1
|
Lee EJ, Choy YJ, Woo RS, Baik TK, Yoo HI, Song DY. Expression of Galectin 3 and Activating Transcription Factor 3 in Nigral Dopaminergic Neurons of 6-Hydroxydopamine Induced Parkinsonian Rat Model. In Vivo 2025; 39:1341-1354. [PMID: 40295017 PMCID: PMC12041963 DOI: 10.21873/invivo.13938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Revised: 01/11/2025] [Accepted: 01/17/2025] [Indexed: 04/30/2025]
Abstract
BACKGROUND/AIM Parkinson's disease (PD) is an age-related neurodegenerative disease marked by the relatively progressive dopaminergic neuronal loss in the substantia nigra (SN). Retrograde degeneration of the nigrostriatal dopaminergic neurons by 6-hydroxydopamine (6-OHDA) has been widely used as a PD animal model, while endogenous 6-OHDA promotes the progression of PD pathology. Galectin 3 (Gal3) and activating transcription factor 3 (ATF3) have been implicated in neurodegenerative processes. The aim of this study was to investigate the expression pattern and roles of Gal3 and ATF3 in a Parkinson's disease animal model induced by 6-OHDA. MATERIALS AND METHODS We investigated temporal and spatial profiles of Gal3 expression in 6-OHDA rat model of PD. Lesions were induced by unilateral stereotactic injections of 6-OHDA into the striatum. Three days prior to 6-OHDA lesion, Fluorogold (FG) was injected at the same coordinates as the subsequent 6-OHDA injection. 6-OHDA induced retrograde degeneration of tyrosine hydroxylase immunopositive and FG immunopositive neurons within SN in a time-dependent manner. RESULTS Activating transcription factor 3 (ATF3) expression was also upregulated in the SN, in a pattern similar to that of Gal3 immunoreactivity. Finally, we confirmed through triple immunofluorescence staining that ATF3 and Gal3 were colocalized in the dopaminergic neurons labeled with FG. These neurons were damaged by 6-OHDA. CONCLUSION Gal3 may play a key role in the signaling pathway of dopaminergic neuronal cell death induced by 6-OHDA. This is the first in vivo demonstration that Gal3 is expressed in dopaminergic neurons injured by 6-OHDA.
Collapse
Affiliation(s)
- Eun-Jin Lee
- Department of Anatomy and Neurosciences, Eulji University School of Medicine, Daejeon, Republic of Korea
| | - Yoon-Jung Choy
- Department of Optometry, Eulji University, Seongnam, Republic of Korea
| | - Ran-Sook Woo
- Department of Anatomy and Neurosciences, Eulji University School of Medicine, Daejeon, Republic of Korea
| | - Tai-Kyoung Baik
- Department of Anatomy and Neurosciences, Eulji University School of Medicine, Daejeon, Republic of Korea
| | - Hong-Il Yoo
- Department of Anatomy and Neurosciences, Eulji University School of Medicine, Daejeon, Republic of Korea;
| | - Dae-Yong Song
- Department of Anatomy and Neurosciences, Eulji University School of Medicine, Daejeon, Republic of Korea;
| |
Collapse
|
2
|
Saponjic J, Mejías R, Nikolovski N, Dragic M, Canak A, Papoutsopoulou S, Gürsoy-Özdemir Y, Fladmark KE, Ntavaroukas P, Bayar Muluk N, Zeljkovic Jovanovic M, Fontán-Lozano Á, Comi C, Marino F. Experimental Models to Study Immune Dysfunction in the Pathogenesis of Parkinson's Disease. Int J Mol Sci 2024; 25:4330. [PMID: 38673915 PMCID: PMC11050170 DOI: 10.3390/ijms25084330] [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: 01/31/2024] [Revised: 03/25/2024] [Accepted: 03/26/2024] [Indexed: 04/28/2024] Open
Abstract
Parkinson's disease (PD) is a chronic, age-related, progressive multisystem disease associated with neuroinflammation and immune dysfunction. This review discusses the methodological approaches used to study the changes in central and peripheral immunity in PD, the advantages and limitations of the techniques, and their applicability to humans. Although a single animal model cannot replicate all pathological features of the human disease, neuroinflammation is present in most animal models of PD and plays a critical role in understanding the involvement of the immune system (IS) in the pathogenesis of PD. The IS and its interactions with different cell types in the central nervous system (CNS) play an important role in the pathogenesis of PD. Even though culture models do not fully reflect the complexity of disease progression, they are limited in their ability to mimic long-term effects and need validation through in vivo studies. They are an indispensable tool for understanding the interplay between the IS and the pathogenesis of this disease. Understanding the immune-mediated mechanisms may lead to potential therapeutic targets for the treatment of PD. We believe that the development of methodological guidelines for experiments with animal models and PD patients is crucial to ensure the validity and consistency of the results.
Collapse
Affiliation(s)
- Jasna Saponjic
- Department of Neurobiology, Institute of Biological Research “Sinisa Stankovic”, National Institute of the Republic of Serbia, University of Belgrade, 11108 Belgrade, Serbia
| | - Rebeca Mejías
- Department of Physiology, School of Biology, University of Seville, 41012 Seville, Spain; (R.M.); (Á.F.-L.)
- Instituto de Biomedicina de Sevilla, IBiS, Hospital Universitario Virgen del Rocío, CSIC, Universidad de Sevilla, 41013 Seville, Spain
| | - Neda Nikolovski
- Department of Immunology, Institute for Biological Research “Siniša Stanković”, National Institute of the Republic of Serbia, University of Belgrade, 11108 Belgrade, Serbia;
| | - Milorad Dragic
- Laboratory for Neurobiology, Department for General Physiology and Biophysics, Faculty of Biology, University of Belgrade, 11000 Belgrade, Serbia; (M.D.); (M.Z.J.)
- Department of Molecular Biology and Endocrinology, Vinca Institute of Nuclear Sciences–National Institute of the Republic of Serbia, University of Belgrade, 11351 Belgrade, Serbia
| | - Asuman Canak
- Department of Medical Services and Techniques, Vocational School of Health Services, Recep Tayyip Erdogan University, Rize 53100, Turkey;
| | - Stamatia Papoutsopoulou
- Department of Biochemistry and Biotechnology, Faculty of Health Sciences, University of Thessaly, Biopolis, 41500 Larisa, Greece; (S.P.); (P.N.)
| | | | - Kari E. Fladmark
- Department of Biological Science, University of Bergen, 5020 Bergen, Norway;
| | - Panagiotis Ntavaroukas
- Department of Biochemistry and Biotechnology, Faculty of Health Sciences, University of Thessaly, Biopolis, 41500 Larisa, Greece; (S.P.); (P.N.)
| | - Nuray Bayar Muluk
- Department of Otorhinolaryngology, Faculty of Medicine, Kirikkale University, Kirikkale 71450, Turkey;
| | - Milica Zeljkovic Jovanovic
- Laboratory for Neurobiology, Department for General Physiology and Biophysics, Faculty of Biology, University of Belgrade, 11000 Belgrade, Serbia; (M.D.); (M.Z.J.)
| | - Ángela Fontán-Lozano
- Department of Physiology, School of Biology, University of Seville, 41012 Seville, Spain; (R.M.); (Á.F.-L.)
- Instituto de Biomedicina de Sevilla, IBiS, Hospital Universitario Virgen del Rocío, CSIC, Universidad de Sevilla, 41013 Seville, Spain
| | - Cristoforo Comi
- Neurology Unit, Department of Translational Medicine, S. Andrea Hospital, University of Piemonte Orientale, 13100 Vercelli, Italy;
| | - Franca Marino
- Center for Research in Medical Pharmacology, School of Medicine, University of Insubria, 21100 Varese, Italy;
| |
Collapse
|
3
|
Cordeiro Y, Freire MHO, Wiecikowski AF, do Amaral MJ. (Dys)functional insights into nucleic acids and RNA-binding proteins modulation of the prion protein and α-synuclein phase separation. Biophys Rev 2023; 15:577-589. [PMID: 37681103 PMCID: PMC10480379 DOI: 10.1007/s12551-023-01067-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 05/22/2023] [Indexed: 09/09/2023] Open
Abstract
Prion diseases are prototype of infectious diseases transmitted by a protein, the prion protein (PrP), and are still not understandable at the molecular level. Heterogenous species of aggregated PrP can be generated from its monomer. α-synuclein (αSyn), related to Parkinson's disease, has also shown a prion-like pathogenic character, and likewise PrP interacts with nucleic acids (NAs), which in turn modulate their aggregation. Recently, our group and others have characterized that NAs and/or RNA-binding proteins (RBPs) modulate recombinant PrP and/or αSyn condensates formation, and uncontrolled condensation might precede pathological aggregation. Tackling abnormal phase separation of neurodegenerative disease-related proteins has been proposed as a promising therapeutic target. Therefore, understanding the mechanism by which polyanions, like NAs, modulate phase transitions intracellularly, is key to assess their role on toxicity promotion and neuronal death. Herein we discuss data on the nucleic acids binding properties and phase separation ability of PrP and αSyn with a special focus on their modulation by NAs and RBPs. Furthermore, we provide insights into condensation of PrP and/or αSyn in the light of non-trivial subcellular locations such as the nuclear and cytosolic environments.
Collapse
Affiliation(s)
- Yraima Cordeiro
- Faculty of Pharmacy, Universidade Federal do Rio de Janeiro, Av Carlos Chagas Filho 373, bloco B, subsolo Sala 36, Rio de Janeiro, RJ 21941-902 Brazil
| | - Maria Heloisa O. Freire
- Faculty of Pharmacy, Universidade Federal do Rio de Janeiro, Av Carlos Chagas Filho 373, bloco B, subsolo Sala 36, Rio de Janeiro, RJ 21941-902 Brazil
| | - Adalgisa Felippe Wiecikowski
- Faculty of Pharmacy, Universidade Federal do Rio de Janeiro, Av Carlos Chagas Filho 373, bloco B, subsolo Sala 36, Rio de Janeiro, RJ 21941-902 Brazil
| | - Mariana Juliani do Amaral
- Faculty of Pharmacy, Universidade Federal do Rio de Janeiro, Av Carlos Chagas Filho 373, bloco B, subsolo Sala 36, Rio de Janeiro, RJ 21941-902 Brazil
| |
Collapse
|
4
|
Tiwari S, Singh A, Gupta P, K A, Singh S. UBA52 Attunes VDAC1-Mediated Mitochondrial Dysfunction and Dopaminergic Neuronal Death. ACS Chem Neurosci 2023; 14:839-850. [PMID: 36755387 DOI: 10.1021/acschemneuro.2c00579] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2023] Open
Abstract
Mitochondrial homeostasis regulates energy metabolism, calcium buffering, cell function, and apoptosis. The present study has been conducted to investigate the implications of the ubiquitin-encoding gene UBA52 in mitochondrial physiology. Transient expression of Myc-UBA52 in neurons significantly inhibited the rotenone-induced increase in reactive oxygen species generation, nitrite level, and depleted glutathione level. Mass spectrometric and coimmunoprecipitation data suggested the profound interaction of UBA52 with mitochondrial outer membrane channel protein, VDAC1 in both the wild-type and Myc-α-synuclein overexpressed neuronal cells and in the Parkinson's disease (PD)-specific substantia nigra and striatal region of the rat brain. In vitro ubiquitylation assay revealed that UBA52 participates in the ubiquitylation of VDAC1 through E3 ligase CHIP. Myc-UBA52 overexpression in neurons further improved the mitochondrial functionality and cell viability by preventing the alteration in mitochondrial membrane potential, mitochondrial complex I activity, and translocation of cytochrome c and p-Nrf2 along with the effect on intracellular calcium uptake, thus collectively inhibiting the opening of mitochondrial permeability transition pore. Additionally, Myc-UBA52 expression in neuronal cells offered protection against apoptotic and autophagic cell death. Altogether, our findings delineate a functional association between UBA52 and mitochondrial homeostasis, providing new insights into the deterrence of dopaminergic cell death during acute PD pathogenesis.
