1
|
Montaño-Rodriguez AR, Schorling T, Andressoo JO. Striatal GDNF Neurons Chemoattract RET-Positive Dopamine Axons at Seven Times Farther Distance Than Medium Spiny Neurons. Cells 2024; 13:1059. [PMID: 38920687 PMCID: PMC11202212 DOI: 10.3390/cells13121059] [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: 05/16/2024] [Revised: 06/08/2024] [Accepted: 06/12/2024] [Indexed: 06/27/2024] Open
Abstract
Glial cell line-derived neurotrophic factor (GDNF) is among the strongest dopamine neuron function- and survival-promoting factors known. Due to this reason, it has clinical relevance in dopamine disorders such as Parkinson's disease and schizophrenia. In the striatum, GDNF is exclusively expressed in interneurons, which make up only about 0.6% of striatal cells. Despite clinical significance, histological analysis of striatal GDNF system arborization and relevance to incoming dopamine axons, which bear its receptor RET, has remained enigmatic. This is mainly due to the lack of antibodies able to visualize GDNF- and RET-positive cellular processes; here, we overcome this problem by using knock-in marker alleles. We find that GDNF neurons chemoattract RET+ axons at least seven times farther in distance than medium spiny neurons (MSNs), which make up 95% of striatal neurons. Furthermore, we provide evidence that tyrosine hydroxylase, the rate-limiting enzyme in dopamine synthesis, is enriched towards GDNF neurons in the dopamine axons. Finally, we find that GDNF neuron arborizations occupy approximately only twelve times less striatal volume than 135 times more abundant MSNs. Collectively, our results improve our understanding of how endogenous GDNF affects striatal dopamine system function.
Collapse
Affiliation(s)
- Ana Rosa Montaño-Rodriguez
- Department of Pharmacology, Faculty of Medicine, Helsinki Institute of Life Science, University of Helsinki, 00290 Helsinki, Finland; (A.R.M.-R.); (T.S.)
| | - Tabea Schorling
- Department of Pharmacology, Faculty of Medicine, Helsinki Institute of Life Science, University of Helsinki, 00290 Helsinki, Finland; (A.R.M.-R.); (T.S.)
| | - Jaan-Olle Andressoo
- Department of Pharmacology, Faculty of Medicine, Helsinki Institute of Life Science, University of Helsinki, 00290 Helsinki, Finland; (A.R.M.-R.); (T.S.)
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society (NVS), Karolinska Institutet, 17177 Stockholm, Sweden
| |
Collapse
|
2
|
Primak A, Bozov K, Rubina K, Dzhauari S, Neyfeld E, Illarionova M, Semina E, Sheleg D, Tkachuk V, Karagyaur M. Morphogenetic theory of mental and cognitive disorders: the role of neurotrophic and guidance molecules. Front Mol Neurosci 2024; 17:1361764. [PMID: 38646100 PMCID: PMC11027769 DOI: 10.3389/fnmol.2024.1361764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Accepted: 03/04/2024] [Indexed: 04/23/2024] Open
Abstract
Mental illness and cognitive disorders represent a serious problem for the modern society. Many studies indicate that mental disorders are polygenic and that impaired brain development may lay the ground for their manifestation. Neural tissue development is a complex and multistage process that involves a large number of distant and contact molecules. In this review, we have considered the key steps of brain morphogenesis, and the major molecule families involved in these process. The review provides many indications of the important contribution of the brain development process and correct functioning of certain genes to human mental health. To our knowledge, this comprehensive review is one of the first in this field. We suppose that this review may be useful to novice researchers and clinicians wishing to navigate the field.
Collapse
Affiliation(s)
- Alexandra Primak
- Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia
| | - Kirill Bozov
- Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia
| | - Kseniya Rubina
- Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia
| | - Stalik Dzhauari
- Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia
| | - Elena Neyfeld
- Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia
- Federal State Budgetary Educational Institution of the Higher Education “A.I. Yevdokimov Moscow State University of Medicine and Dentistry” of the Ministry of Healthcare of the Russian Federation, Moscow, Russia
| | - Maria Illarionova
- Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia
| | - Ekaterina Semina
- Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia
| | - Dmitriy Sheleg
- Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia
- Federal State Budgetary Educational Institution of the Higher Education “A.I. Yevdokimov Moscow State University of Medicine and Dentistry” of the Ministry of Healthcare of the Russian Federation, Moscow, Russia
| | - Vsevolod Tkachuk
- Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia
- Institute for Regenerative Medicine, Medical Research and Education Center, Lomonosov Moscow State University, Moscow, Russia
| | - Maxim Karagyaur
- Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia
- Institute for Regenerative Medicine, Medical Research and Education Center, Lomonosov Moscow State University, Moscow, Russia
| |
Collapse
|
3
|
Vincent B, Shukla M. The Common Denominators of Parkinson's Disease Pathogenesis and Methamphetamine Abuse. Curr Neuropharmacol 2024; 22:2113-2156. [PMID: 37691228 PMCID: PMC11337683 DOI: 10.2174/1570159x21666230907151226] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 07/24/2023] [Accepted: 07/25/2023] [Indexed: 09/12/2023] Open
Abstract
The pervasiveness and mortality associated with methamphetamine abuse have doubled during the past decade, suggesting a possible worldwide substance use crisis. Epitomizing the pathophysiology and toxicology of methamphetamine abuse proclaims severe signs and symptoms of neurotoxic and neurobehavioral manifestations in both humans and animals. Most importantly, chronic use of this drug enhances the probability of developing neurodegenerative diseases manifolds. Parkinson's disease is one such neurological disorder, which significantly and evidently not only shares a number of toxic pathogenic mechanisms induced by methamphetamine exposure but is also interlinked both structurally and genetically. Methamphetamine-induced neurodegeneration involves altered dopamine homeostasis that promotes the aggregation of α-synuclein protofibrils in the dopaminergic neurons and drives these neurons to make them more vulnerable to degeneration, as recognized in Parkinson's disease. Moreover, the pathologic mechanisms such as mitochondrial dysfunction, oxidative stress, neuroinflammation and decreased neurogenesis detected in methamphetamine abusers dramatically resemble to what is observed in Parkinson's disease cases. Therefore, the present review comprehensively cumulates a holistic illustration of various genetic and molecular mechanisms putting across the notion of how methamphetamine administration and intoxication might lead to Parkinson's disease-like pathology and Parkinsonism.
Collapse
Affiliation(s)
- Bruno Vincent
- Institute of Molecular and Cellular Pharmacology, Laboratory of Excellence DistALZ, Université Côte d'Azur, INSERM, CNRS, Sophia-Antipolis, 06560, Valbonne, France
| | - Mayuri Shukla
- Chulabhorn Graduate Institute, Chulabhorn Royal Academy, 10210, Bangkok, Thailand
| |
Collapse
|
4
|
Blackburn JK, Jamwal S, Wang W, Elsworth JD. Pioglitazone transiently stimulates paraoxonase-2 expression in male nonhuman primate brain: Implications for sex-specific therapeutics in neurodegenerative disorders. Neurochem Int 2022; 152:105222. [PMID: 34767873 PMCID: PMC8712400 DOI: 10.1016/j.neuint.2021.105222] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 10/17/2021] [Accepted: 11/07/2021] [Indexed: 01/03/2023]
Abstract
Paraoxonase-2 (PON2) enhances mitochondria function and protects against oxidative stress. Stimulating its expression has therapeutic potential for diseases where oxidative stress plays a significant role in the pathology, such as Parkinson's disease. Clinical and preclinical evidence suggest that the anti-diabetic drug pioglitazone may provide neuroprotection in Parkinson's disease, Alzheimer's disease, and stroke, but the biochemical pathway(s) responsible has not been fully elucidated. To determine the effect of pioglitazone on PON2 expression we treated male African green monkeys with oral pioglitazone (5 mg/kg/day) for 1 and 3 weeks. We found that pioglitazone increased PON2 mRNA and protein expression in brain following 1 week of treatment, however, by 3 weeks of treatment PON2 expression had returned to baseline. This transient increase was detected in substantia nigra, striatum, hippocampus, and dorsolateral prefrontal cortex The short-term impact of pioglitazone on PON2 expression in striatum may contribute to the discrepancy in the potency of the drug between short-term animal models and clinical trials for Parkinson's disease. Both PON2 and pioglitazone's receptor, peroxisome proliferator-activated receptor gamma (PPARγ), possess sex- and brain region-dependent expression, which may play a role in the short-term effect of pioglitazone and provide clues to extending the beneficial effects of PON2 activation.
Collapse
Affiliation(s)
- Jennifer K. Blackburn
- Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut, 06511, USA
| | - Sumit Jamwal
- Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut, 06511, USA
| | - Weiwei Wang
- MS & Proteomics Resource of WM Keck Foundation Biotechnology Resource Laboratory, Yale University, New Haven, CT
| | - John D. Elsworth
- Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut, 06511, USA., Corresponding author for post-publication: John D Elsworth, Ph.D., , Yale University School of Medicine, Department of Psychiatry, 300 George Street, Suite 901, New Haven, CT 06511, Tel: 203-785-6768
| |
Collapse
|
5
|
Elevated endogenous GDNF induces altered dopamine signalling in mice and correlates with clinical severity in schizophrenia. Mol Psychiatry 2022; 27:3247-3261. [PMID: 35618883 PMCID: PMC9708553 DOI: 10.1038/s41380-022-01554-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 03/24/2022] [Accepted: 03/25/2022] [Indexed: 11/08/2022]
Abstract
Presynaptic increase in striatal dopamine is the primary dopaminergic abnormality in schizophrenia, but the underlying mechanisms are not understood. Here, we hypothesized that increased expression of endogenous GDNF could induce dopaminergic abnormalities that resemble those seen in schizophrenia. To test the impact of GDNF elevation, without inducing adverse effects caused by ectopic overexpression, we developed a novel in vivo approach to conditionally increase endogenous GDNF expression. We found that a 2-3-fold increase in endogenous GDNF in the brain was sufficient to induce molecular, cellular, and functional changes in dopamine signalling in the striatum and prefrontal cortex, including increased striatal presynaptic dopamine levels and reduction of dopamine in prefrontal cortex. Mechanistically, we identified adenosine A2a receptor (A2AR), a G-protein coupled receptor that modulates dopaminergic signalling, as a possible mediator of GDNF-driven dopaminergic abnormalities. We further showed that pharmacological inhibition of A2AR with istradefylline partially normalised striatal GDNF and striatal and cortical dopamine levels in mice. Lastly, we found that GDNF levels are increased in the cerebrospinal fluid of first episode psychosis patients, and in post-mortem striatum of schizophrenia patients. Our results reveal a possible contributor for increased striatal dopamine signalling in a subgroup of schizophrenia patients and suggest that GDNF-A2AR crosstalk may regulate dopamine function in a therapeutically targetable manner.
