1
|
Lin J, Zhang G, Lou B, Sun Y, Jia X, Wang M, Zhou J, Xia Z. Identification of copper metabolism-related markers in Parkinson's disease. Ann Med 2024; 56:2425064. [PMID: 39552415 PMCID: PMC11574951 DOI: 10.1080/07853890.2024.2425064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 05/16/2024] [Accepted: 08/30/2024] [Indexed: 11/19/2024] Open
Abstract
OBJECTIVES This study aimed to identify key genes related to copper metabolism in Parkinson's disease (PD), providing insight into their roles in disease progression. METHODS Using bioinformatic analyses, the study identified hub genes related to copper metabolism in PD patients. Differentially expressed genes (DEGs) were identified using the limma package, and copper-metabolism-related genes (CMRGs) were sourced from the Genecard database. Immune cell-related genes were derived through immune infiltration and Weighted Gene Co-expression Network Analysis (WGCNA). Hub genes were pinpointed by integrating DEGs, CMRGs, and immune cell-related genes. Functional analyses included Receiver Operating Characteristic (ROC) analysis, Ingenuity Pathway Analysis (IPA), and networks for miRNA-mRNA-transcription factor (TF), Competitive Endogenous RNA (ceRNA), and hub gene-drug interactions. Validation was performed in cerebrospinal fluid (CSF) samples from PD patients, while in vitro experiments utilized GBE1- overexpressing SH-SY5Y cells to examine cell proliferation, migration, and viability. RESULTS Nine hub genes (HPRT1, GLS, SNCA, MDH1, GBE1, DDC, STXBP1, ACHE, and AGTR1) were identified from 753 CMRGs, 416 DEGs, and 951 immune cell-related genes. ROC analysis showed high predictive accuracy for PD, and principal component analysis (PCA) effectively distinguished PD patients from controls. IPA identified 20 significant pathways, and various networks highlighted miRNA, TF, and drug interactions with the hub genes. Hub gene expression was validated in PD CSF samples. GBE1-overexpressing cells displayed enhanced proliferation, migration, and viability. CONCLUSIONS The study identified nine copper metabolism-related genes as potential therapeutic targets in PD, highlighting their relevance in PD pathology and possible treatment pathways.
Collapse
Affiliation(s)
- Jie Lin
- Department of Joint Laboratory for Translational Medicine Research, Liaocheng People's Hospital, Liaocheng, P.R. China
- School of Basic Medicine Sciences, Shandong University, Jinan,P.R. China
| | - Guifeng Zhang
- Department of Neurology, Liaocheng People's Hospital and Liaocheng Hospital Affiliated to Shandong First Medical University, Liaocheng, P.R. China
| | - Bo Lou
- Department of Neurology, The Third People's Hospital of Liaocheng, Liaocheng, P.R. China
| | - Yi Sun
- Department of Sports Medicine, Peking University Shenzhen Hospital, Shenzhen, P.R. China
| | - Xiaodong Jia
- Department of Joint Laboratory for Translational Medicine Research, Liaocheng People's Hospital, Liaocheng, P.R. China
| | - Meidan Wang
- Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Jing Zhou
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, P.R. China
| | - Zhangyong Xia
- Department of Neurology, Liaocheng People's Hospital, Shandong University, Jinan, P.R. China
- Department of Neurology, The Second People's Hospital of Liaocheng, Liaocheng, P.R. China
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, Shandong Sub-Centre, Liaocheng, P.R. China
| |
Collapse
|
2
|
Shnayder NA, Grechkina VV, Khasanova AK, Bochanova EN, Dontceva EA, Petrova MM, Asadullin AR, Shipulin GA, Altynbekov KS, Al-Zamil M, Nasyrova RF. Therapeutic and Toxic Effects of Valproic Acid Metabolites. Metabolites 2023; 13:metabo13010134. [PMID: 36677060 PMCID: PMC9862929 DOI: 10.3390/metabo13010134] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 01/11/2023] [Accepted: 01/12/2023] [Indexed: 01/18/2023] Open
Abstract
Valproic acid (VPA) and its salts are psychotropic drugs that are widely used in neurological diseases (epilepsy, neuropathic pain, migraine, etc.) and psychiatric disorders (schizophrenia, bipolar affective disorder, addiction diseases, etc.). In addition, the indications for the appointment of valproate have been expanding in recent years in connection with the study of new mechanisms of action of therapeutic and toxic metabolites of VPA in the human body. Thus, VPA is considered a component of disease-modifying therapy for multiple tumors, neurodegenerative diseases (Huntington's disease, Parkinson's disease, Duchenne progressive dystrophy, etc.), and human immunodeficiency syndrome. The metabolism of VPA is complex and continues to be studied. Known pathways of VPA metabolism include: β-oxidation in the tricarboxylic acid cycle (acetylation); oxidation with the participation of cytochrome P-450 isoenzymes (P-oxidation); and glucuronidation. The complex metabolism of VPA explains the diversity of its active and inactive metabolites, which have therapeutic, neutral, or toxic effects. It is known that some active metabolites of VPA may have a stronger clinical effect than VPA itself. These reasons explain the relevance of this narrative review, which summarizes the results of studies of blood (serum, plasma) and urinary metabolites of VPA from the standpoint of the pharmacogenomics and pharmacometabolomics. In addition, a new personalized approach to assessing the cumulative risk of developing VPA-induced adverse reactions is presented and ways for their correction are proposed depending on the patient's pharmacogenetic profile and the level of therapeutic and toxic VPA metabolites in the human body fluids (blood, urine).
Collapse
Affiliation(s)
- Natalia A. Shnayder
- Institute of Personalized Psychiatry and Neurology, Shared Core Facilities, V.M. Bekhterev National Medical Research Centre for Psychiatry and Neurology, 192019 Saint Petersburg, Russia
- Shared Core Facilities “Molecular and Cell Technologies”, V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, 660022 Krasnoyarsk, Russia
- Correspondence: (N.A.S.); (R.F.N.); Tel.: +7-(812)-620-0222 (N.A.S. & R.F.N.)
| | - Violetta V. Grechkina
- Institute of Personalized Psychiatry and Neurology, Shared Core Facilities, V.M. Bekhterev National Medical Research Centre for Psychiatry and Neurology, 192019 Saint Petersburg, Russia
| | - Aiperi K. Khasanova
- Department of Psychiatry, Russian Medical Academy for Continual Professional Education, 125993 Moscow, Russia
| | - Elena N. Bochanova
- Shared Core Facilities “Molecular and Cell Technologies”, V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, 660022 Krasnoyarsk, Russia
| | - Evgenia A. Dontceva
- Shared Core Facilities “Molecular and Cell Technologies”, V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, 660022 Krasnoyarsk, Russia
| | - Marina M. Petrova
- Shared Core Facilities “Molecular and Cell Technologies”, V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, 660022 Krasnoyarsk, Russia
| | - Azat R. Asadullin
- Department of Psychiatry and Addiction, Bashkir State Medical University, 45000 Ufa, Russia
| | - German A. Shipulin
- Centre for Strategic Planning and Management of Biomedical Health Risks, 119121 Moscow, Russia
| | - Kuanysh S. Altynbekov
- Republican Scientific and Practical Center of Mental Health, Almaty 050022, Kazakhstan
- Department of Psychiatry and Narcology, S.D. Asfendiarov Kazakh National Medical University, Almaty 050022, Kazakhstan
| | - Mustafa Al-Zamil
- Department of Physiotherapy, Faculty of Continuing Medical Education, Peoples’ Friendship University of Russia, 11798 Moscow, Russia
| | - Regina F. Nasyrova
- Institute of Personalized Psychiatry and Neurology, Shared Core Facilities, V.M. Bekhterev National Medical Research Centre for Psychiatry and Neurology, 192019 Saint Petersburg, Russia
- Correspondence: (N.A.S.); (R.F.N.); Tel.: +7-(812)-620-0222 (N.A.S. & R.F.N.)
| |
Collapse
|
3
|
Sharma M, Malim FM, Goswami A, Sharma N, Juvvalapalli SS, Chatterjee S, Kate AS, Khairnar A. Neuroprotective Effect of Swertiamarin in a Rotenone Model of Parkinson's Disease: Role of Neuroinflammation and Alpha-Synuclein Accumulation. ACS Pharmacol Transl Sci 2022; 6:40-51. [PMID: 36654754 PMCID: PMC9841796 DOI: 10.1021/acsptsci.2c00120] [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: 06/23/2022] [Indexed: 12/15/2022]
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disease with no permanent cure affecting around 1% of the population over 65. There is an urgency to search for a disease-modifying agent with fewer untoward effects. PD pathology involves the accumulation of toxic alpha-synuclein (α-syn) and neuronal inflammation leading to the degeneration of dopaminergic (DAergic) neurons. Swertiamarin (SWE), a well-studied natural product, possesses a strong anti-inflammatory effect. It is a secoiridoid glycoside isolated from Enicostemma littorale Blume. SWE showed a reversal effect on the α-syn accumulation in the 6-hydroxydopamine (6-OHDA)-induced Caenorhabditis elegans model of PD. However, there are no reports in the literature citing the effect of SWE as a neuroprotective agent in rodents. The present study aimed to evaluate the anti-inflammatory activity of SWE against lipopolysaccharide (LPS)-induced C6 glial cell activation and its neuroprotective effect in the intrastriatal rotenone mouse PD model. SWE treatment showed a significant reduction in interleukin-6 (IL-6), tumor necrosis factor-α (TNF-α), and interleukin-1β (IL-1β) levels in LPS-induced C6 glial cell activation. Further, our studies demonstrated the suppression of microglial and astroglial activation in substantia nigra (SN) after administration of SWE (100 mg/kg, intraperitoneally) in a rotenone mouse model. Moreover, SWE alleviated the rotenone-induced α-syn overexpression in the striatum and SN. SWE ameliorated the motor impairment against rotenone-induced neurotoxicity and mitigated the loss of DAergic neurons in the nigrostriatal pathway. Therefore, SWE has the potential to develop as an adjunct therapy for PD, but it warrants further mechanistic studies.
Collapse
Affiliation(s)
- Monika Sharma
- Department
of Pharmacology and Toxicology, National
Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gujarat 382355 India
| | - Fehmina Mushtaque Malim
- Department
of Pharmacology and Toxicology, National
Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gujarat 382355 India
| | - Ashutosh Goswami
- Department
of Natural Products, National Institute
of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gujarat 382355 India
| | - Nishant Sharma
- Department
of Pharmacology and Toxicology, National
Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gujarat 382355 India
| | - Sai Sowmya Juvvalapalli
- Department
of Natural Products, National Institute
of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gujarat 382355 India
| | - Sayan Chatterjee
- Department
of Pharmacology and Toxicology, National
Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gujarat 382355 India
| | - Abhijeet S. Kate
- Department
of Natural Products, National Institute
of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gujarat 382355 India,; . Phone: +79 66745555
| | - Amit Khairnar
- Department
of Pharmacology and Toxicology, National
Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gujarat 382355 India,International
Clinical Research Center, St. Anne’s
University Hospital Brno, Brno 656 91 Czech Republic,..