Collapse
Affiliation(s)
- Shubhangini Tiwari
- Division of Toxicology and Experimental Medicine, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Abhishek Singh
- Division of Toxicology and Experimental Medicine, CSIR-Central Drug Research Institute, Lucknow 226031, India.,Academy of Scientific & Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Parul Gupta
- Division of Toxicology and Experimental Medicine, CSIR-Central Drug Research Institute, Lucknow 226031, India.,Academy of Scientific & Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Amrutha K
- Division of Toxicology and Experimental Medicine, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Sarika Singh
- Division of Toxicology and Experimental Medicine, CSIR-Central Drug Research Institute, Lucknow 226031, India.,Academy of Scientific & Innovative Research (AcSIR), Ghaziabad 201002, India
| |
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: 1.5] [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
|
Shao J, Liu X, Lian M, Mao Y. Citronellol Prevents 6-OHDA-Induced Oxidative Stress, Mitochondrial Dysfunction, and Apoptosis in Parkinson Disease Model of SH-SY5Y Cells via Modulating ROS-NO, MAPK/ERK, and PI3K/Akt Signaling Pathways. Neurotox Res 2022; 40:2221-2237. [PMID: 36097250 DOI: 10.1007/s12640-022-00558-8] [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: 04/26/2022] [Revised: 07/13/2022] [Accepted: 08/09/2022] [Indexed: 12/31/2022]
Abstract
Parkinson disease is a neurodegenerative disorder distinguished by dopaminergic shortage in the striatum and the accumulation of α-synuclein neuronal aggregates in the brains of patients. Since, there is no accurate treatment available for Parkinson disease, researches are designed to alleviate the pathognomonic symptoms such as neuroinflammation, oxidative stress, mitochondrial dysfunction, and apoptosis. Accordingly, a number of compounds have been reported to inhibit these pathognomonic symptoms. In this study, we have assessed the neuroprotective potential of citronellol against 6-OHDA-induced neurotoxicity in SH-SY5Y cells. The results found that citronellol treatment effectively hindered the cell death caused by 6-OHDA and thereby maintaining the cell viability in SH-SY5Y cells at 50 µg/mL concentration. As expected, the citronellol treatment significantly reduced the 6-OHDA-induced secretion of inflammatory factors (IL-1β, IL-6, and TNF-α), which was obtained through ELISA technique. Similarly, citronellol hindered the 6-OHDA-induced oxidative stress by lowering the intracellular ROS and NO level and MDA leakage along with increased expression of SOD level in SH-SY5Y cells. The JC-1 staining showed that 6-OHDA increased the number of green fluorescent dots with ruptured mitochondrial membrane potential, while citronellol increased the amount of red fluorescent, showing the rescue potential against the 6-OHDA-induced mitochondrial dysfunction. Furthermore, citronellol hampered the 6-OHDA-induced apoptosis via the suppression of Bcl-2/Bax pathway. The western blotting results hypothesized that citronellol rescued SH-SY5Y cells from 6-OHDA-induced neurotoxicity via modulating ROS-NO, MAPK/ERK, and PI3K/Akt signaling pathways. However, further clinical trials are required to verify the anti-Parkinson efficacy.
Collapse
Affiliation(s)
- Jiahui Shao
- Department of Neurology, The First People's Hospital of Wenling, Zhejiang Province, Wenling, 317500, China
| | - Xuan Liu
- Department of Neurology, The First People's Hospital of Wenling, Zhejiang Province, Wenling, 317500, China
| | - Mengjia Lian
- Department of Neurology, The First People's Hospital of Wenling, Zhejiang Province, Wenling, 317500, China
| | - Youbing Mao
- Department of Special Inspection Section, The First People's Hospital of Wenling, No. 333, Chuanan South Road, Chengxi StreetZhejiang Province, Wenling, 317500, China.
| |
Collapse
|
7
|
Mallet D, Goutaudier R, Barbier EL, Carnicella S, Colca JR, Fauvelle F, Boulet S. Re-routing Metabolism by the Mitochondrial Pyruvate Carrier Inhibitor MSDC-0160 Attenuates Neurodegeneration in a Rat Model of Parkinson's Disease. Mol Neurobiol 2022; 59:6170-6182. [PMID: 35895232 DOI: 10.1101/2022.01.17.476616] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 07/10/2022] [Indexed: 05/25/2023]
Abstract
A growing body of evidence supports the idea that mitochondrial dysfunction might represent a key feature of Parkinson's disease (PD). Central regulators of energy production, mitochondria, are also involved in several other essential functions such as cell death pathways and neuroinflammation which make them a potential therapeutic target for PD management. Interestingly, recent studies related to PD have reported a neuroprotective effect of targeting mitochondrial pyruvate carrier (MPC) by the insulin sensitizer MSDC-0160. As the sole point of entry of pyruvate into the mitochondrial matrix, MPC plays a crucial role in energetic metabolism which is impacted in PD. This study therefore aimed at providing insights into the mechanisms underlying the neuroprotective effect of MSDC-0160. We investigated behavioral, cellular, and metabolic impact of chronic MSDC-0160 treatment in unilateral 6-OHDA PD rats. We evaluated mitochondrially related processes through the expression of pivotal mitochondrial enzymes in dorsal striatal biopsies and the level of metabolites in serum samples using nuclear magnetic resonance spectroscopy (NMR)-based metabolomics. MSDC-0160 treatment in unilateral 6-OHDA rats improved motor behavior, decreased dopaminergic denervation, and reduced mTOR activity and neuroinflammation. Concomitantly, MSDC-0160 administration strongly modified energy metabolism as revealed by increased ketogenesis, beta oxidation, and glutamate oxidation to satisfy energy needs and maintain energy homeostasis. MSDC-0160 exerts its neuroprotective effect through reorganization of multiple pathways connected to energy metabolism.
Collapse
Affiliation(s)
- David Mallet
- Université Grenoble Alpes Inserm, U1216, Grenoble Institut Neurosciences, 38000, Grenoble, France
| | - Raphael Goutaudier
- Université Grenoble Alpes Inserm, U1216, Grenoble Institut Neurosciences, 38000, Grenoble, France
| | - Emmanuel L Barbier
- Université Grenoble Alpes Inserm, U1216, Grenoble Institut Neurosciences, 38000, Grenoble, France
- Université Grenoble Alpes Inserm, US17, CNRS, UMS, 3552, CHU Grenoble Alpes IRMaGe, Grenoble, France
| | - Sebastien Carnicella
- Université Grenoble Alpes Inserm, U1216, Grenoble Institut Neurosciences, 38000, Grenoble, France
| | - Jerry R Colca
- Metabolic Solutions Development Company, Kalamazoo, MI, 49007, USA
| | - Florence Fauvelle
- Université Grenoble Alpes Inserm, U1216, Grenoble Institut Neurosciences, 38000, Grenoble, France
- Université Grenoble Alpes Inserm, US17, CNRS, UMS, 3552, CHU Grenoble Alpes IRMaGe, Grenoble, France
| | - Sabrina Boulet
- Université Grenoble Alpes Inserm, U1216, Grenoble Institut Neurosciences, 38000, Grenoble, France.
| |
Collapse
|
8
|
Tiwari S, Gupta P, Singh A, Chaturvedi S, Wahajuddin M, Mishra A, Singh S. 4-Phenylbutyrate Mitigates the Motor Impairment and Dopaminergic Neuronal Death During Parkinson's Disease Pathology via Targeting VDAC1 Mediated Mitochondrial Function and Astrocytes Activation. Neurochem Res 2022; 47:3385-3401. [PMID: 35922743 DOI: 10.1007/s11064-022-03691-0] [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: 03/02/2022] [Revised: 06/15/2022] [Accepted: 07/12/2022] [Indexed: 10/16/2022]
Abstract
Parkinson's disease (PD) is a progressive motor neurodegenerative disorder significantly associated with protein aggregation related neurodegenerative mechanisms. In view of no disease modifying drugs, the present study was targeted to investigate the therapeutic effects of pharmacological agent 4-phenylbutyric acid (4PBA) in PD pathology. 4PBA is an FDA approved monocarboxylic acid with inhibitory activity towards histone deacetylase and clinically treats urea cycle disorder. First, we observed the significant protective effects of 4PBA on PD specific neuromuscular coordination, level of tyrosine hydroxylase, α-synuclein level and neurotransmitter dopamine in both substantia nigra and striatal regions of the experimental rat model of PD. Further results revealed that treatment with 4PBA drug exhibited significant protection against disease related oxidative stress and augmented nitrite levels. The disease pathology-related depletion in mitochondrial membrane potential and augmented level of calcium as well as mitochondrion membrane located VDAC1 protein level and cytochrome-c translocation were also significantly attenuated with 4PBA administration. Inhibited neuronal apoptosis and restored neuronal morphology were also observed with 4PBA treatment as measured by level of pro-apoptotic proteins t-Bid, Bax and cleaved caspase-3 along with cresyl violet staining in both substantia nigra and striatal regions. Lastly, PD-linked astrocyte activation was significantly inhibited with 4PBA treatment. Altogether, our findings suggest that 4PBA exerts broad-spectrum neuroprotective effects in PD animal model.
Collapse
Affiliation(s)
- Shubhangini Tiwari
- Division of Neuroscience and Ageing Biology, Division of Toxicology and Experimental Medicine, CSIR-Central Drug Research Institute, Lucknow, 226031, India
| | - Parul Gupta
- Division of Neuroscience and Ageing Biology, Division of Toxicology and Experimental Medicine, CSIR-Central Drug Research Institute, Lucknow, 226031, India.,Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, India
| | - Abhishek Singh
- Division of Neuroscience and Ageing Biology, Division of Toxicology and Experimental Medicine, CSIR-Central Drug Research Institute, Lucknow, 226031, India.,Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, India
| | - Swati Chaturvedi
- Division of Pharmaceutics & Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, 226031, India
| | - M Wahajuddin
- Division of Pharmaceutics & Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, 226031, India
| | - Amit Mishra
- Cellular and Molecular Neurobiology Unit, Indian Institute of Technology, Jodhpur, Rajasthan, 342011, India
| | - Sarika Singh
- Division of Neuroscience and Ageing Biology, Division of Toxicology and Experimental Medicine, CSIR-Central Drug Research Institute, Lucknow, 226031, India. .,Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, India.
| |
Collapse
|
9
|
Mallet D, Goutaudier R, Barbier EL, Carnicella S, Colca JR, Fauvelle F, Boulet S. Re-routing Metabolism by the Mitochondrial Pyruvate Carrier Inhibitor MSDC-0160 Attenuates Neurodegeneration in a Rat Model of Parkinson's Disease. Mol Neurobiol 2022; 59:6170-6182. [PMID: 35895232 DOI: 10.1007/s12035-022-02962-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 07/10/2022] [Indexed: 11/29/2022]
Abstract
A growing body of evidence supports the idea that mitochondrial dysfunction might represent a key feature of Parkinson's disease (PD). Central regulators of energy production, mitochondria, are also involved in several other essential functions such as cell death pathways and neuroinflammation which make them a potential therapeutic target for PD management. Interestingly, recent studies related to PD have reported a neuroprotective effect of targeting mitochondrial pyruvate carrier (MPC) by the insulin sensitizer MSDC-0160. As the sole point of entry of pyruvate into the mitochondrial matrix, MPC plays a crucial role in energetic metabolism which is impacted in PD. This study therefore aimed at providing insights into the mechanisms underlying the neuroprotective effect of MSDC-0160. We investigated behavioral, cellular, and metabolic impact of chronic MSDC-0160 treatment in unilateral 6-OHDA PD rats. We evaluated mitochondrially related processes through the expression of pivotal mitochondrial enzymes in dorsal striatal biopsies and the level of metabolites in serum samples using nuclear magnetic resonance spectroscopy (NMR)-based metabolomics. MSDC-0160 treatment in unilateral 6-OHDA rats improved motor behavior, decreased dopaminergic denervation, and reduced mTOR activity and neuroinflammation. Concomitantly, MSDC-0160 administration strongly modified energy metabolism as revealed by increased ketogenesis, beta oxidation, and glutamate oxidation to satisfy energy needs and maintain energy homeostasis. MSDC-0160 exerts its neuroprotective effect through reorganization of multiple pathways connected to energy metabolism.