Collapse
|
6
|
Limanaqi F, Busceti CL, Celli R, Biagioni F, Fornai F. Autophagy as a gateway for the effects of methamphetamine: From neurotransmitter release and synaptic plasticity to psychiatric and neurodegenerative disorders. Prog Neurobiol 2021; 204:102112. [PMID: 34171442 DOI: 10.1016/j.pneurobio.2021.102112] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 02/27/2021] [Accepted: 06/18/2021] [Indexed: 02/07/2023]
Abstract
As a major eukaryotic cell clearing machinery, autophagy grants cell proteostasis, which is key for neurotransmitter release, synaptic plasticity, and neuronal survival. In line with this, besides neuropathological events, autophagy dysfunctions are bound to synaptic alterations that occur in mental disorders, and early on, in neurodegenerative diseases. This is also the case of methamphetamine (METH) abuse, which leads to psychiatric disturbances and neurotoxicity. While consistently altering the autophagy machinery, METH produces behavioral and neurotoxic effects through molecular and biochemical events that can be recapitulated by autophagy blockade. These consist of altered physiological dopamine (DA) release, abnormal stimulation of DA and glutamate receptors, as well as oxidative, excitotoxic, and neuroinflammatory events. Recent molecular insights suggest that METH early impairs the autophagy machinery, though its functional significance remains to be investigated. Here we discuss evidence suggesting that alterations of DA transmission and autophagy are intermingled within a chain of events underlying behavioral alterations and neurodegenerative phenomena produced by METH. Understanding how METH alters the autophagy machinery is expected to provide novel insights into the neurobiology of METH addiction sharing some features with psychiatric disorders and parkinsonism.
Collapse
Affiliation(s)
- Fiona Limanaqi
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Roma, 55, 56126, Pisa, PI, Italy
| | | | - Roberta Celli
- IRCCS Neuromed, Via Atinense 18, 86077 Pozzilli, IS, Italy
| | | | - Francesco Fornai
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Roma, 55, 56126, Pisa, PI, Italy; IRCCS Neuromed, Via Atinense 18, 86077 Pozzilli, IS, Italy.
| |
Collapse
|
7
|
Pingale T, Gupta GL. Classic and evolving animal models in Parkinson's disease. Pharmacol Biochem Behav 2020; 199:173060. [PMID: 33091373 DOI: 10.1016/j.pbb.2020.173060] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 10/10/2020] [Accepted: 10/12/2020] [Indexed: 02/07/2023]
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disease with motor and non-motor symptoms. PD is characterized by the degeneration of dopaminergic neurons in the substantia nigra pars compacta (SNpc) and deficiency of dopamine in the striatal region. The primary objective in PD research is to understand the pathogenesis, targets, and development of therapeutic interventions to control the progress of the disease. The anatomical and physiological resemblances between humans and animals gathered the researcher's attention towards the use of animals in PD research. Due to varying age of onset, symptoms, and progression rate, PD becomes heterogeneous which demands the variety of animal models to study diverse features of the disease. Parkinson is a multifactorial disorder, selection of models become important as not a single model shows all the biochemical features of the disease. Currently, conventional pharmacological, neurotoxin-induced, genetically modified and cellular models are available for PD research, but none of them recapitulate all the biochemical characteristics of the disease. In this review, we included the updated knowledge on the main features of currently available in vivo and in vitro models as well as their strengths and weaknesses.
Collapse
Affiliation(s)
- Tanvi Pingale
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM'S NMIMS, V.L. Mehta Road, Vile Parle (W), Mumbai 400 056, India
| | - Girdhari Lal Gupta
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM'S NMIMS, V.L. Mehta Road, Vile Parle (W), Mumbai 400 056, India; School of Pharmacy & Technology Management, SVKM'S NMIMS, Shirpur, Maharashtra, India.
| |
Collapse
|
8
|
Ferreira C, Almeida C, Tenreiro S, Quintas A. Neuroprotection or Neurotoxicity of Illicit Drugs on Parkinson's Disease. Life (Basel) 2020; 10:life10060086. [PMID: 32545328 PMCID: PMC7344445 DOI: 10.3390/life10060086] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Revised: 06/08/2020] [Accepted: 06/09/2020] [Indexed: 12/20/2022] Open
Abstract
Parkinson's Disease (PD) is currently the most rapid growing neurodegenerative disease and over the past generation, its global burden has more than doubled. The onset of PD can arise due to environmental, sporadic or genetic factors. Nevertheless, most PD cases have an unknown etiology. Chemicals, such as the anthropogenic pollutant 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) and amphetamine-type stimulants, have been associated with the onset of PD. Conversely, cannabinoids have been associated with the treatment of the symptoms'. PD and medical cannabis is currently under the spotlight, and research to find its benefits on PD is on-going worldwide. However, the described clinical applications and safety of pharmacotherapy with cannabis products are yet to be fully supported by scientific evidence. Furthermore, the novel psychoactive substances are currently a popular alternative to classical drugs of abuse, representing an unknown health hazard for young adults who may develop PD later in their lifetime. This review addresses the neurotoxic and neuroprotective impact of illicit substance consumption in PD, presenting clinical evidence and molecular and cellular mechanisms of this association. This research area is utterly important for contemporary society since illicit drugs' legalization is under discussion which may have consequences both for the onset of PD and for the treatment of its symptoms.
Collapse
Affiliation(s)
- Carla Ferreira
- Molecular Pathology and Forensic Biochemistry Laboratory, Centro de Investigação Interdisciplinar Egas Moniz, P-2825-084 Caparica, Portugal; (C.F.); (C.A.)
- Laboratório de Ciências Forenses e Psicológicas Egas Moniz, Campus Universitário–Quinta da Granja, Monte de Caparica, P-2825-084 Caparica, Portugal
- Faculty of Medicine of Porto University, Al. Prof. Hernâni Monteiro, P-4200–319 Porto, Portugal
| | - Catarina Almeida
- Molecular Pathology and Forensic Biochemistry Laboratory, Centro de Investigação Interdisciplinar Egas Moniz, P-2825-084 Caparica, Portugal; (C.F.); (C.A.)
| | - Sandra Tenreiro
- CEDOC–Chronic Diseases Research Center, Faculdade de Ciências Médicas, Universidade Nova de Lisboa, P-1150-082 Lisboa, Portugal;
| | - Alexandre Quintas
- Molecular Pathology and Forensic Biochemistry Laboratory, Centro de Investigação Interdisciplinar Egas Moniz, P-2825-084 Caparica, Portugal; (C.F.); (C.A.)
- Laboratório de Ciências Forenses e Psicológicas Egas Moniz, Campus Universitário–Quinta da Granja, Monte de Caparica, P-2825-084 Caparica, Portugal
- Correspondence:
| |
Collapse
|
9
|
Parkinson's disease treatment: past, present, and future. J Neural Transm (Vienna) 2020; 127:785-791. [PMID: 32172471 DOI: 10.1007/s00702-020-02167-1] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Accepted: 03/03/2020] [Indexed: 12/15/2022]
Abstract
The substantial contributions of Dr. Gerald Stern to past and current treatments for Parkinson's disease patients are reviewed, which form the foundation for an evaluation of future options to control symptoms and halt progression of the disease. These opportunities will depend on a greater understanding of the relative contributions of the environment, genetic and epigenetic influences to disease onset, and promise to emerge as strategies for improving mitochondrial function, halting accumulation of synuclein and neuromelanin, in addition to refinement of stem cell and gene therapies. Such advances will be achieved through deployment of improved models for the disease.
Collapse
|
10
|
Valian N, Heravi M, Ahmadiani A, Dargahi L. Effect of methamphetamine on rat primary midbrain cells; mitochondrial biogenesis as a compensatory response. Neuroscience 2019; 406:278-289. [DOI: 10.1016/j.neuroscience.2019.03.016] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2018] [Revised: 02/25/2019] [Accepted: 03/07/2019] [Indexed: 01/07/2023]
|
11
|
Ball N, Teo WP, Chandra S, Chapman J. Parkinson's Disease and the Environment. Front Neurol 2019; 10:218. [PMID: 30941085 PMCID: PMC6433887 DOI: 10.3389/fneur.2019.00218] [Citation(s) in RCA: 227] [Impact Index Per Article: 45.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Accepted: 02/20/2019] [Indexed: 12/13/2022] Open
Abstract
Parkinson's disease (PD) is a heterogeneous neurodegenerative disorder that affects an estimated 10 million sufferers worldwide. The two forms of PD include familial and sporadic, and while the etiology of PD is still largely unknown, the condition is likely to be multifactorial with genetic and environmental factors contributing to disease genesis. Diagnosis of the condition is attained through the observation of cardinal clinical manifestations including resting tremor, muscle rigidity, slowness or loss of movement, and postural instability. Unfortunately, by the time these features become apparent extensive neurological damage has already occurred. A cure for PD has not been identified and the current therapy options are pharmaceutical- and/or surgical-based interventions to treat condition symptoms. There is no specific test for PD and most diagnoses are confirmed by a combination of clinical symptoms and positive responses to dopaminergic drug therapies. The prevalence and incidence of PD vary worldwide influenced by several factors such as age, gender, ethnicity, genetic susceptibilities, and environmental exposures. Here, we will present environmental factors implicated in sporadic PD onset. By understanding the mechanisms in which environmental factors interact with, and affect the brain we can stride toward finding the underlying cause(s) of PD.
