1
|
Yang WY, Wang J, Li XH, Xu B, Yang YW, Yu L, Zhang B, Feng JF. Analysis of non-targeted serum metabolomics in patients with chronic kidney disease and hyperuricemia. Biotechnol Genet Eng Rev 2024; 40:4013-4039. [PMID: 37098873 DOI: 10.1080/02648725.2023.2204715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 04/13/2023] [Indexed: 04/27/2023]
Abstract
Hyperuricemia (HUA) is a common complication of chronic kidney disease (CKD). Conversely, HUA can promote the disease progression of CKD. However, the molecular mechanism of HUA in CKD development remains unclear. In the present study, we applied ultra-performance liquid chromatography-tandem mass spectrometry (UPLC-MS/MS) to analyze the serum metabolite profiling of 47 HUA patients, 41 non-hyperuricemic CKD (NUA-CKD) patients, and 51 CKD and HUA (HUA-CKD) patients, and then subjected to multivariate statistical analysis, metabolic pathway analysis and diagnostic performance evaluation. Metabolic profiling of serums showed that 40 differential metabolites (fold-change threshold (FC) > 1.5 or<2/3, variable importance in projection (VIP) > 1, and p < 0.05) were screened in HUA-CKD and HUA patients, and 24 differential metabolites (FC > 1.2 or<0.83, VIP>1, and p < 0.05) were screened in HUA-CKD and NUA-CKD patients. According to the analysis of metabolic pathways, significant changes existed in three metabolic pathways (compared with the HUA group) and two metabolic pathways (compared with the HUA-CKD group) in HUA-CKD patients. Glycerophospholipid metabolism was a significant pathway in HUA-CKD. Our findings show that the metabolic disorder in HUA-CKD patients was more serious than that in NUA-CKD or HUA patients. A theoretical basis is provided for HUA to accelerate CKD progress.
Collapse
Affiliation(s)
- Wen-Yu Yang
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jun Wang
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiao-Han Li
- Department of Medical Laboratory, Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Bei Xu
- Department of Clinical Laboratory, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, China
| | - Yu-Wei Yang
- Department of Clinical Laboratory, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, China
| | - Lin Yu
- Department of Clinical Laboratory, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, China
| | - Bin Zhang
- Department of Clinical Laboratory, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, China
| | - Jia-Fu Feng
- Department of Clinical Laboratory, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, China
| |
Collapse
|
2
|
Yi LX, Tan EK, Zhou ZD. Tyrosine Hydroxylase Inhibitors and Dopamine Receptor Agonists Combination Therapy for Parkinson's Disease. Int J Mol Sci 2024; 25:4643. [PMID: 38731862 PMCID: PMC11083272 DOI: 10.3390/ijms25094643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 04/11/2024] [Accepted: 04/22/2024] [Indexed: 05/13/2024] Open
Abstract
There are currently no disease-modifying therapies for Parkinson's disease (PD), a progressive neurodegenerative disorder associated with dopaminergic neuronal loss. There is increasing evidence that endogenous dopamine (DA) can be a pathological factor in neurodegeneration in PD. Tyrosine hydroxylase (TH) is the key rate-limiting enzyme for DA generation. Drugs that inhibit TH, such as alpha-methyltyrosine (α-MT), have recently been shown to protect against neurodegeneration in various PD models. DA receptor agonists can activate post-synaptic DA receptors to alleviate DA-deficiency-induced PD symptoms. However, DA receptor agonists have no therapeutic effects against neurodegeneration. Thus, a combination therapy with DA receptor agonists plus TH inhibitors may be an attractive therapeutic approach. TH inhibitors can protect and promote the survival of remaining dopaminergic neurons in PD patients' brains, whereas DA receptor agonists activate post-synaptic DA receptors to alleviate PD symptoms. Additionally, other PD drugs, such as N-acetylcysteine (NAC) and anticholinergic drugs, may be used as adjunctive medications to improve therapeutic effects. This multi-drug cocktail may represent a novel strategy to protect against progressive dopaminergic neurodegeneration and alleviate PD disease progression.
Collapse
Affiliation(s)
- Ling Xiao Yi
- National Neuroscience Institute of Singapore, 11 Jalan Tan Tock Seng, Singapore 308433, Singapore;
| | - Eng King Tan
- National Neuroscience Institute of Singapore, 11 Jalan Tan Tock Seng, Singapore 308433, Singapore;
- Department of Neurology, Singapore General Hospital, Outram Road, Singapore 169608, Singapore
- Signature Research Program in Neuroscience and Behavioral Disorders, Duke-NUS Graduate Medical School Singapore, 8 College Road, Singapore 169857, Singapore
| | - Zhi Dong Zhou
- National Neuroscience Institute of Singapore, 11 Jalan Tan Tock Seng, Singapore 308433, Singapore;
- Signature Research Program in Neuroscience and Behavioral Disorders, Duke-NUS Graduate Medical School Singapore, 8 College Road, Singapore 169857, Singapore
| |
Collapse
|
3
|
Azevedo MD, Prince N, Humbert-Claude M, Mesa-Infante V, Jeanneret C, Golzne V, De Matos K, Jamot BB, Magara F, Gonzalez-Hernandez T, Tenenbaum L. Oxidative stress induced by sustained supraphysiological intrastriatal GDNF delivery is prevented by dose regulation. Mol Ther Methods Clin Dev 2023; 31:101106. [PMID: 37766790 PMCID: PMC10520444 DOI: 10.1016/j.omtm.2023.09.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Accepted: 09/06/2023] [Indexed: 09/29/2023]
Abstract
Despite its established neuroprotective effect on dopaminergic neurons and encouraging phase I results, intraputaminal GDNF administration failed to demonstrate significant clinical benefits in Parkinson's disease patients. Different human GDNF doses were delivered in the striatum of rats with a progressive 6-hydroxydopamine lesion using a sensitive doxycycline-regulated AAV vector. GDNF treatment was applied either continuously or intermittently (2 weeks on/2 weeks off) during 17 weeks. Stable reduction of motor impairments as well as increased number of dopaminergic neurons and striatal innervation were obtained with a GDNF dose equivalent to 3- and 10-fold the rat endogenous level. In contrast, a 20-fold increased GDNF level only temporarily provided motor benefits and neurons were not spared. Strikingly, oxidized DNA in the substantia nigra increased by 50% with 20-fold, but not 3-fold GDNF treatment. In addition, only low-dose GDNF allowed to preserve dopaminergic neuron cell size. Finally, aberrant dopaminergic fiber sprouting was observed with 20-fold GDNF but not at lower doses. Intermittent 20-fold GDNF treatment allowed to avoid toxicity and spare dopaminergic neurons but did not restore their cell size. Our data suggest that maintaining GDNF concentration under a threshold generating oxidative stress is a pre-requisite to obtain significant symptomatic relief and neuroprotection.
Collapse
Affiliation(s)
- Marcelo Duarte Azevedo
- Laboratory of Cellular and Molecular Neurotherapies, Center for Neuroscience Research, Clinical Neurosciences Department, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), 1011 Lausanne, Switzerland
| | - Naika Prince
- Laboratory of Cellular and Molecular Neurotherapies, Center for Neuroscience Research, Clinical Neurosciences Department, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), 1011 Lausanne, Switzerland
| | - Marie Humbert-Claude
- Laboratory of Cellular and Molecular Neurotherapies, Center for Neuroscience Research, Clinical Neurosciences Department, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), 1011 Lausanne, Switzerland
| | - Virginia Mesa-Infante
- Departamento de Ciencias Médicas Básicas, Facultad de Ciencias de la Salud, Instituto de Tecnologías Biomédicas (ITB), Universidad de La Laguna, La Laguna, 38200 Tenerife, Spain
| | - Cheryl Jeanneret
- Laboratory of Cellular and Molecular Neurotherapies, Center for Neuroscience Research, Clinical Neurosciences Department, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), 1011 Lausanne, Switzerland
| | - Valentine Golzne
- Laboratory of Cellular and Molecular Neurotherapies, Center for Neuroscience Research, Clinical Neurosciences Department, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), 1011 Lausanne, Switzerland
| | - Kevin De Matos
- Laboratory of Cellular and Molecular Neurotherapies, Center for Neuroscience Research, Clinical Neurosciences Department, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), 1011 Lausanne, Switzerland
| | - Benjamin Boury Jamot
- Center for the Study of Behaviour, Department of Psychiatry, Lausanne University Hospital and University of Lausanne (CHUV-UNIL), 1008 Lausanne, Switzerland
| | - Fulvio Magara
- Center for the Study of Behaviour, Department of Psychiatry, Lausanne University Hospital and University of Lausanne (CHUV-UNIL), 1008 Lausanne, Switzerland
| | - Tomas Gonzalez-Hernandez
- Departamento de Ciencias Médicas Básicas, Facultad de Ciencias de la Salud, Instituto de Tecnologías Biomédicas (ITB), Universidad de La Laguna, La Laguna, 38200 Tenerife, Spain
| | - Liliane Tenenbaum
- Laboratory of Cellular and Molecular Neurotherapies, Center for Neuroscience Research, Clinical Neurosciences Department, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), 1011 Lausanne, Switzerland
| |
Collapse
|
4
|
Zhou ZD, Yi LX, Wang DQ, Lim TM, Tan EK. Role of dopamine in the pathophysiology of Parkinson's disease. Transl Neurodegener 2023; 12:44. [PMID: 37718439 PMCID: PMC10506345 DOI: 10.1186/s40035-023-00378-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 09/08/2023] [Indexed: 09/19/2023] Open
Abstract
A pathological feature of Parkinson's disease (PD) is the progressive loss of dopaminergic neurons and decreased dopamine (DA) content in the substantia nigra pars compacta in PD brains. DA is the neurotransmitter of dopaminergic neurons. Accumulating evidence suggests that DA interacts with environmental and genetic factors to contribute to PD pathophysiology. Disturbances of DA synthesis, storage, transportation and metabolism have been shown to promote neurodegeneration of dopaminergic neurons in various PD models. DA is unstable and can undergo oxidation and metabolism to produce multiple reactive and toxic by-products, including reactive oxygen species, DA quinones, and 3,4-dihydroxyphenylacetaldehyde. Here we summarize and highlight recent discoveries on DA-linked pathophysiologic pathways, and discuss the potential protective and therapeutic strategies to mitigate the complications associated with DA.
Collapse
Affiliation(s)
- Zhi Dong Zhou
- National Neuroscience Institute of Singapore, 11 Jalan Tan Tock Seng, Singapore, 308433, Singapore.
- Signature Research Program in Neuroscience and Behavioral Disorders, Duke-NUS Graduate Medical School Singapore, 8 College Road, Singapore, 169857, Singapore.
| | - Ling Xiao Yi
- National Neuroscience Institute of Singapore, 11 Jalan Tan Tock Seng, Singapore, 308433, Singapore
| | - Dennis Qing Wang
- Department of Neurology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Tit Meng Lim
- Department of Biological Science, National University of Singapore, Singapore, 119077, Singapore
| | - Eng King Tan
- National Neuroscience Institute of Singapore, 11 Jalan Tan Tock Seng, Singapore, 308433, Singapore.
- Department of Neurology, Singapore General Hospital, Outram Road, Singapore, 169608, Singapore.
- Signature Research Program in Neuroscience and Behavioral Disorders, Duke-NUS Graduate Medical School Singapore, 8 College Road, Singapore, 169857, Singapore.
| |
Collapse
|
5
|
Ranganath L, Khedr M, Milan AM, Davison AS, Norman BP, Janssen MCH, Lock E, Bou‐Gharios G, Gallagher JA. Increased prevalence of Parkinson's disease in alkaptonuria. JIMD Rep 2023; 64:282-292. [PMID: 37404676 PMCID: PMC10315388 DOI: 10.1002/jmd2.12367] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 04/27/2023] [Indexed: 07/06/2023] Open
Abstract
Amongst a cohort of 88 alkaptonuria (AKU) patients attending the United Kingdom National Alkaptonuria Centre (NAC), four unrelated patients had co-existing Parkinson's disease (PD). Two of the NAC patients developed PD before receiving nitisinone (NIT) while the other two developed overt PD during NIT therapy. NIT lowers redox-active homogentisic acid (HGA) and profoundly increases tyrosine (TYR). A further unpublished case of a Dutch patient with AKU and PD on deep brain stimulation is included in this report. A Pubmed search revealed a further five AKU patients with PD, all without NIT usage. The prevalence of PD in AKU in the NAC appears to be nearly 20-times higher than in the non-AKU population (p < 0.001) even when adjusted for age. We propose that life-long exposure to redox-active HGA may account for the higher prevalence of PD in AKU. Furthermore, the appearance of PD in AKU patients during NIT therapy may be due to unmasking dopamine deficiency in susceptible individuals, as a result of the tyrosinaemia during NIT therapy inhibiting the rate-limiting brain tyrosine hydroxylase.