Collapse
Affiliation(s)
- David Mallet
- Université Grenoble Alpes Inserm, U1216, Grenoble Institut Neurosciences, 38000, Grenoble, France
| | - Raphael Goutaudier
- Université Grenoble Alpes Inserm, U1216, Grenoble Institut Neurosciences, 38000, Grenoble, France
| | - Emmanuel L Barbier
- Université Grenoble Alpes Inserm, U1216, Grenoble Institut Neurosciences, 38000, Grenoble, France.,Université Grenoble Alpes Inserm, US17, CNRS, UMS, 3552, CHU Grenoble Alpes IRMaGe, Grenoble, France
| | - Sebastien Carnicella
- Université Grenoble Alpes Inserm, U1216, Grenoble Institut Neurosciences, 38000, Grenoble, France
| | - Jerry R Colca
- Metabolic Solutions Development Company, Kalamazoo, MI, 49007, USA
| | - Florence Fauvelle
- Université Grenoble Alpes Inserm, U1216, Grenoble Institut Neurosciences, 38000, Grenoble, France.,Université Grenoble Alpes Inserm, US17, CNRS, UMS, 3552, CHU Grenoble Alpes IRMaGe, Grenoble, France
| | - Sabrina Boulet
- Université Grenoble Alpes Inserm, U1216, Grenoble Institut Neurosciences, 38000, Grenoble, France.
| |
Collapse
|
10
|
K A, Mishra A, Singh S. Implications of intracellular protein degradation pathways in Parkinson's disease and therapeutics. J Neurosci Res 2022; 100:1834-1844. [PMID: 35819247 DOI: 10.1002/jnr.25101] [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: 09/21/2021] [Revised: 05/31/2022] [Accepted: 06/18/2022] [Indexed: 12/20/2022]
Abstract
Parkinson's disease (PD) pathology is the most common motor neurodegenerative disease that occurs due to the progressive degeneration of dopaminergic neurons of the nigrostriatal pathway of the brain. The histopathological hallmark of the disease is fibrillary aggregate called Lewy bodies which majorly contain α-synuclein, suggesting the critical implication of diminished protein degradation mechanisms in disease pathogenesis. This α-synuclein-containing Lewy bodies are evident in both experimental models as well as in postmortem PD brain and are speculated to be pathogenic but still, the lineal association between these aggregates and the complexity of disease pathology is not yet well established and needs further attention. However, it has been reported that α-synuclein aggregates have consorted with the declined proteasome and lysosome activities. Therefore, in this review, we reappraise intracellular protein degradation mechanisms during PD pathology. This article focused on the findings of the last two decades suggesting the implications of protein degradation mechanisms in disease pathogenesis and based on shreds of evidence, some of the approaches are also suggested which may be adopted to find out the novel therapeutic targets for the management of PD patients.
Collapse
Affiliation(s)
- Amrutha K
- Division of Toxicology and Experimental Medicine, CSIR-Central Drug Research Institute, Lucknow, India
| | - Amit Mishra
- Cellular and Molecular Neurobiology Unit, Indian Institute of Technology Jodhpur, Jodhpur, India
| | - Sarika Singh
- Division of Toxicology and Experimental Medicine, CSIR-Central Drug Research Institute, Lucknow, India.,Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, India
| |
Collapse
|
11
|
Zou XG, Xu MT, Dong XL, Ying YM, Guan RF, Wu WC, Yang K, Sun PL. Solid-state-cultured mycelium of Antrodia camphorata exerts potential neuroprotective activities against 6-hydroxydopamine-induced toxicity in PC12 cells. J Food Biochem 2022; 46:e14208. [PMID: 35467031 DOI: 10.1111/jfbc.14208] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Revised: 03/20/2022] [Accepted: 03/28/2022] [Indexed: 12/17/2022]
Abstract
Antrodia camphorata (A. camphorata) is an edible fungus containing various bioactive compounds generally used for health benefits. This study aimed to explore the potential neuroprotective activities of solid-state-cultured mycelium of A. camphorata (SCMAC) against Parkinson's disease (PD), as well as the underlying mechanism using an in vitro 6-hydroxydopamine (6-OHDA)-induced PC12 cell model. The results showed that SCMAC extracts alleviated cell toxicity induced by 6-OHDA and the loss of dopaminergic neurons, which was confirmed by the increase of cell viabilities, inhibition of cell apoptosis, the upregulation of tyrosine hydroxylase (TH) and dopamine transporter (DAT) levels and the downregulation of α-Synuclein level. After purification, 11 compounds were identified by the NMR technique, including a quinone, four phenolic acid derivatives, three ubiquinone derivatives, two alkaloids, and a triterpenoid. The present study suggests that SCMAC could be an attractive candidate for the prevention or treatment of PD. PRACTICAL APPLICATIONS: Parkinson's disease seriously affects the lifetime and quality of the elder population for a long history. Long-term consumption of L-DOPA will result in side effects, such as developing abnormal involuntary movements called dyskinesia. This study showed that natural SCMAC extracts could be a potential therapeutic agent for the treatment of neurodegenerative disorder.
Collapse
Affiliation(s)
- Xian-Guo Zou
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou, P.R. China.,Key Laboratory of Food Macromolecular Resources Processing Technology Research (Zhejiang University of Technology), China National Light Industry, Beijing, China
| | - Meng-Ting Xu
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou, P.R. China
| | - Xiao-Li Dong
- Food Safety and Technology Research Center, Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hong Kong SAR, China.,Key Laboratory of Food Biological Safety Control, The Hong Kong Polytechnic University Shenzhen Institute, Shenzhen, P.R. China
| | - You-Min Ying
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, P.R. China
| | - Rong-Fa Guan
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou, P.R. China.,Key Laboratory of Food Macromolecular Resources Processing Technology Research (Zhejiang University of Technology), China National Light Industry, Beijing, China
| | - Wei-Cheng Wu
- Food Science Institute, Zhejiang Academy of Agricultural Sciences, Hangzhou, China
| | - Kai Yang
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou, P.R. China.,Key Laboratory of Food Macromolecular Resources Processing Technology Research (Zhejiang University of Technology), China National Light Industry, Beijing, China
| | - Pei-Long Sun
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou, P.R. China.,Key Laboratory of Food Macromolecular Resources Processing Technology Research (Zhejiang University of Technology), China National Light Industry, Beijing, China
| |
Collapse
|
12
|
Ganesan S, Parvathi VD. Deconstructing the molecular genetics behind the PINK1/Parkin axis in Parkinson’s disease using Drosophila melanogaster as a model organism. EGYPTIAN JOURNAL OF MEDICAL HUMAN GENETICS 2021. [DOI: 10.1186/s43042-021-00208-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Abstract
Background
Parkinson’s disease (PD) is a multifactorial neurodegenerative disorder marked by the death of nigrostriatal dopaminergic neurons in response to the compounding effects of oxidative stress, mitochondrial dysfunction and protein aggregation. Transgenic Drosophila models have been used extensively to decipher the underlying genetic interactions that exacerbate neural health in PD. Autosomal recessive forms of the disease have been linked to mutations in the serine/threonine kinase PINK1(PTEN-Induced Putative Kinase 1) and E3 ligase Parkin, which function in an axis that is conserved in flies. This review aims to probe the current understanding of PD pathogenesis via the PINK1/Parkin axis while underscoring the importance of several molecular and pharmacologic rescues brought to light through studies in Drosophila.
Main body
Mutations in PINK1 and Parkin have been shown to affect the axonal transport of mitochondria within dopaminergic neurons and perturb the balance between mitochondrial fusion/fission resulting in abnormal mitochondrial morphology. As per studies in flies, ectopic expression of Fwd kinase and Atg-1 to promote fission and mitophagy while suppressing fusion via MUL1 E3 ligase may aid to halt mitochondrial aggregation and prolong the survival of dopaminergic neurons. Furthermore, upregulation of Hsp70/Hsp90 chaperone systems (Trap1, CHIP) to target misfolded mitochondrial respiratory complexes may help to preserve their bioenergetic capacity. Accumulation of reactive oxygen species as a consequence of respiratory complex dysfunction or antioxidant enzyme deficiency further escalates neural death by inducing apoptosis, lipid peroxidation and DNA damage. Fly studies have reported the induction of canonical Wnt signalling to enhance the activity of transcriptional co-activators (PGC1α, FOXO) which induce the expression of antioxidant enzymes. Enhancing the clearance of free radicals via uncoupling proteins (UCP4) has also been reported to ameliorate oxidative stress-induced cell death in PINK1/Parkin mutants.
Conclusion
While these novel mechanisms require validation through mammalian studies, they offer several explanations for the factors propagating dopaminergic death as well as promising insights into the therapeutic importance of transgenic fly models in PD.
Collapse
|
13
|
Singh A, Yadawa AK, Chaturvedi S, Wahajuddin M, Mishra A, Singh S. Mechanism for antiParkinsonian effect of resveratrol: Involvement of transporters, synaptic proteins, dendrite arborization, biochemical alterations, ER stress and apoptosis. Food Chem Toxicol 2021; 155:112433. [PMID: 34302886 DOI: 10.1016/j.fct.2021.112433] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 07/15/2021] [Accepted: 07/17/2021] [Indexed: 11/21/2022]
Abstract
The present study was undertaken to evaluate the mechanism for antiParkinsonian effect of resveratrol employing 6-hydroxydopamine (6-OHDA) induced experimental model of Parkinson's disease (PD). Resveratrol treatment significantly protects the PD related pathological markers like level of tyrosine hydroxylase, dopamine and apoptotic proteins (Bax and cleaved caspase-3). Disease pathology involves significantly decreased level of dopamine transporter, synaptophysin and postsynaptic density protein 95 (PSD-95) along with augmented level of vesicular monoamine transporter and considerably affected the dendrite arborization. Such affected neuronal communication was significantly restored with resveratrol treatment. Biochemical alterations include the depleted level of glutathione (GSH), mitochondrial complex-I activity with concomitant increased level of lipid peroxidation, nitrite level and calcium levels, which were also significantly inhibited with resveratrol treatment. Altered calcium level induces the endoplasmic reticulum (ER) stress related signalling and phosphorylated Nuclear factor erythroid 2-related factor 2 (Nrf2), and with resveratrol treatment the level of phosphorylated Nrf2 was further increased. The concurrent depleted level of proteasome activity was observed which was attenuated with resveratrol treatment. Proinflammatory cytokines and activated astrocytes were observed which was inhibited with resveratrol treatment. In conclusion, findings suggested that resveratrol exhibits the interference in neuronal communication, oxidative stress, mitochondrial pathophysiology, ER stress, protein degradation mechanism and inflammatory responses and could be utilize in clinics to treat the PD patients.
Collapse
Affiliation(s)
- Ashish Singh
- Division of Toxicology and Experimental Medicine, Central Drug Research Institute, Lucknow, 226031, India
| | - Arun Kumar Yadawa
- Division of Toxicology and Experimental Medicine, Central Drug Research Institute, Lucknow, 226031, India
| | - Swati Chaturvedi
- Division of Pharmacokinetics and Pharmaceutics, Central Drug Research Institute, Lucknow, 226031, India
| | - M Wahajuddin
- Division of Pharmacokinetics and Pharmaceutics, Central Drug Research Institute, Lucknow, 226031, India
| | - Amit Mishra
- Cellular and Molecular Neurobiology Unit, Indian Institute of Technology, Jodhpur, Rajasthan, 342011, India
| | - Sarika Singh
- Division of Toxicology and Experimental Medicine, Central Drug Research Institute, Lucknow, 226031, India.
| |
Collapse
|
14
|
Rivastigmine attenuates the Alzheimer's disease related protein degradation and apoptotic neuronal death signalling. Biochem J 2021; 478:1435-1451. [PMID: 33660768 DOI: 10.1042/bcj20200754] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 03/02/2021] [Accepted: 03/04/2021] [Indexed: 12/13/2022]
Abstract
Rivastigmine is a clinical drug for patients of Alzheimer's disease (AD) exerting its inhibitory effect on acetylcholinesterase activity however, its effect on other disease-related pathological mechanisms are not yet known. This study was conducted to evaluate the effect of rivastigmine on protein aggregation and degradation related mechanisms employing streptozotocin (STZ) induced experimental rat model. The known inhibitory effect of rivastigmine on cognition and acetylcholinesterase activity was observed in both cortex and hippocampus and further its effect on tau level, amyloid aggregation, biochemical alterations, endoplasmic reticulum (ER) stress, calcium homeostasis, proteasome activity and apoptosis was estimated. STZ administration in rat brain caused significant cognitive impairment, augmented acetylcholinesterase activity, tau phosphorylation and amyloid aggregation which were significantly inhibited with rivastigmine treatment. STZ also caused significant biochemical alterations which were attenuated with rivastigmine treatment. Since AD pathology is related to protein aggregation and we have found disease-related amyloid aggregation, further the investigation was done to decipher the ER functionality and apoptotic signalling. STZ caused significantly altered level of ER stress related markers (GRP78, GADD153 and caspase-12) which were significantly inhibited with rivastigmine treatment. Furthermore, the effect of rivastigmine was estimated on proteasome activity in both regions. Rivastigmine treatment significantly enhances the proteasome activity and may contributes in removal of amyloid aggregation. In conclusion, findings suggested that along with inhibitory effect of rivastigmine on acetylcholinesterase activity and up to some extent on cognition, it has significant effect on disease-related biochemical alterations, ER functionality, protein degradation machinery and neuronal apoptosis.