Phone: +91 9284349396
| |
Collapse
|
4
|
Ilie OD, Duta R, Balmus IM, Savuca A, Petrovici A, Nita IB, Antoci LM, Jijie R, Mihai CT, Ciobica A, Nicoara M, Popescu R, Dobrin R, Solcan C, Trifan A, Stanciu C, Doroftei B. Assessing the Neurotoxicity of a Sub-Optimal Dose of Rotenone in Zebrafish ( Danio rerio) and the Possible Neuroactive Potential of Valproic Acid, Combination of Levodopa and Carbidopa, and Lactic Acid Bacteria Strains. Antioxidants (Basel) 2022; 11:2040. [PMID: 36290763 PMCID: PMC9598446 DOI: 10.3390/antiox11102040] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 10/03/2022] [Accepted: 10/13/2022] [Indexed: 11/20/2022] Open
Abstract
Parkinson's disease (PD) is an enigmatic neurodegenerative disorder that is currently the subject of extensive research approaches aiming at deepening the understanding of its etiopathophysiology. Recent data suggest that distinct compounds used either as anticonvulsants or agents usually used as dopaminergic agonists or supplements consisting of live active lactic acid bacteria strains might alleviate and improve PD-related phenotypes. This is why we aimed to elucidate how the administration of rotenone (ROT) disrupts homeostasis and the possible neuroactive potential of valproic acid (VPA), antiparkinsonian agents (levodopa and carbidopa - LEV+CARB), and a mixture of six Lactobacillus and three Bifidobacterium species (PROBIO) might re-establish the optimal internal parameters. ROT causes significant changes in the central nervous system (CNS), notably reduced neurogenesis and angiogenesis, by triggering apoptosis, reflected by the increased expression of PARKIN and PINK1 gene(s), low brain dopamine (DA) levels, and as opposed to LRRK2 and SNCA compared with healthy zebrafish. VPA, LEV/CARB, and PROBIO sustain neurogenesis and angiogenesis, manifesting a neuroprotective role in diminishing the effect of ROT in zebrafish. Interestingly, none of the tested compounds influenced oxidative stress (OS), as reflected by the level of malondialdehyde (MDA) level and superoxide dismutase (SOD) enzymatic activity revealed in non-ROT-exposed zebrafish. Overall, the selected concentrations were enough to trigger particular behavioral patterns as reflected by our parameters of interest (swimming distance (mm), velocity (mm/s), and freezing episodes (s)), but sequential testing is mandatory to decipher whether they exert an inhibitory role following ROT exposure. In this way, we further offer data into how ROT may trigger a PD-related phenotype and the possible beneficial role of VPA, LEV+CARB, and PROBIO in re-establishing homeostasis in Danio rerio.
Collapse
Affiliation(s)
- Ovidiu-Dumitru Ilie
- Department of Biology, Faculty of Biology, “Alexandru Ioan Cuza” University, Carol I Avenue, no 20A, 700505 Iasi, Romania
| | - Raluca Duta
- Department of Biology, Faculty of Biology, “Alexandru Ioan Cuza” University, Carol I Avenue, no 20A, 700505 Iasi, Romania
| | - Ioana-Miruna Balmus
- Department of Exact and Natural Sciences, Institute of Interdisciplinary Research, “Alexandru Ioan Cuza” University, Carol I Avenue, no 11, 700506 Iasi, Romania
- Doctoral School of Biology, Faculty of Biology, “Alexandru Ioan Cuza” University, Carol I Avenue, 20A, 700506 Iasi, Romania
| | - Alexandra Savuca
- Doctoral School of Geosciences, Faculty of Geography-Geology, “Alexandru Ioan Cuza” University, Carol I Avenue, no 20A, 700505 Iasi, Romania
| | - Adriana Petrovici
- Department of Molecular Biology, Histology and Embryology, Faculty of Veterinary Medicine, University of Life Sciences “Ion Ionescu de la Brad”, Mihail Sadoveanu Street, no 3, 700490 Iasi, Romania
| | - Ilinca-Bianca Nita
- Faculty of Medicine, University of Medicine and Pharmacy “Grigore T. Popa”, University Street, no 16, 700115 Iasi, Romania
| | - Lucian-Mihai Antoci
- Department of Medical Genetics, University of Medicine and Pharmacy “Grigore T. Popa”, University Street, no 16, 700115 Iasi, Romania
| | - Roxana Jijie
- Research Center on Advanced Materials and Technologies, Department of Exact and Natural Sciences, Institute of Inderdisciplinary Research, “Alexandru Ioan Cuza” University, Carol I Avenue, no 11, 700506 Iasi, Romania
| | - Cosmin-Teodor Mihai
- Advanced Research and Development Center for Experimental Medicine (CEMEX), University of Medicine and Pharmacy “Grigore T. Popa”, University Street, no 16, 700115 Iasi, Romania
| | - Alin Ciobica
- Department of Biology, Faculty of Biology, “Alexandru Ioan Cuza” University, Carol I Avenue, no 20A, 700505 Iasi, Romania
| | - Mircea Nicoara
- Department of Biology, Faculty of Biology, “Alexandru Ioan Cuza” University, Carol I Avenue, no 20A, 700505 Iasi, Romania
- Doctoral School of Geosciences, Faculty of Geography-Geology, “Alexandru Ioan Cuza” University, Carol I Avenue, no 20A, 700505 Iasi, Romania
| | - Roxana Popescu
- Department of Medical Genetics, University of Medicine and Pharmacy “Grigore T. Popa”, University Street, no 16, 700115 Iasi, Romania
- Department of Medical Genetics, “Saint Mary” Emergency Children’s Hospital, Vasile Lupu Street, no 62, 700309 Iasi, Romania
| | - Romeo Dobrin
- Department of Psychiatry, University of Medicine and Pharmacy “Grigore T. Popa”, University Street, no 16, 700115 Iasi, Romania
| | - Carmen Solcan
- Department of Molecular Biology, Histology and Embryology, Faculty of Veterinary Medicine, University of Life Sciences “Ion Ionescu de la Brad”, Mihail Sadoveanu Street, no 3, 700490 Iasi, Romania
| | - Anca Trifan
- Department of Gastroenterology, University of Medicine and Pharmacy “Grigore T. Popa”, University Street, no 16, 700115 Iasi, Romania
- Institute of Gastroenterology and Hepatology, “St. Spiridon” Emergency Hospital, Independence Avenue, no 1, 700111 Iasi, Romania
| | - Carol Stanciu
- Department of Gastroenterology, University of Medicine and Pharmacy “Grigore T. Popa”, University Street, no 16, 700115 Iasi, Romania
- Institute of Gastroenterology and Hepatology, “St. Spiridon” Emergency Hospital, Independence Avenue, no 1, 700111 Iasi, Romania
| | - Bogdan Doroftei
- Faculty of Medicine, University of Medicine and Pharmacy “Grigore T. Popa”, University Street, no 16, 700115 Iasi, Romania
| |
Collapse
|
5
|
Assessing Anti-Social and Aggressive Behavior in a Zebrafish ( Danio rerio) Model of Parkinson's Disease Chronically Exposed to Rotenone. Brain Sci 2022; 12:brainsci12070898. [PMID: 35884705 PMCID: PMC9313068 DOI: 10.3390/brainsci12070898] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Revised: 07/03/2022] [Accepted: 07/05/2022] [Indexed: 11/17/2022] Open
Abstract
Background: Rotenone (ROT) is currently being used in various research fields, especially neuroscience. Separated from other neurotoxins, ROT induces a Parkinson’s disease (PD)-related phenotype that mimics the associated clinical spectrum by directly entering the central nervous system (CNS). It easily crosses through the blood−brain barrier (BBB) and accumulates in mitochondria. Unfortunately, most of the existing data focus on locomotion. This is why the present study aimed to bring novel evidence on how ROT alone or in combination with different potential ant(agonists) might influence the social and aggressive behavior using the counterclockwise rotation as a neurological pointer. Material and Methods: Thus, we exposed zebrafish to ROT—2.5 µg/L, valproic acid (VPA)—0.5 mg/mL, anti-parkinsonian drugs (LEV/CARB)—250 mg + 25 mg, and probiotics (PROBIO)—3 g for 32 days by assessing the anti-social profile and mirror tests and counterclockwise rotation every 4 days to avoid chronic stress. Results: We observed an abnormal pattern in the counterclockwise rotation only in the (a) CONTROL, (c) LEV/CARB, and (d) PROBIO groups, from both the top and side views, this indicating a reaction to medication and supplements administered or a normal intrinsic feature due to high levels of stress/anxiety (p < 0.05). Four out of eight studied groups—(b) VPA, (c) LEV/CARB, (e) ROT, and (f) ROT + VPA—displayed an impaired, often antithetical behavior demonstrated by long periods of time on distinct days spent on the right and the central arm (p < 0.05, 0.005, and 0.0005). Interestingly, groups (d) PROBIO, (g) ROT + LEV/CARB, and (h) ROT + PROBIO registered fluctuations but not significant ones in contrast with the above groups (p > 0.05). Except for groups (a) CONTROL and (d) PROBIO, where a normalized trend in terms of behavior was noted, the rest of the experimental groups exhibited exacerbated levels of aggression (p < 0.05, 0.005, and 0.001) not only near the mirror but as an overall reaction (p < 0.05, 0.005, and 0.001). Conclusions: The (d) PROBIO group showed a significant improvement compared with (b) VPA, (c) LEV/CARB, and ROT-treated zebrafish (e−h). Independently of the aggressive-like reactions and fluctuations among the testing day(s) and groups, ROT disrupted the social behavior, while VPA promoted a specific typology in contrast with LEV/CARB.
Collapse
|
6
|
Makav M, Eroğlu HA. Recuperative effect of estrogen on rotenone-induced experimental model of Parkinson's disease in rats. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2021; 28:21266-21275. [PMID: 33410082 DOI: 10.1007/s11356-020-11985-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Accepted: 12/06/2020] [Indexed: 06/12/2023]
Abstract
Parkinson's disease (PD) is described as the loss of dopaminergic neurons located in the substantia nigra (SN) region of the brain and a progressive motor failure. Increased frequency of PD in women, especially after menopause, suggests the effect of estrogen. This view has been supported with empirical studies. Therefore, the effect of estrogen in an experimental model of Parkinson's disease induced by rotenone was investigated. A total of 32 female Wistar Albino rats were randomly assigned to four groups (control group, ovariectomy group, Parkinson's group, Parkinson's + estrogen group). The Parkinson's group received rotenone subcutanously at the dose of 2.5 mg/kg bw, on the 1st, 2nd, 3rd 4th, 6th, 9th, 12th, 15th, 18th, and 21st days animals in the Parkinson's + estrogen group received retonon as in the Parkinson's group and was additionally subcutaneously given estrogen (implant containing 0.5 mg 17 β-estradiol lasting for 21 days). The rats were subjected to rotarod, pole, and swimming tests at the end of the experiment for comparison of their motor activities, and then, histopathological and biochemical analyses were performed on the tissues that were extracted. The rotarod results revealed that Parkinson's group had the shortest time (32.33 ± 3.98 sn) than the groups of control (92.50 ± 12.60 s) ovariectomy (71.42 ± 10.58 s), and Parkinson's + estrogen (71.37 ± 9.26 s). The results of pole disclosed that return and landing time prolonged for Parkinson's group when compared with other groups (return time for control 2.98 ± 0.38 s, ovariectomy 3.02 ± 0.75 s, Parkinson 5.91 ± 0.33 s, Parkinson's + estrogen 3.48 ± 0.42 s and landing time for control 5.30 ± 0.59 s, ovariectomy 5.45 ± 0.73 s, Parkinson 9.80 ± 0.90 s, Parkinson's + estrogen 5.37 ± 1.02 s). Parkinson's group had longest (90.71 ± 12.56 s) swimming time to reach the target when compared with control (33.16 ± 8.68 s), ovariectomy (47.37 ± 12.19 s), and Parkinson's + estrogen (49.82 ± 5.78 s). Histopathological examination indicated a significant difference in tyrosine hydroxylase-stained cells (dopaminergic neurons and dopamine) between the Parkinson's + estrogen group and the Parkinson's group. The biochemical analyses of Caspas-3 activation in SN and striatum (STR) was significantly different between the Parkinson's + estrogen group and the Parkinson's group, but this difference was not observed in STR while evaluating Bcl-2. The results of this study suggested that estrogen may have a recuperative effect on PD.