Collapse
Affiliation(s)
- Nicole Ball
- School of Health, Medical and Applied Sciences, Central Queensland University, Rockhampton, QLD, Australia
| | - Wei-Peng Teo
- School of Exercise and Nutrition Sciences, Institute for Physical Activity and Nutrition, Deakin University, Melbourne, VIC, Australia.,Physical Education and Sports Science Academic Group, National Institute of Education, Nanyang Technological University, Singapore, Singapore
| | - Shaneel Chandra
- School of Health, Medical and Applied Sciences, Central Queensland University, Rockhampton, QLD, Australia
| | - James Chapman
- School of Science, RMIT University, Melbourne, VIC, Australia
| |
Collapse
|
12
|
Ramesh J, Ronsard L, Gao A, Venugopal B. Autophagy Intertwines with Different Diseases-Recent Strategies for Therapeutic Approaches. Diseases 2019; 7:diseases7010015. [PMID: 30717078 PMCID: PMC6473623 DOI: 10.3390/diseases7010015] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 01/22/2019] [Accepted: 01/28/2019] [Indexed: 12/12/2022] Open
Abstract
Autophagy is a regular and substantial “clear-out process” that occurs within the cell and that gets rid of debris that accumulates in membrane-enclosed vacuoles by using enzyme-rich lysosomes, which are filled with acids that degrade the contents of the vacuoles. This machinery is well-connected with many prevalent diseases, including cancer, HIV, and Parkinson’s disease. Considering that autophagy is well-known for its significant connections with a number of well-known fatal diseases, a thorough knowledge of the current findings in the field is essential in developing therapies to control the progression rate of diseases. Thus, this review summarizes the critical events comprising autophagy in the cellular system and the significance of its key molecules in manifesting this pathway in various diseases for down- or upregulation. We collectively reviewed the role of autophagy in various diseases, mainly neurodegenerative diseases, cancer, inflammatory diseases, and renal disorders. Here, some collective reports on autophagy showed that this process might serve as a dual performer: either protector or contributor to certain diseases. The aim of this review is to help researchers to understand the role of autophagy-regulating genes encoding functional open reading frames (ORFs) and its connection with diseases, which will eventually drive better understanding of both the progression and suppression of different diseases at various stages. This review also focuses on certain novel therapeutic strategies which have been published in the recent years based on targeting autophagy key proteins and its interconnecting signaling cascades.
Collapse
Affiliation(s)
- Janani Ramesh
- Department of Medical Biochemistry, Dr. A.L.M. Post Graduate Institute of Basic Medical Sciences, University of Madras, Chennai 600113, India.
- Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| | - Larance Ronsard
- The Ragon Institute of Massachusetts General Hospital, The Massachusetts Institute of Technology and Harvard University, 400 Technology Square, Cambridge, MA 02140, USA.
| | - Anthony Gao
- Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| | - Bhuvarahamurthy Venugopal
- Department of Medical Biochemistry, Dr. A.L.M. Post Graduate Institute of Basic Medical Sciences, University of Madras, Chennai 600113, India.
| |
Collapse
|
13
|
Zhu LN, Qiao HH, Chen L, Sun LP, Hui JL, Lian YL, Xie WB, Ding JY, Meng YL, Zhu BF, Qiu PM. SUMOylation of Alpha-Synuclein Influences on Alpha-Synuclein Aggregation Induced by Methamphetamine. Front Cell Neurosci 2018; 12:262. [PMID: 30197588 PMCID: PMC6117395 DOI: 10.3389/fncel.2018.00262] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2018] [Accepted: 07/31/2018] [Indexed: 12/13/2022] Open
Abstract
Methamphetamine (METH) is an illegal and widely abused psychoactive stimulant. METH abusers are at high risk of neurodegenerative disorders, including Parkinson’s disease (PD). Previous studies have demonstrated that METH causes alpha-synuclein (α-syn) aggregation in the both laboratory animal and human. In this study, exposure to high METH doses increased the expression of α-syn and the small ubiquitin-related modifier 1 (SUMO-1). Therefore, we hypothesized that SUMOylation of α-syn is involved in high-dose METH-induced α-syn aggregation. We measured the levels of α-syn SUMOylation and these enzymes involved in the SUMOylation cycle in SH-SY5Y human neuroblastoma cells (SH-SY5Y cells), in cultures of C57 BL/6 primary mouse neurons and in brain tissues of mice exposure to METH. We also demonstrated the effect of α-syn SUMOylation on α-syn aggregation after METH exposure by overexpressing the key enzyme of the SUMOylation cycle or silencing SUMO-1 expression in vitro. Then, we make introduced mutations in the major SUMOylation acceptor sites of α-syn by transfecting a lentivirus containing the sequence of WT α-syn or K96/102R α-syn into SH-SY5Y cells and injecting an adenovirus containing the sequence of WT α-syn or K96/102R α-syn into the mouse striatum. Levels of the ubiquitin-proteasome system (UPS)-related makers ubiquitin (Ub) and UbE1, as well as the autophagy-lysosome pathway (ALP)-related markers LC3, P62 and lysosomal associated membrane protein 2A (LAMP2A), were also measured in SH-SY5Y cells transfected with lentivirus and mice injected with adenovirus. The results showed that METH exposure decreases the SUMOylation level of α-syn, although the expression of α-syn and SUMO-1 are increased. One possible cause is the reduction of UBC9 level. The increase in α-syn SUMOylation by UBC9 overexpression relieves METH-induced α-syn overexpression and aggregation, whereas the decrease in α-syn SUMOylation by SUMO-1 silencing exacerbates the same pathology. Furthermore, mutations in the major SUMOylation acceptor sites of α-syn also aggravate α-syn overexpression and aggregation by impairing degradation through the UPS and the ALP in vitro and in vivo. These results suggest that SUMOylation of α-syn plays a fundamental part in α-syn overexpression and aggregation induced by METH and could be a suitable target for the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Lin-Nan Zhu
- School of Forensic Medicine, Southern Medical University, Guangzhou, China
| | - Hong-Hua Qiao
- School of Forensic Medicine, Southern Medical University, Guangzhou, China
| | - Ling Chen
- School of Forensic Medicine, Southern Medical University, Guangzhou, China
| | - Le-Ping Sun
- School of Forensic Medicine, Southern Medical University, Guangzhou, China
| | - Jia-Liang Hui
- First Clinical Medical College, Southern Medical University, Guangzhou, China
| | - Yong-Ling Lian
- School of Forensic Medicine, Southern Medical University, Guangzhou, China
| | - Wei-Bing Xie
- School of Forensic Medicine, Southern Medical University, Guangzhou, China
| | - Jiu-Yang Ding
- School of Forensic Medicine, Southern Medical University, Guangzhou, China.,Department of Anatomy, Zunyi Medical College, Zunyi, China
| | - Yun-le Meng
- School of Forensic Medicine, Southern Medical University, Guangzhou, China
| | - Bo-Feng Zhu
- School of Forensic Medicine, Southern Medical University, Guangzhou, China
| | - Ping-Ming Qiu
- School of Forensic Medicine, Southern Medical University, Guangzhou, China
| |
Collapse
|
14
|
Epigenetic Effects Induced by Methamphetamine and Methamphetamine-Dependent Oxidative Stress. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:4982453. [PMID: 30140365 PMCID: PMC6081569 DOI: 10.1155/2018/4982453] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Accepted: 06/10/2018] [Indexed: 12/21/2022]
Abstract
Methamphetamine is a widely abused drug, which possesses neurotoxic activity and powerful addictive effects. Understanding methamphetamine toxicity is key beyond the field of drug abuse since it allows getting an insight into the molecular mechanisms which operate in a variety of neuropsychiatric disorders. In fact, key alterations produced by methamphetamine involve dopamine neurotransmission in a way, which is reminiscent of spontaneous neurodegeneration and psychiatric schizophrenia. Thus, understanding the molecular mechanisms operated by methamphetamine represents a wide window to understand both the addicted brain and a variety of neuropsychiatric disorders. This overlapping, which is already present when looking at the molecular and cellular events promoted immediately after methamphetamine intake, becomes impressive when plastic changes induced in the brain of methamphetamine-addicted patients are considered. Thus, the present manuscript is an attempt to encompass all the molecular events starting at the presynaptic dopamine terminals to reach the nucleus of postsynaptic neurons to explain how specific neurotransmitters and signaling cascades produce persistent genetic modifications, which shift neuronal phenotype and induce behavioral alterations. A special emphasis is posed on disclosing those early and delayed molecular events, which translate an altered neurotransmitter function into epigenetic events, which are derived from the translation of postsynaptic noncanonical signaling into altered gene regulation. All epigenetic effects are considered in light of their persistent changes induced in the postsynaptic neurons including sensitization and desensitization, priming, and shift of neuronal phenotype.