Collapse
Affiliation(s)
- Lakshminarayan Ranganath
- Department of Clinical Biochemistry and Metabolic MedicineRoyal Liverpool University HospitalLiverpoolUK
- Department of Musculoskeletal and Ageing Science, Institute of Life Course and Medical SciencesUniversity of LiverpoolLiverpoolUK
| | - Milad Khedr
- Department of Clinical Biochemistry and Metabolic MedicineRoyal Liverpool University HospitalLiverpoolUK
| | - Anna M. Milan
- Department of Clinical Biochemistry and Metabolic MedicineRoyal Liverpool University HospitalLiverpoolUK
| | - Andrew S. Davison
- Department of Clinical Biochemistry and Metabolic MedicineRoyal Liverpool University HospitalLiverpoolUK
| | - Brendan P. Norman
- Department of Musculoskeletal and Ageing Science, Institute of Life Course and Medical SciencesUniversity of LiverpoolLiverpoolUK
| | - Mirian C. H. Janssen
- Departments of Internal Medicine & PediatricsRadboud University Nijmegen Medical CentreNijmegenNetherlands
| | - Edward Lock
- School of Pharmacy and Biomolecular SciencesLiverpool John Moores UniversityLiverpoolUK
| | - George Bou‐Gharios
- Department of Musculoskeletal and Ageing Science, Institute of Life Course and Medical SciencesUniversity of LiverpoolLiverpoolUK
| | - James A. Gallagher
- Department of Musculoskeletal and Ageing Science, Institute of Life Course and Medical SciencesUniversity of LiverpoolLiverpoolUK
| |
Collapse
|
6
|
Xiong C, Zhu Y, Luo Q, Phan CW, Huo Y, Li P, Li Q, Jin X, Huang W. Neuroprotective effects of a novel peptide from Lignosus rhinocerotis against 6-hydroxydopamine-induced apoptosis in PC12 cells by inhibiting NF-κB activation. Food Sci Nutr 2023; 11:2152-2165. [PMID: 37181320 PMCID: PMC10171544 DOI: 10.1002/fsn3.3050] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Revised: 08/07/2022] [Accepted: 08/22/2022] [Indexed: 11/11/2022] Open
Abstract
According to previous studies, oxidative stress is a leading cause of dopaminergic neuron death and may contribute to the pathogenesis of Parkinson's disease (PD). In the current study, we used chromatography of gel filtration to identify a novel peptide (Lignosus rhinocerotis peptide [LRP]) from the sclerotium of Lignosus rhinocerotis (Cooke) Ryvarden. Its neuroprotective effect was evaluated using an in vitro PD model constructed by 6-hydroxydopamine (6-OHDA)-stimulated to apoptosis in PC12 cells. The molecular weight of LRP is determined as 1532 Da and the secondary structure is irregular. The simple amino acid sequence of LRP is Thr-Leu-Ala-Pro-Thr-Phe-Leu-Ser-Ser-Leu-Gly-Pro-Cys-Leu-Leu. Notably, LRP has the ability to significantly boost the viability of PC12 cells after exposure to 6-OHDA, as well as enhance the cellular activity of antioxidative enzymes like superoxide dismutase (SOD), catalase (CAT), and glutathione peroxidase (GSH-Px). LRP also lowers the level of malondialdehyde (MDA), decreases the activation performance of Caspase-3, and reduces 6-OHDA-induced apoptosis via inhibition of nuclear factor-kappa B (NF-κB) activation. These data indicate that LRP may have the potential to act as a neuroprotective agent.
Collapse
Affiliation(s)
- Chuan Xiong
- Biotechnology and Nuclear Technology Research InstituteSichuan Academy of Agricultural SciencesChengduChina
| | - Yu Zhu
- Biotechnology and Nuclear Technology Research InstituteSichuan Academy of Agricultural SciencesChengduChina
| | - Qiang Luo
- The Second Affiliated HospitalChongqing Medical UniversityChongqingChina
| | - Chia Wei Phan
- Mushroom Research CentreUniversiti MalayaKuala LumpurMalaysia
- Department of Pharmaceutical Life SciencesFaculty of PharmacyUniversiti MalayaKuala LumpurMalaysia
| | - Yujie Huo
- Yunnan Plateau Characteristic Agricultural Industry Research InstituteYunnan Agricultural UniversityKunmingChina
| | - Ping Li
- Biotechnology and Nuclear Technology Research InstituteSichuan Academy of Agricultural SciencesChengduChina
| | - Qiang Li
- College of Food and Biological EngineeringChengdu UniversityChengduChina
| | - Xin Jin
- Biotechnology and Nuclear Technology Research InstituteSichuan Academy of Agricultural SciencesChengduChina
| | - Wenli Huang
- Biotechnology and Nuclear Technology Research InstituteSichuan Academy of Agricultural SciencesChengduChina
| |
Collapse
|
7
|
Dutta A, Phukan BC, Roy R, Mazumder MK, Paul R, Choudhury A, Kumar D, Bhattacharya P, Nath J, Kumar S, Borah A. Garcinia morella extract confers dopaminergic neuroprotection by mitigating mitochondrial dysfunctions and inflammation in mouse model of Parkinson's disease. Metab Brain Dis 2022; 37:1887-1900. [PMID: 35622265 DOI: 10.1007/s11011-022-01001-9] [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: 02/16/2022] [Accepted: 05/04/2022] [Indexed: 11/24/2022]
Abstract
Dopaminergic neuroprotection is the main interest in designing novel therapeutics against Parkinson's disease (PD). In the process of dopaminergic degeneration, mitochondrial dysfunctions and inflammation are significant. While the existing drugs provide symptomatic relief against PD, a therapy conferring total neuroprotection by targeting multiple degenerative pathways is still lacking. Garcinia morella is a common constituent of Ayurvedic medication and has been used for the treatment of inflammatory disorders. The present study investigates whether administration of G. morella fruit extract (GME) in MPTP mouse model of PD protects against dopaminergic neurodegeneration, including the underlying pathophysiologies, and reverses the motor behavioural abnormalities. Administration of GME prevented the loss of dopaminergic cell bodies in the substantia nigra and its terminals in the corpus striatum of PD mice. Subsequently, reversal of parkinsonian behavioural abnormalities, viz. akinesia, catalepsy, and rearing, was observed along with the recovery of striatal dopamine and its metabolites in the experimental model. Furthermore, reduced activity of the mitochondrial complex II in the nigrostriatal pathway of brain of the mice was restored after the administration of GME. Also, MPTP-induced enhanced activation of Glial fibrillary acidic protein (GFAP) and neuronal nitric oxide synthase (nNOS) in the nigrostriatal pathway, which are the markers of inflammatory stress, were found to be ameliorated on GME treatment. Thus, our study presented a novel mode of dopaminergic neuroprotection by G. morella in PD by targeting the mitochondrial dysfunctions and neuroinflammation, which are considered to be intricately associated with the loss of dopaminergic neurons.
Collapse
Affiliation(s)
- Ankumoni Dutta
- Department of Life Science and Bioinformatics, Assam University, Silchar, 788011, Assam, India
- Department of Zoology, Pandit Deendayal Upadhyaya Adarsha Mahavidyalaya (PDUAM), Behali, Biswanath, Assam, India
| | - Banashree Chetia Phukan
- Department of Life Science and Bioinformatics, Assam University, Silchar, 788011, Assam, India
| | - Rubina Roy
- Department of Life Science and Bioinformatics, Assam University, Silchar, 788011, Assam, India
| | | | - Rajib Paul
- Department of Zoology, Pandit Deendayal Upadhyaya Adarsha Mahavidyalaya (PDUAM), Eraligool, Karimganj, Assam, India
| | | | - Diwakar Kumar
- Department of Microbiology, Assam University, Silchar, Assam, India
| | - Pallab Bhattacharya
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gandhinagar, Gujarat, India
| | - Joyobrato Nath
- Department of Zoology, Cachar College, Silchar, Assam, India
| | - Sanjeev Kumar
- Department of Life Science and Bioinformatics, Assam University, Silchar, 788011, Assam, India.
| | - Anupom Borah
- Department of Life Science and Bioinformatics, Assam University, Silchar, 788011, Assam, India.
| |
Collapse
|
8
|
Elabi OF, Pass R, Sormonta I, Nolbrant S, Drummond N, Kirkeby A, Kunath T, Parmar M, Lane EL. Human Embryonic Stem Cell-Derived Dopaminergic Grafts Alleviate L-DOPA Induced Dyskinesia. JOURNAL OF PARKINSON'S DISEASE 2022; 12:1881-1896. [PMID: 35466951 DOI: 10.3233/jpd-212920] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
BACKGROUND First-in-human studies to test the efficacy and safety of human embryonic stem cells (hESC)-derived dopaminergic cells in the treatment of Parkinson's disease (PD) are imminent. Pre-clinical studies using hESC-derived dopamine neuron transplants in rat models have indicated that the benefits parallel those shown with fetal tissue but have thus far failed to consider how ongoing L-DOPA administration might impact on the graft. OBJECTIVE To determine whether L-DOPA impacts on survival and functional recovery following grafting of hESC-derived dopaminergic neurons. METHODS Unilateral 6-OHDA lesioned rats were administered with either saline or L-DOPA prior to, and for 18 weeks following surgical implantation of dopaminergic neural progenitors derived from RC17 hESCs according to two distinct protocols in independent laboratories. RESULTS Grafts from both protocols elicited reduction in amphetamine-induced rotations. Reduced L-DOPA-induced dyskinesia preceded the improvement in amphetamine-induced rotations. Furthermore, L-DOPA had no effect on overall survival (HuNu) or dopaminergic neuron content of the graft (TH positive cells) but did lead to an increase in the number of GIRK2 positive neurons. CONCLUSION Critically, we found that L-DOPA was not detrimental to graft function, potentially enhancing graft maturation and promoting an A9 phenotype. Early improvement of L-DOPA-induced dyskinesia suggests that grafts may support the handling of exogenously supplied dopamine earlier than improvements in amphetamine-induced behaviours indicate. Given that one of the protocols will be employed in the production of cells for the European STEM-PD clinical trial, this is vital information for the management of patients and achieving optimal outcomes following transplantation of hESC-derived grafts for PD.
Collapse
Affiliation(s)
- Osama F Elabi
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, UK
| | - Rachel Pass
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, UK
| | - Irene Sormonta
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, UK
| | - Sara Nolbrant
- Developmental and Regenerative Neurobiology, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Nicola Drummond
- Centre for Regenerative Medicine, Institute for Stem Cell Research, School of Biological Sciences, University of Edinburgh, Edinburgh, UK
| | - Agnete Kirkeby
- Developmental and Regenerative Neurobiology, Department of Experimental Medical Science, Lund University, Lund, Sweden
- Department of Neuroscience and The Novo Nordisk Foundation Center for Stem Cell Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Tilo Kunath
- Centre for Regenerative Medicine, Institute for Stem Cell Research, School of Biological Sciences, University of Edinburgh, Edinburgh, UK
| | - Malin Parmar
- Developmental and Regenerative Neurobiology, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Emma L Lane
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, UK
| |
Collapse
|
9
|
Aboushady Y, Gabr M, ElHady AK, Salah M, Abadi AH, Wilms G, Becker W, Abdel-Halim M, Engel M. Discovery of Hydroxybenzothiazole Urea Compounds as Multitargeted Agents Suppressing Major Cytotoxic Mechanisms in Neurodegenerative Diseases. ACS Chem Neurosci 2021; 12:4302-4318. [PMID: 34726394 DOI: 10.1021/acschemneuro.1c00475] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Multiple factors are causally responsible and/or contribute to the progression of Alzheimer's and Parkinson's diseases. The protein kinase Dyrk1A was identified as a promising target as it phosphorylates tau protein, α-synuclein, and parkin. The first goal of our study was to optimize our previously identified Dyrk1A inhibitors of the 6-hydroxy benzothiazole urea chemotype in terms of potency and selectivity. Our efforts led to the development of the 3-fluorobenzyl amide derivative 16b, which displayed the highest potency against Dyrk1A (IC50 = 9.4 nM). In general, the diversification of the benzylamide moiety led to an enhanced selectivity over the most homologous isoform, Dyrk1B, which was a meaningful indicator, as the high selectivity could be confirmed in an extended selectivity profiling of 3b and 16b. Eventually, we identified the novel phenethyl amide derivative 24b as a triple inhibitor of Dyrk1A kinase activity (IC50 = 119 nM) and the aggregation of tau and α-syn oligomers. We provide evidence that the novel combination of selective Dyrk1A inhibition and suppression of tau and α-syn aggregations of our new lead compound confers efficacy in several established cellular models of neurotoxic mechanisms relevant to neurodegenerative diseases, including α-syn- and 6-hydroxydopamine-induced cytotoxicities.