Collapse
Affiliation(s)
- Mustafa Makav
- Department of Physiology, Faculty of Veterinary Medicine, Kafkas University, Paşaçayırı Campus, TR-36100, Kars, Turkey.
| | - Hüseyin Avni Eroğlu
- Department of Physiology, Faculty of Medicine, Çanakkale Onsekiz Mart University, Çanakkale, Turkey
| |
Collapse
|
7
|
Li M, Hu K, Lin D, Wang Z, Xu M, Huang J, Chen Z, Zhang Y, Yin L, You R, Li CH, Guan YQ. Synthesis of Double Interfering Biodegradable Nano-MgO Micelle Composites and Their Effect on Parkinson's Disease. ACS Biomater Sci Eng 2021; 7:1216-1229. [PMID: 33560819 DOI: 10.1021/acsbiomaterials.0c01474] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Although gene therapy targeting the α-synuclein gene (SNCA) has achieved outstanding results in the treatment of Parkinson's disease (PD), the lack of a suitable gene delivery system and inadequate therapeutic effects remains a tremendous obstacle for RNAi therapy. Here, a degradable nano-MgO micelle composite (MgO(pDNA)-INS-Plu-mRNA-NGF) with double interference (mediated by RNAi and α-synuclein (α-syn)-targeted mRNA) was constructed. Binding mRNA treatment significantly increased the inhibitory effect compared to the reduction of α-syn expression by RNAi alone. Moreover, the cell experiments demonstrated that the viability of the PD cell model can be significantly improved by nano-MgO micelle composite treatment. More importantly, the composite has the ability to penetrate the blood brain barrier and deliver genes and mRNA to neurons through endocytosis mediated by the nerve growth factor and its receptors, thus significantly downregulating the expression of α-syn in the PD mice model without causing damage to other major organs. Overall, this work provides a novel insight into the design of biomaterials for gene therapy for PD.
Collapse
Affiliation(s)
- Mingchao Li
- School of Life Science, South China Normal University, Guangzhou 510631, China
| | - Kaikai Hu
- Guangzhou Key Laboratory of Spectral Analysis and Functional Probes, College of Biophotonics, South China Normal University, Guangzhou 510631, China
| | - Danmin Lin
- School of Life Science, South China Normal University, Guangzhou 510631, China
| | - Zhen Wang
- School of Life Science, South China Normal University, Guangzhou 510631, China
| | - Mingze Xu
- School of Life Science, South China Normal University, Guangzhou 510631, China
| | - Jinpeng Huang
- School of Life Science, South China Normal University, Guangzhou 510631, China.,South China Normal University-Panyu Central Hospital Joint Laboratory of Translational Medical Research, Panyu Central Hospital, Guangzhou 511400, China
| | - Zhan Chen
- School of Life Science, South China Normal University, Guangzhou 510631, China
| | - Yi Zhang
- School of Life Science, South China Normal University, Guangzhou 510631, China
| | - Liang Yin
- School of Life Science, South China Normal University, Guangzhou 510631, China
| | - Rong You
- School of Life Science, South China Normal University, Guangzhou 510631, China
| | - Chu-Hua Li
- School of Life Science, South China Normal University, Guangzhou 510631, China
| | - Yan-Qing Guan
- School of Life Science, South China Normal University, Guangzhou 510631, China.,South China Normal University-Panyu Central Hospital Joint Laboratory of Translational Medical Research, Panyu Central Hospital, Guangzhou 511400, China.,Guangzhou Key Laboratory of Spectral Analysis and Functional Probes, College of Biophotonics, South China Normal University, Guangzhou 510631, China
| |
Collapse
|
8
|
Differential Expression of Multiple Disease-Related Protein Groups Induced by Valproic Acid in Human SH-SY5Y Neuroblastoma Cells. Brain Sci 2020; 10:brainsci10080545. [PMID: 32806546 PMCID: PMC7465595 DOI: 10.3390/brainsci10080545] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 08/07/2020] [Accepted: 08/08/2020] [Indexed: 12/23/2022] Open
Abstract
Valproic acid (VPA) is a multifunctional medication used for the treatment of epilepsy, mania associated with bipolar disorder, and migraine. The pharmacological effects of VPA involve a variety of neurotransmitter and cell signaling systems, but the molecular mechanisms underlying its clinical efficacy is to date largely unknown. In this study, we used the isobaric tags for relative and absolute quantitation shotgun proteomic analysis to screen differentially expressed proteins in VPA-treated SH-SY5Y cells. We identified changes in the expression levels of multiple proteins involved in Alzheimer’s disease, Parkinson’s disease, chromatin remodeling, controlling gene expression via the vitamin D receptor, ribosome biogenesis, ubiquitin-mediated proteolysis, and the mitochondrial oxidative phosphorylation and electron transport chain. Our data indicate that VPA may modulate the differential expression of proteins involved in mitochondrial function and vitamin D receptor-mediated chromatin transcriptional regulation and proteins implicated in the pathogenesis of neurodegenerative diseases.
Collapse
|
9
|
Hsu SW, Hsu PC, Chang WS, Yu CC, Wang YC, Yang JS, Tsai FJ, Chen KY, Tsai CW, Bau DT. Protective effects of valproic acid on 6-hydroxydopamine-induced neuroinjury. ENVIRONMENTAL TOXICOLOGY 2020; 35:840-848. [PMID: 32167238 DOI: 10.1002/tox.22920] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 02/27/2020] [Accepted: 03/01/2020] [Indexed: 06/10/2023]
Abstract
Oxidative stress may play critically important roles in the etiology of Parkinson's disease (PD). 6-Hydroxydopamine (6-OHDA) is a physiological neurotoxin reported to induce oxidative-induced apoptosis of dopaminergic neurons in PD mice models. Valproic acid (VPA), a clinical mood stabilizer, is a HDAC inhibitor with neuroprotective capacities. In the study, we aim at examining the feasibility of VPA as a protector for dopaminergic neurons against damage from 6-OHDA, and the intracellular mechanisms. The 6-OHDA-induced neurotoxicity to the human dopaminergic cell line SH-SY5Y was applied for examining VPA protective effects. Pretreatment with VPA was able to improve cell viability and reduce 6-OHDA-induced reactive oxygen species. Furthermore, a significant suppression of apoptotic caspases including cleaved caspase-3, caspase-7, and caspase-9 was observed. The results also revealed VPA decreased the 6-OHDA-induced Bax/Bcl2 ratio, as measured at protein level. These novel findings indicate that VPA may be capable of protecting the SH-SY5Y dopaminergic neuronal cells from 6-OHDA-induced toxicity via the deceasing of apoptotic caspases (cleaved caspase-3, caspase-7, and caspase-9) and reducing of the Bax/Bcl2 ratio. Very possibly, VPA could serve as not only a mood stabilizer but also a potential antidote for PD prevention.
Collapse
Affiliation(s)
- Shih-Wei Hsu
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
- National Defense Medical Center, Taipei, Taiwan
- Taichung Armed Forces General Hospital, Taichung, Taiwan
| | - Pei-Chen Hsu
- Department of Pediatrics, Taoyuan General Hospital, Ministry of Health and Welfare, Taoyuan, Taiwan
| | - Wen-Shin Chang
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
- Terry Fox Cancer Research Laboratory, Department of Medical Research, China Medical University Hospital, Taichung, Taiwan
| | - Chien-Chih Yu
- School of Pharmacy, China Medical University, Taichung, Taiwan
| | - Yun-Chi Wang
- Terry Fox Cancer Research Laboratory, Department of Medical Research, China Medical University Hospital, Taichung, Taiwan
| | - Jai-Sing Yang
- Terry Fox Cancer Research Laboratory, Department of Medical Research, China Medical University Hospital, Taichung, Taiwan
| | - Fuu-Jen Tsai
- Terry Fox Cancer Research Laboratory, Department of Medical Research, China Medical University Hospital, Taichung, Taiwan
| | - Kai-Yuan Chen
- Terry Fox Cancer Research Laboratory, Department of Medical Research, China Medical University Hospital, Taichung, Taiwan
| | - Chia-Wen Tsai
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
- Terry Fox Cancer Research Laboratory, Department of Medical Research, China Medical University Hospital, Taichung, Taiwan
| | - Da-Tian Bau
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
- Terry Fox Cancer Research Laboratory, Department of Medical Research, China Medical University Hospital, Taichung, Taiwan
- Department of Bioinformatics and Medical Engineering, Asia University, Taichung, Taiwan
| |
Collapse
|
10
|
Ádám Á, Kemecsei R, Company V, Murcia-Ramón R, Juarez I, Gerecsei LI, Zachar G, Echevarría D, Puelles E, Martínez S, Csillag A. Gestational Exposure to Sodium Valproate Disrupts Fasciculation of the Mesotelencephalic Dopaminergic Tract, With a Selective Reduction of Dopaminergic Output From the Ventral Tegmental Area. Front Neuroanat 2020; 14:29. [PMID: 32581730 PMCID: PMC7290005 DOI: 10.3389/fnana.2020.00029] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 05/11/2020] [Indexed: 01/14/2023] Open
Abstract
Gestational exposure to valproic acid (VPA) is known to cause behavioral deficits of sociability, matching similar alterations in human autism spectrum disorder (ASD). Available data are scarce on the neuromorphological changes in VPA-exposed animals. Here, we focused on alterations of the dopaminergic system, which is implicated in motivation and reward, with relevance to social cohesion. Whole brains from 7-day-old mice born to mothers given a single injection of VPA (400 mg/kg b.wt.) on E13.5 were immunostained against tyrosine hydroxylase (TH). They were scanned using the iDISCO method with a laser light-sheet microscope, and the reconstructed images were analyzed in 3D for quantitative morphometry. A marked reduction of mesotelencephalic (MT) axonal fascicles together with a widening of the MT tract were observed in VPA treated mice, while other major brain tracts appeared anatomically intact. We also found a reduction in the abundance of dopaminergic ventral tegmental (VTA) neurons, accompanied by diminished tissue level of DA in ventrobasal telencephalic regions (including the nucleus accumbens (NAc), olfactory tubercle, BST, substantia innominata). Such a reduction of DA was not observed in the non-limbic caudate-putamen. Conversely, the abundance of TH+ cells in the substantia nigra (SN) was increased, presumably due to a compensatory mechanism or to an altered distribution of TH+ neurons occupying the SN and the VTA. The findings suggest that defasciculation of the MT tract and neuronal loss in VTA, followed by diminished dopaminergic input to the ventrobasal telencephalon at a critical time point of embryonic development (E13-E14) may hinder the patterning of certain brain centers underlying decision making and sociability.