Collapse
|
15
|
Gupta S, Mishra A, Singh S. Cardinal role of eukaryotic initiation factor 2 (eIF2α) in progressive dopaminergic neuronal death & DNA fragmentation: Implication of PERK:IRE1α:ATF6 axis in Parkinson's pathology. Cell Signal 2021; 81:109922. [PMID: 33484794 DOI: 10.1016/j.cellsig.2021.109922] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 01/05/2021] [Accepted: 01/05/2021] [Indexed: 12/14/2022]
Abstract
The study was conducted to assess the role of eukaryotic initiation factor 2 (eIF2α) in progressive dopaminergic neuronal death employing various interventions (YM08, 4μ8C, AEBSF, salubrinal, ursolic acid) of endoplasmic reticulum (ER) stress signaling. The protein level of all the ER stress related signaling factors (GRP78, IRE1α, ATF6, eIF2α, ATF4, XBP-1, GADD153) were estimated after 3 and 7 day of experiment initiation. Findings with single administration of interventions showed that salubrinal exhibited significant protection against rotenone induced adverse alterations in comparison to other interventions. Therefore, further study was expanded with repeat dose of salubrinal. Rotenone administration in rat brain caused the significant biochemical alterations, dose dependent progressive neuronal apoptosis and altered neuronal morphology which was significantly attenuated with salubrinal treatment. In conclusion, findings showed that rotenone administration caused the dose dependent progressive neuronal death including cardinal role of eIF2α, suggesting the potential pharmacological utilization of salubrinal or salubrinal like molecules in therapeutics of Parkinson's diseases.
Collapse
Affiliation(s)
- Sonam Gupta
- Department of Neuroscience and Ageing Biology, Division of Toxicology and Experimental Medicine, CSIR-Central Drug Research Institute, Lucknow 226031, India; Academy of Scientific & Innovative Research (AcSIR), CSIR-Central Drug Research Institute, India
| | - Amit Mishra
- Cellular and Molecular Neurobiology Unit, Indian Institute of Technology, Jodhpur, Rajasthan 342011, India
| | - Sarika Singh
- Department of Neuroscience and Ageing Biology, Division of Toxicology and Experimental Medicine, CSIR-Central Drug Research Institute, Lucknow 226031, India; Academy of Scientific & Innovative Research (AcSIR), CSIR-Central Drug Research Institute, India.
| |
Collapse
|
16
|
Satta N, Weppe R, Pagano S, Frias M, Juillard C, Vuilleumier N. Auto-antibodies against apolipoprotein A-1 block cancer cells proliferation and induce apoptosis. Oncotarget 2020; 11:4266-4280. [PMID: 33245719 PMCID: PMC7679029 DOI: 10.18632/oncotarget.27814] [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: 07/03/2020] [Accepted: 10/27/2020] [Indexed: 11/25/2022] Open
Abstract
Auto-antibodies against apoA-1 (anti-apoA-1 IgGs) have been identified as important actors of atherosclerosis development through pro-inflammatory and pro-atherogenic properties and to also induce apoptosis in tumoral neuronal and lymphocyte derived cell lines through unknown mechanisms. The purpose of this study was to explore the cellular pathways involved in tumoral cell survival modulated by anti-apoA-1 antibodies. We observed that anti-apoA-1 antibodies induce growth arrest (in G2/M phase) and cell apoptosis through caspase 3 activation, accompanied by a selective p53 phosphorylation on serine 15. RNA sequencing indicated that anti-apoA-1 IgGs affect the expression of more than 950 genes belonging to five major groups of genes and respectively involved in i) cell proliferation inhibition, ii) p53 stabilisation and regulation, iii) apoptosis regulation, iv) inflammation regulation, and v) oxidative stress. In conclusion, anti-apoA-1 antibodies seem to have a role in blocking tumoral cell proliferation and survival, by activating a major tumor suppressor protein and by modulating the inflammatory and oxidative stress response. Further investigations are needed to explore a possible anti-cancer therapeutic approach of these antibodies in very specific and circumscribed conditions.
Collapse
Affiliation(s)
- Nathalie Satta
- Division of Laboratory Medicine, Department of Diagnostic, Geneva University Hospitals, Geneva, Switzerland.,Department of Medicine, Medical Faculty, Geneva University, Geneva, Switzerland
| | - Rémy Weppe
- Division of Laboratory Medicine, Department of Diagnostic, Geneva University Hospitals, Geneva, Switzerland.,Department of Medicine, Medical Faculty, Geneva University, Geneva, Switzerland
| | - Sabrina Pagano
- Division of Laboratory Medicine, Department of Diagnostic, Geneva University Hospitals, Geneva, Switzerland.,Department of Medicine, Medical Faculty, Geneva University, Geneva, Switzerland
| | - Miguel Frias
- Division of Laboratory Medicine, Department of Diagnostic, Geneva University Hospitals, Geneva, Switzerland.,Department of Medicine, Medical Faculty, Geneva University, Geneva, Switzerland
| | - Catherine Juillard
- Division of Laboratory Medicine, Department of Diagnostic, Geneva University Hospitals, Geneva, Switzerland.,Department of Medicine, Medical Faculty, Geneva University, Geneva, Switzerland
| | - Nicolas Vuilleumier
- Division of Laboratory Medicine, Department of Diagnostic, Geneva University Hospitals, Geneva, Switzerland.,Department of Medicine, Medical Faculty, Geneva University, Geneva, Switzerland
| |
Collapse
|
17
|
Intranigral Administration of β-Sitosterol- β-D-Glucoside Elicits Neurotoxic A1 Astrocyte Reactivity and Chronic Neuroinflammation in the Rat Substantia Nigra. J Immunol Res 2020; 2020:5907591. [PMID: 33282962 PMCID: PMC7685831 DOI: 10.1155/2020/5907591] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 06/01/2020] [Accepted: 06/09/2020] [Indexed: 12/23/2022] Open
Abstract
Chronic consumption of β-sitosterol-β-D-glucoside (BSSG), a neurotoxin contained in cycad seeds, leads to Parkinson's disease in humans and rodents. Here, we explored whether a single intranigral administration of BSSG triggers neuroinflammation and neurotoxic A1 reactive astrocytes besides dopaminergic neurodegeneration. We injected 6 μg BSSG/1 μL DMSO or vehicle into the left substantia nigra and immunostained with antibodies against tyrosine hydroxylase (TH) together with markers of microglia (OX42), astrocytes (GFAP, S100β, C3), and leukocytes (CD45). We also measured nitric oxide (NO), lipid peroxidation (LPX), and proinflammatory cytokines (TNF-α, IL-1β, IL-6). The Evans blue assay was used to explore the blood-brain barrier (BBB) permeability. We found that BSSG activates NO production on days 15 and 30 and LPX on day 120. Throughout the study, high levels of TNF-α were present in BSSG-treated animals, whereas IL-1β was induced until day 60 and IL-6 until day 30. Immunoreactivity of activated microglia (899.0 ± 80.20%) and reactive astrocytes (651.50 ± 11.28%) progressively increased until day 30 and then decreased to remain 251.2 ± 48.8% (microglia) and 91.02 ± 39.8 (astrocytes) higher over controls on day 120. C3(+) cells were also GFAP and S100β immunoreactive, showing they were neurotoxic A1 reactive astrocytes. BBB remained permeable until day 15 when immune cell infiltration was maximum. TH immunoreactivity progressively declined, reaching 83.6 ± 1.8% reduction on day 120. Our data show that BSSG acute administration causes chronic neuroinflammation mediated by activated microglia, neurotoxic A1 reactive astrocytes, and infiltrated immune cells. The severe neuroinflammation might trigger Parkinson's disease in BSSG intoxication.
Collapse
|
18
|
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: 17] [Impact Index Per Article: 3.4] [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
|
19
|
Tiwari S, Singh S. Reciprocal Upshot of Nitric Oxide, Endoplasmic Reticulum Stress, and Ubiquitin Proteasome System in Parkinson's Disease Pathology. Neuroscientist 2020; 27:340-354. [PMID: 32713286 DOI: 10.1177/1073858420942211] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Parkinson's disease (PD) pathology involves degeneration of nigrostriatal pathway, postulating symptoms associated with age, environment, and genetic anomalies, including nonlinear disease progression. Hallmark characteristics of PD include dopaminergic neuronal degeneration and death, which may also be exhibited by other neurological diseases, making the diagnosis of the disease intricate at early stage. Such obscure diagnosis of the disease, limited symptomatic improvements with available therapeutics, and their inability to modify the disease status instigate us to appraise the past research and formulate the colligating comprehensive insights. This review is accentuating on the role of nitric oxide, endoplasmic reticulum stress, and their association with the ubiquitin proteasome system (UPS) during PD pathology involving focus on ubiquitin ligases due to their regulatory functions. Meticulous understanding of these major disease-related pathological events and their functional alliance may render novel dimensions for better understanding of disease etiology, related mechanisms, as well as direction toward witnessing of new therapeutic targets for the management of Parkinson's patients.
Collapse
Affiliation(s)
- Shubhangini Tiwari
- Department of Neurosciences and Ageing Biology and Division of Toxicology and Experimental Medicine, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, India
| | - Sarika Singh
- Department of Neurosciences and Ageing Biology and Division of Toxicology and Experimental Medicine, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, India
| |
Collapse
|
20
|
Tripathi MK, Kartawy M, Amal H. The role of nitric oxide in brain disorders: Autism spectrum disorder and other psychiatric, neurological, and neurodegenerative disorders. Redox Biol 2020; 34:101567. [PMID: 32464501 PMCID: PMC7256645 DOI: 10.1016/j.redox.2020.101567] [Citation(s) in RCA: 100] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 05/04/2020] [Accepted: 05/05/2020] [Indexed: 12/21/2022] Open
Abstract
Nitric oxide (NO) is a multifunctional signalling molecule and a neurotransmitter that plays an important role in physiological and pathophysiological processes. In physiological conditions, NO regulates cell survival, differentiation and proliferation of neurons. It also regulates synaptic activity, plasticity and vesicle trafficking. NO affects cellular signalling through protein S-nitrosylation, the NO-mediated posttranslational modification of cysteine thiols (SNO). SNO can affect protein activity, protein-protein interaction and protein localization. Numerous studies have shown that excessive NO and SNO can lead to nitrosative stress in the nervous system, contributing to neuropathology. In this review, we summarize the role of NO and SNO in the progression of neurodevelopmental, psychiatric and neurodegenerative disorders, with special attention to autism spectrum disorder (ASD). We provide mechanistic insights into the contribution of NO in diverse brain disorders. Finally, we suggest that pharmacological agents that can inhibit or augment the production of NO as well as new approaches to modulate the formation of SNO-proteins can serve as a promising approach for the treatment of diverse brain disorders.
Collapse
Affiliation(s)
- Manish Kumar Tripathi
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Maryam Kartawy
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Haitham Amal
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel.
| |
Collapse
|
21
|
Promising Polyphenols in Parkinson’s Disease Therapeutics. Neurochem Res 2020; 45:1731-1745. [DOI: 10.1007/s11064-020-03058-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 04/15/2020] [Accepted: 05/19/2020] [Indexed: 12/11/2022]
|
22
|
Nitroxide Radical-Containing Redox Nanoparticles Protect Neuroblastoma SH-SY5Y Cells against 6-Hydroxydopamine Toxicity. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:9260748. [PMID: 32377313 PMCID: PMC7196160 DOI: 10.1155/2020/9260748] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 02/06/2020] [Accepted: 04/01/2020] [Indexed: 12/19/2022]
Abstract
Parkinson's disease (PD) patients can benefit from antioxidant supplementation, and new efficient antioxidants are needed. The aim of this study was to evaluate the protective effect of selected nitroxide-containing redox nanoparticles (NRNPs) in a cellular model of PD. Antioxidant properties of NRNPs were studied in cell-free systems by protection of dihydrorhodamine 123 against oxidation by 3-morpholino-sydnonimine and protection of fluorescein against bleaching by 2,2-azobis(2-amidinopropane) hydrochloride and sodium hypochlorite. Model blood-brain barrier penetration was studied using hCMEC/D3 cells. Human neuroblastoma SH-SY5Y cells, exposed to 6-hydroxydopamine (6-OHDA), were used as an in vitro model of PD. Cells were preexposed to NRNPs or free nitroxides (TEMPO or 4-amino-TEMPO) for 2 h and treated with 6-OHDA for 1 h and 24 h. The reactive oxygen species (ROS) level was estimated with dihydroethidine 123 and Fluorimetric Mitochondrial Superoxide Activity Assay Kit. Glutathione level (GSH) was measured with ortho-phtalaldehyde, ATP by luminometry, changes in mitochondrial membrane potential with JC-1, and mitochondrial mass with 10-Nonyl-Acridine Orange. NRNP1, TEMPO, and 4-amino-TEMPO (25-150 μM) protected SH-SY5Y cells from 6-OHDA-induced viability loss; the protection was much higher for NRNP1 than for free nitroxides. NRNP1 were better antioxidants in vitro and permeated better the model BBB than free nitroxides. Exposure to 6-OHDA decreased the GSH level after 1 h and increased it considerably after 24 h (apparently a compensatory overresponse); NRNPs and free nitroxides prevented this increase. NRNP1 and free nitroxides prevented the decrease in ATP level after 1 h and increased it after 24 h. 6-OHDA increased the intracellular ROS level and mitochondrial superoxide level. Studied antioxidants mostly decreased ROS and superoxide levels. 6-OHDA decreased the mitochondrial potential and mitochondrial mass; both effects were prevented by NRNP1 and nitroxides. These results suggest that the mitochondria are the main site of 6-OHDA-induced cellular damage and demonstrate a protective effect of NRNP1 in a cellular model of PD.