Collapse
Affiliation(s)
- Youssef Aboushady
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo 11835, Egypt
| | - Moustafa Gabr
- Department of Radiology, Stanford University, Stanford, California 94305, United States
| | - Ahmed K. ElHady
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo 11835, Egypt
- School of Life and Medical Sciences, University of Hertfordshire Hosted By Global Academic Foundation, New Administrative Capital, Cairo 11311, Egypt
| | - Mohamed Salah
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, October University for Modern Sciences and Arts, Cairo 12451, Egypt
| | - Ashraf H. Abadi
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo 11835, Egypt
| | - Gerrit Wilms
- Institute of Pharmacology and Toxicology, Medical Faculty of the RWTH Aachen University, Wendlingweg 2, Aachen 52074, Germany
| | - Walter Becker
- Institute of Pharmacology and Toxicology, Medical Faculty of the RWTH Aachen University, Wendlingweg 2, Aachen 52074, Germany
| | - Mohammad Abdel-Halim
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo 11835, Egypt
| | - Matthias Engel
- Pharmaceutical and Medicinal Chemistry, Saarland University, Campus C2.3 Saarbrücken D-66123, Germany
| |
Collapse
|
10
|
Wang L, Sheng W, Tan Z, Ren Q, Wang R, Stoika R, Liu X, Liu K, Shang X, Jin M. Treatment of Parkinson's disease in Zebrafish model with a berberine derivative capable of crossing blood brain barrier, targeting mitochondria, and convenient for bioimaging experiments. Comp Biochem Physiol C Toxicol Pharmacol 2021; 249:109151. [PMID: 34343700 DOI: 10.1016/j.cbpc.2021.109151] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 06/23/2021] [Accepted: 07/23/2021] [Indexed: 12/25/2022]
Abstract
Berberine is a famous alkaloid extracted from Berberis plants and has been widely used as medications and functional food additives. Recent studies reveal that berberine exhibits neuroprotective activity in animal models of Parkinson's disease (PD), the second most prevalent neurodegenerative disorders all over the world. However, the actual site of anti-PD action of berberine remains largely unknown. To this end, we employed a fluorescently labeled berberine derivative BBRP to investigate the subcellular localization and blood brain barrier (BBB) permeability in a cellular model of PD and zebrafish PD model. Biological investigations revealed that BBRP retained the neuroprotective activity of berberine against PD-like symptoms in PC12 cells and zebrafish, such as protecting 6-OHDA induced cell death, relieving MPTP induced PD-like behavior and increasing dopaminergic neuron loss in zebrafish. We also found that BBRP could readily penetrate BBB and function in the brain of zebrafish suffering from PD. Subcellular localization study indicated that BBRP could rapidly and specifically accumulate in mitochondria of PC12 cells when it exerted anti-PD effect. In addition, BBRP could suppress accumulation of Pink1 protein and inhibit the overexpression of LC3 protein in 6-OHDA damaged cells. All these results suggested that the potential site of action of berberine is mitochondria in the brain under the PD condition. Therefore, the findings described herein would be useful for further development of berberine as an anti-PD drug.
Collapse
Affiliation(s)
- Lizhen Wang
- Biology Institute, Qilu University of Technology, Shandong Academy of Sciences, Jinan 250103, Shandong Province, China; Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Jinan 250103, Shandong Province, China
| | - Wenlong Sheng
- Biology Institute, Qilu University of Technology, Shandong Academy of Sciences, Jinan 250103, Shandong Province, China; Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Jinan 250103, Shandong Province, China
| | - Zhaoshun Tan
- School of Bioengineering, Qilu University of Technology, Shandong Academy of Sciences, 3501 Daxue Road, Jinan 250353, Shandong Province, China
| | - Qingyu Ren
- School of Psychology and mental health, North China University of Science and Technology, 21 Bohai Road, Tang'shan 063210, Hebei Province, China
| | - Rongchun Wang
- Biology Institute, Qilu University of Technology, Shandong Academy of Sciences, Jinan 250103, Shandong Province, China; Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Jinan 250103, Shandong Province, China
| | - Rostyslav Stoika
- Department of Regulation of Cell Proliferation and Apoptosis, Institute of Cell Biology, National Academy of Sciences of Ukraine, Lviv 79005, Ukraine
| | - Xuedong Liu
- Department of Chemistry and Biochemistry, University of Colorado at Boulder, Boulder, CO 80303, United States of America
| | - Kechun Liu
- Biology Institute, Qilu University of Technology, Shandong Academy of Sciences, Jinan 250103, Shandong Province, China; Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Jinan 250103, Shandong Province, China
| | - Xueliang Shang
- School of Psychology and mental health, North China University of Science and Technology, 21 Bohai Road, Tang'shan 063210, Hebei Province, China.
| | - Meng Jin
- Biology Institute, Qilu University of Technology, Shandong Academy of Sciences, Jinan 250103, Shandong Province, China; Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Jinan 250103, Shandong Province, China.
| |
Collapse
|
11
|
Yonesi M, Garcia-Nieto M, Guinea GV, Panetsos F, Pérez-Rigueiro J, González-Nieto D. Silk Fibroin: An Ancient Material for Repairing the Injured Nervous System. Pharmaceutics 2021; 13:429. [PMID: 33806846 PMCID: PMC8004633 DOI: 10.3390/pharmaceutics13030429] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 03/16/2021] [Accepted: 03/17/2021] [Indexed: 12/25/2022] Open
Abstract
Silk refers to a family of natural fibers spun by several species of invertebrates such as spiders and silkworms. In particular, silkworm silk, the silk spun by Bombyx mori larvae, has been primarily used in the textile industry and in clinical settings as a main component of sutures for tissue repairing and wound ligation. The biocompatibility, remarkable mechanical performance, controllable degradation, and the possibility of producing silk-based materials in several formats, have laid the basic principles that have triggered and extended the use of this material in regenerative medicine. The field of neural soft tissue engineering is not an exception, as it has taken advantage of the properties of silk to promote neuronal growth and nerve guidance. In addition, silk has notable intrinsic properties and the by-products derived from its degradation show anti-inflammatory and antioxidant properties. Finally, this material can be employed for the controlled release of factors and drugs, as well as for the encapsulation and implantation of exogenous stem and progenitor cells with therapeutic capacity. In this article, we review the state of the art on manufacturing methodologies and properties of fiber-based and non-fiber-based formats, as well as the application of silk-based biomaterials to neuroprotect and regenerate the damaged nervous system. We review previous studies that strategically have used silk to enhance therapeutics dealing with highly prevalent central and peripheral disorders such as stroke, Alzheimer's disease, Parkinson's disease, and peripheral trauma. Finally, we discuss previous research focused on the modification of this biomaterial, through biofunctionalization techniques and/or the creation of novel composite formulations, that aim to transform silk, beyond its natural performance, into more efficient silk-based-polymers towards the clinical arena of neuroprotection and regeneration in nervous system diseases.
Collapse
Affiliation(s)
- Mahdi Yonesi
- Center for Biomedical Technology, Universidad Politécnica de Madrid, 28223 Pozuelo de Alarcón, Spain; (M.Y.); (G.V.G.)
- Silk Biomed SL, 28260 Madrid, Spain;
| | | | - Gustavo V. Guinea
- Center for Biomedical Technology, Universidad Politécnica de Madrid, 28223 Pozuelo de Alarcón, Spain; (M.Y.); (G.V.G.)
- Silk Biomed SL, 28260 Madrid, Spain;
- Departamento de Ciencia de Materiales, ETSI Caminos, Canales y Puertos, Universidad Politécnica de Madrid, 28040 Madrid, Spain
- Biomedical Research Networking Center in Bioengineering Biomaterials and Nanomedicine (CIBER-BBN), 28029 Madrid, Spain
| | - Fivos Panetsos
- Silk Biomed SL, 28260 Madrid, Spain;
- Neurocomputing and Neurorobotics Research Group, Faculty of Biology and Faculty of Optics, Universidad Complutense de Madrid, 28040 Madrid, Spain
- Innovation Group, Institute for Health Research San Carlos Clinical Hospital (IdISSC), 28040 Madrid, Spain
| | - José Pérez-Rigueiro
- Center for Biomedical Technology, Universidad Politécnica de Madrid, 28223 Pozuelo de Alarcón, Spain; (M.Y.); (G.V.G.)
- Silk Biomed SL, 28260 Madrid, Spain;
- Departamento de Ciencia de Materiales, ETSI Caminos, Canales y Puertos, Universidad Politécnica de Madrid, 28040 Madrid, Spain
- Biomedical Research Networking Center in Bioengineering Biomaterials and Nanomedicine (CIBER-BBN), 28029 Madrid, Spain
| | - Daniel González-Nieto
- Center for Biomedical Technology, Universidad Politécnica de Madrid, 28223 Pozuelo de Alarcón, Spain; (M.Y.); (G.V.G.)
- Silk Biomed SL, 28260 Madrid, Spain;
- Biomedical Research Networking Center in Bioengineering Biomaterials and Nanomedicine (CIBER-BBN), 28029 Madrid, Spain
- Departamento de Tecnología Fotónica y Bioingeniería, ETSI Telecomunicaciones, Universidad Politécnica de Madrid, 28040 Madrid, Spain
| |
Collapse
|
12
|
Chen SH, Kuo CW, Lin TK, Tsai MH, Liou CW. Dopamine Therapy and the Regulation of Oxidative Stress and Mitochondrial DNA Copy Number in Patients with Parkinson's Disease. Antioxidants (Basel) 2020; 9:antiox9111159. [PMID: 33233852 PMCID: PMC7699910 DOI: 10.3390/antiox9111159] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 11/06/2020] [Accepted: 11/08/2020] [Indexed: 11/25/2022] Open
Abstract
Few studies have reported on changes to oxidative stress and mitochondrial DNA copy numbers in patients with Parkinson’s disease (PD), particularly those undergoing long-term dopamine therapy. This study measured mitochondrial copy numbers, thiobarbituric acid reactive substances (TBARS), and thiols in 725 PD patients and 744 controls. The total prescribed dopamine dose was calculated for each PD patient. A decreased mitochondrial copy number and antioxidant thiols level, but an elevated oxidative TBARS level presented in PD patients. Stratification into age subgroups revealed a consistently lower mitochondrial copy number and thiols in all PD subgroups, but increased TBARS levels compared with those of the controls. Further study found an association between lower serum TBARS and dopamine administration. There appears to be an indirect relationship with the mitochondrial copy number, where a decrease in TBARS was found to diminish the effect of pathogenetic and age-related decrease in mitochondrial copy number in PD patients. Follow-up evaluations noted more significant decreases of mitochondrial copy numbers in PD patients over time; meanwhile, dopamine administration was associated with an initial decrease of the TBARS level which attenuated with high-dose and long-term therapy. Our study provides evidence that moderate dopamine dose therapy benefits PD patients through attenuation of oxidative stress and manipulation of the mitochondrial copy number.
Collapse
Affiliation(s)
- Shih-Hsuan Chen
- Department of Neurology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan; (S.-H.C.); (T.-K.L.); (M.-H.T.)
| | - Chung-Wen Kuo
- Core Laboratory for Phenomics and Diagnostics, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan;
| | - Tsu-Kung Lin
- Department of Neurology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan; (S.-H.C.); (T.-K.L.); (M.-H.T.)
- Center for Mitochondrial Research and Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan
| | - Meng-Han Tsai
- Department of Neurology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan; (S.-H.C.); (T.-K.L.); (M.-H.T.)
- Center for Mitochondrial Research and Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan
| | - Chia-Wei Liou
- Department of Neurology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan; (S.-H.C.); (T.-K.L.); (M.-H.T.)