Collapse
Affiliation(s)
- Ágota Ádám
- Department of Anatomy, Histology, and Embryology, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Róbert Kemecsei
- Department of Anatomy, Histology, and Embryology, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Verónica Company
- Institute of Neuroscience (UMH-CSIC), University of Miguel Hernández, Alicante, Spain
| | - Raquel Murcia-Ramón
- Institute of Neuroscience (UMH-CSIC), University of Miguel Hernández, Alicante, Spain
| | - Iris Juarez
- Institute of Neuroscience (UMH-CSIC), University of Miguel Hernández, Alicante, Spain
| | - László I Gerecsei
- Department of Anatomy, Histology, and Embryology, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Gergely Zachar
- Department of Anatomy, Histology, and Embryology, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Diego Echevarría
- Institute of Neuroscience (UMH-CSIC), University of Miguel Hernández, Alicante, Spain
| | - Eduardo Puelles
- Institute of Neuroscience (UMH-CSIC), University of Miguel Hernández, Alicante, Spain
| | - Salvador Martínez
- Institute of Neuroscience (UMH-CSIC), University of Miguel Hernández, Alicante, Spain
| | - András Csillag
- Department of Anatomy, Histology, and Embryology, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| |
Collapse
|
11
|
Hong GU, Cho JW, Kim SY, Shin JH, Ro JY. Inflammatory mediators resulting from transglutaminase 2 expressed in mast cells contribute to the development of Parkinson's disease in a mouse model. Toxicol Appl Pharmacol 2018; 358:10-22. [PMID: 30195017 DOI: 10.1016/j.taap.2018.09.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2017] [Revised: 09/01/2018] [Accepted: 09/04/2018] [Indexed: 12/29/2022]
Abstract
This study aimed to investigate the role of transglutaminase 2 (TG2) expressed in mast cells in substantia nigra (SN) in Parkinson's disease (PD) model or human PD patients. C57BL/6 mice received 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) by ip injection to induce PD. Bone marrow-derived mast cells (BMMCs) were adoptively transferred to TG2 knockout (KO or TG2-/-) mice by iv injection 1 day before MPTP injection or stimulated by 1 methyl-4-phenylpyridinium (MMP+). KO-MPTP mice showed reduced expression of tyrosine hydroxylase (TH) and dopamine (DA) transporter (DAT) and loss of TH+ DA neurons, and expression of markers (c-kit, tryptase, FcεRI), mediators' release (histamine, leukotrienes, cytokines), and TG2 related to mast cells, and co-localization of DA neuronal cells and mast cells in SN tissues or release of mediators and TG2 activity in SN tissues and sera versus those in WT (wild type)-MPTP or BM + KO-MPTP mice. KO-MPTP mice reversed the alterations of behavior. KO-BMMCs-transferred KO-MPTP (BM + KO-MPTP) mice had restoration of all the responses versus the KO-MPTP mice. MPP+-stimulated BMMCs had increased mediators' release, which were inhibited by TG2 inhibitor (R2 peptide). All the mediators and TG2 activity were also increased in the sera of human PD patients. The data suggest that TG2 expressed in mast cells recruited into SN tissues might contribute to neuroinflammation, which is known as one of the important features in pathogenesis of PD, via up-regulating the release of various mediators.
Collapse
Affiliation(s)
- Gwan Ui Hong
- Department of Pharmacology, SBRI, Sungkyunkwan University School of Medicine, Suwon, Republic of Korea
| | - Jin Whan Cho
- Department of Neurology, SBRI, Sungkyunkwan University School of Medicine, Suwon, Republic of Korea
| | - Soo Youl Kim
- Cancer Cell and Molecular Biology Branch, Division of Cancer Biology, Research Institute, National Cancer Center, Goyang, Republic of Korea
| | - Joo Ho Shin
- Department of Pharmacology, SBRI, Sungkyunkwan University School of Medicine, Suwon, Republic of Korea
| | - Jai Youl Ro
- Department of Pharmacology, SBRI, Sungkyunkwan University School of Medicine, Suwon, Republic of Korea.
| |
Collapse
|
12
|
Phillipson OT. Alpha-synuclein, epigenetics, mitochondria, metabolism, calcium traffic, & circadian dysfunction in Parkinson's disease. An integrated strategy for management. Ageing Res Rev 2017; 40:149-167. [PMID: 28986235 DOI: 10.1016/j.arr.2017.09.006] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Accepted: 09/14/2017] [Indexed: 12/15/2022]
Abstract
The motor deficits which characterise the sporadic form of Parkinson's disease arise from age-related loss of a subset of dopamine neurons in the substantia nigra. Although motor symptoms respond to dopamine replacement therapies, the underlying disease process remains. This review details some features of the progressive molecular pathology and proposes deployment of a combination of nutrients: R-lipoic acid, acetyl-l-carnitine, ubiquinol, melatonin (or receptor agonists) and vitamin D3, with the collective potential to slow progression of these features. The main nutrient targets include impaired mitochondria and the associated oxidative/nitrosative stress, calcium stress and impaired gene transcription induced by pathogenic forms of alpha- synuclein. Benefits may be achieved via nutrient influence on epigenetic signaling pathways governing transcription factors for mitochondrial biogenesis, antioxidant defences and the autophagy-lysosomal pathway, via regulation of the metabolic energy sensor AMP activated protein kinase (AMPK) and the mammalian target of rapamycin mTOR. Nutrients also benefit expression of the transcription factor for neuronal survival (NR4A2), trophic factors GDNF and BDNF, and age-related calcium signals. In addition a number of non-motor related dysfunctions in circadian control, clock genes and associated metabolic, endocrine and sleep-wake activity are briefly addressed, as are late-stage complications in respect of cognitive decline and osteoporosis. Analysis of the network of nutrient effects reveals how beneficial synergies may counter the accumulation and promote clearance of pathogenic alpha-synuclein.
Collapse
|
13
|
Green AL, Zhan L, Eid A, Zarbl H, Guo GL, Richardson JR. Valproate increases dopamine transporter expression through histone acetylation and enhanced promoter binding of Nurr1. Neuropharmacology 2017; 125:189-196. [PMID: 28743636 DOI: 10.1016/j.neuropharm.2017.07.020] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Revised: 07/17/2017] [Accepted: 07/18/2017] [Indexed: 12/19/2022]
Abstract
The dopamine transporter (DAT) is the key regulator of dopaminergic transmission and is a target of several xenobiotics, including pesticides and pharmacological agents. Previously, we identified a prominent role for histone deacetylases in the regulation of DAT expression. Here, we utilized a rat dopaminergic cell line (N27) to probe the responsiveness of DAT mRNA expression to inhibitors of histone acetylation. Inhibition of histone deacetylases (HDACs) by valproate, butyrate and Trichostatin A led to a 3-10-fold increase in DAT mRNA expression, a 50% increase in protein levels, which were accompanied by increased H3 acetylation levels. To confirm the mechanism of valproate-mediated increase in DAT mRNA, chromatin immunoprecipitation (ChIP) assays were used and demonstrated a significant increase in enrichment of acetylation of histone 3 on lysines 9 and 14 (H3K9/K14ac) in the DAT promoter. Expression of Nurr1 and Pitx3, key regulators of DAT expression, were increased following valproate treatment and Nurr1 binding was enriched in the DAT promoter. Together, these results indicate that histone acetylation and subsequent enhancement of transcription factor binding are plausible mechanisms for DAT regulation by valproate and, perhaps, by other xenobiotics.
Collapse
Affiliation(s)
- Ashley L Green
- Environmental and Occupational Health Sciences Institute, Department of Environmental and Occupational Medicine, Rutgers Robert Wood Johnson Medical School, Piscataway, NJ, USA
| | - Le Zhan
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University Piscataway, NJ, USA
| | - Aseel Eid
- Department of Pharmaceutical Sciences, Center for Neurodegenerative Disease and Aging, Northeast Ohio Medical University, Rootstown, OH, USA
| | - Helmut Zarbl
- Environmental and Occupational Health Sciences Institute, Department of Environmental and Occupational Medicine, Rutgers Robert Wood Johnson Medical School, Piscataway, NJ, USA
| | - Grace L Guo
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University Piscataway, NJ, USA
| | - Jason R Richardson
- Environmental and Occupational Health Sciences Institute, Department of Environmental and Occupational Medicine, Rutgers Robert Wood Johnson Medical School, Piscataway, NJ, USA; Department of Pharmaceutical Sciences, Center for Neurodegenerative Disease and Aging, Northeast Ohio Medical University, Rootstown, OH, USA.
| |
Collapse
|
14
|
Valproic Acid Induces Endocytosis-Mediated Doxorubicin Internalization and Shows Synergistic Cytotoxic Effects in Hepatocellular Carcinoma Cells. Int J Mol Sci 2017; 18:ijms18051048. [PMID: 28498322 PMCID: PMC5454960 DOI: 10.3390/ijms18051048] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 05/03/2017] [Accepted: 05/08/2017] [Indexed: 02/07/2023] Open
Abstract
Valproic acid (VPA), a well-known histone deacetylase (HDAC) inhibitor, is used as an anti-cancer drug for various cancers, but the synergistic anti-cancer effect of VPA and doxorubicin (DOX) combination treatment and its potential underlying mechanism in hepatocellular carcinoma (HCC) remain to be elucidated. Here, we evaluate the mono- and combination-therapy effects of VPA and DOX in HCC and identify a specific and efficient, synergistic anti-proliferative effect of the VPA and DOX combination in HCC cells, especially HepG2 cells; this effect was not apparent in MIHA cells, a normal hepatocyte cell line. The calculation of the coefficient of drug interaction confirmed the significant synergistic effect of the combination treatment. Concurrently, the synergistic apoptotic cell death caused by the VPA and DOX combination treatment was confirmed by Hoechst nuclear staining and Western blot analysis of caspase-3 and poly (ADP-ribose) polymerase (PARP) activation. Co-treatment with VPA and DOX enhanced reactive oxygen species (ROS) generation and autophagy, which were clearly attenuated by ROS and autophagy inhibitors, respectively. Furthermore, as an indication of the mechanism underlying the synergistic effect, we observed that DOX internalization, which was induced in the VPA and DOX combination-treated group, occurred via by the caveolae-mediated endocytosis pathway. Taken together, our study uncovered the potential effect of the VPA and DOX combination treatment with regard to cell death, including induction of cellular ROS, autophagy, and the caveolae-mediated endocytosis pathway. Therefore, these results present novel implications in drug delivery research for the treatment of HCC.