Collapse
|
23
|
Salubrinal attenuates nitric oxide mediated PERK:IRE1α: ATF-6 signaling and DNA damage in neuronal cells. Neurochem Int 2019; 131:104581. [DOI: 10.1016/j.neuint.2019.104581] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 10/09/2019] [Accepted: 10/15/2019] [Indexed: 01/14/2023]
|
24
|
Gupta P, Singh A, Tiwari S, Mishra A, Maurya R, Singh S. Ulmosides A: Flavonoid 6-C-glycosides from Ulmus wallichiana attenuates lipopolysacchride induced oxidative stress, apoptosis and neuronal death. Neurotoxicology 2019; 73:100-111. [PMID: 30857974 DOI: 10.1016/j.neuro.2019.02.017] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 01/09/2019] [Accepted: 02/25/2019] [Indexed: 01/04/2023]
Abstract
Extract of Ulmus wallichiana is being used as traditional medicine used for the treatment of fractured bones however the effect of its individual flavonols is not known. The present study was conducted to investigate the effect of its novel flavonol, (2S, 3S)-(+)-30, 40, 5, 7-tetrahydroxydihydroflavonol-6-C-b-d-glucopyranoside named as Ulmoside A (UA), on lipopolysaccharides (LPS) treated neurons. LPS treatment to neuronal cells caused significant cytotoxicity, reactive oxygen species generation, depletion in glutathione and mitochondrial impairment which were significantly inhibited with UA treatment. LPS treatment also caused significant translocation of cytochrome-c, decreased level of Bcl2, increased level of Bax and cleaved caspase-3 in neuronal cells reflecting the involvement of intrinsic apoptotic pathway in neuronal death which was attenuated with UA treatment. Since LPS is a well known pro-inflammatory agent it also offered the significant increase in proinflammatory cytokines (tumor necrosis factors-α & interleukin 1-beta) however, UA treatment did not exhibit significant inhibition against LPS induced inflammatory response. LPS also caused the augmented level of inducible nitric oxide synthase (iNOS) which was also not inhibited with co treatment of UA. We have also observed the significant DNA fragmentation and augmented level of cleaved Poly (ADP-Ribose) polymerase 1 after LPS treatment which was significantly reverted with UA treatment. Findings suggested that UA acts through mitochondria and exhibited its anti-oxidative and anti-apoptotic activities in neuronal cells while no significant anti-inflammatory activity and effect on iNOS were observed.
Collapse
Affiliation(s)
- Parul Gupta
- Toxicology and Experimental Medicine Division, CSIR-Central Drug Research Institute, Lucknow, 226031, UP, India; Academy of Scientific & Innovative Research (AcSIR), India
| | - Abhishek Singh
- Toxicology and Experimental Medicine Division, CSIR-Central Drug Research Institute, Lucknow, 226031, UP, India; Academy of Scientific & Innovative Research (AcSIR), India
| | - Shubhangini Tiwari
- Toxicology and Experimental Medicine Division, CSIR-Central Drug Research Institute, Lucknow, 226031, UP, India
| | - Amit Mishra
- Cellular and Molecular Neurobiology Unit, Indian Institute of Technology Jodhpur, Rajasthan, 342011, India
| | - Rakesh Maurya
- Toxicology and Experimental Medicine Division, CSIR-Central Drug Research Institute, Lucknow, 226031, UP, India
| | - Sarika Singh
- Toxicology and Experimental Medicine Division, CSIR-Central Drug Research Institute, Lucknow, 226031, UP, India; Academy of Scientific & Innovative Research (AcSIR), India.
| |
Collapse
|
25
|
Verma DK, Gupta S, Biswas J, Joshi N, Sivarama Raju K, Wahajuddin M, Singh S. Metabolic Enhancer Piracetam Attenuates the Translocation of Mitochondrion-Specific Proteins of Caspase-Independent Pathway, Poly [ADP-Ribose] Polymerase 1 Up-regulation and Oxidative DNA Fragmentation. Neurotox Res 2018; 34:198-219. [PMID: 29532444 DOI: 10.1007/s12640-018-9878-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Revised: 01/29/2018] [Accepted: 01/31/2018] [Indexed: 11/28/2022]
Abstract
Piracetam, a nootropic drug, has been clinically used for decades; however, its mechanism of action still remains enigmatic. The present study was undertaken to evaluate the role of mitochondrion-specific factors of caspase-independent pathway like apoptotic-inducing factor (AIF) and endonuclease-G (endo-G) in piracetam-induced neuroprotection. N2A cells treated with lipopolysaccharide (LPS) exhibited significant cytotoxicity, impaired mitochondrial activity, and reactive oxygen species generation which was significantly attenuated with piracetam co-treatment. Cells co-treated with LPS and piracetam exhibited significant uptake of piracetam in comparison to only piracetam-treated cells as estimated by liquid chromatography-mass spectrometry (LC-MSMS). LPS treatment caused significant translocation of AIF and endonuclease-G in neuronal N2A cells which were significantly attenuated with piracetam co-treatment. Significant over-expression of proinflammatory cytokines was also observed after treatment of LPS to cells which was inhibited with piracetam co-treatment demonstrating its anti-inflammatory property. LPS-treated cells exhibited significant oxidative DNA fragmentation and poly [ADP-ribose] polymerase-1 (PARP-1) up-regulation in nucleus, both of which were attenuated with piracetam treatment. Antioxidant melatonin but not z-VAD offered the inhibited LPS-induced DNA fragmentation indicating the involvement of oxidative DNA fragmentation. Further, we did not observe the altered caspase-3 level after LPS treatment initially while at a later time point, significantly augmented level of caspase-3 was observed which was not inhibited with piracetam treatment. In total, our findings indicate the interference of piracetam in mitochondrion-mediated caspase-independent pathway, as well as its anti-inflammatory and antioxidative properties. Graphical Abstract Graphical abstract indicating the novel interference of metabolic enhancer piracetam (P) in neuronal death mechanisms.
Collapse
Affiliation(s)
- Dinesh Kumar Verma
- Toxicology Division, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, 226031, India
| | - Sonam Gupta
- Toxicology Division, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, 226031, India
| | - Joyshree Biswas
- Toxicology Division, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, 226031, India
| | - Neeraj Joshi
- Department of Biochemistry and Biophysics, Helen Diller Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA
| | - K Sivarama Raju
- Pharmacokinetics & Metabolism Division, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, 226031, India
| | - Mu Wahajuddin
- Pharmacokinetics & Metabolism Division, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, 226031, India
| | - Sarika Singh
- Toxicology Division, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, 226031, India.
| |
Collapse
|
26
|
Verma DK, Gupta S, Biswas J, Joshi N, Singh A, Gupta P, Tiwari S, Sivarama Raju K, Chaturvedi S, Wahajuddin M, Singh S. New therapeutic activity of metabolic enhancer piracetam in treatment of neurodegenerative disease: Participation of caspase independent death factors, oxidative stress, inflammatory responses and apoptosis. Biochim Biophys Acta Mol Basis Dis 2018; 1864:2078-2096. [DOI: 10.1016/j.bbadis.2018.03.014] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 02/26/2018] [Accepted: 03/13/2018] [Indexed: 12/12/2022]
|
27
|
Verma DK, Singh DK, Gupta S, Gupta P, Singh A, Biswas J, Singh S. Minocycline diminishes the rotenone induced neurotoxicity and glial activation via suppression of apoptosis, nitrite levels and oxidative stress. Neurotoxicology 2018; 65:9-21. [PMID: 29360531 DOI: 10.1016/j.neuro.2018.01.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 01/18/2018] [Accepted: 01/19/2018] [Indexed: 01/10/2023]
Abstract
The study was conducted to evaluate the effect of minocycline against pesticide rotenone induced adverse effects in different rat brain regions. Assessment of oxidative stress, nitrite levels, degenerating neurons and level of cleaved caspase-3 was done in frontal cortex, mid brain, hippocampus and striatum regions of rat brain. In addition the expression profile of neuronal (MAP2), astrocytes (GFAP) and microglia (cd11b) markers was done after treatments. Rotenone induced DNA fragmentation was also assessed in all studied rat brain regions by utilizing comet assay. Rotenone administration caused significantly decreased level of glutathione along with increased level of nitrite and lipid peroxidation. Significant oxidative and nitrosative stress was also observed after rotenone administration which was considerably inhibited in minocycline treated rats in time dependent manner. Fluorojade staining and levels of cleaved caspase 3 showed the degeneration of neurons and apoptosis respectively in studied rat brain regions which were further inhibited with minocycline treatment. Rotenone administration caused significantly increased reactivity of astrocytes, microglia and altered neuronal morphology in rat brain regions which was also partially restored with minocycline treatment. In conclusion, present study showed that minocycline treatment attenuated the rotenone induced oxidative stress, nitrite level, degeneration of neurons, augmented glial reactivity and apoptosis.
Collapse
Affiliation(s)
- Dinesh Kumar Verma
- Experimental Medicine and Toxicology Division, CSIR-Central Drug Research Institute, Lucknow-226031, UP, India
| | - Dhirendra Kumar Singh
- Experimental Medicine and Toxicology Division, CSIR-Central Drug Research Institute, Lucknow-226031, UP, India
| | - Sonam Gupta
- Experimental Medicine and Toxicology Division, CSIR-Central Drug Research Institute, Lucknow-226031, UP, India
| | - Parul Gupta
- Experimental Medicine and Toxicology Division, CSIR-Central Drug Research Institute, Lucknow-226031, UP, India
| | - Abhishek Singh
- Experimental Medicine and Toxicology Division, CSIR-Central Drug Research Institute, Lucknow-226031, UP, India
| | - Joyshree Biswas
- Experimental Medicine and Toxicology Division, CSIR-Central Drug Research Institute, Lucknow-226031, UP, India
| | - Sarika Singh
- Experimental Medicine and Toxicology Division, CSIR-Central Drug Research Institute, Lucknow-226031, UP, India.
| |
Collapse
|
28
|
Joshi N, Singh S. Updates on immunity and inflammation in Parkinson disease pathology. J Neurosci Res 2017; 96:379-390. [DOI: 10.1002/jnr.24185] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Revised: 09/05/2017] [Accepted: 09/13/2017] [Indexed: 01/13/2023]
Affiliation(s)
- Neeraj Joshi
- Department of Biochemistry and Biophysics; Helen Diller Comprehensive Cancer Center; San Francisco California
| | - Sarika Singh
- Toxicology and Experimental Medicine Division, CSIR-Central Drug Research Institute; Lucknow India
| |
Collapse
|
29
|
Hernandez-Baltazar D, Zavala-Flores L, Villanueva-Olivo A. The 6-hydroxydopamine model and parkinsonian pathophysiology: Novel findings in an older model. NEUROLOGÍA (ENGLISH EDITION) 2017. [DOI: 10.1016/j.nrleng.2015.06.019] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
|
30
|
Biswas J, Gupta S, Verma DK, Singh S. Streptozotocin alters glucose transport, connexin expression and endoplasmic reticulum functions in neurons and astrocytes. Neuroscience 2017; 356:151-166. [PMID: 28527957 DOI: 10.1016/j.neuroscience.2017.05.018] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Revised: 05/09/2017] [Accepted: 05/09/2017] [Indexed: 12/25/2022]
Abstract
The study was undertaken to explore the cell-specific streptozotocin (STZ)-induced mechanistic alterations. STZ-induced rodent model is a well-established experimental model of Alzheimer's disease (AD) and in our previous studies we have established it as an in vitro screening model of AD by employing N2A neuronal cells. Therefore, STZ was selected in the present study to understand the STZ-induced cell-specific alterations by utilizing neuronal N2A and astrocytes C6 cells. Both neuronal and astrocyte cells were treated with STZ at 10, 50, 100 and 1000μM concentrations for 48h. STZ exposure caused significant decline in cellular viability and augmented cytotoxicity of cells involving astrocytes activation. STZ treatment also disrupted the energy metabolism by altered glucose uptake and its transport in both cells as reflected with decreased expression of glucose transporters (GLUT) 1/3. The consequent decrease in ATP level and decreased mitochondrial membrane potential was also observed in both the cells. STZ caused increased intracellular calcium which could cause the initiation of endoplasmic reticulum (ER) stress. Significant upregulation of ER stress-related markers were observed in both cells after STZ treatment. The cellular communication of astrocytes and neurons was altered as reflected by increased expression of connexin 43 along with DNA fragmentation. STZ-induced apoptotic death was evaluated by elevated expression of caspase-3 and PI/Hoechst staining of cells. In conclusion, study showed that STZ exert alike biochemical alterations, ER stress and cellular apoptosis in both neuronal and astrocyte cells.