Collapse
|
15
|
Valian N, Ahmadiani A, Dargahi L. Increasing methamphetamine doses inhibit glycogen synthase kinase 3β activity by stimulating the insulin signaling pathway in substantia nigra. J Cell Biochem 2018; 119:8522-8530. [PMID: 30011098 DOI: 10.1002/jcb.27082] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Accepted: 04/26/2018] [Indexed: 01/01/2023]
Abstract
Methamphetamine (MA), a highly abused psychostimulant, exerts neurotoxic effects on the dopaminergic system via several neurotoxicity mechanisms in the long-term administration. Since the effect of MA on the signaling insulin pathway is less studied, the current study was designed to evaluate the effect of escalating an MA regimen on different insulin signaling elements in substantia nigra (SN) and striatum of a rat. Increasing MA doses (1-14 mg/kg) were administrated intraperitoneally twice a day for 14 days in rats. In the control group, normal saline was injected in the same volume. On days 1, 14, 28, and 60 after MA discontinuation, molecular assessments were performed. Insulin receptor (IR) and insulin receptor substrate (IRS) 1 and 2 gene expression were evaluated using real-time polymerase chain reaction, and protein levels of phosphatidylinositol-4,5-bisphosphate 3-kinase (PI3K), phospho-PI3K, Akt, phospho-Akt, glycogen synthase kinase 3β (GSK3β), and phospho-GSK3β were measured by the Western blot analysis in SN and striatum. Messenger RNA levels of IR and insulin receptor substrate 2 were increased in SN, 1 day after the last injection. Although no changes were observed in PI3K, phospho-PI3K, Akt, phospho-Akt, and GSK3β levels, increase in the level of inactive form of GSK3β (phosphorylated on serine 9) was indicated in SN on day 28. In striatum, decreases in IR and phospho-Akt were demonstrated, without any change in other elements. Repeated escalating regimen of MA activated the insulin signaling pathway and inhibited GSK3β activity in SN. This response, which did not occur in striatum, may act as an adaptive mechanism to prevent MA-induced neurotoxicity in dopaminergic cell bodies.
Collapse
Affiliation(s)
- Neda Valian
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Abolhassan Ahmadiani
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Leila Dargahi
- Neurobiology Research Center, Shahid Beheshti University ofMedical Sciences, Tehran, Iran
| |
Collapse
|
16
|
Li L, Chen S, Wang Y, Yue X, Xu J, Xie W, Qiu P, Liu C, Wang A, Wang H. Role of GSK3β/α-synuclein axis in methamphetamine-induced neurotoxicity in PC12 cells. Toxicol Res (Camb) 2018; 7:221-234. [PMID: 30090577 PMCID: PMC6062219 DOI: 10.1039/c7tx00189d] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2017] [Accepted: 12/22/2017] [Indexed: 12/22/2022] Open
Abstract
Methamphetamine (METH) is well-known as a potent psychostimulant of abuse worldwide. METH administration can cause neurotoxicity and neurodegenerative injury, which are similar to the two prevalent neurodegenerative disorders Alzheimer's disease (AD) and Parkinson's disease (PD). Recent results suggested that METH exposure increased the level of α-synuclein (α-syn) that could be a possible cause of neurotoxicity. However, the mechanism of METH-induced neurodegeneration remains unclear. This study was aimed at examining the effects of glycogen synthase kinase3β (GSK3β), α-syn, and tau on METH-induced neurotoxicity. Our results indicated that P-GSK3β (Tyr216), P-Tau (Ser396), α-syn, and P-α-syn (Ser129) levels were increased after METH administration in dose- and time-dependent manners. Upon inhibiting the GSK3β activity with LiCl or GSK3β-siRNA, these protein expressions were significantly decreased. We observed that LiCl protected the cells from METH-caused cytotoxicity by weakening the cell morphological damage and preventing cell apoptosis and death. We also found that P-GSK3β colocalized with P-Tau and α-syn by the immunofluorescence method. Further, METH disrupted the cellular autophagy by upregulation of LC3-II and P62 proteins, and the cellular autophagy was restored by LiCl and GSK3β-siRNA. The expressions of the α-syn-specific degradative enzyme glucocerebrosidase (GCase) with its regulator lysosomal integral membrane protein type-2 (LIMP-2) decreased inversely with the doses of METH treatment. The GCase inhibitor conduritol-β-epoxide (CβE) increased the α-syn levels, and LiCl restored GCase and LIMP-2 expressions disrupted by the METH treatment. In summary, we conclude that GSK3β plays key roles in METH-induced neurotoxicity and neurodegenerative injury by promoting abnormal protein phosphorylation and α-syn accumulation, blocking the autophagy-lysosomal degradation pathway, and finally leading to cell apoptosis and death. GSK3β may be a potential target to prevent METH-induced neurodegeneration.
Collapse
Affiliation(s)
- Lizeng Li
- School of Forensic Medicine , Southern Medical University , Guangzhou 510515 , People's Republic of China . ; ; Tel: +86 2062789101
| | - Si Chen
- School of Forensic Medicine , Southern Medical University , Guangzhou 510515 , People's Republic of China . ; ; Tel: +86 2062789101
| | - Yue Wang
- School of Forensic Medicine , Southern Medical University , Guangzhou 510515 , People's Republic of China . ; ; Tel: +86 2062789101
| | - Xia Yue
- School of Forensic Medicine , Southern Medical University , Guangzhou 510515 , People's Republic of China . ; ; Tel: +86 2062789101
| | - Jingtao Xu
- School of Forensic Medicine , Southern Medical University , Guangzhou 510515 , People's Republic of China . ; ; Tel: +86 2062789101
| | - Weibing Xie
- School of Forensic Medicine , Southern Medical University , Guangzhou 510515 , People's Republic of China . ; ; Tel: +86 2062789101
| | - Pingming Qiu
- School of Forensic Medicine , Southern Medical University , Guangzhou 510515 , People's Republic of China . ; ; Tel: +86 2062789101
| | - Chao Liu
- Guangzhou Forensic Science Institute , Guangzhou 510030 , People's Republic of China
| | - AiFeng Wang
- School of Forensic Medicine , Southern Medical University , Guangzhou 510515 , People's Republic of China . ; ; Tel: +86 2062789101
| | - Huijun Wang
- School of Forensic Medicine , Southern Medical University , Guangzhou 510515 , People's Republic of China . ; ; Tel: +86 2062789101
| |
Collapse
|
17
|
Dang DK, Shin EJ, Kim DJ, Tran HQ, Jeong JH, Jang CG, Ottersen OP, Nah SY, Hong JS, Nabeshima T, Kim HC. PKCδ-dependent p47phox activation mediates methamphetamine-induced dopaminergic neurotoxicity. Free Radic Biol Med 2018; 115:318-337. [PMID: 29269308 PMCID: PMC7074955 DOI: 10.1016/j.freeradbiomed.2017.12.018] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Revised: 11/29/2017] [Accepted: 12/15/2017] [Indexed: 12/11/2022]
Abstract
Protein kinase C (PKC) has been recognized to activate NADPH oxidase (PHOX). However, the interaction between PKC and PHOX in vivo remains elusive. Treatment with methamphetamine (MA) resulted in a selective increase in PKCδ expression out of PKC isoforms. PKCδ co-immunoprecipitated with p47phox, and facilitated phosphorylation and membrane translocation of p47phox. MA-induced increases in PHOX activity and reactive oxygen species were attenuated by knockout of p47phox or PKCδ. In addition, MA-induced impairments in the Nrf-2-related glutathione synthetic system were also mitigated by knockout of p47phox or PKCδ. Glutathione-immunoreactivity was co-localized in Iba-1-labeled microglial cells and in NeuN-labeled neurons, but not in GFAP-labeled astrocytes, reflecting the necessity for self-protection against oxidative stress by mainly microglia. Buthionine-sulfoximine, an inhibitor of glutathione biosynthesis, potentiated microglial activation and pro-apoptotic changes, leading to dopaminergic losses. These neurotoxic processes were attenuated by rottlerin, a pharmacological inhibitor of PKCδ, genetic inhibitions of PKCδ [i.e., PKCδ knockout mice (KO) and PKCδ antisense oligonucleotide (ASO)], or genetic inhibition of p47phox (i.e., p47phox KO or p47phox ASO). Rottlerin did not exhibit any additive effects against the protective activity offered by genetic inhibition of p47phox. Therefore, we suggest that PKCδ is a critical regulator for p47phox activation induced by MA, and that Nrf-2-dependent GSH induction via inhibition of PKCδ or p47phox, is important for dopaminergic protection against MA insult.
Collapse
Affiliation(s)
- Duy-Khanh Dang
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon 24341, Republic of Korea
| | - Eun-Joo Shin
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon 24341, Republic of Korea
| | - Dae-Joong Kim
- Department of Anatomy and Cell Biology, Medical School, Kangwon National University, Chunchon 24341, Republic of Korea
| | - Hai-Quyen Tran
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon 24341, Republic of Korea
| | - Ji Hoon Jeong
- Department of Pharmacology, College of Medicine, Chung-Ang University, Seoul, Republic of Korea
| | - Choon-Gon Jang
- Department of Pharmacology, School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Ole Petter Ottersen
- Laboratory of Molecular Neuroscience, Division of Anatomy, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, N-0317 Oslo, Norway
| | - Seung-Yeol Nah
- Ginsentology Research Laboratory and Department of Physiology, College of Veterinary Medicine, Konkuk University, Seoul 05029, Republic of Korea
| | - Jau-Shyong Hong
- Neuropharmacology Section, Laboratory of Toxicology and Pharmacology, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | - Toshitaka Nabeshima
- Nabeshima Laboratory, Graduate School of Pharmaceutical Sciences, Meijo University, Nagoya 468-8503, Japan
| | - Hyoung-Chun Kim
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon 24341, Republic of Korea.
| |
Collapse
|
18
|
Valian N, Ahmadiani A, Dargahi L. Does Repeated Methamphetamine Exposure at Different Regimens Cause Parkinsonian-Like Behavior in Rats? IRANIAN JOURNAL OF PHARMACEUTICAL RESEARCH : IJPR 2018; 17:543-552. [PMID: 29881412 PMCID: PMC5985172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/01/2022]
Abstract
Methamphetamine (MA), a highly addictive psychostimulant, produces long-lasting neurotoxic effects well proven in nigrostriatal dopaminergic neurons. Considering the similarities between pathological profile of MA neurotoxicity and Parkinson's disease (PD), some reports show that previous MA abusers will be at greater risk of PD-like motor deficits. To answer the question if repeated MA exposure causes parkinsonian-like behavior in rats, we used three regimens of MA administration and assessed the motor performance parameters immediately and over a long period after MA discontinuation. Male Wistar rats in two experimental groups were treated with escalating paradigms consisting of twice daily intraperitoneal injection of either 1-7 mg/kg or 1-14 mg/kg of MA over 14 days. The third group received twice-daily doses of 15 mg/kg of MA every other day for total number of 7 days. At the 1st, 7th, 14th, 21st, 28th, and 60th days after last injections, motor activities were evaluated using narrow beam, pole, and rotarod tests. Locomotor activity was also evaluated using open field test. Repeated-measures ANOVA indicated that over the two months period following MA exposure, drug-treated rats perform beam, pole, and rotarod tests equally well as their corresponding vehicle-treated controls. Comparison of the locomotor activity didn't show significant differences between groups. These data indicated that MA at these regimens does not cause PD-related motor deficits in rats. Since MA doses, exposure duration, and dosing intervals have been shown to affect MA-induced dopaminergic toxicity, it can be concluded that none of these regimens; are strong enough to produce measurable behavioral motor deficits in rat.