Collapse
|
15
|
Liu B, Lv C, Zhang J, Liu Y, Sun J, Cheng X, Mao W, Ma Y, Li S. Effects of eldepryl on glial cell proliferation and activation in the substantia nigra and striatum in a rat model of Parkinson’s disease. Neurol Res 2017; 39:459-467. [PMID: 28276259 DOI: 10.1080/01616412.2017.1297911] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Affiliation(s)
- Bin Liu
- First Department of Neurology, Hospital Affiliated to North China University of Science and Technology, Tangshan, China
| | - Chaonan Lv
- First Department of Neurology, Hospital Affiliated to North China University of Science and Technology, Tangshan, China
| | - Jinxia Zhang
- First Department of Neurology, Hospital Affiliated to North China University of Science and Technology, Tangshan, China
| | - Ying Liu
- First Department of Neurology, Hospital Affiliated to North China University of Science and Technology, Tangshan, China
| | - Jing Sun
- First Department of Neurology, Hospital Affiliated to North China University of Science and Technology, Tangshan, China
| | - Xiaohua Cheng
- First Department of Neurology, Hospital Affiliated to North China University of Science and Technology, Tangshan, China
| | - Wenjing Mao
- First Department of Neurology, Hospital Affiliated to North China University of Science and Technology, Tangshan, China
| | - Yuanyuan Ma
- First Department of Neurology, Hospital Affiliated to North China University of Science and Technology, Tangshan, China
| | - Shiying Li
- First Department of Neurology, Hospital Affiliated to North China University of Science and Technology, Tangshan, China
| |
Collapse
|
16
|
Perez-Pardo P, Dodiya HB, Broersen LM, Douna H, van Wijk N, Lopes da Silva S, Garssen J, Keshavarzian A, Kraneveld AD. Gut-brain and brain-gut axis in Parkinson's disease models: Effects of a uridine and fish oil diet. Nutr Neurosci 2017; 21:391-402. [PMID: 28276272 DOI: 10.1080/1028415x.2017.1294555] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Recent investigations have focused on the potential role of gastrointestinal (GI) abnormalities in the pathogenesis of Parkinson's disease (PD). The 'dual-hit' hypothesis of PD speculates that a putative pathogen enters the brain via two routes: the olfactory system and the GI system. Here, we investigated (1) whether local exposures of the neurotoxin rotenone in the gut or the brain of mice could induce PD-like neurological and GI phenotypes as well as a characteristic neuropathology in accordance with this 'dual-hit hypothesis' and (2) the effects of a diet containing uridine and fish oil providing docosahexaenoic acid (DHA), in both models. Mice were given rotenone either orally or by an injection in the striatum. Dietary interventions were started 1 week before rotenone exposures. We found that (1) both oral and intrastriatal administration of rotenone induced similar PD-like motor deficits, dopaminergic cell loss, delayed intestinal transit, inflammation, and alpha-synuclein accumulation in the colon; (2) the uridine and DHA containing diet prevented rotenone-induced motor and GI dysfunctions in both models. The models suggest possible bidirectional communication between the gut and the brain for the genesis of PD-like phenotype and pathology. The dietary intervention may provide benefits in the prevention of motor and non-motor symptoms in PD.
Collapse
Affiliation(s)
- Paula Perez-Pardo
- a Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University , Universiteitsweg 99, 3584 CG Utrecht, The Netherlands
| | - Hemraj B Dodiya
- b Department of Internal Medicine, Division of Gastroenterology , Rush University Medical Center , 1725 West Harrison Street, Chicago , IL 60612 , USA
| | - Laus M Broersen
- a Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University , Universiteitsweg 99, 3584 CG Utrecht, The Netherlands.,c Nutricia Research , Uppsalalaan 12, 3584 CT Utrecht, The Netherlands
| | - Hidde Douna
- a Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University , Universiteitsweg 99, 3584 CG Utrecht, The Netherlands
| | - Nick van Wijk
- c Nutricia Research , Uppsalalaan 12, 3584 CT Utrecht, The Netherlands
| | | | - Johan Garssen
- a Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University , Universiteitsweg 99, 3584 CG Utrecht, The Netherlands.,c Nutricia Research , Uppsalalaan 12, 3584 CT Utrecht, The Netherlands
| | - Ali Keshavarzian
- a Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University , Universiteitsweg 99, 3584 CG Utrecht, The Netherlands.,b Department of Internal Medicine, Division of Gastroenterology , Rush University Medical Center , 1725 West Harrison Street, Chicago , IL 60612 , USA
| | - Aletta D Kraneveld
- a Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University , Universiteitsweg 99, 3584 CG Utrecht, The Netherlands
| |
Collapse
|
17
|
Semmler A, Frisch C, Bleul C, Smith D, Bigler L, Prost JC, Blom H, Linnebank M. Intrauterine valproate exposure is associated with alterations in hippocampal cell numbers and folate metabolism in a rat model of valproate teratogenicity. Seizure 2017; 46:7-12. [DOI: 10.1016/j.seizure.2017.01.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2016] [Revised: 01/14/2017] [Accepted: 01/19/2017] [Indexed: 01/20/2023] Open
|
18
|
Li Z, Wu F, Zhang X, Chai Y, Chen D, Yang Y, Xu K, Yin J, Li R, Shi H, Wang Z, Li X, Xiao J, Zhang H. Valproate Attenuates Endoplasmic Reticulum Stress-Induced Apoptosis in SH-SY5Y Cells via the AKT/GSK3β Signaling Pathway. Int J Mol Sci 2017; 18:ijms18020315. [PMID: 28208696 PMCID: PMC5343851 DOI: 10.3390/ijms18020315] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Revised: 01/12/2017] [Accepted: 01/27/2017] [Indexed: 12/22/2022] Open
Abstract
Endoplasmic reticulum (ER) stress-induced apoptosis plays an important role in a range of neurological disorders, such as neurodegenerative diseases, spinal cord injury, and diabetic neuropathy. Valproate (VPA), a typical antiepileptic drug, is commonly used in the treatment of bipolar disorder and epilepsy. Recently, VPA has been reported to exert neurotrophic effects and promote neurite outgrowth, but its molecular mechanism is still unclear. In the present study, we investigated whether VPA inhibited ER stress and promoted neuroprotection and neuronal restoration in SH-SY5Y cells and in primary rat cortical neurons, respectively, upon exposure to thapsigargin (TG). In SH-SY5Y cells, cell viability was detected by the 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2-H-tetrazolium bromide (MTT) assay, and the expression of ER stress-related apoptotic proteins such as glucose‑regulated protein (GRP78), C/EBP homologous protein (CHOP), and cleaved caspase-12/-3 were analyzed with Western blot analyses and immunofluorescence assays. To explore the pathway involved in VPA-induced cell proliferation, we also examined p-AKT, GSK3β, p-JNK and MMP-9. Moreover, to detect the effect of VPA in primary cortical neurons, immunofluorescence staining of β-III tubulin and Anti-NeuN was analyzed in primary cultured neurons exposed to TG. Our results demonstrated that VPA administration improved cell viability in cells exposed to TG. In addition, VPA increased the levels of GRP78 and p-AKT and decreased the levels of ATF6, XBP-1, GSK3β, p-JNK and MMP-9. Furthermore, the levels of the ER stress-induced apoptosis response proteins CHOP, cleaved caspase-12 and cleaved caspase-3 were inhibited by VPA treatment. Meanwhile, VPA administration also increased the ratio of Bcl-2/Bax. Moreover, VPA can maintain neurite outgrowth of primary cortical neurons. Collectively, the neurotrophic effect of VPA is related to the inhibition of ER stress-induced apoptosis in SH-SY5Y cells and the maintenance of neuronal growth. Collectively, our results suggested a new approach for the therapeutic function of VPA in neurological disorders and neuroprotection.
Collapse
Affiliation(s)
- Zhengmao Li
- Key Laboratory of Biotechnology and Pharmaceutical Engineering, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China.
| | - Fenzan Wu
- Science and Education division, Cixi People's Hospital, Wenzhou Medical University, Ningbo 315300, China.
| | - Xie Zhang
- Ningbo Medical Treatment Center, Li Huili Hospital, Ningbo 315000, China.
| | - Yi Chai
- Department of neurosurgery, The second Affiliated Hospital, Nanchang University, Nanchang 330006, China.
| | - Daqing Chen
- Emergency Department, The Second Affiliated Hospital, Wenzhou Medical University, Wenzhou 325035, China.
| | - Yuetao Yang
- Emergency Department, The Second Affiliated Hospital, Wenzhou Medical University, Wenzhou 325035, China.
| | - Kebin Xu
- Key Laboratory of Biotechnology and Pharmaceutical Engineering, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China.
| | - Jiayu Yin
- Key Laboratory of Biotechnology and Pharmaceutical Engineering, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China.
| | - Rui Li
- Key Laboratory of Biotechnology and Pharmaceutical Engineering, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China.
| | - Hongxue Shi
- Key Laboratory of Biotechnology and Pharmaceutical Engineering, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China.
| | - Zhouguang Wang
- Key Laboratory of Biotechnology and Pharmaceutical Engineering, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China.
| | - Xiaokun Li
- Key Laboratory of Biotechnology and Pharmaceutical Engineering, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China.
- Institute of Life Sciences, Wenzhou University, Wenzhou 325035, China.
| | - Jian Xiao
- Key Laboratory of Biotechnology and Pharmaceutical Engineering, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China.
| | - Hongyu Zhang
- Key Laboratory of Biotechnology and Pharmaceutical Engineering, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China.
| |
Collapse
|
19
|
Carriere CH, Kang NH, Niles LP. Bilateral upregulation of α-synuclein expression in the mouse substantia nigra by intracranial rotenone treatment. ACTA ACUST UNITED AC 2016; 69:109-114. [PMID: 27986376 DOI: 10.1016/j.etp.2016.12.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Accepted: 12/09/2016] [Indexed: 01/08/2023]
Abstract
The pesticide rotenone has been shown to cause systemic inhibition of mitochondrial complex I activity, with consequent degeneration of dopamine neurons along the nigrostriatal pathway, as observed in Parkinson's disease (PD). Recently, intracranial infusion of rotenone was found to increase the protein levels of the Lewy body constituents, α-synuclein and small ubiquitin-related modifier-1(SUMO-1), in the lesioned hemisphere of the mouse brain. These findings are supportive of a mouse model of PD, but information about the dopamine-synthesizing enzyme, tyrosine hydroxylase (TH), an essential marker of dopaminergic status, was not reported. Clarification of this issue is important because an intracranial rotenone mouse model of Parkinson's disease has not been established. Towards this end, the present study examined the effects of intracranial rotenone treatment on TH and α-synuclein immunohistochemistry in addition to forelimb motor function. Mice were unilaterally infused with either vehicle or rotenone (2μg/site) in both the medial forebrain bundle and the substantia nigra. The forelimb asymmetry (cylinder) test indicated a significant decrease in use of the contralateral forelimb in lesioned animals as compared to the sham group. Densitometric analysis revealed a significant depletion of TH immunofluorescence within the ipsilateral striatum and substantia nigra of lesioned animals. Moreover, a significant bilateral increase in α-synuclein immunofluorescence was found in the substantia nigra of lesioned mice, as compared to control animals. These findings indicate that this intracranial rotenone mouse model will be useful for studies of neurodegenerative disorders such as PD.
