitochondrial bioenergetics. The present results indicate a brain-region specific response against 6-OHDA, providing new insights into brain mitochondrial bioenergetic function in PD. These findings may contribute to the development of future therapies with a target on energy metabolism.
Collapse
Affiliation(s)
- Débora F Gonçalves
- Universidade Federal de Santa Maria, Centro de Ciências Naturais e Exatas, Departamento de Bioquímica e Biologia Molecular, Programa de Pós-graduação em Ciências Biológicas: Bioquímica Toxicológica, Camobi, 97105-900, Santa Maria, RS, Brazil
| | - Aline A Courtes
- Universidade Federal de Santa Maria, Centro de Ciências Naturais e Exatas, Departamento de Bioquímica e Biologia Molecular, Programa de Pós-graduação em Ciências Biológicas: Bioquímica Toxicológica, Camobi, 97105-900, Santa Maria, RS, Brazil
| | - Diane D Hartmann
- Universidade Federal de Santa Maria, Centro de Ciências Naturais e Exatas, Departamento de Bioquímica e Biologia Molecular, Programa de Pós-graduação em Ciências Biológicas: Bioquímica Toxicológica, Camobi, 97105-900, Santa Maria, RS, Brazil
| | - Pamela C da Rosa
- Universidade Federal de Santa Maria, Centro de Ciências Naturais e Exatas, Departamento de Bioquímica e Biologia Molecular, Programa de Pós-graduação em Ciências Biológicas: Bioquímica Toxicológica, Camobi, 97105-900, Santa Maria, RS, Brazil
| | - Débora M Oliveira
- Universidade Federal de Santa Maria, Centro de Ciências Naturais e Exatas, Departamento de Bioquímica e Biologia Molecular, Programa de Pós-graduação em Ciências Biológicas: Bioquímica Toxicológica, Camobi, 97105-900, Santa Maria, RS, Brazil
| | - Félix A A Soares
- Universidade Federal de Santa Maria, Centro de Ciências Naturais e Exatas, Departamento de Bioquímica e Biologia Molecular, Programa de Pós-graduação em Ciências Biológicas: Bioquímica Toxicológica, Camobi, 97105-900, Santa Maria, RS, Brazil
| | - Cristiane L Dalla Corte
- Universidade Federal de Santa Maria, Centro de Ciências Naturais e Exatas, Departamento de Bioquímica e Biologia Molecular, Programa de Pós-graduação em Ciências Biológicas: Bioquímica Toxicológica, Camobi, 97105-900, Santa Maria, RS, Brazil; Universidade Federal do Pampa - Campus Caçapava do Sul, 96570-000, Caçapava do Sul, RS, Brazil.
| |
Collapse
|
19
|
Thirtamara-Rajamani K, Li P, Escobar Galvis ML, Labrie V, Brundin P, Brundin L. Is the Enzyme ACMSD a Novel Therapeutic Target in Parkinson's Disease? JOURNAL OF PARKINSON'S DISEASE 2017; 7:577-587. [PMID: 29103054 PMCID: PMC5676848 DOI: 10.3233/jpd-171240] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Accepted: 10/13/2017] [Indexed: 02/07/2023]
Abstract
Several large genome wide association studies have identified a locus in close proximity to the gene encoding the enzyme aminocarboxymuconate-semialdehyde-decarboxylase (ACMSD) to be associated with the risk for Parkinson's disease (PD), tentatively suggesting that this enzyme might influence PD pathogenesis. Further support for this comes from the recent identification of a disease-segregating stop codon mutation in ACMSD in a family with Parkinsonism, and a missense mutation in the ACMSD gene predicted to disrupt enzyme function in an individual with typical PD. ACMSD is part of the kynurenine pathway, responsible for the catalytic breakdown of tryptophan into NAD+, generating several neuroactive metabolites in the process. The enzyme is located at a key branch-point of the pathway, limiting the production of the neurotoxin quinolinic acid, which has excitotoxic and inflammatory properties. In this review, we discuss the genetic findings in light of the functions of ACMSD and its potential involvement in PD pathogenesis.
Collapse
Affiliation(s)
| | - Peipei Li
- Center for Neurodegenerative Science, Van Andel Research Institute, Grand Rapids, MI, USA
| | | | - Viviane Labrie
- Center for Neurodegenerative Science, Van Andel Research Institute, Grand Rapids, MI, USA
| | - Patrik Brundin
- Center for Neurodegenerative Science, Van Andel Research Institute, Grand Rapids, MI, USA
| | - Lena Brundin
- Center for Neurodegenerative Science, Van Andel Research Institute, Grand Rapids, MI, USA
| |
Collapse
|