Collapse
Affiliation(s)
- Neda Valian
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Abolhassan Ahmadiani
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Leila Dargahi
- Neurobiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Corresponding author: E-mail:
| |
Collapse
|
19
|
Effects of methamphetamine in the hippocampus of cynomolgus monkeys according to age. BIOCHIP JOURNAL 2017. [DOI: 10.1007/s13206-017-1403-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
20
|
Park JH, Seo YH, Jang JH, Jeong CH, Lee S, Park B. Asiatic acid attenuates methamphetamine-induced neuroinflammation and neurotoxicity through blocking of NF-kB/STAT3/ERK and mitochondria-mediated apoptosis pathway. J Neuroinflammation 2017; 14:240. [PMID: 29228978 PMCID: PMC5725763 DOI: 10.1186/s12974-017-1009-0] [Citation(s) in RCA: 101] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Accepted: 11/22/2017] [Indexed: 02/04/2023] Open
Abstract
Background Methamphetamine (METH) is a commonly abused drug that may result in neurotoxic effects. Recent studies have suggested that involvement of neuroinflammatory processes in brain dysfunction is induced by misuse of this drug. However, the mechanism underlying METH-induced inflammation and neurotoxicity in neurons is still unclear. In this study, we investigated whether asiatic acid (AA) effected METH-mediated neuroinflammation and neurotoxicity in dopaminergic neuronal cells. And we further determined whether the effect involved in the nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) and signal transducer and activator of transcription (STAT)3 and extracellular signal-regulated kinase (ERK) pathway. Methods We used the human dopaminergic neuroblastoma SH-SY5Y cell line, murine microglial BV2 cell line, and primary culture of rat embryo mesencephalic neurons. Pro-inflammatory cytokine production was monitored by ELISA and RT/real-time PCR. The cell cycle distribution and mitochondrial membrane integrity was analyzed by flow cytometry. We used immunoblotting, DNA-binding activity, and immunofluorescence staining to analyze the effect of AA on activation of the NF-κB, STAT3, MAPK-ERK, and apoptosis signaling pathways. Results METH induced TNF receptor (TNFR) expression and led to morphological changes of cells. Additionally, this drug increased pro-inflammatory cytokine (TNFα and IL-6) expression. AA significantly suppressed METH-induced TNFR expression in concentration dependent. Increased secretion of TNFα and IL-6 was inhibited in METH-stimulated neuronal cells by AA administration. AA showed significant protection against METH-induced translocation of NF-κB/STAT3 and ERK phosphorylation. AA inhibited METH-induced proteolytic fragmentation of caspase-3 and PARP. The pro-apoptotic protein Bax was significantly decreased, while the anti-apoptotic protein Bcl-xL was increased by AA treatment in METH-stimulated cells. A similar protective effect of AA on mitochondrial membrane integrity was also confirmed by flow cytometry and immunofluorescence staining. Conclusions Based on the literatures and our findings, AA is a promising candidate for an anti-neurotoxic agent, and it can potentially be used for the prevention and treatment of various neurological disorders. Electronic supplementary material The online version of this article (10.1186/s12974-017-1009-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ji-Hyun Park
- College of Pharmacy, Keimyung University, 1095 Dalgubeoldaero, Dalseo-Gu, Daegu, 42601, Republic of Korea
| | - Young Ho Seo
- College of Pharmacy, Keimyung University, 1095 Dalgubeoldaero, Dalseo-Gu, Daegu, 42601, Republic of Korea
| | - Jung-Hee Jang
- Department of Pharmacology, School of Medicine, Keimyung University, 1095 Dalgubeoldaero, Dalseo-Gu, Daegu, 42601, Republic of Korea
| | - Chul-Ho Jeong
- College of Pharmacy, Keimyung University, 1095 Dalgubeoldaero, Dalseo-Gu, Daegu, 42601, Republic of Korea
| | - Sooyeun Lee
- College of Pharmacy, Keimyung University, 1095 Dalgubeoldaero, Dalseo-Gu, Daegu, 42601, Republic of Korea
| | - Byoungduck Park
- College of Pharmacy, Keimyung University, 1095 Dalgubeoldaero, Dalseo-Gu, Daegu, 42601, Republic of Korea.
| |
Collapse
|
21
|
Prenatal methamphetamine exposure is associated with reduced subcortical volumes in neonates. Neurotoxicol Teratol 2017; 65:51-59. [PMID: 29069607 DOI: 10.1016/j.ntt.2017.10.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Revised: 10/19/2017] [Accepted: 10/20/2017] [Indexed: 11/20/2022]
Abstract
OBJECTIVES Prenatal exposure to methamphetamine is associated with a range of neuropsychological, behavioural and cognitive deficits. A small number of imaging studies suggests that these may be mediated by neurostructural changes, including reduced volumes of specific brain regions. This study investigated potential volumetric changes in the brains of neonates with prenatal methamphetamine exposure. To our knowledge no previous studies have examined methamphetamine effects on regional brain volumes at this age. STUDY DESIGN Mothers were recruited antenatally and interviewed regarding methamphetamine use during pregnancy. Mothers in the exposure group reported using methamphetamine≥twice/month during pregnancy; control infants had no exposure to methamphetamine or other drugs and minimal exposure to alcohol. MRI scans were performed in the first postnatal month, following which anatomical images were processed using FreeSurfer. Subcortical and cerebellar regions were manually segmented and their volumes determined using FreeView. Pearson correlations were used to analyse potential associations between methamphetamine exposure and regional volumes. The associations between methamphetamine exposure and regional volumes were then examined adjusting for potential confounding variables. RESULTS Methamphetamine exposure was associated with reduced left and right caudate and thalamus volumes. The association in the right caudate remained significant following adjustment for potential confounding variables. CONCLUSIONS Our findings showing reduced caudate and thalamus volumes in neonates with prenatal methamphetamine exposure are consistent with previous findings in older exposed children, and demonstrate that these changes are already detectable in neonates. Continuing research is warranted to examine whether reduced subcortical volumes are predictive of cognitive, behavioural and affective impairment in older children.
Collapse
|
22
|
Valian N, Ahmadiani A, Dargahi L. Escalating Methamphetamine Regimen Induces Compensatory Mechanisms, Mitochondrial Biogenesis, and GDNF Expression, in Substantia Nigra. J Cell Biochem 2017; 118:1369-1378. [PMID: 27862224 DOI: 10.1002/jcb.25795] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2016] [Accepted: 11/14/2016] [Indexed: 12/22/2022]
Abstract
Methamphetamine (MA) produces long-lasting deficits in dopaminergic neurons in the long-term use via several neurotoxic mechanisms. The effects of MA on mitochondrial biogenesis is less studied currently. So, we evaluated the effects of repeated escalating MA regimen on transcriptional factors involved in mitochondrial biogenesis and glial-derived neurotrophic factor (GDNF) expression in substantia nigra (SN) and striatum of rat. In male Wistar rats, increasing doses of MA (1-14 mg/kg) were administrated twice a day for 14 days. At the 1st, 14th, 28th, and 60th days after MA discontinuation, we measured the PGC1α, TFAM and NRF1 mRNA levels, indicator of mitochondrial biogenesis, and GDNF expression in SN and striatum. Furthermore, we evaluated the glial fibrillary acidic protein (GFAP) and Iba1 mRNA levels, and the levels of tyrosine hydroxylase (TH) and α-synuclein (α-syn) using immunohistochemistry and real-time polymerase chain reaction (PCR). We detected increments in PGC1α and TFAM mRNA levels in SN, but not striatum, and elevations in GDNF levels in SN immediately after MA discontinuation. We also observed increases in GFAP and Iba1 mRNA levels in SN on day 1 and increases in Iba1 mRNA on days 1 and 14 in striatum. Data analysis revealed that the number of TH+ cells in the SN did not reduce in any time points, though TH mRNA levels was increased on day 1 after MA discontinuation in SN. These data show that repeated escalating MA induces several compensatory mechanisms, such as mitochondrial biogenesis and elevation in GDNF in SN. These mechanisms can reverse MA-induced neuroinflammation and prevent TH-immunoreactivity reduction in nigrostriatal pathway. J. Cell. Biochem. 118: 1369-1378, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Neda Valian
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Abolhassan Ahmadiani
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Leila Dargahi
- Neurobiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
23
|
Gonzalez R, Garitaonandia I, Poustovoitov M, Abramihina T, McEntire C, Culp B, Attwood J, Noskov A, Christiansen-Weber T, Khater M, Mora-Castilla S, To C, Crain A, Sherman G, Semechkin A, Laurent LC, Elsworth JD, Sladek J, Snyder EY, Redmond DE, Kern RA. Neural Stem Cells Derived from Human Parthenogenetic Stem Cells Engraft and Promote Recovery in a Nonhuman Primate Model of Parkinson's Disease. Cell Transplant 2016; 25:1945-1966. [DOI: 10.3727/096368916x691682] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Cell therapy has attracted considerable interest as a promising therapeutic alternative for patients with Parkinson's disease (PD). Clinical studies have shown that grafted fetal neural tissue can achieve considerable biochemical and clinical improvements in PD. However, the source of fetal tissue grafts is limited and ethically controversial. Human parthenogenetic stem cells offer a good alternative because they are derived from unfertilized oocytes without destroying potentially viable human embryos and can be used to generate an unlimited supply of neural cells for transplantation. We have previously reported that human parthenogenetic stem cell-derived neural stem cells (hpNSCs) successfully engraft, survive long term, and increase brain dopamine (DA) levels in rodent and nonhuman primate models of PD. Here we report the results of a 12-month transplantation study of hpNSCs in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-lesioned African green monkeys with moderate to severe clinical parkinsonian symptoms. The hpNSCs manufactured under current good manufacturing practice (cGMP) conditions were injected bilaterally into the striatum and substantia nigra of immunosuppressed monkeys. Transplantation of hpNSCs was safe and well tolerated by the animals with no dyskinesia, tumors, ectopic tissue formation, or other test article-related serious adverse events. We observed that hpNSCs promoted behavioral recovery; increased striatal DA concentration, fiber innervation, and number of dopaminergic neurons; and induced the expression of genes and pathways downregulated in PD compared to vehicle control animals. These results provide further evidence for the clinical translation of hpNSCs and support the approval of the world's first pluripotent stem cell-based phase I/IIa study for the treatment of PD (Clinical Trial Identifier NCT02452723).