Collapse
Affiliation(s)
- Candace H Carriere
- Department of Psychiatry and Behavioural Neurosciences, Faculty of Health Sciences, McMaster University, 1200 Main Street West, Hamilton, ON, L8N 3Z5, Canada.
| | - Na Hyea Kang
- Department of Psychiatry and Behavioural Neurosciences, Faculty of Health Sciences, McMaster University, 1200 Main Street West, Hamilton, ON, L8N 3Z5, Canada.
| | - Lennard P Niles
- Department of Psychiatry and Behavioural Neurosciences, Faculty of Health Sciences, McMaster University, 1200 Main Street West, Hamilton, ON, L8N 3Z5, Canada.
| |
Collapse
|
20
|
The neuroprotection of Sinomenine against ischemic stroke in mice by suppressing NLRP3 inflammasome via AMPK signaling. Int Immunopharmacol 2016; 40:492-500. [PMID: 27769021 DOI: 10.1016/j.intimp.2016.09.024] [Citation(s) in RCA: 117] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Revised: 09/22/2016] [Accepted: 09/24/2016] [Indexed: 12/21/2022]
Abstract
Neuroinflammation remains the primary cause of morbidity and mortality in stroke-induced secondary brain injury. The NOD-like receptor pyrin 3 (NLRP3) inflammasome is involved in diverse inflammatory diseases, including cerebral ischemia, and is thus considered an effective therapeutic target. In the present study, we investigated the neuroprotection of Sinomenine (SINO), a potent natural anti-apoptotic and anti-inflammatory molecule, against cerebral ischemia in a mouse model of middle cerebral artery occlusion (MCAO) in vivo and in an oxygen glucose deprivation (OGD)-treated astrocytes/microglia model in vitro. SINO administration intraperitoneally alleviated the cerebral infarction, brain edema, neuronal apoptosis, and neurological deficiency after MCAO induction. SINO also attenuated astrocytic and microglial activation in the ischemic hemisphere. NLRP3 inflammasome activation after MCAO and OGD induction, with the up-regulation of NLRP3, apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC), cleaved caspase-1 and pro-inflammatory cytokines, was significantly inhibited by SINO treatment both in vivo and in vitro. In addition, SINO reversed the OGD-induced inhibition of AMPK phosphorylation in vitro. Further, the suppressive effect of SINO on NLRP3 inflammasomes was blocked by an AMPK inhibitor, Compound C. Our findings demonstrate that SINO exerts a neuroprotective effect in ischemic stroke by inhibiting NLRP3 inflammasomes via the AMPK pathway, which also provides evidence of a novel treatment for clinical stroke therapy.
Collapse
|
21
|
Paliperidone increases spontaneous and evoked firing of mesocortical dopaminergic neurons by activating a hyperpolarization-activated inward current. Schizophr Res 2016; 176:252-258. [PMID: 27435059 DOI: 10.1016/j.schres.2016.07.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2014] [Revised: 06/29/2016] [Accepted: 07/04/2016] [Indexed: 11/21/2022]
Abstract
Mesocortical dopaminergic (DA) subtype neurons specifically project to the prefrontal cortex, which is closely related with schizophrenia. Mesocortical DA neurons have unique physiological characteristics that are different from those of mesostriatal and mesolimbic DA neurons. Paliperidone, an atypical antipsychotic, is currently used to treat schizophrenia and has better therapeutic effects than typical antipsychotics. However, the underlying physiological mechanism remains unclear. To explore the effects of paliperidone on mesocortical DA neuron activity, here, we retrogradely labeled these cells with fluorescent microsphere retrobeads, and the electrophysiological changes were recorded in whole-cell recordings in rat midbrain slices with or without paliperidone. The data showed that paliperidone (20μmol/L) increased the spontaneous firing rates of labeled mesocortical neurons (P<0.05). Moreover, paliperidone also increased the frequency of evoked action potentials by current injection stimulation (P<0.05), whereas the accompanying amplitude decreased. Furthermore, to explore the mechanisms of paliperidone's effect, Ih currents were detected, and the results showed that hyperpolarizing voltage pulses evoked instantaneous Ih inward currents and paliperidone increased the maximum Ih current. In addition, paliperidone decreased the spontaneous inhibitory postsynaptic currents. Thus, paliperidone increased the spontaneous and evoked firing of mesocortical neurons, possibly by activating the Ih inward current and reducing the inhibitory synaptic transmission, which provides an underlying mechanism of paliperidone's application in schizophrenia.
Collapse
|
22
|
Kang NH, Carriere CH, Bahna SG, Niles LP. Altered melatonin MT 1 receptor expression in the ventral midbrain following 6-hydroxydopamine lesions in the rat medial forebrain bundle. Brain Res 2016; 1652:89-96. [PMID: 27693415 DOI: 10.1016/j.brainres.2016.09.036] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Revised: 09/21/2016] [Accepted: 09/26/2016] [Indexed: 10/20/2022]
Abstract
The indoleamine hormone melatonin protects dopamine neurons in the rat nigrostriatal pathway following 6-hydroxydopamine lesioning, and an increase in striatal melatonin levels has been detected in this model of Parkinson's disease. Melatonin induces the expression of tyrosine hydroxylase, the rate-limiting enzyme for dopamine synthesis, in the ventral midbrain, where G protein-coupled melatonin receptors are present. Based on the interaction between the melatonergic and dopaminergic systems, we hypothesized that 6-hydroxydopamine-induced degeneration of dopamine neurons would affect the expression of melatonin receptors in the nigrostriatal pathway. Following unilateral injection of 6-hydroxydopamine into the rat striatum or medial forebrain bundle, there was a significant increase in apomorphine-induced contralateral rotations in lesioned animals as compared to sham controls. A loss of tyrosine hydroxylase immunoreactivity and/or immunofluorescence in the striatum and substantia nigra was seen in animals lesioned in either the striatum or medial forebrain bundle, indicating degeneration of dopamine neurons. There were no significant differences in melatonin MT1 receptor protein expression in the striatum or substantia nigra, between intrastriatally lesioned animals and sham controls. In contrast, lesions in the medial forebrain bundle caused a significant increase in MT1 receptor mRNA expression (p<0.03) on the lesioned side of the ventral midbrain, as compared with the contralateral side. Given the presence of MT1 receptors on neurons in the ventral midbrain, these results suggest that a compensatory increase in MT1 transcription occurs to maintain expression of this receptor and neuroprotective melatonergic signaling in the injured brain.
Collapse
Affiliation(s)
- Na Hyea Kang
- Department of Psychiatry and Behavioural Neurosciences, Faculty of Health Sciences, McMaster University, HSC-4N77, 1280 Main Street West, Hamilton, ON, Canada L8S 4L8.
| | - Candace H Carriere
- Department of Psychiatry and Behavioural Neurosciences, Faculty of Health Sciences, McMaster University, HSC-4N77, 1280 Main Street West, Hamilton, ON, Canada L8S 4L8.
| | - Sarra G Bahna
- Department of Psychiatry and Behavioural Neurosciences, Faculty of Health Sciences, McMaster University, HSC-4N77, 1280 Main Street West, Hamilton, ON, Canada L8S 4L8.
| | - Lennard P Niles
- Department of Psychiatry and Behavioural Neurosciences, Faculty of Health Sciences, McMaster University, HSC-4N77, 1280 Main Street West, Hamilton, ON, Canada L8S 4L8.
| |
Collapse
|
23
|
Leng Y, Wang J, Wang Z, Liao HM, Wei M, Leeds P, Chuang DM. Valproic Acid and Other HDAC Inhibitors Upregulate FGF21 Gene Expression and Promote Process Elongation in Glia by Inhibiting HDAC2 and 3. Int J Neuropsychopharmacol 2016; 19:pyw035. [PMID: 27207921 PMCID: PMC5006201 DOI: 10.1093/ijnp/pyw035] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Accepted: 04/18/2016] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Fibroblast growth factor 21, a novel regulator of glucose and lipid metabolism, has robust protective properties in neurons. However, its expression and function in glia are unknown. Valproic acid, a mood stabilizer and anticonvulsant, is a histone deacetylase inhibitor and a dynamic gene regulator. We investigated whether histone deacetylase inhibition by valproic acid and other inhibitors upregulates fibroblast growth factor 21 expression and, if so, sought to identify the histone deacetylase isoform(s) involved and their role in altering glial cell morphology. METHODS C6 glioma or primary cortical glial cultures were treated with histone deacetylase inhibitors, and fibroblast growth factor 21 levels and length of cell processes were subsequently measured. Histone deacetylase 1, 2, or 3 was also knocked down to detect which isoform was involved in regulating fibroblast growth factor 21 mRNA levels. Finally, knockdown and overexpression of fibroblast growth factor 21 were performed to determine whether it played a role in regulating cell process length. RESULTS Treatment of C6 cells or primary glial cultures with valproic acid elevated fibroblast growth factor 21 mRNA levels, extended cell process length, and markedly increased acetylated histone-H3 levels. Other histone deacetylase inhibitors including pan- and class I-specific inhibitors, or selective knockdown of histone deacetylase 2 or 3 isoform produced similar effects. Knockdown or overexpression of fibroblast growth factor 21 significantly decreased or increased C6 cell process length, respectively. CONCLUSIONS In glial cell line and primary glia, using pharmacological inhibition and selective gene silencing of histone deacetylases to boost fibroblast growth factor 21 mRNA levels results in elongation of cell processes. Our study provides a new mechanism via which histone deacetylase 2 and 3 participate in upregulating fibroblast growth factor 21 transcription and extending process outgrowth in glia.
Collapse
Affiliation(s)
- Yan Leng
- Molecular Neurobiology Section, National Institute of Mental Health, National Institutes of Health, Bethesda, MD.
| | | | | | | | | | | | - De-Maw Chuang
- Molecular Neurobiology Section, National Institute of Mental Health, National Institutes of Health, Bethesda, MD.
| |
Collapse
|
24
|
Jiang H, Wang J, Song L, Cao X, Yao X, Tang F, Yue Y. GC×GC-TOFMS Analysis of Essential Oils Composition from Leaves, Twigs and Seeds of Cinnamomum camphora L. Presl and Their Insecticidal and Repellent Activities. Molecules 2016; 21:423. [PMID: 27043503 PMCID: PMC6274170 DOI: 10.3390/molecules21040423] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Revised: 03/18/2016] [Accepted: 03/23/2016] [Indexed: 12/02/2022] Open
Abstract
Interest in essential oils with pesticidal activity against insects and pests is growing. In this study, essential oils from different parts (leaves, twigs and seeds) of Cinnamomum camphora L. Presl were investigated for their chemical composition, and insecticidal and repellent activities against the cotton aphid. The essential oils, obtained by hydrodistillation, were analyzed by GC×GC-TOFMS. A total of 96 components were identified in the essential oils and the main constituents found in the leaves and twigs were camphor, eucalyptol, linalool and 3,7-dimethyl-1,3,7-octatriene. The major components found in the seeds were eucalyptol (20.90%), methyleugenol (19.98%), linalool (14.66%) and camphor (5.5%). In the contact toxicity assay, the three essential oils of leaves, twigs and seeds exhibited a strong insecticidal activity against cotton aphids with LC50 values of 245.79, 274.99 and 146.78 mg/L (after 48 h of treatment), respectively. In the repellent assay, the highest repellent rate (89.86%) was found in the seed essential oil at the concentration of 20 μL/mL after 24 h of treatment. Linalool was found to be a significant contributor to the insecticidal and repellent activities. The results indicate that the essential oils of C. camphora might have the potential to be developed into a natural insecticide or repellent for controlling cotton aphids.
Collapse
Affiliation(s)
- Hao Jiang
- College of Plant Protection, Anhui Agricultural University, Hefei 230036, China.
- SFA Key Laboratory of Bamboo and Rattan Science and Technology, International Centre for Bamboo and Rattan, No. 8 Futong Dongdajie, Wangjing, Chaoyang District, Beijing 100102, China.