- Center for Mitochondrial Research and Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan
- Correspondence: ; Tel.: +886-7-7317123 (ext. 2285); Fax: +886-7-7318762
| |
Collapse
|
13
|
Biju KC, Evans RC, Shrestha K, Carlisle DCB, Gelfond J, Clark RA. Methylene Blue Ameliorates Olfactory Dysfunction and Motor Deficits in a Chronic MPTP/Probenecid Mouse Model of Parkinson's Disease. Neuroscience 2018; 380:111-122. [PMID: 29684508 DOI: 10.1016/j.neuroscience.2018.04.008] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Revised: 04/03/2018] [Accepted: 04/09/2018] [Indexed: 12/30/2022]
Abstract
Mitochondrial dysfunction and oxidative stress are very prominent and early features in Parkinson's disease (PD) and in animal models of PD. Thus, antioxidant therapy for PD has been proposed, but in clinical trials such strategies have met with very limited success. Methylene blue (MB), a small-molecule synthetic heterocyclic organic compound that acts as a renewable electron cycler in the mitochondrial electron transport chain, manifesting robust antioxidant and cell energetics-enhancing properties, has recently been shown to have significant beneficial effects in reducing nigrostriatal dopaminergic loss and motor impairment in acute toxin models of PD. However, no studies have investigated the impact of this promising agent in chronic models or for olfactory dysfunction, an early non-motor feature of PD. To test the efficacy of low-dose MB for olfactory dysfunction, motor symptoms, and dopaminergic neurodegeneration, mice were injected with ten subcutaneous doses of 25 mg/kg MPTP, plus 250 mg/kg intraperitoneal probenecid or saline/probenecid at 3.5-day intervals. Following the onset of olfactory dysfunction, MPTP/probenecid (MPTP/p) and saline/probenecid mice were provided drinking water with or without 1 mg/kg/day MB. Oral delivery of low-dose MB significantly ameliorated MPTP/p-induced deficits in motor coordination, as well as degeneration of tyrosine hydroxylase (TH)-positive neurons of the substantia nigra and TH-positive terminals in the striatum. Importantly, olfactory dysfunction was ameliorated by MB treatment, whereas this benefit is not observed with currently available anti-Parkinsonian medications. These results indicate that low-dose MB is a promising neuroprotective intervention for both motor and non-motor features of PD.
Collapse
Affiliation(s)
- K C Biju
- Department of Medicine, University of Texas Health Science Center, 7703 Floyd Curl Drive, San Antonio, TX 78229, United States
| | - Robert C Evans
- Department of Medicine, University of Texas Health Science Center, 7703 Floyd Curl Drive, San Antonio, TX 78229, United States
| | - Kripa Shrestha
- Department of Medicine, University of Texas Health Science Center, 7703 Floyd Curl Drive, San Antonio, TX 78229, United States
| | - Daniel C B Carlisle
- Department of Medicine, University of Texas Health Science Center, 7703 Floyd Curl Drive, San Antonio, TX 78229, United States
| | - Jonathan Gelfond
- Department of Epidemiology and Biostatistics, University of Texas Health Science Center, 7703 Floyd Curl Drive, San Antonio, TX 78229, United States
| | - Robert A Clark
- Department of Medicine, University of Texas Health Science Center, 7703 Floyd Curl Drive, San Antonio, TX 78229, United States; South Texas Veterans Health Care System, 7400 Merton Minter Blvd, San Antonio, TX 78229, United States.
| |
Collapse
|
14
|
Mapanao R, Kuo HW, Chang CC, Liu KF, Cheng W. L-3,4-Dihydroxyphenylalanine (l-DOPA) induces neuroendocrinological, physiological, and immunological regulation in white shrimp, Litopenaeus vannamei. FISH & SHELLFISH IMMUNOLOGY 2018; 74:162-169. [PMID: 29305987 DOI: 10.1016/j.fsi.2017.12.063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 12/18/2017] [Accepted: 12/31/2017] [Indexed: 06/07/2023]
Abstract
L-3,4-Dihydroxyphenylalanine (l-DOPA) is a precursor for dopamine (DA) synthesis. Assessments were conducted to analyze the effects of l-DOPA on mediating regulation of neuroendocrinological, immunological, and physiological parameters in the shrimp, Litopenaeus vannamei when they were individually injected with 0.01 N HCl or l-DOPA at 0.5 or 1.0 μmol shrimp-1 for 60, 120, and 240 min. For catecholamine synthesis evaluation, tyrosine hydroxylase (TH) and DA beta hydroxylase (DBH) activities, l-DOPA, DA, and norepinephrine (NE) levels in hemolymph were determined. The total hemocyte count (THC), differential hemocyte count (DHC), phenoloxidase (PO) activity, respiratory bursts (RBs), superoxide dismutase (SOD) activity, phagocytic activity, and clearance efficiency in response to the pathogen, Vibrio alginolyticus were assessed for immune responses, and plasma glucose and lactate levels were for physiological response. Results showed that the TH activity, THC, hyaline cells (HCs), and semigranular cells (SGCs) at 120 min, DA levels at 60-240 min, PO activity in hemocytes per 50 μL of hemolymph at 60-120 min, and PO activity per granulocyte (granular cells (GCs) + SGCs) at 60 min significantly increased, but TH activity, l-DOPA levels, GCs, SGCs, and respiratory bursts in hemocytes per 10 μL of hemolymph at 60 min, respiratory bursts per hemocyte and SOD activity at 120 min, phagocytic activity at 60-240 min, and the clearance efficiency at 60-120 min significantly decreased in shrimp injected with l-DOPA at 1.0 μmol shrimp-1. In another experiment, 60 min after shrimp had received l-DOPA at 0.5 or 1.0 μmol shrimp-1, they were challenged with an injection of V. alginolyticus at 2 × 105 colony-forming units (cfu) shrimp-1. The injection of l-DOPA at 1.0 μmol shrimp-1 also significantly increased the cumulative mortality of shrimp by 16.7%, compared to the HCl-challenged control after 120 h. These results suggest that l-DOPA administration at 1.0 μmol shrimp-1 can mediate the transient regulation of neuroendocrinological, immunological, and physiologic responses resulting in immunosuppression, which in turn promoted the susceptibility of L. vannamei to V. alginolyticus.
Collapse
Affiliation(s)
- Ratchaneegorn Mapanao
- Faculty of Applied Science and Engineering, Khon Kaen University, Nong Khai Campus, Nong Khai 43000, Thailand
| | - Hsin-Wei Kuo
- Department of Aquaculture, National Pingtung University of Science and Technology, Pingtung 91201, Taiwan, ROC
| | - Chin-Chuan Chang
- Department of Aquaculture, National Pingtung University of Science and Technology, Pingtung 91201, Taiwan, ROC
| | - Kuan-Fu Liu
- Tungkang Biotechnology Research Center, Fisheries Research Institute, C.O.A., Pingtung 92845, Taiwan, ROC.
| | - Winton Cheng
- Department of Aquaculture, National Pingtung University of Science and Technology, Pingtung 91201, Taiwan, ROC.
| |
Collapse
|
15
|
Choi H, Koh SH. Understanding the role of glycogen synthase kinase-3 in L-DOPA-induced dyskinesia in Parkinson’s disease. Expert Opin Drug Metab Toxicol 2017; 14:83-90. [DOI: 10.1080/17425255.2018.1417387] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Hojin Choi
- Department of Neurology, Hanyang University College of Medicine, Seoul, South Korea
| | - Seong-Ho Koh
- Department of Neurology, Hanyang University College of Medicine, Seoul, South Korea
| |
Collapse
|
16
|
Yadav SK, Pandey S, Singh B. Role of estrogen and levodopa in 1-methyl-4-pheny-l-1, 2, 3, 6-tetrahydropyridine (mptp)-induced cognitive deficit in Parkinsonian ovariectomized mice model: A comparative study. J Chem Neuroanat 2017; 85:50-59. [DOI: 10.1016/j.jchemneu.2017.07.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Revised: 07/01/2017] [Accepted: 07/06/2017] [Indexed: 12/30/2022]
|
17
|
Jang EY, Yang CH, Hedges DM, Kim SP, Lee JY, Ekins TG, Garcia BT, Kim HY, Nelson AC, Kim NJ, Steffensen SC. The role of reactive oxygen species in methamphetamine self-administration and dopamine release in the nucleus accumbens. Addict Biol 2017; 22:1304-1315. [PMID: 27417190 DOI: 10.1111/adb.12419] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Revised: 05/10/2016] [Accepted: 05/13/2016] [Indexed: 12/30/2022]
Abstract
Methamphetamine (METH) markedly increases dopamine (DA) release in the mesolimbic DA system, which plays an important role in mediating the reinforcing effects of METH. METH-induced DA release results in the formation of reactive oxygen species (ROS), leading to oxidative damage. We have recently reported that ROS are implicated in behavior changes and DA release in the nucleus accumbens (NAc) following cocaine administration. The aim of this study was to evaluate the involvement of ROS in METH-induced locomotor activity, self-administration and enhancement of DA release in the NAc. Systemic administration of a non-specific ROS scavenger, N-tert-butyl-α-phenylnitrone (PBN; 0, 50 and 75 mg/kg, IP) or a superoxide-selective scavenger, 4-hydroxy-2,2,6,6-tetramethylpiperidine-1-oxyl (TEMPOL; 0, 50 and 100 mg/kg, IP), attenuated METH-induced locomotor activity without affecting generalized behavior in METH-naïve rats. PBN and TEMPOL significantly attenuated METH self-administration without affecting food intake. Increased oxidative stress was found in neurons, but not astrocytes, microglia or oligodendrocytes, in the NAc of METH self-administering rats. In addition, TEMPOL significantly decreased METH enhancement of DA release in the NAc. Taken together, these results suggest that enhancement of ROS in the NAc contributes to the reinforcing effect of METH.
Collapse
Affiliation(s)
- Eun Young Jang
- Department of Psychology and Center for Neuroscience; Brigham Young University; Provo UT USA
- College of Korean Medicine; Daegu Haany University; Daegu South Korea
| | - Chae Ha Yang
- College of Korean Medicine; Daegu Haany University; Daegu South Korea
| | - David M. Hedges
- Department of Psychology and Center for Neuroscience; Brigham Young University; Provo UT USA
| | - Soo Phil Kim
- College of Korean Medicine; Daegu Haany University; Daegu South Korea
| | - Jun Yeon Lee
- College of Korean Medicine; Daegu Haany University; Daegu South Korea
| | - Tyler G. Ekins
- Department of Psychology and Center for Neuroscience; Brigham Young University; Provo UT USA
| | - Brandon T. Garcia
- Department of Psychology and Center for Neuroscience; Brigham Young University; Provo UT USA
| | - Hee Young Kim
- College of Korean Medicine; Daegu Haany University; Daegu South Korea
| | - Ashley C. Nelson
- Department of Psychology and Center for Neuroscience; Brigham Young University; Provo UT USA
| | - Nam Jun Kim
- College of Korean Medicine; Daegu Haany University; Daegu South Korea
| | - Scott C. Steffensen
- Department of Psychology and Center for Neuroscience; Brigham Young University; Provo UT USA
| |
Collapse
|
18
|
Smith ES, Clark ME, Hardy GA, Kraan DJ, Biondo E, Gonzalez-Lima F, Cormack LK, Monfils M, Lee HJ. Daily consumption of methylene blue reduces attentional deficits and dopamine reduction in a 6-OHDA model of Parkinson's disease. Neuroscience 2017; 359:8-16. [PMID: 28694175 DOI: 10.1016/j.neuroscience.2017.07.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Revised: 06/01/2017] [Accepted: 07/02/2017] [Indexed: 01/05/2023]
Abstract
Recently, alternative drug therapies for Parkinson's disease (PD) have been investigated as there are many shortcomings of traditional dopamine-based therapies including difficulties in treating cognitive and attentional dysfunction. A promising therapeutic avenue is to target mitochondrial dysfunction and oxidative stress in PD. One option might be the use of methylene blue (MB), an antioxidant and metabolic enhancer. MB has been shown to improve cognitive function in both intact rodents and rodent disease models. Therefore, we investigated whether MB might treat attentional deficits in a rat model of PD induced by 6-hydroxydopamine (6-OHDA). MB also has neuroprotective capabilities against neurotoxic insult, so we also assessed the ability of MB to provide neuroprotection in our PD model. The results show that MB could preserve some dopamine neurons in the substantia nigra par compacta when 6-OHDA was infused into the medial forebrain bundle. This neuroprotection did not yield a significant behavioral improvement when motor functions were measured. However, MB significantly improved attentional performance in the five-choice task designed to measure selective and sustained attention. In conclusion, MB might be useful in improving some attentional function and preserving dopaminergic cells in this model. Future work should continue to study and optimize the abilities of MB for the treatment of PD.