Collapse
Affiliation(s)
| | | | | | | | | | - Ben Culp
- Axion Research Foundation, Hamden, CT, USA
| | | | | | | | - Marwa Khater
- Department of Reproductive Medicine, University of California San Diego, La Jolla, CA, USA
| | - Sergio Mora-Castilla
- Department of Reproductive Medicine, University of California San Diego, La Jolla, CA, USA
| | - Cuong To
- Department of Reproductive Medicine, University of California San Diego, La Jolla, CA, USA
| | - Andrew Crain
- Stem Cell Research Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Glenn Sherman
- International Stem Cell Corporation, Carlsbad, CA, USA
| | | | - Louise C. Laurent
- Department of Reproductive Medicine, University of California San Diego, La Jolla, CA, USA
| | - John D. Elsworth
- Department of Psychiatry and Neurosurgery, Yale University School of Medicine, New Haven, CT, USA
| | - John Sladek
- Department of Neurology, Pediatrics and Neuroscience, University of Colorado School of Medicine, Aurora, CO, USA
| | - Evan Y. Snyder
- Stem Cell Research Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - D. Eugene Redmond
- Axion Research Foundation, Hamden, CT, USA
- Department of Psychiatry and Neurosurgery, Yale University School of Medicine, New Haven, CT, USA
| | | |
Collapse
|
24
|
Garrick JM, Dao K, de Laat R, Elsworth J, Cole TB, Marsillach J, Furlong CE, Costa LG. Developmental expression of paraoxonase 2. Chem Biol Interact 2016; 259:168-174. [PMID: 27062895 DOI: 10.1016/j.cbi.2016.04.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2016] [Revised: 03/10/2016] [Accepted: 04/04/2016] [Indexed: 02/07/2023]
Abstract
Paraoxonase 2 (PON2) is a member of the paraoxonase gene family also comprising PON1 and PON3. PON2 functions as a lactonase and exhibits anti-bacterial as well as antioxidant properties. At the cellular level, PON2 localizes to the mitochondrial and endoplasmic reticulum membranes where it scavenges reactive oxygen species. PON2 is of particular interest as it is the only paraoxonase expressed in brain tissue and appears to play a critical role in mitigating oxidative stress in the brain. The aim of this study was to investigate the expression of PON2 at the protein and mRNA level in the brain and liver of mice through development to identify potential age windows of susceptibility to oxidative stress, as well as to compare expression of hepatic PON2 to expression of PON1 and PON3. Overall, PON2 expression in the brain was lower in neonatal mice and increased with age up to postnatal day (PND) 21, with a significant decrease observed at PND 30 and 60. In contrast, the liver showed continuously increasing levels of PON2 with age, similar to the patterns of PON1 and PON3. PON2 protein levels were also investigated in brain samples from non-human primates, with PON2 increasing with age up to the infant stage and decreasing at the juvenile stage, mirroring the results observed in the mouse brain. These variable expression levels of PON2 suggest that neonatal and young adult animals may be more susceptible to neurological insult by oxidants due to lower levels of PON2 in the brain.
Collapse
Affiliation(s)
- Jacqueline M Garrick
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA, United States.
| | - Khoi Dao
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA, United States
| | - Rian de Laat
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA, United States
| | - John Elsworth
- Department of Psychiatry, Yale University, New Haven, CT, United States
| | - Toby B Cole
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA, United States; Center on Human Development and Disabilities, University of Washington, United States
| | - Judit Marsillach
- Depts. of Medicine (Div. Medical Genetics) and of Genome Sciences, University of Washington, United States
| | - Clement E Furlong
- Depts. of Medicine (Div. Medical Genetics) and of Genome Sciences, University of Washington, United States
| | - Lucio G Costa
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA, United States; Dept. of Neuroscience, University of Parma, Italy
| |
Collapse
|
25
|
Dang DK, Shin EJ, Nam Y, Ryoo S, Jeong JH, Jang CG, Nabeshima T, Hong JS, Kim HC. Apocynin prevents mitochondrial burdens, microglial activation, and pro-apoptosis induced by a toxic dose of methamphetamine in the striatum of mice via inhibition of p47phox activation by ERK. J Neuroinflammation 2016; 13:12. [PMID: 26780950 PMCID: PMC4717833 DOI: 10.1186/s12974-016-0478-x] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Accepted: 01/11/2016] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Activation of NADPH oxidase (PHOX) plays a critical role in mediating dopaminergic neuroinflammation. In the present study, we investigated the role of PHOX in methamphetamine (MA)-induced neurotoxic and inflammatory changes in mice. METHODS We examined changes in mitogen-activated protein kinases (MAPKs), mitochondrial function [i.e., mitochondrial membrane potential, intramitochondrial Ca(2+) accumulation, mitochondrial oxidative burdens, mitochondrial superoxide dismutase expression, and mitochondrial translocation of the cleaved form of protein kinase C delta type (cleaved PKCδ)], microglial activity, and pro-apoptotic changes [i.e., cytosolic cytochrome c release, cleaved caspase 3, and terminal deoxynucleotidyl transferase dUDP nick-end labeling (TUNEL) positive populations] after a neurotoxic dose of MA in the striatum of mice to achieve a better understanding of the effects of apocynin, a non-specific PHOX inhibitor, or genetic inhibition of p47phox (by using p47phox knockout mice or p47phox antisense oligonucleotide) against MA-induced dopaminergic neurotoxicity. RESULTS Phosphorylation of extracellular signal-regulated kinases (ERK1/2) was most pronounced out of MAPKs after MA. We observed MA-induced phosphorylation and membrane translocation of p47phox in the striatum of mice. The activation of p47phox promoted mitochondrial stresses followed by microglial activation into the M1 phenotype, and pro-apoptotic changes, and led to dopaminergic impairments. ERK activated these signaling pathways. Apocynin or genetic inhibition of p47phox significantly protected these signaling processes induced by MA. ERK inhibitor U0126 did not exhibit any additional positive effects against protective activity mediated by apocynin or p47phox genetic inhibition, suggesting that ERK regulates p47phox activation, and ERK constitutes the crucial target for apocynin-mediated inhibition of PHOX activation. CONCLUSIONS Our results indicate that the neuroprotective mechanism of apocynin against MA insult is via preventing mitochondrial burdens, microglial activation, and pro-apoptotic signaling process by the ERK-dependent activation of p47phox.
Collapse
Affiliation(s)
- Duy-Khanh Dang
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon, South Korea.
| | - Eun-Joo Shin
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon, South Korea.
| | - Yunsung Nam
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon, South Korea.
| | - Sungwoo Ryoo
- Department of Biological Sciences, College of Natural Sciences, Kangwon National University, Chunchon, South Korea.
| | - Ji Hoon Jeong
- Department of Pharmacology, College of Medicine, Chung-Ang University, Seoul, South Korea.
| | - Choon-Gon Jang
- Department of Pharmacology, School of Pharmacy, Sungkyunkwan University, Suwon, South Korea.
| | - Toshitaka Nabeshima
- Department of Regional Pharmaceutical Care and Sciences, Graduate School of Pharmaceutical Sciences, Meijo University, Nagoya, Japan. .,NPO, Japanese Drug Organization of Appropriate Use and Research, Nagoya, Japan.
| | - Jau-Shyong Hong
- Neuropharmacology Section, Laboratory of Toxicology and Pharmacology, National Institute of Environmental Health Sciences, Research Triangle Park, Durham, NC, USA.
| | - Hyoung-Chun Kim
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon, South Korea.
| |
Collapse
|
26
|
Gubellini P, Kachidian P. Animal models of Parkinson's disease: An updated overview. Rev Neurol (Paris) 2015; 171:750-61. [PMID: 26343921 DOI: 10.1016/j.neurol.2015.07.011] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Revised: 07/20/2015] [Accepted: 07/22/2015] [Indexed: 12/21/2022]
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disorder whose etiology, besides a minority of genetic cases, is still largely unknown. Animal models have contributed to elucidate PD etiology and pathogenesis, as well as its cellular and molecular mechanisms, leading to the general hypothesis that this neurological disorder is due to complex interactions between environmental and genetic factors. However, the full understanding of PD is still very far from being achieved, and new potential treatments need to be tested to further improve patients' quality of life and, possibly, slow down the neurodegenerative process. In this context, animal models of PD are required to address all these issues. "Classic" models are based on neurotoxins that selectively target catecholaminergic neurons (such as 6-hydroxydopamine, 1-methyl-1,2,3,6-tetrahydropiridine, agricultural pesticides, etc.), while more recent models employ genetic manipulations that either introduce mutations similar to those find in familial cases of PD (α-synuclein, DJ-1, PINK1, Parkin, etc.) or selectively disrupt nigrostriatal neurons (MitoPark, Pitx3, Nurr1, etc.). Each one of these models has its own advantages and limitations, thus some are better suited for studying PD pathogenesis, while others are more pertinent to test therapeutic treatments. Here, we provide a critical and updated review of the most used PD models.