- State Key Laboratory of Tea Plant Biology and Utilization, Anhui Agricultural University, Hefei 230036, China.
| | - Jin Wang
- SFA Key Laboratory of Bamboo and Rattan Science and Technology, International Centre for Bamboo and Rattan, No. 8 Futong Dongdajie, Wangjing, Chaoyang District, Beijing 100102, China.
| | - Li Song
- SFA Key Laboratory of Bamboo and Rattan Science and Technology, International Centre for Bamboo and Rattan, No. 8 Futong Dongdajie, Wangjing, Chaoyang District, Beijing 100102, China.
| | - Xianshuang Cao
- SFA Key Laboratory of Bamboo and Rattan Science and Technology, International Centre for Bamboo and Rattan, No. 8 Futong Dongdajie, Wangjing, Chaoyang District, Beijing 100102, China.
| | - Xi Yao
- SFA Key Laboratory of Bamboo and Rattan Science and Technology, International Centre for Bamboo and Rattan, No. 8 Futong Dongdajie, Wangjing, Chaoyang District, Beijing 100102, China.
| | - Feng Tang
- SFA Key Laboratory of Bamboo and Rattan Science and Technology, International Centre for Bamboo and Rattan, No. 8 Futong Dongdajie, Wangjing, Chaoyang District, Beijing 100102, China.
| | - Yongde Yue
- SFA Key Laboratory of Bamboo and Rattan Science and Technology, International Centre for Bamboo and Rattan, No. 8 Futong Dongdajie, Wangjing, Chaoyang District, Beijing 100102, China.
| |
Collapse
|
25
|
Lauterbach EC. Six psychotropics for pre-symptomatic & early Alzheimer's (MCI), Parkinson's, and Huntington's disease modification. Neural Regen Res 2016; 11:1712-1726. [PMID: 28123400 PMCID: PMC5204212 DOI: 10.4103/1673-5374.194708] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The quest for neuroprotective drugs to slow the progression of neurodegenerative diseases (NDDs), including Alzheimer's disease (AD), Parkinson's disease (PD), and Huntington's disease (HD), has been largely unrewarding. Preclinical evidence suggests that repurposing quetiapine, lithium, valproate, fluoxetine, donepezil, and memantine for early and pre-symptomatic disease-modification in NDDs may be promising and can spare regulatory barriers. The literature of these psychotropics in early stage and pre-symptomatic AD, PD, and HD is reviewed and propitious findings follow. Mild cognitive impairment (MCI) phase of AD: salutary human randomized controlled trial findings for low-dose lithium and, in selected patients, donepezil await replication. Pre-symptomatic AD: human epidemiological data indicate that lithium reduces AD risk. Animal model studies (AMS) reveal encouraging results for quetiapine, lithium, donepezil, and memantine. Early PD: valproate AMS findings show promise. Pre-symptomatic PD: lithium and valproate AMS findings are encouraging. Early HD: uncontrolled clinical data indicate non-progression with lithium, fluoxetine, donepezil, and memantine. Pre-symptomatic HD: lithium and valproate are auspicious in AMS. Many other promising findings awaiting replication (valproate in MCI; lithium, valproate, fluoxetine in pre-symptomatic AD; lithium in early PD; lithium, valproate, fluoxetine in pre-symptomatic PD; donepezil in early HD; lithium, fluoxetine, memantine in pre-symptomatic HD) are reviewed. Dose- and stage-dependent effects are considered. Suggestions for signal-enhancement in human trials are provided for each NDD stage.
Collapse
Affiliation(s)
- Edward C Lauterbach
- Professor Emeritus of Psychiatry and Neurology, Mercer University School of Medicine, Macon, GA, USA
| |
Collapse
|
26
|
Zhang Z, Hou L, Li X, Ju C, Zhang J, Li X, Wang X, Liu C, Lv Y, Wang Y. Neuroprotection of inositol hexaphosphate and changes of mitochondrion mediated apoptotic pathway and α-synuclein aggregation in 6-OHDA induced parkinson's disease cell model. Brain Res 2015; 1633:87-95. [PMID: 26740400 DOI: 10.1016/j.brainres.2015.12.035] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Revised: 12/07/2015] [Accepted: 12/18/2015] [Indexed: 12/19/2022]
Abstract
Animal and cell experiments showed that inositol hexaphosphate (IP6) was protective on neurons in parkinson's disease (PD) model, but the underlying mechanism of this action was not extensively elucidated. To address this question, we established 6-hydroxydopamine (6-OHDA) induced human dopaminergic cell line SH-SY5Y as PD cell model and testified the neuroprotection of IP6. Through hoechst nuclear stain method and flow cytometric analysis, apoptosis induced by 6-OHDA was blocked by IP6 pretreatment. Significant protection against reactive oxygen species (ROS) and lipid peroxidation product malondialdehyde (MDA) was observed in 6-OHDA induced cells pretreated with IP6. To further investigate the mechanism of anti-apoptotic effect of IP6, expression of mediators in mitochondrion dependent apoptotic pathway was detected. Results indicated that loss of mitochondrial membrane potential, cytochrome c releasing, upregulation of Bcl-2-associated X protein (Bax), downregulation of B-cell CLL/lymphoma 2 (Bcl-2) and caspases activation were reversed by IP6. In addition, using flow cytometric method and western blot approach, our data showed that IP6 attenuated the rise of calcium and α-synuclein aggregation in cytosol. Collectively, IP6 exerted its neuroprotection on dopaminergic cells in PD cell model and the mechanism may be associated with changes of mitochondrion mediated apoptotic pathway and α-synuclein aggregation.
Collapse
Affiliation(s)
- Zheng Zhang
- Department of Biochemistry and Molecular Biology, Medical College of Qingdao University, Qingdao, Shandong Province, China
| | - Lin Hou
- Department of Biochemistry and Molecular Biology, Medical College of Qingdao University, Qingdao, Shandong Province, China.
| | - Xianghong Li
- Department of Neonatology, Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Chuanxia Ju
- Department of Pharmacology, Medical College of Qingdao University, Qingdao, Shandong Province, China
| | - Jinyu Zhang
- Department of Biochemistry and Molecular Biology, Medical College of Qingdao University, Qingdao, Shandong Province, China
| | - Xin Li
- Experiment Center of Biochemistry and Molecular Biology, Medical College of Qingdao University, Qingdao, Shandong Province, China
| | - Xiuli Wang
- Experiment Center of Biochemistry and Molecular Biology, Medical College of Qingdao University, Qingdao, Shandong Province, China
| | - Cun Liu
- Laboratory Department of the Third People׳s Hospital of Qingdao, Qingdao, Shandong Province, China
| | - Yuqiang Lv
- Department of Biochemistry and Molecular Biology, Medical College of Qingdao University, Qingdao, Shandong Province, China
| | - Yuehua Wang
- Department of Biochemistry and Molecular Biology, Medical College of Qingdao University, Qingdao, Shandong Province, China
| |
Collapse
|
27
|
Carriere CH, Kang NH, Niles LP. Chronic low-dose melatonin treatment maintains nigrostriatal integrity in an intrastriatal rotenone model of Parkinson's disease. Brain Res 2015; 1633:115-125. [PMID: 26740407 DOI: 10.1016/j.brainres.2015.12.036] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Revised: 11/11/2015] [Accepted: 12/18/2015] [Indexed: 12/21/2022]
Abstract
Parkinson's disease is a major neurodegenerative disorder which primarily involves the loss of dopaminergic neurons in the substantia nigra and related projections in the striatum. The pesticide/neurotoxin, rotenone, has been shown to cause systemic inhibition of mitochondrial complex I activity in nigral dopaminergic neurons, with consequent degeneration of the nigrostriatal pathway, as observed in Parkinson's disease. A novel intrastriatal rotenone model of Parkinson's disease was used to examine the neuroprotective effects of chronic low-dose treatment with the antioxidant indoleamine, melatonin, which can upregulate neurotrophic factors and other protective proteins in the brain. Sham or lesioned rats were treated with either vehicle (0.04% ethanol in drinking water) or melatonin at a dose of 4 µg/mL in drinking water. The right striatum was lesioned by stereotactic injection of rotenone at three sites (4 μg/site) along its rostrocaudal axis. Apomorphine administration to lesioned animals resulted in a significant (p<0.001) increase in ipsilateral rotations, which was suppressed by melatonin. Nine weeks post-surgery, animals were sacrificed by transcardial perfusion. Subsequent immunohistochemical examination revealed a decrease in tyrosine hydroxylase immunoreactivity within the striatum and substantia nigra of rotenone-lesioned animals. Melatonin treatment attenuated the decrease in tyrosine hydroxylase in the striatum and abolished it in the substantia nigra. Stereological cell counts indicated a significant (p<0.05) decrease in dopamine neurons in the substantia nigra of rotenone-lesioned animals, which was confirmed by Nissl staining. Importantly, chronic melatonin treatment blocked the loss of dopamine neurons in rotenone-lesioned animals. These findings strongly support the therapeutic potential of long-term and low-dose melatonin treatment in Parkinson's disease.
Collapse
Affiliation(s)
- Candace H Carriere
- Department of Psychiatry and Behavioural Neurosciences, Faculty of Health Sciences, McMaster University, HSC-4N77, 1200 Main Street West, Hamilton, ON, Canada L8N 3Z5.
| | - Na Hyea Kang
- Department of Psychiatry and Behavioural Neurosciences, Faculty of Health Sciences, McMaster University, HSC-4N77, 1200 Main Street West, Hamilton, ON, Canada L8N 3Z5.
| | - Lennard P Niles
- Department of Psychiatry and Behavioural Neurosciences, Faculty of Health Sciences, McMaster University, HSC-4N77, 1200 Main Street West, Hamilton, ON, Canada L8N 3Z5.
| |
Collapse
|
28
|
Autophagy-related protein expression in the substantia nigra and eldepryl intervention in rat models of Parkinson׳s disease. Brain Res 2015; 1625:180-8. [DOI: 10.1016/j.brainres.2015.08.033] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Revised: 08/18/2015] [Accepted: 08/25/2015] [Indexed: 01/09/2023]
|
29
|
Valproic Acid Neuroprotection in the 6-OHDA Model of Parkinson's Disease Is Possibly Related to Its Anti-Inflammatory and HDAC Inhibitory Properties. JOURNAL OF NEURODEGENERATIVE DISEASES 2015; 2015:313702. [PMID: 26317011 PMCID: PMC4437346 DOI: 10.1155/2015/313702] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Revised: 01/08/2015] [Accepted: 01/11/2015] [Indexed: 12/15/2022]
Abstract
Parkinson's disease is a neurodegenerative disorder where the main hallmark is the dopaminergic neuronal loss. Besides motor symptoms, PD also causes cognitive decline. Although current therapies focus on the restoration of dopamine levels in the striatum, prevention or disease-modifying therapies are urgently needed. Valproic acid (VA) is a wide spectrum antiepileptic drug, exerting many biochemical and physiological effects. It has been shown to inhibit histone deacetylase which seems to be associated with the drug neuroprotective action. The objectives were to study the neuroprotective properties of VA in a model of Parkinson's disease, consisting in the unilateral striatal injection of the neurotoxin 6-OHDA. For that, male Wistar rats (250 g) were divided into the groups: sham-operated (SO), untreated 6-OHDA-lesioned, and 6-OHDA-lesioned treated with VA (25 or 50 mg/kg). Oral treatments started 24 h after the stereotaxic surgery and continued daily for 2 weeks, when the animals were subjected to behavioral evaluations (apomorphine-induced rotations and open-field tests). Then, they were sacrificed and had their mesencephalon, striatum, and hippocampus dissected for neurochemical (DA and DOPAC determinations), histological (Fluoro-Jade staining), and immunohistochemistry evaluations (TH, OX-42, GFAP, TNF-alpha, and HDAC). The results showed that VA partly reversed behavioral and neurochemical alterations observed in the untreated 6-OHDA-lesioned rats. Besides, VA also decreased neuron degeneration in the striatum and reversed the TH depletion observed in the mesencephalon of the untreated 6-OHDA groups. This neurotoxin increased the OX-42 and GFAP immunoreactivities in the mesencephalon, indicating increased microglia and astrocyte reactivities, respectively, which were reversed by VA. In addition, the immunostainings for TNF-alpha and HDAC demonstrated in the untreated 6-OHDA-lesioned rats were also decreased after VA treatments. These results were observed not only in the CA1 and CA3 subfields of the hippocampus, but also in the temporal cortex. In conclusion, we showed that VA partly reversed the behavioral, neurochemical, histological, and immunohistochemical alterations observed in the untreated 6-OHDA-lesioned animals. These effects are probably related to the drug anti-inflammatory activity and strongly suggest that VA is a potential candidate to be included in translational studies for the treatment of neurodegenerative diseases as PD.