Collapse
Affiliation(s)
- Elizabeth S Smith
- The University of Texas at Austin, Department of Psychology, United States
| | - Madeline E Clark
- The University of Texas at Austin, Department of Psychology, United States
| | - Gwendolyn A Hardy
- The University of Texas at Austin, Department of Psychology, United States
| | - David J Kraan
- The University of Texas at Austin, Department of Psychology, United States
| | - Elisa Biondo
- The University of Texas at Austin, Department of Psychology, United States
| | - F Gonzalez-Lima
- The University of Texas at Austin, Department of Psychology, United States
| | - Lawrence K Cormack
- The University of Texas at Austin, Department of Psychology, United States
| | - Marie Monfils
- The University of Texas at Austin, Department of Psychology, United States
| | - Hongjoo J Lee
- The University of Texas at Austin, Department of Psychology, United States.
| |
Collapse
|
19
|
Effects of asarinin on dopamine biosynthesis and 6-hydroxydopamine-induced cytotoxicity in PC12 cells. Arch Pharm Res 2017; 40:631-639. [PMID: 28397192 DOI: 10.1007/s12272-017-0908-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Accepted: 03/08/2017] [Indexed: 02/03/2023]
Abstract
This study investigated the effects of asarinin on dopamine biosynthesis and 6-hydroxydopamine (6-OHDA)-induced cytotoxicity in rat adrenal pheochromocytoma (PC12) cells. Treatment with asarinin (25-50 μM) increased intracellular dopamine levels and enhanced L-DOPA-induced increases in dopamine levels. Asarinin (25 μM) induced cyclic AMP-dependent protein kinase A (PKA) signaling, leading to increased cyclic AMP-response element binding protein (CREB) and tyrosine hydroxylase (TH) phosphorylation, which in turn stimulated dopamine production. Asarinin (25 μM) also activated transient phosphorylation of extracellular signal-regulated kinase (ERK1/2) and Bad phosphorylation at Ser 112, both of which have been shown to promote cell survival. In contrast, asarinin (25 μM) inhibited sustained ERK1/2, Bax, c-Jun N-terminal kinase (JNK1/2) and p38 mitogen-activated protein kinase (p38MAPK) phosphorylation and caspase-3 activity, which were induced by 6-OHDA (100 μM). These results suggest that asarinin induces dopamine biosynthesis via activation of the PKA-CREB-TH system and protects against 6-OHDA-induced cytotoxicity by inhibiting the sustained activation of the ERK-p38MAPK-JNK1/2-caspase-3 system in PC12 cells.
Collapse
|
20
|
Paul R, Borah A. L-DOPA-induced hyperhomocysteinemia in Parkinson's disease: Elephant in the room. Biochim Biophys Acta Gen Subj 2016; 1860:1989-97. [DOI: 10.1016/j.bbagen.2016.06.018] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Revised: 05/20/2016] [Accepted: 06/14/2016] [Indexed: 02/08/2023]
|
21
|
Hargreaves IP, Al Shahrani M, Wainwright L, Heales SJR. Drug-Induced Mitochondrial Toxicity. Drug Saf 2016; 39:661-74. [DOI: 10.1007/s40264-016-0417-x] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
22
|
L-DOPA modulates cell viability through the ERK-c-Jun system in PC12 and dopaminergic neuronal cells. Neuropharmacology 2016; 101:87-97. [DOI: 10.1016/j.neuropharm.2015.09.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Revised: 08/25/2015] [Accepted: 09/03/2015] [Indexed: 01/20/2023]
|
23
|
Ganie SA, Dar TA, Bhat AH, Dar KB, Anees S, Zargar MA, Masood A. Melatonin: A Potential Anti-Oxidant Therapeutic Agent for Mitochondrial Dysfunctions and Related Disorders. Rejuvenation Res 2015; 19:21-40. [PMID: 26087000 DOI: 10.1089/rej.2015.1704] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Mitochondria play a central role in cellular physiology. Besides their classic function of energy metabolism, mitochondria are involved in multiple cell functions, including energy distribution through the cell, energy/heat modulation, regulation of reactive oxygen species (ROS), calcium homeostasis, and control of apoptosis. Simultaneously, mitochondria are the main producer and target of ROS with the result that multiple mitochondrial diseases are related to ROS-induced mitochondrial injuries. Increased free radical generation, enhanced mitochondrial inducible nitric oxide synthase (iNOS) activity, enhanced nitric oxide (NO) production, decreased respiratory complex activity, impaired electron transport system, and opening of mitochondrial permeability transition pores have all been suggested as factors responsible for impaired mitochondrial function. Because of these, neurodegenerative diseases, such as Alzheimer's disease (AD), Parkinson's disease (PD), amyotrophic lateral sclerosis (ALS), Huntington's disease (HD), and aging, are caused by ROS-induced mitochondrial dysfunctions. Melatonin, the major hormone of the pineal gland, also acts as an anti-oxidant and as a regulator of mitochondrial bioenergetic function. Melatonin is selectively taken up by mitochondrial membranes, a function not shared by other anti-oxidants, and thus has emerged as a major potential therapeutic tool for treating neurodegenerative disorders. Multiple in vitro and in vivo experiments have shown the protective role of melatonin for preventing oxidative stress-induced mitochondrial dysfunction seen in experimental models of PD, AD, and HD. With these functions in mind, this article reviews the protective role of melatonin with mechanistic insights against mitochondrial diseases and suggests new avenues for safe and effective treatment modalities against these devastating neurodegenerative diseases. Future insights are also discussed.
Collapse
Affiliation(s)
- Showkat Ahmad Ganie
- 1 Department of Clinical Biochemistry, University of Kashmir Srinagar , India
| | - Tanveer Ali Dar
- 1 Department of Clinical Biochemistry, University of Kashmir Srinagar , India
| | - Aashiq Hussain Bhat
- 1 Department of Clinical Biochemistry, University of Kashmir Srinagar , India
| | - Khalid B Dar
- 1 Department of Clinical Biochemistry, University of Kashmir Srinagar , India
| | - Suhail Anees
- 1 Department of Clinical Biochemistry, University of Kashmir Srinagar , India
| | | | - Akbar Masood
- 2 Department of Biochemistry, University of Kashmir Srinagar , India
| |
Collapse
|
24
|
Tripathy D, Chakraborty J, Mohanakumar KP. Antagonistic pleiotropic effects of nitric oxide in the pathophysiology of Parkinson's disease. Free Radic Res 2015; 49:1129-39. [DOI: 10.3109/10715762.2015.1045505] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
25
|
Orenlili Yaylagul E, Cansev M, Celikler Kasimogullari S. In vivo protective effect of Uridine, a pyrimidine nucleoside, on genotoxicity induced by Levodopa/Carbidopa in mice. Food Chem Toxicol 2015; 82:36-41. [PMID: 25976300 DOI: 10.1016/j.fct.2015.05.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2014] [Revised: 03/16/2015] [Accepted: 05/05/2015] [Indexed: 11/27/2022]
Abstract
Parkinson's disease (PD) is a common neurodegenerative disorder that affects millions of people all over the world. Motor symptoms of PD are most commonly controlled by L-3,4-dihydroxyphenylalanine (Levodopa, L-DOPA), a precursor of dopamine, plus a peripherally-acting aromatic-L-amino-acid decarboxylase (dopa decarboxylase) inhibitor, such as carbidopa. However, chronic treatment with a combination of Levodopa plus carbidopa has been demonstrated to cause a major complication, namely abnormal involuntary movements. On the other hand, the effect of this treatment on bone marrow cells is unknown. Therefore, in this study, we aimed to investigate possible genotoxic effects of Levodopa and Carbidopa using male Balb/C mice. Our results showed that Levodopa alone or in combination with carbidopa caused genotoxicity in in vivo micronucleus test (mouse bone marrow) and Comet assay (blood cells). Furthermore, we showed that simultaneous administration of uridine, a pyrimidine nucleoside, reversed the genotoxic effect of Levodopa and Carbidopa in both assays. Our data show for the first time that Levodopa plus carbidopa combination causes genotoxicity which is reversed by uridine treatment. These findings might enhance our understanding for the complications of a common Parkinson's treatment and confer benefit in terms of reducing a possible genotoxic effect of this treatment.
Collapse
Affiliation(s)
- Esra Orenlili Yaylagul
- Department of Biology, Faculty of Arts and Sciences, Adnan Menderes University, Aydin, Turkey
| | - Mehmet Cansev
- Department of Pharmacology, Faculty of Medicine, Uludag University, Bursa, Turkey
| | | |
Collapse
|
26
|
Park HJ, Zhao TT, Lee KS, Lee SH, Shin KS, Park KH, Choi HS, Lee MK. Effects of (-)-sesamin on 6-hydroxydopamine-induced neurotoxicity in PC12 cells and dopaminergic neuronal cells of Parkinson's disease rat models. Neurochem Int 2015; 83-84:19-27. [DOI: 10.1016/j.neuint.2015.01.003] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Revised: 01/09/2015] [Accepted: 01/12/2015] [Indexed: 10/23/2022]
|
27
|
Masoudi N, Ibanez-Cruceyra P, Offenburger SL, Holmes A, Gartner A. Tetraspanin (TSP-17) protects dopaminergic neurons against 6-OHDA-induced neurodegeneration in C. elegans. PLoS Genet 2014; 10:e1004767. [PMID: 25474638 PMCID: PMC4256090 DOI: 10.1371/journal.pgen.1004767] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2014] [Accepted: 09/21/2014] [Indexed: 12/31/2022] Open
Abstract
Parkinson's disease (PD), the second most prevalent neurodegenerative disease after Alzheimer's disease, is linked to the gradual loss of dopaminergic neurons in the substantia nigra. Disease loci causing hereditary forms of PD are known, but most cases are attributable to a combination of genetic and environmental risk factors. Increased incidence of PD is associated with rural living and pesticide exposure, and dopaminergic neurodegeneration can be triggered by neurotoxins such as 6-hydroxydopamine (6-OHDA). In C. elegans, this drug is taken up by the presynaptic dopamine reuptake transporter (DAT-1) and causes selective death of the eight dopaminergic neurons of the adult hermaphrodite. Using a forward genetic approach to find genes that protect against 6-OHDA-mediated neurodegeneration, we identified tsp-17, which encodes a member of the tetraspanin family of membrane proteins. We show that TSP-17 is expressed in dopaminergic neurons and provide genetic, pharmacological and biochemical evidence that it inhibits DAT-1, thus leading to increased 6-OHDA uptake in tsp-17 loss-of-function mutants. TSP-17 also protects against toxicity conferred by excessive intracellular dopamine. We provide genetic and biochemical evidence that TSP-17 acts partly via the DOP-2 dopamine receptor to negatively regulate DAT-1. tsp-17 mutants also have subtle behavioral phenotypes, some of which are conferred by aberrant dopamine signaling. Incubating mutant worms in liquid medium leads to swimming-induced paralysis. In the L1 larval stage, this phenotype is linked to lethality and cannot be rescued by a dop-3 null mutant. In contrast, mild paralysis occurring in the L4 larval stage is suppressed by dop-3, suggesting defects in dopaminergic signaling. In summary, we show that TSP-17 protects against neurodegeneration and has a role in modulating behaviors linked to dopamine signaling. Parkinson's disease (PD) is characterized by the progressive loss of dopaminergic neurons. While hereditary forms are known, most cases are attributable to a combination of genetic and environmental risk factors. In PD models, dopaminergic neurodegeneration can be triggered by neurotoxins such as 6-hydroxydopamine (6-OHDA). This drug, which is taken up by the presynaptic dopamine reuptake transporter (DAT-1), also causes the selective death of C. elegans dopaminergic neurons. We found that TSP-17, a member of the tetraspanin family of membrane proteins, protects dopaminergic neurons from 6-OHDA-induced degeneration. We provide evidence that TSP-17 inhibits the C. elegans dopamine transporter DAT-1, leading to increased neuronal 6-OHDA uptake in tsp-17 mutants. TSP-17 also protects against toxicity conferred by excessive intracellular dopamine. TSP-17 interacts with the DOP-2 dopamine receptor, possibly as part of a pathway that negatively regulates DAT-1. tsp-17 mutants have subtle behavioral phenotypes that are partly conferred by aberrant dopamine signaling. In summary, we have used C. elegans genetics to model key aspects of PD.