Collapse
Affiliation(s)
- P Gubellini
- Aix-Marseille Université, CNRS, Institut de Biologie du Développement de Marseille (IBDM) UMR7288, case 907, parc scientifique de Luminy, 163, avenue de Luminy, 13009 Marseille, France
| | - P Kachidian
- Aix-Marseille Université, CNRS, Institut de Biologie du Développement de Marseille (IBDM) UMR7288, case 907, parc scientifique de Luminy, 163, avenue de Luminy, 13009 Marseille, France.
| |
Collapse
|
27
|
Elsworth JD, Jentsch JD, Groman SM, Roth RH, Redmond ED, Leranth C. Low circulating levels of bisphenol-A induce cognitive deficits and loss of asymmetric spine synapses in dorsolateral prefrontal cortex and hippocampus of adult male monkeys. J Comp Neurol 2015; 523:1248-57. [PMID: 25557059 PMCID: PMC4390445 DOI: 10.1002/cne.23735] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Revised: 12/22/2014] [Accepted: 12/22/2014] [Indexed: 01/25/2023]
Abstract
Bisphenol-A (BPA) is widely used in the manufacture of plastics, epoxy resins, and certain paper products. A majority of the population in the developed world is routinely exposed to BPA from multiple sources and has significant circulating levels of BPA. Although BPA is categorized as an endocrine disruptor with a growing literature on adverse effects, it is uncertain whether cognitive dysfunction is induced in humans by exposure to BPA. The present study examined the impact of BPA in primate brain by exposing adult male vervet monkeys for 4 weeks continuously to circulating levels of BPA that were in the range measured in studies of humans environmentally exposed to BPA. This regimen of exposure to BPA decreased both working memory accuracy and the number of excitatory synaptic inputs on dendritic spines of pyramidal neurons in two brain regions that are necessary for working memory (prefrontal cortex and hippocampus). These observed behavioral and synaptic effects were ameliorated following withdrawal from BPA. As Old World monkeys (e.g., vervets) and humans share some uniquely primate morphological, endocrine, and cognitive traits, this study indicates the potential for significant cognitive disruption following exposure of humans to BPA.
Collapse
Affiliation(s)
- John D Elsworth
- Department of Psychiatry, Yale University, School of Medicine, New Haven, Connecticut
| | | | | | | | | | | |
Collapse
|
28
|
Abstract
Psychostimulants are a diverse group of substances with their main psychomotor effects resembling those of amphetamine, methamphetamine, cocaine, or cathinone. Due to their potential as drugs of abuse, recreational use of most of these substances is illegal since 1971 Convention on Psychotropic Substances. In recent years, new psychoactive substances have emerged mainly as synthetic cathinones with new molecules frequently complementing the list. Psychostimulant related movement disorders are a known entity often seen in emergency rooms around the world. These admissions are becoming more frequent as are fatalities associated with drug abuse. Still the legal constraints of the novel synthetic molecules are bypassed. At the same time, chronic and permanent movement disorders are much less frequently encountered. These disorders frequently manifest as a combination of movement disorders. The more common symptoms include agitation, tremor, hyperkinetic and stereotypical movements, cognitive impairment, and also hyperthermia and cardiovascular dysfunction. The pathophysiological mechanisms behind the clinical manifestations have been researched for decades. The common denominator is the monoaminergic signaling. Dopamine has received the most attention but further research has demonstrated involvement of other pathways. Common mechanisms linking psychostimulant use and several movement disorders exist.
Collapse
Affiliation(s)
- Andres Asser
- Department of Neurology and Neurosurgery, University of Tartu , Tartu , Estonia
| | - Pille Taba
- Department of Neurology and Neurosurgery, University of Tartu , Tartu , Estonia
| |
Collapse
|
29
|
Huot P, Fox SH, Brotchie JM. Monoamine reuptake inhibitors in Parkinson's disease. PARKINSON'S DISEASE 2015; 2015:609428. [PMID: 25810948 PMCID: PMC4355567 DOI: 10.1155/2015/609428] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Accepted: 12/26/2014] [Indexed: 12/13/2022]
Abstract
The motor manifestations of Parkinson's disease (PD) are secondary to a dopamine deficiency in the striatum. However, the degenerative process in PD is not limited to the dopaminergic system and also affects serotonergic and noradrenergic neurons. Because they can increase monoamine levels throughout the brain, monoamine reuptake inhibitors (MAUIs) represent potential therapeutic agents in PD. However, they are seldom used in clinical practice other than as antidepressants and wake-promoting agents. This review article summarises all of the available literature on use of 50 MAUIs in PD. The compounds are divided according to their relative potency for each of the monoamine transporters. Despite wide discrepancy in the methodology of the studies reviewed, the following conclusions can be drawn: (1) selective serotonin transporter (SERT), selective noradrenaline transporter (NET), and dual SERT/NET inhibitors are effective against PD depression; (2) selective dopamine transporter (DAT) and dual DAT/NET inhibitors exert an anti-Parkinsonian effect when administered as monotherapy but do not enhance the anti-Parkinsonian actions of L-3,4-dihydroxyphenylalanine (L-DOPA); (3) dual DAT/SERT inhibitors might enhance the anti-Parkinsonian actions of L-DOPA without worsening dyskinesia; (4) triple DAT/NET/SERT inhibitors might exert an anti-Parkinsonian action as monotherapy and might enhance the anti-Parkinsonian effects of L-DOPA, though at the expense of worsening dyskinesia.
Collapse
Affiliation(s)
- Philippe Huot
- Toronto Western Research Institute, Toronto Western Hospital, University Health Network, 399 Bathurst Street, Toronto, ON, Canada M5T 2S8
- Division of Neurology, Movement Disorder Clinic, Toronto Western Hospital, University Health Network, University of Toronto, 399 Bathurst Street, Toronto, ON, Canada M5T 2S8
- Department of Pharmacology and Division of Neurology, Faculty of Medicine, Université de Montréal and Centre Hospitalier de l'Université de Montréal, Montréal, QC, Canada
| | - Susan H. Fox
- Toronto Western Research Institute, Toronto Western Hospital, University Health Network, 399 Bathurst Street, Toronto, ON, Canada M5T 2S8
- Division of Neurology, Movement Disorder Clinic, Toronto Western Hospital, University Health Network, University of Toronto, 399 Bathurst Street, Toronto, ON, Canada M5T 2S8
| | - Jonathan M. Brotchie
- Toronto Western Research Institute, Toronto Western Hospital, University Health Network, 399 Bathurst Street, Toronto, ON, Canada M5T 2S8
| |
Collapse
|
30
|
Blesa J, Przedborski S. Parkinson's disease: animal models and dopaminergic cell vulnerability. Front Neuroanat 2014; 8:155. [PMID: 25565980 PMCID: PMC4266040 DOI: 10.3389/fnana.2014.00155] [Citation(s) in RCA: 345] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Accepted: 11/27/2014] [Indexed: 12/18/2022] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder that affects about 1.5% of the global population over 65 years of age. A hallmark feature of PD is the degeneration of the dopamine (DA) neurons in the substantia nigra pars compacta (SNc) and the consequent striatal DA deficiency. Yet, the pathogenesis of PD remains unclear. Despite tremendous growth in recent years in our knowledge of the molecular basis of PD and the molecular pathways of cell death, important questions remain, such as: (1) why are SNc cells especially vulnerable; (2) which mechanisms underlie progressive SNc cell loss; and (3) what do Lewy bodies or α-synuclein reveal about disease progression. Understanding the variable vulnerability of the dopaminergic neurons from the midbrain and the mechanisms whereby pathology becomes widespread are some of the primary objectives of research in PD. Animal models are the best tools to study the pathogenesis of PD. The identification of PD-related genes has led to the development of genetic PD models as an alternative to the classical toxin-based ones, but does the dopaminergic neuronal loss in actual animal models adequately recapitulate that of the human disease? The selection of a particular animal model is very important for the specific goals of the different experiments. In this review, we provide a summary of our current knowledge about the different in vivo models of PD that are used in relation to the vulnerability of the dopaminergic neurons in the midbrain in the pathogenesis of PD.
Collapse
Affiliation(s)
- Javier Blesa
- Department of Pathology and Cell Biology, Center for Motor Neuron Biology and Disease, College of Physicians and Surgeons, Columbia UniversityNew York, NY, USA
| | | |
Collapse
|
31
|
Hypothalamic prolyl endopeptidase (PREP) regulates pancreatic insulin and glucagon secretion in mice. Proc Natl Acad Sci U S A 2014; 111:11876-81. [PMID: 25071172 DOI: 10.1073/pnas.1406000111] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Prolyl endopeptidase (PREP) has been implicated in neuronal functions. Here we report that hypothalamic PREP is predominantly expressed in the ventromedial nucleus (VMH), where it regulates glucose-induced neuronal activation. PREP knockdown mice (Prep(gt/gt)) exhibited glucose intolerance, decreased fasting insulin, increased fasting glucagon levels, and reduced glucose-induced insulin secretion compared with wild-type controls. Consistent with this, central infusion of a specific PREP inhibitor, S17092, impaired glucose tolerance and decreased insulin levels in wild-type mice. Arguing further for a central mode of action of PREP, isolated pancreatic islets showed no difference in glucose-induced insulin release between Prep(gt/gt) and wild-type mice. Furthermore, hyperinsulinemic euglycemic clamp studies showed no difference between Prep(gt/gt) and wild-type control mice. Central PREP regulation of insulin and glucagon secretion appears to be mediated by the autonomic nervous system because Prep(gt/gt) mice have elevated sympathetic outflow and norepinephrine levels in the pancreas, and propranolol treatment reversed glucose intolerance in these mice. Finally, re-expression of PREP by bilateral VMH injection of adeno-associated virus-PREP reversed the glucose-intolerant phenotype of the Prep(gt/gt) mice. Taken together, our results unmask a previously unknown player in central regulation of glucose metabolism and pancreatic function.