Collapse
Affiliation(s)
- Joyshree Biswas
- Toxicology Division, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Sonam Gupta
- Toxicology Division, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Dinesh Kumar Verma
- Toxicology Division, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Sarika Singh
- Toxicology Division, CSIR-Central Drug Research Institute, Lucknow 226031, India.
| |
Collapse
|
31
|
Hosseini M, Rajaei Z, Alaei H, Tajadini M. The Effects of Crocin on 6-OHDA-Induced Oxidative/Nitrosative Damage and Motor Behaviour in Hemiparkinsonian Rats. Malays J Med Sci 2016; 23:35-43. [PMID: 28090177 DOI: 10.21315/mjms2016.23.6.4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2015] [Accepted: 08/07/2016] [Indexed: 10/20/2022] Open
Abstract
BACKGROUND Crocin is considered to prevent oxidative stress-related diseases, such as ischemia and Alzheimer's. The aim of the present investigation was to evaluate the effects of crocin on motor behaviour and 6-OHDA-induced oxidative/nitrosative damage to the striatum in an experimental model of Parkinson's disease. METHODS Left medial forebrain bundle was lesioned by microinjection of 6-OHDA (16μg in 0.2% ascorbate-saline). Crocin (30 and 60 mg/kg) was injected intraperitoneally three days before surgery until six weeks. Rotational behaviour and biochemical analysis were used to evaluate the effect of crocin in a unilateral 6-OHDA-induced model of Parkinson's disease. RESULTS The contralateral rotations induced by apomorphine in 6-OHDA lesioned group were highly significant (P < 0.001) as compared to the sham group. Moreover, chronic administration of crocin at doses of 30 and 60 mg/kg over six weeks did not change the rotations. The TBARS and nitrite levels in the striatum were also significantly (P < 0.05) increased in lesioned group. Treatment with crocin at a dose of 60 mg/kg significantly decreased the nitrite levels (P < 0.05) in the striatum. CONCLUSION Crocin at a dose of 60 mg/kg could be effective in preventing the nitrosative damage in the striatum. Further investigations using higher doses of crocin is suggested to get the full neuroprotective effects of crocin in Parkinson's disease.
Collapse
Affiliation(s)
- Maryam Hosseini
- Department of Physiology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Ziba Rajaei
- Department of Physiology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran; Neurocognitive Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hojjatallah Alaei
- Isfahan Neurosciences Research Center, Alzahra Hospital, Isfahan University of Medical Sciences, Isfahan, Iran
| | | |
Collapse
|
32
|
Dahl R. A new target for Parkinson's disease: Small molecule SERCA activator CDN1163 ameliorates dyskinesia in 6-OHDA-lesioned rats. Bioorg Med Chem 2016; 25:53-57. [PMID: 27776889 DOI: 10.1016/j.bmc.2016.10.008] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Revised: 10/04/2016] [Accepted: 10/07/2016] [Indexed: 12/24/2022]
Abstract
Endoplasmic reticulum (ER) stress is intimately linked to Parkinson's disease (PD) pathophysiology. Disrupted intracellular calcium homeostasis is a major cause of the ER stress seen in dopaminergic neurons, leading to the cell death and subsequent loss of movement and coordination in patients. Dysfunctional calcium handling proteins play a major role in the promulgation of ER stress in PD. Specifically, compromised sarco/endoplasmic reticulum Ca2+-ATPase (SERCA) has been identified as a major cause of ER stress and neuron loss in PD. We have identified a small molecule activator of SERCA that increases ER calcium content, rescues neurons from ER stress-induced cell death in vitro, and shows significant efficacy in the rat 6-hydroxydopamine (6-OHDA) model of PD. Together, these results support targeting SERCA activation as a viable strategy to develop disease-modifying therapeutics for PD.
Collapse
Affiliation(s)
- Russell Dahl
- Neurodon LLC, 5700 Tanager St., Schererville, IN 46375, USA.
| |
Collapse
|
33
|
Del-Bel E, Bortolanza M, Dos-Santos-Pereira M, Bariotto K, Raisman-Vozari R. l-DOPA-induced dyskinesia in Parkinson's disease: Are neuroinflammation and astrocytes key elements? Synapse 2016; 70:479-500. [DOI: 10.1002/syn.21941] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Revised: 09/06/2016] [Accepted: 09/06/2016] [Indexed: 12/22/2022]
Affiliation(s)
- Elaine Del-Bel
- Department of MFPB-Physiology; FORP, Campus USP, University of São Paulo; Av. Café, s/no Ribeirão Preto SP 14040-904 Brazil
- Center for Interdisciplinary Research on Applied Neurosciences (NAPNA); São Paulo Brazil
- Department of Physiology; FMRP; São Paulo Brazil
- Department of Neurology and Behavioral Neuroscience; FMRP, Campus USP, University of São Paulo; Av. Bandeirantes 13400 Ribeirão Preto SP 14049-900 Brazil
| | - Mariza Bortolanza
- Department of MFPB-Physiology; FORP, Campus USP, University of São Paulo; Av. Café, s/no Ribeirão Preto SP 14040-904 Brazil
- Center for Interdisciplinary Research on Applied Neurosciences (NAPNA); São Paulo Brazil
| | - Maurício Dos-Santos-Pereira
- Department of MFPB-Physiology; FORP, Campus USP, University of São Paulo; Av. Café, s/no Ribeirão Preto SP 14040-904 Brazil
- Center for Interdisciplinary Research on Applied Neurosciences (NAPNA); São Paulo Brazil
- Department of Physiology; FMRP; São Paulo Brazil
| | - Keila Bariotto
- Department of MFPB-Physiology; FORP, Campus USP, University of São Paulo; Av. Café, s/no Ribeirão Preto SP 14040-904 Brazil
- Center for Interdisciplinary Research on Applied Neurosciences (NAPNA); São Paulo Brazil
- Department of Neurology and Behavioral Neuroscience; FMRP, Campus USP, University of São Paulo; Av. Bandeirantes 13400 Ribeirão Preto SP 14049-900 Brazil
| | - Rita Raisman-Vozari
- INSERM UMR 1127, CNRS UMR 7225, UPMC; Thérapeutique Expérimentale de la Neurodégénérescence, Hôpital de la Salpetrière-ICM (Institut du cerveau et de la moelle épinière); Paris France
| |
Collapse
|
34
|
Goswami P, Gupta S, Biswas J, Sharma S, Singh S. Endoplasmic Reticulum Stress Instigates the Rotenone Induced Oxidative Apoptotic Neuronal Death: a Study in Rat Brain. Mol Neurobiol 2015; 53:5384-400. [PMID: 26446018 DOI: 10.1007/s12035-015-9463-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Accepted: 09/25/2015] [Indexed: 12/21/2022]
Abstract
The present study was conducted to evaluate the involvement of endoplasmic reticulum stress in rotenone-induced oxidative neuronal death in rat brain. Rotenone (6 μg/3 μl) was administered intranigrally, unilaterally (right side) in SD rat brain. Neuronal morphology, expression level of tyrosine hydroxylase (TH) and endoplasmic reticulum (ER) stress markers like glucose-regulated protein 78 (GRP78), growth arrest and DNA damage-inducible gene 153 (GADD153), eukaryotic translation initiation factor 2α (p-eIF2α/eIF2α) and cleaved caspase-12 were estimated in the rat brain. Levels of reactive oxygen species (ROS), reduced glutathione (GSH) and enzymatic activities of glutathione peroxidase (GPx) and glutathione reductase (GRd) were estimated to assess the rotenone induced oxidative stress. Apoptotic death of neurons was assessed by estimating the mRNA level of caspase-3. Rotenone administration caused altered neuronal morphology, decreased expression of TH, augmented ROS level, decreased level of GSH and decreased activities of GPx and GRd enzymes which were significantly attenuated with the pretreatment of ER stress inhibitor, salubrinal (1 mg/kg, intraperitoneal). Significantly increased levels of GRP78, GADD, dephosphorylated eIF2α and cleaved caspase-12 was also observed after rotenone administration, which was inhibited with the pretreatment of salubrinal. Rotenone-induced increased mRNA level of caspase-3 was also attenuated by pretreatment of salubrinal. Findings suggested that salubrinal treatment significantly inhibited the rotenone-induced neurotoxicity implicating that ER stress initiates the rotenone-induced oxidative stress and neuronal death.
Collapse
Affiliation(s)
- Poonam Goswami
- Toxicology Division, CSIR-CDRI, Lucknow, 226031, Uttar Pradesh, India.,Academy of Scientific and Innovative Research (AcSIR), Lucknow, 226031, Uttar Pradesh, India
| | - Sonam Gupta
- Toxicology Division, CSIR-CDRI, Lucknow, 226031, Uttar Pradesh, India
| | - Joyshree Biswas
- Toxicology Division, CSIR-CDRI, Lucknow, 226031, Uttar Pradesh, India
| | - Sharad Sharma
- Toxicology Division, CSIR-CDRI, Lucknow, 226031, Uttar Pradesh, India
| | - Sarika Singh
- Toxicology Division, CSIR-CDRI, Lucknow, 226031, Uttar Pradesh, India. .,Academy of Scientific and Innovative Research (AcSIR), Lucknow, 226031, Uttar Pradesh, India.
| |
Collapse
|
35
|
Hernandez-Baltazar D, Zavala-Flores LM, Villanueva-Olivo A. The 6-hydroxydopamine model and parkinsonian pathophysiology: Novel findings in an older model. Neurologia 2015; 32:533-539. [PMID: 26304655 DOI: 10.1016/j.nrl.2015.06.011] [Citation(s) in RCA: 81] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2014] [Revised: 06/09/2015] [Accepted: 06/15/2015] [Indexed: 01/09/2023] Open
Abstract
The neurotoxin 6-hydroxydopamine (6-OHDA) is widely used to induce models of Parkinson's disease (PD). We now know that the model induced by 6-OHDA does not include all PD symptoms, although it does reproduce the main cellular processes involved in PD, such as oxidative stress, neurodegeneration, neuroinflammation, and neuronal death by apoptosis. In this review we analyse the factors affecting the vulnerability of dopaminergic neurons as well as the close relationships between neuroinflammation, neurodegeneration, and apoptosis in the 6-OHDA model. Knowledge of the mechanisms involved in neurodegeneration and cell death in this model is the key to identifying potential therapeutic targets for PD.