Collapse
|
30
|
Bollino D, Balan I, Aurelian L. Valproic acid induces neuronal cell death through a novel calpain-dependent necroptosis pathway. J Neurochem 2015; 133:174-86. [PMID: 25581256 DOI: 10.1111/jnc.13029] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2014] [Revised: 12/24/2014] [Accepted: 01/05/2015] [Indexed: 02/06/2023]
Abstract
A growing body of evidence indicates that valproic acid (VPA), a histone deacetylase inhibitor used to treat epilepsy and mood disorders, has histone deacetylase-related and -unrelated neurotoxic activity, the mechanism of which is still poorly understood. We report that VPA induces neuronal cell death through an atypical calpain-dependent necroptosis pathway that initiates with downstream activation of c-Jun N-terminal kinase 1 (JNK1) and increased expression of receptor-interacting protein 1 (RIP-1) and is accompanied by cleavage and mitochondrial release/nuclear translocation of apoptosis-inducing factor, mitochondrial release of Smac/DIABLO, and inhibition of the anti-apoptotic protein X-linked inhibitor of apoptosis (XIAP). Coinciding with apoptosis-inducing factor nuclear translocation, VPA induces phosphorylation of the necroptosis-associated histone H2A family member H2AX, which is known to contribute to lethal DNA degradation. These signals are inhibited in neuronal cells that express constitutively activated MEK/ERK and/or PI3-K/Akt survival pathways, allowing them to resist VPA-induced cell death. The data indicate that VPA has neurotoxic activity and identify a novel calpain-dependent necroptosis pathway that includes JNK1 activation and RIP-1 expression. A growing body of evidence indicates that valproic acid (VPA) has neurotoxic activity, the mechanism of which is still poorly understood. We report, for the first time, that VPA activates a previously unrecognized calpain-dependent necroptosis cascade that initiates with JNK1 activation and involves AIF cleavage/nuclear translocation and H2AX phosphorylation as well as an altered Smac/DIABLO to XIAP balance.
Collapse
Affiliation(s)
- Dominique Bollino
- Department of Pharmacology, University of Maryland, Baltimore, Maryland, USA
| | | | | |
Collapse
|
31
|
Johnson ME, Bobrovskaya L. An update on the rotenone models of Parkinson's disease: their ability to reproduce the features of clinical disease and model gene-environment interactions. Neurotoxicology 2014; 46:101-16. [PMID: 25514659 DOI: 10.1016/j.neuro.2014.12.002] [Citation(s) in RCA: 234] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2014] [Revised: 11/19/2014] [Accepted: 12/03/2014] [Indexed: 12/19/2022]
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disorder that is characterized by two major neuropathological hallmarks: the degeneration of dopaminergic neurons in the substantia nigra (SN) and the presence of Lewy bodies in the surviving SN neurons, as well as other regions of the central and peripheral nervous system. Animal models have been invaluable tools for investigating the underlying mechanisms of the pathogenesis of PD and testing new potential symptomatic, neuroprotective and neurorestorative therapies. However, the usefulness of these models is dependent on how precisely they replicate the features of clinical PD with some studies now employing combined gene-environment models to replicate more of the affected pathways. The rotenone model of PD has become of great interest following the seminal paper by the Greenamyre group in 2000 (Betarbet et al., 2000). This paper reported for the first time that systemic rotenone was able to reproduce the two pathological hallmarks of PD as well as certain parkinsonian motor deficits. Since 2000, many research groups have actively used the rotenone model worldwide. This paper will review rotenone models, focusing upon their ability to reproduce the two pathological hallmarks of PD, motor deficits, extranigral pathology and non-motor symptoms. We will also summarize the recent advances in neuroprotective therapies, focusing on those that investigated non-motor symptoms and review rotenone models used in combination with PD genetic models to investigate gene-environment interactions.
Collapse
Affiliation(s)
- Michaela E Johnson
- School of Pharmacy and Medical Sciences, Sansom Institute for Health Research, University of South Australia, Adelaide, SA 5000, Australia
| | - Larisa Bobrovskaya
- School of Pharmacy and Medical Sciences, Sansom Institute for Health Research, University of South Australia, Adelaide, SA 5000, Australia.
| |
Collapse
|
32
|
Lu J, Wu L, Jiang T, Wang Y, Zhao H, Gao Q, Pan Y, Tian Y, Zhang Y. Angiotensin AT2 receptor stimulation inhibits activation of NADPH oxidase and ameliorates oxidative stress in rotenone model of Parkinson's disease in CATH.a cells. Neurotoxicol Teratol 2014; 47:16-24. [PMID: 25446015 DOI: 10.1016/j.ntt.2014.11.004] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2014] [Revised: 11/07/2014] [Accepted: 11/07/2014] [Indexed: 01/17/2023]
Abstract
Oxidative stress has long been considered as a major contributing factor in the pathogenesis of Parkinson's disease (PD). The brain has an independent local renin-angiotensin system (RAS). Angiotensin II (Ang II) activates NADPH-dependent oxidases, which are a major source of superoxide and are upregulated in major aging-related diseases such as hypertension and neurodegenerative disease. In this study, we firstly examined whether CGP42112, an AT2 receptor (AT2R) agonist, may exert direct protective effects on the rotenone-induced CATH.a cell injury in vitro. We used CATH.a cell line to evaluate changes in cultured dopaminergic neuron levels of superoxide dismutase (SOD), glutathione (GSH) and reactive oxygen species (ROS). We also evaluated expression of NADPH oxidase, AT1 and AT2 receptors in treated with phosphate buffer saline (PBS), rotenone, Ang II, AT2R agonist CGP42112, or AT2R antagonist PD123319, alone and combined (n=6, each group). Quantitative reverse transcriptase PCR (qRT-PCR) and western blot were used to determine messenger RNA (mRNA) and protein levels of the AT1, AT2 receptors and NADPH oxidase. ROS generation was determined by the dichlorodihydrofluorescein diacetate fluorescent probe assay. The levels of SOD and GSH were measured by using available kits. In our study, CGP42112 (100nM) significantly reduced rotenone-induced oxidative stress and elevated the total SOD activity and GSH level. In addition, CGP42112 significantly increased AT2R expression and attenuated Ang II-induced NADPH oxidase activation, and these effects were completely abolished by the AT2R antagonist, PD123319 (1μM). Our results suggest that CGP42112 attenuates rotenone-induced oxidative stress in CATH.a neuron via activating AT2R and suppressing NADPH oxidase expression.
Collapse
Affiliation(s)
- Jie Lu
- Department of Neurology, Nanjing Brain Hospital, Nanjing Medical University, Nanjing, Jiangsu 210029, PR China
| | - Liang Wu
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210006, PR China
| | - Teng Jiang
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210006, PR China
| | - Yao Wang
- Department of Neurology, Nanjing Brain Hospital, Nanjing Medical University, Nanjing, Jiangsu 210029, PR China
| | - Hongrui Zhao
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210006, PR China
| | - Qing Gao
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210006, PR China
| | - Yang Pan
- Department of Neurology, Nanjing Brain Hospital, Nanjing Medical University, Nanjing, Jiangsu 210029, PR China
| | - Youyong Tian
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210006, PR China
| | - Yingdong Zhang
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210006, PR China.
| |
Collapse
|
33
|
Huang S, Zhu M, Wu W, Rashid A, Liang Y, Hou L, Ning Q, Luo X. Valproate pretreatment protects pancreatic β-cells from palmitate-induced ER stress and apoptosis by inhibiting glycogen synthase kinase-3β. J Biomed Sci 2014; 21:38. [PMID: 24884462 PMCID: PMC4084580 DOI: 10.1186/1423-0127-21-38] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2014] [Accepted: 04/29/2014] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Reduction of pancreatic β-cells mass, major secondary to increased β-cells apoptosis, is increasingly recognized as one of the main contributing factors to the pathogenesis of type 2 diabetes (T2D), and saturated free fatty acid palmitate has been shown to induce endoplasmic reticulum (ER) stress that may contribute to promoting β-cells apoptosis. Recent literature suggests that valproate, a diffusely prescribed drug in the treatment of epilepsy and bipolar disorder, can inhibit glycogen synthase kinase-3β (GSK-3β) activity and has cytoprotective effects in neuronal cells and HepG2 cells. Thus, we hypothesized that valproate may protect INS-1 β-cells from palmitate-induced apoptosis via inhibiting GSK-3β. RESULTS Valproate pretreatment remarkable prevented palmitate-mediated cytotoxicity and apoptosis (lipotoxicity) as well as ER distension. Furthermore, palmitate triggered ER stress as evidenced by increased mRNA levels of C/EBP homologous protein (CHOP) and activating transcription factor 4 (ATF4) in a time-dependent fashion. However, valproate not only reduced the mRNA and protein expression of CHOP but also inhibited GSK-3β and caspase-3 activity induced by palmitate, whereas, the mRNA expression of ATF4 was not affected. Interestingly, TDZD-8, a specific GSK-3β inhibitor, also showed the similar effect on lipotoxicity and ER stress as valproate in INS-1 cells. Finally, compared with CHOP knockdown, valproate displayed better cytoprotection against palmitate. CONCLUSIONS Valproate may protect β-cells from palmitate-induced apoptosis and ER stress via GSK-3β inhibition, independent of ATF4/CHOP pathway. Besides, GSK-3β, rather than CHOP, may be a more promising therapeutic target for T2D.
Collapse
Affiliation(s)
- Shan Huang
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095, Jiefang Avenue, Wuhan, Hubei Province 430030, P.R. China
| | - Minghui Zhu
- Department of Neurology, Wuhan Integrated TCM & Western Medicine Hospital, Wuhan, China
| | - Wei Wu
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095, Jiefang Avenue, Wuhan, Hubei Province 430030, P.R. China
| | - Abid Rashid
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095, Jiefang Avenue, Wuhan, Hubei Province 430030, P.R. China
| | - Yan Liang
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095, Jiefang Avenue, Wuhan, Hubei Province 430030, P.R. China
| | - Ling Hou
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095, Jiefang Avenue, Wuhan, Hubei Province 430030, P.R. China
| | - Qin Ning
- Department of Infectious Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoping Luo
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095, Jiefang Avenue, Wuhan, Hubei Province 430030, P.R. China
| |
Collapse
|