Collapse
Affiliation(s)
- Neda Masoudi
- Centre for Gene Regulation and Expression, University of Dundee, Dow Street, Dundee, United Kingdom
| | - Pablo Ibanez-Cruceyra
- Centre for Gene Regulation and Expression, University of Dundee, Dow Street, Dundee, United Kingdom
| | - Sarah-Lena Offenburger
- Centre for Gene Regulation and Expression, University of Dundee, Dow Street, Dundee, United Kingdom
| | - Alexander Holmes
- Centre for Gene Regulation and Expression, University of Dundee, Dow Street, Dundee, United Kingdom
| | - Anton Gartner
- Centre for Gene Regulation and Expression, University of Dundee, Dow Street, Dundee, United Kingdom
- * E-mail:
| |
Collapse
|
28
|
The combination of oral L-DOPA/rimonabant for effective dyskinesia treatment and cytological preservation in a rat model of Parkinson's disease and L-DOPA-induced dyskinesia. Behav Pharmacol 2014; 24:640-52. [PMID: 24196024 DOI: 10.1097/fbp.0000000000000004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Parkinson's disease is the second most prevalent neurodegenerative disease in the world. Its treatment is limited so far to the management of parkinsonian symptoms with L-DOPA (LD). The long-term use of LD is limited by the development of L-DOPA-induced dyskinesias and dystonia. However, recent studies have suggested that pharmacological targeting of the endocannabinoid system may potentially provide a valuable therapeutic tool to suppress these motor alterations. In the present study, we have explored the behavioral (L-DOPA-induced dyskinesias severity) and cytological (substantia nigra compacta neurons and striatum neuropil preservation) effects of the oral coadministration of LD and rimonabant, a selective antagonist of CB1 receptors, in the 6-hydroxydopamine rat model of Parkinson's disease. Oral coadministration of LD (30 mg/kg) and rimonabant (1 mg/kg) significantly decreased abnormal involuntary movements and dystonia, possibly through the conservation of some functional tyrosine hydroxylase-immunoreactive dopaminergic cells, which in turn translates into a well-preserved neuropil of a less denervated striatum. Our results provide anatomical evidence that long-term coadministration of LD with cannabinoid antagonist-based therapy may not only alleviate specific motor symptoms but also delay/arrest the degeneration of striatal and substantia nigra compacta cells.
Collapse
|
29
|
Jang W, Park HH, Lee KY, Lee YJ, Kim HT, Koh SH. 1,25-dyhydroxyvitamin D3 Attenuates l-DOPA-Induced Neurotoxicity in Neural Stem Cells. Mol Neurobiol 2014; 51:558-70. [DOI: 10.1007/s12035-014-8835-1] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Accepted: 07/25/2014] [Indexed: 11/27/2022]
|
30
|
Liu X, Yamada N, Osawa T. Amide-type adduct of dopamine - plausible cause of Parkinson diseases. Subcell Biochem 2014; 77:49-60. [PMID: 24374917 DOI: 10.1007/978-94-007-7920-4_4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Dopamine is the endogenous neurotransmitter produced by nigral neurons. Dopamine loss can trigger not only prominent secondary morphological changes, but also changes in the density and sensitivity of dopamine receptors; therefore, it is a sign of PD development. The reasons for dopamine loss are attributed to dopamine's molecular instability due to it is a member of catecholamine family, whose catechol structure contributes to high oxidative stress through enzymatic and non-enzymatic oxidation. Oxidative stress in the brain easily leads to the lipid peroxidation reaction due to a high concentration of polyunsaturated fatty acids (PUFA), such as docosahexaenoic acid (DHA, C22:6/ω-3) and arachidonic acid (AA, C18:4/ω-6). Recent studies have shown that lipid hydroperoxides, the primary peroxidative products, could non-specifically react with primary amino groups to form N-acyl-type (amide-linkage) adducts. Therefore, based on the NH2-teminals in dopamine's structure, the aims of this chapter are to describes the possibility that reactive LOOH species derived from DHA/AA lipid peroxidation may modify dopamine to form amide-linkage dopamine adducts, which might be related to etiology of Parkinson's diseases.
Collapse
Affiliation(s)
- Xuebo Liu
- The Laboratory of Food and Biodynamics, Graduate School of Bioagricultural Science, Nagoya University, Nagoya, 464-8601, Japan,
| | | | | |
Collapse
|
31
|
Weinreb O, Mandel S, Youdim MBH, Amit T. Targeting dysregulation of brain iron homeostasis in Parkinson's disease by iron chelators. Free Radic Biol Med 2013; 62:52-64. [PMID: 23376471 DOI: 10.1016/j.freeradbiomed.2013.01.017] [Citation(s) in RCA: 152] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2012] [Revised: 01/09/2013] [Accepted: 01/14/2013] [Indexed: 10/27/2022]
Abstract
Brain iron accumulation has been implicated in a host of chronic neurological diseases, including Parkinson's disease (PD). The elevated iron levels observed in the substantia nigra of PD subjects have been suggested to incite the generation of reactive oxygen species and intracellular α-synuclein aggregation, terminating in the oxidative neuronal destruction of this brain area. Thus, elucidation of the molecular mechanisms involved in iron dysregulation and oxidative stress-induced neurodegeneration is a crucial step in deciphering PD pathology and in developing novel iron-complexing compounds aimed at restoring brain iron homeostasis and attenuating neurodegeneration. This review discusses the involvement of dysregulation of brain iron homeostasis in PD pathology, with an emphasis on the potential effectiveness of naturally occurring compounds and novel iron-chelating/antioxidant therapeutic hybrid molecules, exerting a spectrum of neuroprotective interrelated activities: antioxidant/monoamine oxidase inhibition, activation of the hypoxia-inducible factor (HIF)-1 signaling pathway, induction of HIF-1 target iron-regulatory and antioxidative genes, and inhibition of α-synuclein accumulation and aggregation.
Collapse
Affiliation(s)
- Orly Weinreb
- Eve Topf Centers of Excellence for Neurodegenerative Diseases Research, Department of Pharmacology, Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 31096, Israel.
| | - Silvia Mandel
- Eve Topf Centers of Excellence for Neurodegenerative Diseases Research, Department of Pharmacology, Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 31096, Israel
| | - Moussa B H Youdim
- Eve Topf Centers of Excellence for Neurodegenerative Diseases Research, Department of Pharmacology, Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 31096, Israel
| | - Tamar Amit
- Eve Topf Centers of Excellence for Neurodegenerative Diseases Research, Department of Pharmacology, Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 31096, Israel
| |
Collapse
|
32
|
HASHIMOTO K, KINPARA M, UDA Y. Inhibitory Effect of Heat-Treated 3-(3^|^prime;,4^|^prime;-dihydroxyphenyl)-L-alanine (DOPA) on ^|^beta;-glucuronidase Activity. FOOD SCIENCE AND TECHNOLOGY RESEARCH 2013. [DOI: 10.3136/fstr.19.1115] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
|
33
|
Abstract
L-dopa is a precursor for dopamine synthesis and a mainstay treatment for Parkinson's disease. However, l-dopa therapy is not without side effects that may be attributed to non-dopaminergic mechanisms. Synthesized dopamine can be neurotoxic through its enzymatic degradation by monoamine oxidase (MAO) to form the reactive byproduct, hydrogen peroxide and hydroxyl radicals or through auto-oxidation to form highly reactive quinones that can bind proteins and render them non-functional. Since l-dopa could be decarboxylated by aromatic amino acid decarboxylase (AADC) present within both dopamine and serotonin neurons, it was hypothesized that serotonin neurons convert l-dopa into dopamine to generate excessive reactive oxygen species and quinoproteins that ultimately lead to serotonin neuron death. To examine the effects of l-dopa on serotonin neurons, the RN46A-B14 cell line was used. These immortalized serotonergic cell cultures were terminally differentiated and then incubated with varying concentrations of l-dopa. Results show that RN46A-B14 cells contain AADC and can synthesize dopamine after incubation with l-dopa. Furthermore, l-dopa dose-dependently increased intracellular reactive oxygen species (ROS) and cell death. Dopamine, ROS production and cell death were attenuated by co-incubation with the AADC inhibitor, NSD-1015. The MAO inhibitor, pargyline, also attenuated cell death and ROS after l-dopa treatment. Lastly, quinoprotein formation was enhanced significantly by incubation with l-dopa. Taken together, these data illustrate that serotonergic cells can produce dopamine and that the accumulation of dopamine after l-dopa and its subsequent degradation can lead to ROS production and death of RN46A-B14 serotonergic cells.
Collapse
|
34
|
Borah A, Mohanakumar KP. L-DOPA induced-endogenous 6-hydroxydopamine is the cause of aggravated dopaminergic neurodegeneration in Parkinson’s disease patients. Med Hypotheses 2012; 79:271-3. [DOI: 10.1016/j.mehy.2012.05.008] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2012] [Revised: 04/10/2012] [Accepted: 05/07/2012] [Indexed: 12/21/2022]
|
35
|
Park KH, Park HJ, Shin KS, Choi HS, Kai M, Lee MK. Modulation of PC12 cell viability by forskolin-induced cyclic AMP levels through ERK and JNK pathways: an implication for L-DOPA-induced cytotoxicity in nigrostriatal dopamine neurons. Toxicol Sci 2012; 128:247-57. [PMID: 22539619 DOI: 10.1093/toxsci/kfs139] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The intracellular levels of cyclic AMP (cAMP) increase in response to cytotoxic concentrations of L-DOPA in PC12 cells, and forskolin that induces intracellular cAMP levels either protects PC12 cells from L-DOPA-induced cytotoxicity or enhances cytotoxicity in a concentration-dependent manner. This study investigated the effects of cAMP induced by forskolin on cell viability of PC12 cells, relevant to L-DOPA-induced cytotoxicity in Parkinson's disease therapy. The low levels of forskolin (0.01 and 0.1 μM)-induced cAMP increased dopamine biosynthesis and tyrosine hydroxylase (TH) phosphorylation, and induced transient phosphorylation of ERK1/2 within 1 h. However, at the high levels of forskolin (1.0 and 10 μM)-induced cAMP, dopamine biosynthesis and TH phosphorylation did not increase, but rapid differentiation in neurite-like formation was observed with a steady state. The high levels of forskolin-induced cAMP also induced sustained increase in ERK1/2 phosphorylation within 0.25-6 h and then led to apoptosis, which was apparently mediated by JNK1/2 and caspase-3 activation. Multiple treatment of PC12 cells with nontoxic L-DOPA (20 μM) for 4-6 days induced neurite-like formation and decreased intracellular dopamine levels by reducing TH phosphorylation. These results suggest that the low levels of forskolin-induced cAMP increased dopamine biosynthesis in cell survival via transient ERK1/2 phosphorylation. In contrast, the high levels of forskolin-induced cAMP induced differentiation via sustained ERK1/2 phosphorylation and then led to apoptosis. Taken together, the intracellular levels of cAMP play a dual role in cell survival and death through the ERK1/2 and JNK1/2 pathways in PC12 cells.
Collapse
Affiliation(s)
- Keun Hong Park
- College of Pharmacy and Research Center for Bioresource and Health, Chungbuk National University, Cheongju, Republic of Korea
| | | | | | | | | | | |
Collapse
|
36
|
Gutierrez-Valdez AL, Anaya-Martínez V, Ordoñez-Librado JL, García-Ruiz R, Torres-Esquivel C, Moreno-Rivera M, Sánchez-Betancourt J, Montiel-Flores E, Avila-Costa MR. Effect of chronic L-dopa or melatonin treatments after dopamine deafferentation in rats: dyskinesia, motor performance, and cytological analysis. ISRN NEUROLOGY 2012; 2012:360379. [PMID: 22462019 PMCID: PMC3302121 DOI: 10.5402/2012/360379] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/05/2011] [Accepted: 10/20/2011] [Indexed: 12/02/2022]
Abstract
The present study examines the ability of melatonin to protect striatal dopaminergic loss induced by 6-OHDA in a rat model of Parkinson's disease, comparing the results with L-DOPA-treated rats. The drugs were administered orally daily for a month, their therapeutic or dyskinetic effects were assessed by means of abnormal involuntary movements (AIMs) and stepping ability. At the cellular level, the response was evaluated using tyrosine hydroxylase immunoreactivity and striatal ultrastructural changes to compare between L-DOPA-induced AIMs and Melatonin-treated rats. Our findings demonstrated that chronic oral administration of Melatonin improved the alterations caused by the neurotoxin 6-OHDA. Melatonin-treated animals perform better in the motor tasks and had no dyskinetic alterations compared to L-DOPA-treated group. At the cellular level, we found that Melatonin-treated rats showed more TH-positive neurons and their striatal ultrastructure was well preserved. Thus, Melatonin is a useful treatment to delay the cellular and behavioral alterations observed in Parkinson's disease.