Collapse
Affiliation(s)
- D Hernandez-Baltazar
- Cátedra CONACyT, Dirección Adjunta de Desarrollo Científico CONACyT, México, D. F., México; Instituto de Neuroetología, Universidad Veracruzana, Xalapa, Veracruz, México.
| | - L M Zavala-Flores
- Centro de Investigación Biomédica del Noreste, IMSS, Monterrey, Nuevo León, México
| | - A Villanueva-Olivo
- Departamento de Histología, Facultad de Medicina, Universidad Autónoma de Nuevo León, Monterrey, Nuevo León, México
| |
Collapse
|
36
|
Goswami P, Gupta S, Joshi N, Sharma S, Singh S. Astrocyte activation and neurotoxicity: A study in different rat brain regions and in rat C6 astroglial cells. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2015; 40:122-139. [PMID: 26113375 DOI: 10.1016/j.etap.2015.06.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Revised: 05/29/2015] [Accepted: 06/01/2015] [Indexed: 06/04/2023]
Abstract
The present study was conducted to investigate the effect of rotenone on astrocytes activation, their viability and its effect on neuronal death in different brain regions. Rotenone was injected in rat brain by intracerebroventricularly (bilateral) route at dose of 6 μg and 12 μg. In vitro C6 cells were treated with rotenone at concentration of 0.1, 0.25, 0.5, 1 and 2 μM. Rotenone administration to rat brain caused significant astrocytes activation in frontal cortex, cerebellum, cerebellar nucleus, substantia nigra, hypothalamus and hippocampus regions of the rat brain. Rotenone administration also led to significant degeneration of cells in all the studied regions along with altered nuclear morphology assessed by hematoxylin-eosin and cresyl violet staining. Histological staining showed the significantly decreased number of cells in all the studied regions except cerebellar nucleus in dose and time dependant manner. Rotenone administration in the rat brain also caused significant decrease in glutathione levels and augmented nitrite levels. In vitro treatment of rotenone to astrocytic C6 cells caused significantly increased expression of glial fibrillar acidic protein (GFAP) and decreased viability in dose and time dependent manner. Rotenone treatment to C6 cells exhibited significant generation of reactive oxygen species, augmented nitrite level, impaired mitochondrial activity, apoptotic chromatin condensation and DNA damage in comparison to control cells. Findings showed that oxidative stress play a considerable role in rotenone induced astrocyte death that was attenuated with co-treatment of antioxidant melatonin. In conclusion, results showed that rotenone caused significant astrocytes activation, altered nuclear morphology, biochemical alteration and apoptotic cell death in different rat brain regions. In vitro observations in C6 cells showed that rotenone treatment exhibited oxidative stress mediated apoptotic cell death, which was attenuated with co treatment of melatonin.
Collapse
Affiliation(s)
- Poonam Goswami
- Toxicology Division, CSIR-CDRI, Lucknow 226031, India; Academy of Scientific & Innovative Research (AcSIR), India
| | - Sonam Gupta
- Toxicology Division, CSIR-CDRI, Lucknow 226031, India; Academy of Scientific & Innovative Research (AcSIR), India
| | - Neeraj Joshi
- Department of Biochemistry and Biophysics, University of California, San Francisco, CA, USA
| | - Sharad Sharma
- Toxicology Division, CSIR-CDRI, Lucknow 226031, India
| | - Sarika Singh
- Toxicology Division, CSIR-CDRI, Lucknow 226031, India; Academy of Scientific & Innovative Research (AcSIR), India.
| |
Collapse
|
37
|
Pramila B, Kalaivani P, Anita A, Saravana Babu C. L-NAME combats excitotoxicity and recuperates neurological deficits in MCAO/R rats. Pharmacol Biochem Behav 2015; 135:246-53. [PMID: 26093193 DOI: 10.1016/j.pbb.2015.06.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2015] [Revised: 03/21/2015] [Accepted: 06/09/2015] [Indexed: 11/19/2022]
Abstract
PURPOSE OF RESEARCH Since, transient focal cerebral ischaemia exhibits detrimental effect not only during the course of ischaemia but also after the onset of reperfusion, the current study is focussed on identifying the appropriate therapeutic time point at which NG-nitro-l-arginine methyl ester (l-NAME) exerts better neuroprotection. PRINCIPAL RESULTS Pre-ischaemic administration of l-NAME ameliorated neurological deficits much better than the during ischaemic and post-ischaemic groups. Pre-ischaemic l-NAME has also mitigated glutamate excitotoxicity, increased glutamine synthetase activity, ATP and NAD levels, decreased nitrate/nitrite content, down regulated TNF-α and upregulated IL-10 expressions and reduced the cerebral infarction significantly than the during ischaemic and post-ischaemic groups. MAJOR CONCLUSION Current study revealed that l-NAME improved neurological deficit at the pre-ischaemic state in transient focal cerebral ischaemia and has also significantly ameliorated glutamate excitotoxicity. Though l-NAME showed neuroprotective effects when administered at during and post-ischaemia (during reperfusion), it exerts considerable neuroprotection when administered pre-ischaemically.
Collapse
Affiliation(s)
- B Pramila
- Centre for Toxicology and Developmental Research, No.1, Ramachandra Nagar, Sri Ramachandra University, Porur, Chennai 600 116, India; Dr. M.G.R. Educational and Research Institute University, Periyar E.V.R. High Road (NH 4 Highway), Maduravoyal, Chennai 600 095, India.
| | - P Kalaivani
- Centre for Toxicology and Developmental Research, No.1, Ramachandra Nagar, Sri Ramachandra University, Porur, Chennai 600 116, India.
| | - A Anita
- Centre for Toxicology and Developmental Research, No.1, Ramachandra Nagar, Sri Ramachandra University, Porur, Chennai 600 116, India.
| | - C Saravana Babu
- Centre for Toxicology and Developmental Research, No.1, Ramachandra Nagar, Sri Ramachandra University, Porur, Chennai 600 116, India.
| |
Collapse
|
38
|
Gupta S, Goswami P, Biswas J, Joshi N, Sharma S, Nath C, Singh S. 6-Hydroxydopamine and lipopolysaccharides induced DNA damage in astrocytes: involvement of nitric oxide and mitochondria. MUTATION RESEARCH. GENETIC TOXICOLOGY AND ENVIRONMENTAL MUTAGENESIS 2015; 778:22-36. [PMID: 25726145 DOI: 10.1016/j.mrgentox.2014.12.007] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/12/2014] [Revised: 12/22/2014] [Accepted: 12/23/2014] [Indexed: 10/24/2022]
Abstract
The present study was conducted to investigate the effect of the neurotoxins 6-hydroxydopamine and lipopolysaccharide on astrocytes. Rat astrocyte C6 cells were treated with different concentration of 6-hydroxydopamine (6-OHDA)/lipopolysaccharides (LPS) for 24 h. Both neurotoxins significantly decreased the viability of astrocytes, augmented the expression of inducible nitric oxide synthase (iNOS) and the astrocyte marker--glial fibrillar acidic protein. A significantly decreased mitochondrial dehydrogenase activity, mitochondrial membrane potential, augmented reactive oxygen species (ROS) level, caspase-3 mRNA level, chromatin condensation and DNA damage was observed in 6-OHDA/LPS treated astroglial cells. 6-OHDA/LPS treatment also caused the significantly increased expression of iNOS and nitrite level. Findings showed that 6-OHDA/LPS treatment caused mitochondrial dysfunction mediated death of astrocytes, which significantly involve the nitric oxide. Since we have observed significantly increased level of iNOS along with mitochondrial impairment and apoptotic cell death in astrocytes, therefore to validate the role of iNOS, the cells were co-treated with iNOS inhibitor aminoguanidine (AG, 100 μM). Co-treatment of AG significantly attenuated the 6-OHDA/LPS induced cell death, mitochondrial activity, augmented ROS level, chromatin condensation and DNA damage. GFAP and caspase-3 expression were also inhibited with co-treatment of AG, although the extent of inhibition was different in both experimental sets. In conclusion, the findings showed that iNOS mediated increased level of nitric oxide acts as a key regulatory molecule in 6-OHDA/LPS induced mitochondrial dysfunction, DNA damage and apoptotic death of astrocytes.
Collapse
Affiliation(s)
- Sonam Gupta
- Toxicology Division, CSIR-CDRI, Lucknow 226031, India
| | | | | | - Neeraj Joshi
- Center for Gene Regulation in Health and Disease, Department of Biological Sciences, Cleveland State University, Cleveland, OH, USA; Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Sharad Sharma
- Toxicology Division, CSIR-CDRI, Lucknow 226031, India
| | - C Nath
- Toxicology Division, CSIR-CDRI, Lucknow 226031, India
| | - Sarika Singh
- Toxicology Division, CSIR-CDRI, Lucknow 226031, India.
| |
Collapse
|
39
|
Joshi N, Biswas J, Nath C, Singh S. Promising Role of Melatonin as Neuroprotectant in Neurodegenerative Pathology. Mol Neurobiol 2014; 52:330-40. [DOI: 10.1007/s12035-014-8865-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2014] [Accepted: 08/14/2014] [Indexed: 12/14/2022]
|
40
|
Gupta S, Verma DK, Biswas J, Rama Raju KS, Joshi N, Wahajuddin, Singh S. The metabolic enhancer piracetam attenuates mitochondrion-specific endonuclease G translocation and oxidative DNA fragmentation. Free Radic Biol Med 2014; 73:278-90. [PMID: 24882422 DOI: 10.1016/j.freeradbiomed.2014.05.014] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2014] [Revised: 04/13/2014] [Accepted: 05/19/2014] [Indexed: 11/24/2022]
Abstract
This study was performed to investigate the involvement of mitochondrion-specific endonuclease G in piracetam (P)-induced protective mechanisms. Studies have shown the antiapoptotic effects of piracetam but the mechanism of action of piracetam is still an enigma. To assess the involvement of endonuclease G in piracetam-induced protective effects, astrocyte glial cells were treated with lipopolysaccharide (LPS) and piracetam. LPS treatment caused significantly decreased viability, mitochondrial activity, oxidative stress, chromatin condensation, and DNA fragmentation, which were attenuated by piracetam cotreatment. Cotreatment of astrocytes with piracetam showed its significantly time-dependent absorption as observed with high-performance liquid chromatography. Astrocytes treated with piracetam alone showed enhanced mitochondrial membrane potential (MMP) in comparison to control astrocytes. However, in LPS-treated cells no significant alteration in MMP was observed in comparison to control cells. Protein and mRNA levels of the terminal executor of the caspase-mediated pathway, caspase-3, were not altered significantly in LPS or LPS + piracetam-treated astrocytes, whereas endonuclease G was significantly translocated to the nucleus in LPS-treated astrocytes. Piracetam cotreatment attenuated the LPS-induced endonuclease G translocation. In conclusion this study indicates that LPS treatment of astrocytes caused decreased viability, oxidative stress, mitochondrial dysfunction, chromatin condensation, DNA damage, and translocation of endonuclease G to the nucleus, which was inhibited by piracetam cotreatment, confirming that the mitochondrion-specific endonuclease G is one of the factors involved in piracetam-induced protective mechanisms.
Collapse
Affiliation(s)
- Sonam Gupta
- Toxicology Division, CSIR-Central Drug Research Institute, Lucknow 226031, Uttar Pradesh, India
| | - Dinesh Kumar Verma
- Toxicology Division, CSIR-Central Drug Research Institute, Lucknow 226031, Uttar Pradesh, India
| | - Joyshree Biswas
- Toxicology Division, CSIR-Central Drug Research Institute, Lucknow 226031, Uttar Pradesh, India
| | - K Siva Rama Raju
- Pharmacokinetics & Metabolism Division, CSIR-Central Drug Research Institute, Lucknow 226031, Uttar Pradesh, India
| | - Neeraj Joshi
- Center for Gene Regulation in Health and Disease, Department of Biological Sciences, Cleveland State University, Cleveland, Ohio, USA
| | - Wahajuddin
- Pharmacokinetics & Metabolism Division, CSIR-Central Drug Research Institute, Lucknow 226031, Uttar Pradesh, India
| | - Sarika Singh
- Toxicology Division, CSIR-Central Drug Research Institute, Lucknow 226031, Uttar Pradesh, India.
| |
Collapse
|
41
|
Baier CJ, Pallarés ME, Adrover E, Katunar MR, Raisman-Vozari R, Antonelli MC. Intrastriatal 6-OHDA Lesion Differentially Affects Dopaminergic Neurons in the Ventral Tegmental Area of Prenatally Stressed Rats. Neurotox Res 2014; 26:274-84. [DOI: 10.1007/s12640-014-9479-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2013] [Revised: 05/09/2014] [Accepted: 05/27/2014] [Indexed: 01/07/2023]
|
42
|
MSTMP, a Stilbene Derivative, Protects SH-SY5Y Cells Against Oxidative Stress. Can J Neurol Sci 2014; 41:382-8. [PMID: 24718825 DOI: 10.1017/s0317167100017340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
OBJECTIVE the protective effects of a novel stilbene derivative, (e)-2-(3,4,5- trimethoxystyryl)-3,5,6-trimethylpyrazine (MStMp), on hydrogen peroxide (h2o2)-induced human derived neuroblastoma cell (Sh-Sy5y) damage and its molecular mechanisms were investigated. METHODS Sh-Sy5y cells were exposed to 200 μmol.l-1 h2o2 for 12 h. the effect of MStMp on cell viability and apoptosis was assessed by 3-(4,5-dimethyl- thiazol-2-yl)-2,5-diphenyl tetrazolium bromide (Mtt) assay and flow cytometry method. the activities of lactate dehydrogenase (ldh), superoxide dismutase (Sod) and nitric oxide synthetase (noS) and the content of malondialdehyde (Mda), reduced glutathione (gSh) and nitric oxide (no) in cells were determined by commercial kits. the expressions of pro-apoptotic factor caspase-3, caspase-9 and inducible noS (inoS) were detected by Western blotting. intracellular formation of reactive oxygen species (roS) was assessed using 6-carboxy-2’,7’-dichlorofluorescin diacetate (dCfh-da) fluorescent probe. RESULTS MStMp increased the Sh-Sy5y cell viability by inhibition of cell apoptosis induced by h2o2. these effects were accompanied by an increase of Sod activity, gSh level, and a decrease of Mda content. Moreover, MStMp showed stronger effects on inhibition of ldh leakage, apoptotic cells, intracellular roS level and the expression of caspase-3 and caspase-9 than tMp. furthermore, MStMp induced a decrease of no level and the activity of inoS, tnoS in a time-dependent manner. CONCLUSIONS MStMp prevents h2o2-induced cell injury through anti-oxidation and anti-apoptosis via roS-no pathway.
Collapse
|
43
|
Does Restraining Nitric Oxide Biosynthesis Rescue from Toxins-Induced Parkinsonism and Sporadic Parkinson's Disease? Mol Neurobiol 2013; 49:262-75. [DOI: 10.1007/s12035-013-8517-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2013] [Accepted: 07/16/2013] [Indexed: 12/21/2022]
|
44
|
Rao VS, Carvalho AC, Trevisan MTS, Andrade GM, Nobre HV, Moraes MO, Iury HIMH, Morais TC, Santos FA. Mangiferin ameliorates 6-hydroxydopamineinduced cytotoxicity and oxidative stress in ketamine model of schizophrenia. Pharmacol Rep 2012; 64:848-56. [DOI: 10.1016/s1734-1140(12)70879-4] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2011] [Revised: 04/23/2012] [Indexed: 12/19/2022]
|
45
|
Abstract
SIGNIFICANCE Parkinson's disease (PD) is a neurodegenerative disorder characterized, in part, by the progressive and selective loss of dopaminergic neuron cell bodies within the substantia nigra pars compacta (SNpc) and the associated deficiency of the neurotransmitter dopamine (DA) in the striatum, which gives rise to the typical motor symptoms of PD. The mechanisms that contribute to the induction and progressive cell death of dopaminergic neurons in PD are multi-faceted and remain incompletely understood. Data from epidemiological studies in humans and molecular studies in genetic, as well as toxin-induced animal models of parkinsonism, indicate that mitochondrial dysfunction occurs early in the pathogenesis of both familial and idiopathic PD. In this review, we provide an overview of toxin models of mitochondrial dysfunction in experimental Parkinson's disease and discuss mitochondrial mechanisms of neurotoxicity. RECENT ADVANCES A new toxin model using the mitochondrial toxin trichloroethylene was recently described and novel methods, such as intranasal exposure to toxins, have been explored. Additionally, recent research conducted in toxin models of parkinsonism provides an emerging emphasis on extranigral aspects of PD pathology. CRITICAL ISSUES Unfortunately, none of the existing animal models of experimental PD completely mimics the etiology, progression, and pathology of human PD. FUTURE DIRECTIONS Continued efforts to optimize established animal models of parkinsonism, as well as the development and characterization of new animal models are essential, as there still remains a disconnect in terms of translating mechanistic observations in animal models of experimental PD into bona fide disease-modifying therapeutics for human PD patients.
Collapse
Affiliation(s)
- Terina N Martinez
- Pittsburgh Institute for Neurodegenerative Diseases, Department of Neurology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | | |
Collapse
|
46
|
Butler EK, Voigt A, Lutz AK, Toegel JP, Gerhardt E, Karsten P, Falkenburger B, Reinartz A, Winklhofer KF, Schulz JB. The mitochondrial chaperone protein TRAP1 mitigates α-Synuclein toxicity. PLoS Genet 2012; 8:e1002488. [PMID: 22319455 PMCID: PMC3271059 DOI: 10.1371/journal.pgen.1002488] [Citation(s) in RCA: 113] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2011] [Accepted: 12/02/2011] [Indexed: 01/13/2023] Open
Abstract
Overexpression or mutation of α-Synuclein is associated with protein aggregation and interferes with a number of cellular processes, including mitochondrial integrity and function. We used a whole-genome screen in the fruit fly Drosophila melanogaster to search for novel genetic modifiers of human [A53T]α-Synuclein-induced neurotoxicity. Decreased expression of the mitochondrial chaperone protein tumor necrosis factor receptor associated protein-1 (TRAP1) was found to enhance age-dependent loss of fly head dopamine (DA) and DA neuron number resulting from [A53T]α-Synuclein expression. In addition, decreased TRAP1 expression in [A53T]α-Synuclein-expressing flies resulted in enhanced loss of climbing ability and sensitivity to oxidative stress. Overexpression of human TRAP1 was able to rescue these phenotypes. Similarly, human TRAP1 overexpression in rat primary cortical neurons rescued [A53T]α-Synuclein-induced sensitivity to rotenone treatment. In human (non)neuronal cell lines, small interfering RNA directed against TRAP1 enhanced [A53T]α-Synuclein-induced sensitivity to oxidative stress treatment. [A53T]α-Synuclein directly interfered with mitochondrial function, as its expression reduced Complex I activity in HEK293 cells. These effects were blocked by TRAP1 overexpression. Moreover, TRAP1 was able to prevent alteration in mitochondrial morphology caused by [A53T]α-Synuclein overexpression in human SH-SY5Y cells. These results indicate that [A53T]α-Synuclein toxicity is intimately connected to mitochondrial dysfunction and that toxicity reduction in fly and rat primary neurons and human cell lines can be achieved using overexpression of the mitochondrial chaperone TRAP1. Interestingly, TRAP1 has previously been shown to be phosphorylated by the serine/threonine kinase PINK1, thus providing a potential link of PINK1 via TRAP1 to α-Synuclein.
Collapse
Affiliation(s)
- Erin K. Butler
- Department of Neurology, University Medical Center, RWTH Aachen, Aachen, Germany
- Department of Neurodegeneration and Restorative Research, Center Molecular Physiology of the Brain (CMPB), Georg-August University Göttingen, Göttingen, Germany
- Göttingen Graduate School for Neurosciences and Molecular Biology (GGNB), Göttingen, Germany
| | - Aaron Voigt
- Department of Neurology, University Medical Center, RWTH Aachen, Aachen, Germany
| | - A. Kathrin Lutz
- Neurobiochemistry, Adolf-Butenandt-Institute, Ludwig Maximilians University, Munich, Germany
| | - Jane P. Toegel
- Department of Neurology, University Medical Center, RWTH Aachen, Aachen, Germany
| | - Ellen Gerhardt
- Department of Neurodegeneration and Restorative Research, Center Molecular Physiology of the Brain (CMPB), Georg-August University Göttingen, Göttingen, Germany
| | - Peter Karsten
- Department of Neurology, University Medical Center, RWTH Aachen, Aachen, Germany
| | - Björn Falkenburger
- Department of Neurology, University Medical Center, RWTH Aachen, Aachen, Germany
| | - Andrea Reinartz
- Department of Pathology, University Medical Center, RWTH Aachen, Aachen, Germany
| | - Konstanze F. Winklhofer
- Neurobiochemistry, Adolf-Butenandt-Institute, Ludwig Maximilians University, Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Jörg B. Schulz
- Department of Neurology, University Medical Center, RWTH Aachen, Aachen, Germany
- Jülich-Aachen Research Alliance (JARA) Brain, Jülich/Aachen, Germany
- * E-mail:
| |
Collapse
|
47
|
Singh S, Swarnkar S, Goswami P, Nath C. Astrocytes and microglia: responses to neuropathological conditions. Int J Neurosci 2011; 121:589-97. [PMID: 21827229 DOI: 10.3109/00207454.2011.598981] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Activated astrocytes and microglia, hallmark of neurodegenerative diseases release different factors like array of pro and anti-inflammatory cytokines, free radicals, anti-oxidants, and neurotrophic factors during neurodegeneration which further contribute to neuronal death as well as in survival mechanisms. Astrocytes act as double-edged sword exerting both detrimental and neuroprotective effects while microglial cells are attributed more in neurodegenerative mechanisms. The dual and insufficient knowledge about the precise role of glia in neurodegeneration showed the need for further investigations and thorough review of the function of glia in neurodegeneration. In this review, we consolidate and categorize the glia-released factors which contribute in degenerative and protective mechanisms during neuropathological conditions.
Collapse
Affiliation(s)
- Sarika Singh
- Toxicology Division, Central Drug Research Institute-CSIR-CDRI, Lucknow, India.
| | | | | | | |
Collapse
|
48
|
Tufekci KU, Genc S, Genc K. The endotoxin-induced neuroinflammation model of Parkinson's disease. PARKINSON'S DISEASE 2011; 2011:487450. [PMID: 21331154 PMCID: PMC3034925 DOI: 10.4061/2011/487450] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/14/2010] [Revised: 11/18/2010] [Accepted: 12/16/2010] [Indexed: 01/22/2023]
Abstract
Parkinson's disease (PD) is a common neurodegenerative disorder characterized by the progressive loss of dopaminergic (DA) neurons in the substantia nigra. Although the exact cause of the dopaminergic neurodegeneration remains elusive, recent postmortem and experimental studies have revealed an essential role for neuroinflammation that is initiated and driven by activated microglial and infiltrated peripheral immune cells and their neurotoxic products (such as proinflammatory cytokines, reactive oxygen species, and nitric oxide) in the pathogenesis of PD. A bacterial endotoxin-based experimental model of PD has been established, representing a purely inflammation-driven animal model for the induction of nigrostriatal dopaminergic neurodegeneration. This model, by itself or together with genetic and toxin-based animal models, provides an important tool to delineate the precise mechanisms of neuroinflammation-mediated dopaminergic neuron loss. Here, we review the characteristics of this model and the contribution of neuroinflammatory processes, induced by the in vivo administration of bacterial endotoxin, to neurodegeneration. Furthermore, we summarize the recent experimental therapeutic strategies targeting endotoxin-induced neuroinflammation to elicit neuroprotection in the nigrostriatal dopaminergic system. The potential of the endotoxin-based PD model in the development of an early-stage specific diagnostic biomarker is also emphasized.
Collapse
Affiliation(s)
- Kemal Ugur Tufekci
- Department of Neuroscience, Health Science Institute, Dokuz Eylul University, Inciralti, 35340 Izmir, Turkey
| | - Sermin Genc
- Department of Neuroscience, Health Science Institute, Dokuz Eylul University, Inciralti, 35340 Izmir, Turkey
| | - Kursad Genc
- Department of Neuroscience, Health Science Institute, Dokuz Eylul University, Inciralti, 35340 Izmir, Turkey
| |
Collapse
|
49
|
Kim MK, Kim SC, Kang JI, Hyun JH, Boo HJ, Eun SY, Park DB, Yoo ES, Kang HK, Kang JH. 6-Hydroxydopamine-induced PC12 cell death is mediated by MEF2D down-regulation. Neurochem Res 2010; 36:223-31. [PMID: 21057871 DOI: 10.1007/s11064-010-0309-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/25/2010] [Indexed: 12/15/2022]
Abstract
Recently, it was reported that in a 4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) model, neuronal cell death is associated with the cdk5-mediated hyperphosphorylation of myocyte enhancer factor 2 (MEF2), a transcription factor that is critically required for neuronal survival. In the present study, we investigated the possible involvement of cdk5-mediated MEF2D down-regulation on 6-hydroxydopamine (6-OHDA)-induced PC12 cell death. 6-OHDA was found to significantly increase nitric oxide (NO) production and to induce apoptosis in a time-dependent manner in PC12 cells. Furthermore, 6-OHDA was found to markedly reduce MEF2D levels under conditions that could induce PC12 cell apoptosis. In addition, PC12 cell death and MEF2D degradation by 6-OHDA were prevented by the cdk5 inhibitor roscovitine, but roscovitine could not restore the 6-OHDA-induced inactivation of Akt. These results suggest that the cell death and MEF2D degradation caused by 6-OHDA are dependent on cdk5 activity. On the other hand, roscovitine enhanced the 6-OHDA-induced activations of ERK1/2 and JNK, but reduced the 6-OHDA-induced activation of p38. These results suggest that PC12 cell death by 6-OHDA appears to be regulated by the down-regulation of MEF2D via some interaction between cdk5 and MAP kinase.
Collapse
Affiliation(s)
- Min-Kyoung Kim
- Department of Pharmacology, School of Medicine, Institute of Medical Sciences, Jeju National University, Jeju, South Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|