Collapse
Affiliation(s)
- Ana Luisa Gutierrez-Valdez
- Laboratorio de Neuromorfologia, Departamento de Neurociencias, Facultad de Estudios Superiores Iztacala, UNAM, Avenida de los Barrios 1, Los Reyes Iztacala, 54090 Tlalnepantla, MEX, Mexico
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Onzawa Y, Kimura Y, Uzuhashi K, Shirasuna M, Hirosawa T, Taogoshi T, Kihira K. Effects of 3- O-Methyldopa, L-3,4-Dihydroxyphenylalanine Metabolite, on Locomotor Activity and Dopamine Turnover in Rats. Biol Pharm Bull 2012; 35:1244-8. [DOI: 10.1248/bpb.b110714] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Yoritaka Onzawa
- Division of Clinical Pharmaceutical Sciences, Graduate School of Biomedical Sciences, Hiroshima University
| | - Yasuhiro Kimura
- Department of Pharmaceutical Services, Hiroshima University Hospital
- Division of Clinical Pharmacotherapeutics, Graduate School of Biomedical Sciences, Hiroshima University
| | - Kengo Uzuhashi
- Division of Clinical Pharmaceutical Sciences, Graduate School of Biomedical Sciences, Hiroshima University
| | - Megumi Shirasuna
- Division of Clinical Pharmaceutical Sciences, Graduate School of Biomedical Sciences, Hiroshima University
| | - Tasuku Hirosawa
- Division of Clinical Pharmaceutical Sciences, Graduate School of Biomedical Sciences, Hiroshima University
| | - Takanori Taogoshi
- Division of Clinical Pharmaceutical Sciences, Graduate School of Biomedical Sciences, Hiroshima University
- Department of Pharmaceutical Services, Hiroshima University Hospital
| | - Kenji Kihira
- Division of Clinical Pharmaceutical Sciences, Graduate School of Biomedical Sciences, Hiroshima University
- Department of Pharmaceutical Services, Hiroshima University Hospital
| |
Collapse
|
38
|
Srinivasan V, Cardinali DP, Srinivasan US, Kaur C, Brown GM, Spence DW, Hardeland R, Pandi-Perumal SR. Therapeutic potential of melatonin and its analogs in Parkinson's disease: focus on sleep and neuroprotection. Ther Adv Neurol Disord 2011; 4:297-317. [PMID: 22010042 DOI: 10.1177/1756285611406166] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Sleep disorders constitute major nonmotor features of Parkinson's disease (PD) that have a substantial effect on patients' quality of life and can be related to the progression of the neurodegenerative disease. They can also serve as preclinical markers for PD, as it is the case for rapid eye movement (REM)-associated sleep behavior disorder (RBD). Although the etiology of sleep disorders in PD remains undefined, the assessment of the components of the circadian system, including melatonin secretion, could give therapeutically valuable insight on their pathophysiopathology. Melatonin is a regulator of the sleep/wake cycle and also acts as an effective antioxidant and mitochondrial function protector. A reduction in the expression of melatonin MT(1) and MT(2) receptors has been documented in the substantia nigra of PD patients. The efficacy of melatonin for preventing neuronal cell death and for ameliorating PD symptoms has been demonstrated in animal models of PD employing neurotoxins. A small number of controlled trials indicate that melatonin is useful in treating disturbed sleep in PD, in particular RBD. Whether melatonin and the recently developed melatonergic agents (ramelteon, tasimelteon, agomelatine) have therapeutic potential in PD is also discussed.
Collapse
Affiliation(s)
- Venkatramanujam Srinivasan
- Sri Sathya Sai Medical Educational and Research Foundation, Prasanthi Nilayam, Plot-40, Kovai Thirunagar, Coimbatore 641014, India
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Lipski J, Nistico R, Berretta N, Guatteo E, Bernardi G, Mercuri NB. L-DOPA: a scapegoat for accelerated neurodegeneration in Parkinson's disease? Prog Neurobiol 2011; 94:389-407. [PMID: 21723913 DOI: 10.1016/j.pneurobio.2011.06.005] [Citation(s) in RCA: 84] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2011] [Revised: 06/17/2011] [Accepted: 06/17/2011] [Indexed: 12/21/2022]
Abstract
There is consensus that amelioration of the motor symptoms of Parkinson's disease is most effective with L-DOPA (levodopa). However, this necessary therapeutic step is biased by an enduring belief that L-DOPA is toxic to the remaining substantia nigra dopaminergic neurons by itself, or by specific metabolites such as dopamine. The concept of L-DOPA toxicity originated from pre-clinical studies conducted mainly in cell culture, demonstrating that L-DOPA or its derivatives damage dopaminergic neurons due to oxidative stress and other mechanisms. However, the in vitro data remain controversial as some studies showed neuroprotective, rather than toxic action of the drug. The relevance of this debate needs to be considered in the context of the studies conducted on animals and in clinical trials that do not provide convincing evidence for L-DOPA toxicity in vivo. This review presents the current views on the pathophysiology of Parkinson's disease, focusing on mitochondrial dysfunction and oxidative/proteolytic stress, the factors that can be affected by L-DOPA or its metabolites. We then critically discuss the evidence supporting the two opposing views on the effects of L-DOPA in vitro, as well as the animal and human data. We also address the problem of inadequate experimental models used in these studies. L-DOPA remains the symptomatic 'hero' of Parkinson's disease. Whether it contributes to degeneration of nigral dopaminergic neurons, or is a 'scapegoat' for explaining undesirable or unexpected effects of the treatment, remains a hotly debated topic.
Collapse
Affiliation(s)
- Janusz Lipski
- Department of Physiology and Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, 85 Park Rd., Auckland 1142, New Zealand.
| | | | | | | | | | | |
Collapse
|
40
|
Park KH, Choi NY, Koh SH, Park HH, Kim YS, Kim MJ, Lee SJ, Yu HJ, Lee KY, Lee YJ, Kim HT. L-DOPA neurotoxicity is prevented by neuroprotective effects of erythropoietin. Neurotoxicology 2011; 32:879-87. [PMID: 21683736 DOI: 10.1016/j.neuro.2011.05.009] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2011] [Revised: 04/21/2011] [Accepted: 05/19/2011] [Indexed: 01/27/2023]
Abstract
The neurotoxicity of L-3,4-dihydroxyphenylalanine (L-DOPA), one of the most important drugs for the treatment of Parkinson's disease, still remains controversial, although much more data on L-DOPA neurotoxicity have been presented. Considering the well known neuroprotective effects of erythropoietin (EPO), the inhibitory effects of EPO on L-DOPA neurotoxicity need to be evaluated. Neuronally differentiated PC12 (nPC12) cells were treated with different concentrations of L-DOPA and/or EPO for 24h. Cell viability was evaluated using trypan blue, 4',6-diamidino-2-phenylindole (DAPI) and TUNEL staining, and cell counting. Free radicals and intracellular signaling protein levels were measured with 2',7'-dichlorodihydrofluorescein diacetate (DCFH-DA) and Western blotting, respectively. L-DOPA reduced nPC12 cell viability at higher concentrations, but combined treatment with EPO and L-DOPA significantly restored cell viability. Free radicals and hydroxyl radical levels increased by L-DOPA were decreased after combined treatment of L-DOPA and EPO. Levels of survival-related intracellular signaling proteins decreased in nPC12 cells treated with 200 μM L-DOPA but increased significantly in cells treated with 200μM L-DOPA and 5 μM EPO. However, cleaved caspase-3, a death-related protein, increased in nPC12 cells treated with 200 μM L-DOPA but decreased significantly in cells treated with 200 μM L-DOPA and 5 μM EPO. Pretreatment with LY294002, a phosphatidylinositol 3-kinase inhibitor, prior to combined treatment with EPO and L-DOPA almost completely blocked the protective effects of EPO. These results indicate that EPO can prevent L-DOPA neurotoxicity by activating the PI3K pathway as well as reducing oxidative stress.
Collapse
Affiliation(s)
- Kee Hyung Park
- Department of Neurology, Gachon University Gil Hospital, Inchon, Republic of Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Shiba T, Yamato M, Kudo W, Watanabe T, Utsumi H, Yamada KI. In vivo imaging of mitochondrial function in methamphetamine-treated rats. Neuroimage 2011; 57:866-72. [PMID: 21624473 DOI: 10.1016/j.neuroimage.2011.05.041] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2011] [Revised: 05/10/2011] [Accepted: 05/14/2011] [Indexed: 10/18/2022] Open
Abstract
Abuse of the powerfully addictive psychostimulant, methamphetamine, occurs worldwide. Recent studies have suggested that methamphetamine-induced dopaminergic neurotoxicity is related to oxidative stress. In response to nerve activation, the mitochondrial respiratory chain is rapidly activated. The enhancement of mitochondrial respiratory chain activation may induce oxidative stress in the brain. However, there is little experimental evidence regarding the mitochondrial function after methamphetamine administration in vivo. Here, we evaluated whether a single administration of methamphetamine induces ATP consumption and overactivation of mitochondria. We measured mitochondrial function in two different ways: by monitoring oxygen partial pressure using an oxygen-selective electrode, and by imaging of redox reactions using a nitroxyl radical (i.e., nitroxide) coupled with Overhauser-enhanced magnetic resonance imaging (OMRI). A single administration of methamphetamine to Wistar rats induced dopaminergic nerve activation, ATP consumption and an increase in mitochondrial respiratory chain function in both the striatum and cortex. Furthermore, antioxidant TEMPOL prevented the increase in mitochondrial oxidative damage and methamphetamine-induced sensitization. These findings suggest that energy-supplying reactions after dopaminergic nerve activation are associated with oxidative stress in both the striatum and cortex, leading to abnormal behavior.
Collapse
Affiliation(s)
- Takeshi Shiba
- Department of REDOX Medicinal Science, Faculty of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | | | | | | | | | | |
Collapse
|
42
|
Protective effects of statins on l-DOPA neurotoxicity due to the activation of phosphatidylinositol 3-kinase and free radical scavenging in PC12 cell culture. Brain Res 2011; 1370:53-63. [DOI: 10.1016/j.brainres.2010.11.021] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2010] [Revised: 10/18/2010] [Accepted: 11/05/2010] [Indexed: 01/08/2023]
|
43
|
Kwon IH, Choi HS, Shin KS, Lee BK, Lee CK, Hwang BY, Lim SC, Lee MK. Effects of berberine on 6-hydroxydopamine-induced neurotoxicity in PC12 cells and a rat model of Parkinson's disease. Neurosci Lett 2010; 486:29-33. [PMID: 20851167 DOI: 10.1016/j.neulet.2010.09.038] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2010] [Revised: 09/03/2010] [Accepted: 09/14/2010] [Indexed: 10/19/2022]
Abstract
Protoberberine isoquinoline alkaloids including berberine inhibit dopamine biosynthesis and aggravate l-DOPA-induced cytotoxicity in PC12 cells. In this study, the effects of berberine on 6-hydroxydopamine (6-OHDA)-induced cytotoxicity in PC12 cells and on unilateral 6-OHDA-lesioned rats were investigated. In PC12 cells, berberine at 10 and 30μM associated with 6-OHDA (10, 20, and 50μM) enhanced cytotoxicity at 48h compared to 6-OHDA alone, indicated by an increase in apoptotic cell death. In addition, treatment with berberine (5 and 30mg/kg, i.p.) for 21 days in 6-OHDA-lesioned rats markedly depleted tyrosine hydroxylase-immunopositive cells in the substantia nigra as compared to berberine-untreated rats. Further, the levels of dopamine and norepinephrine were also significantly decreased by berberine administration (5 and 30mg/kg) in the striatal regions of 6-OHDA-lesioned rats. These results suggested that berberine aggravated 6-OHDA-induced cytotoxicity in PC12 cells, and led to the degeneration of dopaminergic neuronal cells in the substantia nigra of 6-OHDA-lesioned rats. It is, therefore, suggested that the use of long-term l-DOPA therapy with isoquinoline derivatives including berberine may need to be examined for the presence of adverse symptoms.
Collapse
Affiliation(s)
- Ik Hyun Kwon
- College of Pharmacy, Chungbuk National University, Heungduk-gu, Cheongju 361-763, Republic of Korea
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Weinreb O, Amit T, Mandel S, Kupershmidt L, Youdim MBH. Neuroprotective multifunctional iron chelators: from redox-sensitive process to novel therapeutic opportunities. Antioxid Redox Signal 2010; 13:919-49. [PMID: 20095867 DOI: 10.1089/ars.2009.2929] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Accumulating evidence suggests that many cytotoxic signals occurring in the neurodegenerative brain can initiate neuronal death processes, including oxidative stress, inflammation, and accumulation of iron at the sites of the neuronal deterioration. Neuroprotection by iron chelators has been widely recognized with respect to their ability to prevent hydroxyl radical formation in the Fenton reaction by sequestering redox-active iron. An additional neuroprotective mechanism of iron chelators is associated with their ability to upregulate or stabilize the transcriptional activator, hypoxia-inducible factor-1alpha (HIF-1alpha). HIF-1alpha stability within the cells is under the control of a class of iron-dependent and oxygen-sensor enzymes, HIF prolyl-4-hydroxylases (PHDs) that target HIF-1alpha for degradation. Thus, an emerging novel target for neuroprotection is associated with the HIF system to promote stabilization of HIF-1alpha and increase transcription of HIF-1-related survival genes, which have been reported to be regulated in patient's brains afflicted with diverse neurodegenerative diseases. In accordance, a new potential therapeutic strategy for neurodegenerative diseases is explored, by which iron chelators would inhibit PHDs, target the HIF-1-signaling pathway and ultimately activate HIF-1-dependent neuroprotective genes. This review discusses two interrelated approaches concerning therapy targets in neurodegeneration, sharing in common the implementation of iron chelation activity: antioxidation and HIF-1-pathway activation.
Collapse
Affiliation(s)
- Orly Weinreb
- Eve Topf Centers of Excellence for Neurodegenerative Diseases Research and Department of Pharmacology, Rappaport Family Research Institute, Technion-Faculty of Medicine, Haifa, Israel.
| | | | | | | | | |
Collapse
|
45
|
Reiter RJ, Manchester LC, Tan DX. Neurotoxins: free radical mechanisms and melatonin protection. Curr Neuropharmacol 2010; 8:194-210. [PMID: 21358970 PMCID: PMC3001213 DOI: 10.2174/157015910792246236] [Citation(s) in RCA: 127] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2010] [Revised: 05/21/2010] [Accepted: 05/30/2010] [Indexed: 12/15/2022] Open
Abstract
Toxins that pass through the blood-brain barrier put neurons and glia in peril. The damage inflicted is usually a consequence of the ability of these toxic agents to induce free radical generation within cells but especially at the level of the mitochondria. The elevated production of oxygen and nitrogen-based radicals and related non-radical products leads to the oxidation of essential macromolecules including lipids, proteins and DNA. The resultant damage is referred to as oxidative and nitrosative stress and, when the molecular destruction is sufficiently severe, it causes apoptosis or necrosis of neurons and glia. Loss of brain cells compromises the functions of the central nervous system expressed as motor, sensory and cognitive deficits and psychological alterations. In this survey we summarize the publications related to the following neurotoxins and the protective actions of melatonin: aminolevulinic acid, cyanide, domoic acid, kainic acid, metals, methamphetamine, polychlorinated biphenyls, rotenone, toluene and 6-hydroxydopamine. Given the potent direct free radical scavenging activities of melatonin and its metabolites, their ability to indirectly stimulate antioxidative enzymes and their efficacy in reducing electron leakage from mitochondria, it would be expected that these molecules would protect the brain from oxidative and nitrosative molecular mutilation. The studies summarized in this review indicate that this is indeed the case, an action that is obviously assisted by the fact that melatonin readily crosses the blood brain barrier.
Collapse
Affiliation(s)
- Russel J. Reiter
- Department of Cellular and Structural Biology, University of Texas Health Science Center, San Antonio, Texas
| | | | | |
Collapse
|
46
|
Borah A, Mohanakumar KP. Salicylic acid protects against chronic l-DOPA-induced 6-OHDA generation in experimental model of parkinsonism. Brain Res 2010; 1344:192-9. [DOI: 10.1016/j.brainres.2010.05.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2010] [Revised: 05/01/2010] [Accepted: 05/04/2010] [Indexed: 10/19/2022]
|
47
|
Choi HS, Park MS, Kim SH, Hwang BY, Lee CK, Lee MK. Neuroprotective effects of herbal ethanol extracts from Gynostemma pentaphyllum in the 6-hydroxydopamine-lesioned rat model of Parkinson's disease. Molecules 2010; 15:2814-24. [PMID: 20428081 PMCID: PMC6257318 DOI: 10.3390/molecules15042814] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2010] [Revised: 03/25/2010] [Accepted: 04/01/2010] [Indexed: 11/16/2022] Open
Abstract
6-Hydroxydopamine administration for 28 days (8 microg/2 microL) reduced the number of tyrosine hydroxylase (TH)-immunopositive neurons to 40.2% in the substantia nigra compared to the intact contralateral side. Dopamine, 3,4-dihydroxyphenylacetic acid, homovanillic acid and norepinephrine levels were reduced to 19.1%, 52.3%, 47.1% and 67.4% in the striatum of 6-hydroxydopamine-lesioned rats compared to the control group, respectively. However, an oral administration of herbal ethanol extracts from Gynostemma pentaphyllum (GP-EX) (10 mg/kg and 30 mg/kg) starting on day 3 post-lesion for 28 days markedly ameliorated the reduction of TH-immunopositive neurons induced by 6-hydroxydopamine-lesioned rat brain from 40.2% to 67.4% and 75.8% in the substantia nigra. GP-EX administration (10 and 30 mg/kg) also recovered the levels of dopamine, 3,4-dihydroxyphenylacetic acid, homovanillic acid and norepinephrine in post-lesion striatum to 64.1% and 65.0%, 77.9% and 89.7%, 82.6% and 90.2%, and 88.1% and 89.2% of the control group. GP-EX at the given doses did not produce any sign of toxicity such as weight loss, diarrhea and vomiting in rats during the 28 day treatment period and four gypenoside derivatives, gynosaponin TN-1, gynosaponin TN-2, gypenoside XLV and gypenoside LXXIV were identified from GP-EX. These results suggest that GP-EX might be helpful in the prevention of Parkinson's disease.
Collapse
Affiliation(s)
- Hyun Sook Choi
- College of Pharmacy and Research Center for Bioresource and Health, Chungbuk National University, Cheongju 361-763, Korea; E-Mails: (H.S.C); (M.S.P); (B.Y.H); (C.K.L)
| | - Mi Sook Park
- College of Pharmacy and Research Center for Bioresource and Health, Chungbuk National University, Cheongju 361-763, Korea; E-Mails: (H.S.C); (M.S.P); (B.Y.H); (C.K.L)
| | - Seung Hwan Kim
- College of Physical Education, Kyunghee University, Youngin 449-701, Korea; E-Mail: (S.H.K)
| | - Bang Yeon Hwang
- College of Pharmacy and Research Center for Bioresource and Health, Chungbuk National University, Cheongju 361-763, Korea; E-Mails: (H.S.C); (M.S.P); (B.Y.H); (C.K.L)
| | - Chong Kil Lee
- College of Pharmacy and Research Center for Bioresource and Health, Chungbuk National University, Cheongju 361-763, Korea; E-Mails: (H.S.C); (M.S.P); (B.Y.H); (C.K.L)
| | - Myung Koo Lee
- College of Pharmacy and Research Center for Bioresource and Health, Chungbuk National University, Cheongju 361-763, Korea; E-Mails: (H.S.C); (M.S.P); (B.Y.H); (C.K.L)
| |
Collapse
|
48
|
Borah A, Mohanakumar KP. l-DOPA-induced 6-hydroxydopamine production in the striata of rodents is sensitive to the degree of denervation. Neurochem Int 2010; 56:357-62. [DOI: 10.1016/j.neuint.2009.11.008] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2009] [Revised: 11/10/2009] [Accepted: 11/11/2009] [Indexed: 11/17/2022]
|
49
|
Park HH, Lee KY, Kim SH, Lee YJ, Koh SH. L-DOPA-induced neurotoxicity is reduced by the activation of the PI3K signaling pathway. Toxicology 2009; 265:80-6. [PMID: 19786063 DOI: 10.1016/j.tox.2009.09.011] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2009] [Revised: 09/17/2009] [Accepted: 09/18/2009] [Indexed: 11/30/2022]
Abstract
L-3,4-Dihydroxyphenylalanine (L-DOPA) is one of the most important drugs for the treatment of Parkinson's disease (PD). Although neurotoxicity of L-DOPA remains controversial, there are many reports suggesting that L-DOPA causes neuronal death. We investigated whether the neurotoxic effect of L-DOPA could be inhibited by the activation of the phosphatidylinositol 3-kinase (PI3K) pathway. Cell counting kit-8, trypan blue staining, and DAPI staining all showed that L-DOPA decreased nPC12 cell viability at high concentrations. However, combined treatment with the PI3K activator and L-DOPA significantly increased the viability of nPC12 cells when compared with treatment with L-DOPA only. Phosphorylated Akt (Ser473), phosphorylated glycogen synthase kinase-3beta (GSK-3beta) (Ser9), and heat shock transcription factor-1, which are survival-related signaling proteins, were decreased in nPC12 cells treated with 200 microM L-DOPA, but were significantly increased with combined treatment with the PI3K activator in a concentration-dependent manner. However, treatment of L-DOPA significantly increased expressions of cytosolic cytochrome c and cleaved caspase-3, which are death-related signaling proteins, in nPC12 cells, but combined treatment with the PI3K activator reduced those expressions. To confirm whether the effect of the PI3K activator is associated with direct activation of PI3K, LY294002, a PI3K inhibitor, was used to pretreat the nPC12 cells prior to combined treatment with the PI3K activator and L-DOPA. The protective effect of the PI3K activator was almost completely blocked. Together, these results suggest that L-DOPA neurotoxicity can be prevented by PI3K activation.
Collapse
Affiliation(s)
- Hyun-Hee Park
- Department of Neurology, Institute of Biomedical Science, Hanyang University College of Medicine, Seoul, Republic of Korea
| | | | | | | | | |
Collapse
|
50
|
Borah A, Mohanakumar KP. Melatonin inhibits 6-hydroxydopamine production in the brain to protect against experimental parkinsonism in rodents. J Pineal Res 2009; 47:293-300. [PMID: 19796048 DOI: 10.1111/j.1600-079x.2009.00713.x] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
We tested the hypothesis that melatonin regulates formation of 6-hydroxydopamine (6-OHDA) in the brain and thereby protects animals from dopaminergic neurotoxicity and the development of parkinsonism in animals. Employing a ferrous-ascorbate-dopamine (FAD) hydroxyl radical ((*)OH) generating system, in the present study we demonstrate a dose-dependent attenuation of 6-OHDA generation by melatonin in vitro. Intra-median forebrain bundle infusion of FAD caused significant depletion of striatal dopamine (DA), which was blocked by melatonin. Per-oral administration of l-3,4-dihydroxyphenylalanine (L-DOPA) for 7 days caused a dose-dependent increase in the formation of 6-OHDA in the mouse striatum, which was increased synergistically by the systemic administration of the parkinsonian neurotoxin, 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) on the 7th day of L-DOPA treatment. Melatonin treatment significantly attenuated both the L-DOPA and MPTP-induced increases in the levels of striatal 6-OHDA, and protected against striatal DA depletion caused by the neurotoxin. These observations suggest a novel mode of melatonin-induced dopaminergic neuroprotection in two models of Parkinson's disease, and suggest the possible therapeutic use of this well-known antioxidant indoleamine neurohormone in parkinsonism.
Collapse
Affiliation(s)
- Anupom Borah
- Division of Cell Biology and Physiology, Laboratory of Clinical & Experimental Neuroscience, Indian Institute of Chemical Biology, Kolkata, West Bengal, India
| | | |
Collapse
|