Collapse
|
32
|
Kwiatkowski MA, Roos A, Stein DJ, Thomas KGF, Donald K. Effects of prenatal methamphetamine exposure: a review of cognitive and neuroimaging studies. Metab Brain Dis 2014; 29:245-54. [PMID: 24370774 DOI: 10.1007/s11011-013-9470-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2013] [Accepted: 12/06/2013] [Indexed: 10/25/2022]
Abstract
Prenatal methamphetamine exposure (PME) is a significant problem in several parts of the world and poses important health risks for the developing fetus. Research on the short- and long-term outcomes of PME is scarce, however. Here, we summarize present knowledge on the cognitive and behavioral outcomes of PME, based on a review of the neuroimaging, neuropsychology, and neuroscience literature published in the past 15 years. Several studies have reported that the behavioral and cognitive sequelae of PME include broad deficits in the domains of attention, memory, and visual-motor integration. Knowledge regarding brain-behavior relationships is poor, however, in large part because imaging studies are rare. Hence, the effects of PME on developing neurocircuitry and brain architecture remain speculative, and are largely deductive. Some studies have implicated the dopamine-rich fronto-striatal pathways; however, cognitive deficits (e.g., impaired visual-motor integration) that should be associated with damage to those pathways are not manifested consistently across studies. We conclude by discussing challenges endemic to research on prenatal drug exposure, and argue that they may account for some of the inconsistencies in the extant research on PME. Studies confirming predicted brain-behavior relationships in PME, and exploring possible mechanisms underlying those relationships, are needed if neuroscience is to address the urgency of this growing public health problem.
Collapse
|
33
|
Viola TW, Tractenberg SG, Levandowski ML, Pezzi JC, Bauer ME, Teixeira AL, Grassi-Oliveira R. Neurotrophic factors in women with crack cocaine dependence during early abstinence: the role of early life stress. J Psychiatry Neurosci 2014; 39:206-14. [PMID: 24331739 PMCID: PMC3997606 DOI: 10.1503/jpn.130027] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2013] [Revised: 06/26/2013] [Accepted: 10/11/2013] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Neurotrophic factors have been investigated in the pathophysiology of alcohol and drug dependence and have been related to early life stress driving developmental programming of neuroendocrine systems. METHODS We conducted a follow-up study that aimed to assess the plasma levels of glial cell line-derived neurotrophic factor (GDNF), brain-derived neurotrophic factor (BDNF), nerve growth factor (NGF), neurotrophin-3 (NT3) and neurotrophin-4/5 (NT4/5) in crack users during 3 weeks of early abstinence in comparison with healthy controls. We performed a comprehensive clinical assessment in female inpatients with crack cocaine dependence (separated into 2 groups: participants with (CSA+) and without (CSA-) a history of childhood sexual abuse) and a group of nonuser control participants. RESULTS Our sample included 104 women with crack cocaine dependence and 22 controls; of the women who used crack cocaine, 22 had a history of childhood sexual abuse and 82 did not. The GDNF plasma levels in the CSA+ group increased dramatically during 3 weeks of detoxification. In contrast, those in the CSA- group showed lower and stable levels of GDNF under the same conditions. Compared with the control group, BDNF plasma levels remained elevated and NGF levels were reduced during early abstinence. We found no differences in NT3 and NT4/5 between the patients and controls. However, within-group analyses showed that the CSA+ group exhibited higher levels of NT4/5 than the CSA- group at the end of detoxification. LIMITATIONS Some of the participants were using neuroleptics, mood stabilizers or antidepressants; our sample included only women; memory bias could not be controlled; and we did not investigate the possible confounding effects of other forms of stress during childhood. CONCLUSION This study supports the association between early life stress and peripheral neurotrophic factor levels in crack cocaine users. During early abstinence, plasmastic GDNF and NT4/5 were the only factors to show changes associated with a history of childhood sexual abuse.
Collapse
Affiliation(s)
- Thiago Wendt Viola
- Viola, Tractenberg, Levandowski, Bauer, Grassi-Oliveira — Centre of Studies and Research in Traumatic Stress, PostGraduate Program in Psychology, Pontifical Catholic University of the Rio Grande do Sul (PUCRS), Porto Alegre, Brazil; Pezzi — Post-Graduate Program in Health Sciences, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, Brazil; Bauer, Grassi-Oliveira — Laboratory of Immunosenescence, Institute of Biomedical Research, Pontifical Catholic University of the Rio Grande do Sul (PUCRS), Porto Alegre, Brazil; Teixeira — Neuroscience Program, Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Saulo Gantes Tractenberg
- Viola, Tractenberg, Levandowski, Bauer, Grassi-Oliveira — Centre of Studies and Research in Traumatic Stress, PostGraduate Program in Psychology, Pontifical Catholic University of the Rio Grande do Sul (PUCRS), Porto Alegre, Brazil; Pezzi — Post-Graduate Program in Health Sciences, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, Brazil; Bauer, Grassi-Oliveira — Laboratory of Immunosenescence, Institute of Biomedical Research, Pontifical Catholic University of the Rio Grande do Sul (PUCRS), Porto Alegre, Brazil; Teixeira — Neuroscience Program, Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Mateus Luz Levandowski
- Viola, Tractenberg, Levandowski, Bauer, Grassi-Oliveira — Centre of Studies and Research in Traumatic Stress, PostGraduate Program in Psychology, Pontifical Catholic University of the Rio Grande do Sul (PUCRS), Porto Alegre, Brazil; Pezzi — Post-Graduate Program in Health Sciences, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, Brazil; Bauer, Grassi-Oliveira — Laboratory of Immunosenescence, Institute of Biomedical Research, Pontifical Catholic University of the Rio Grande do Sul (PUCRS), Porto Alegre, Brazil; Teixeira — Neuroscience Program, Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Júlio Carlos Pezzi
- Viola, Tractenberg, Levandowski, Bauer, Grassi-Oliveira — Centre of Studies and Research in Traumatic Stress, PostGraduate Program in Psychology, Pontifical Catholic University of the Rio Grande do Sul (PUCRS), Porto Alegre, Brazil; Pezzi — Post-Graduate Program in Health Sciences, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, Brazil; Bauer, Grassi-Oliveira — Laboratory of Immunosenescence, Institute of Biomedical Research, Pontifical Catholic University of the Rio Grande do Sul (PUCRS), Porto Alegre, Brazil; Teixeira — Neuroscience Program, Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Moisés Evandro Bauer
- Viola, Tractenberg, Levandowski, Bauer, Grassi-Oliveira — Centre of Studies and Research in Traumatic Stress, PostGraduate Program in Psychology, Pontifical Catholic University of the Rio Grande do Sul (PUCRS), Porto Alegre, Brazil; Pezzi — Post-Graduate Program in Health Sciences, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, Brazil; Bauer, Grassi-Oliveira — Laboratory of Immunosenescence, Institute of Biomedical Research, Pontifical Catholic University of the Rio Grande do Sul (PUCRS), Porto Alegre, Brazil; Teixeira — Neuroscience Program, Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Antonio Lúcio Teixeira
- Viola, Tractenberg, Levandowski, Bauer, Grassi-Oliveira — Centre of Studies and Research in Traumatic Stress, PostGraduate Program in Psychology, Pontifical Catholic University of the Rio Grande do Sul (PUCRS), Porto Alegre, Brazil; Pezzi — Post-Graduate Program in Health Sciences, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, Brazil; Bauer, Grassi-Oliveira — Laboratory of Immunosenescence, Institute of Biomedical Research, Pontifical Catholic University of the Rio Grande do Sul (PUCRS), Porto Alegre, Brazil; Teixeira — Neuroscience Program, Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Rodrigo Grassi-Oliveira
- Viola, Tractenberg, Levandowski, Bauer, Grassi-Oliveira — Centre of Studies and Research in Traumatic Stress, PostGraduate Program in Psychology, Pontifical Catholic University of the Rio Grande do Sul (PUCRS), Porto Alegre, Brazil; Pezzi — Post-Graduate Program in Health Sciences, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, Brazil; Bauer, Grassi-Oliveira — Laboratory of Immunosenescence, Institute of Biomedical Research, Pontifical Catholic University of the Rio Grande do Sul (PUCRS), Porto Alegre, Brazil; Teixeira — Neuroscience Program, Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil
| |
Collapse
|
34
|
Jumnongprakhon P, Govitrapong P, Tocharus C, Tungkum W, Tocharus J. Protective effect of melatonin on methamphetamine-induced apoptosis in glioma cell line. Neurotox Res 2013; 25:286-94. [PMID: 23975636 DOI: 10.1007/s12640-013-9419-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2013] [Revised: 08/06/2013] [Accepted: 08/09/2013] [Indexed: 12/24/2022]
Abstract
Methamphetamine (METH) is a highly addictive drug causing neurodegenerative diseases. METH has been known to be neurotoxic by inducing oxidative stress, free radical, and pro-inflammatory cytokines. Previous studies have shown that METH could induce neuron and glial cell death, especially inducing glial cell-mediated neurotoxicity that plays a critical role in stress-induced central nervous system damage. Therefore, the aim of the present study is to explore the mechanisms of METH-induced cell death in the glial cell. METH-induced glial cells death is mediated via mitochondrial damage pathway. METH activates the upregulation of the Bax, cytochrome c, cleavage caspase 9 and 3 proteins, and downregulation of Bcl-XL protein in cascade. Pretreatment with melatonin, a neurohormone secreted by the pineal gland, effectively reduced glial cell death. Moreover, melatonin increased the Bcl-XL/Bax ratio but reduced the level of cytochrome c, cleavage caspase 9 and 3 proteins. Therefore, these results demonstrated that melatonin could reduce the cytotoxic effect of METH by decreasing the mitochondrial death pathway activation in glial cells. This outcome suggests that melatonin might be beneficial as the neuroprotection in neurodegenerative diseases caused by METH or other pathogens.
Collapse
Affiliation(s)
- Pichaya Jumnongprakhon
- Department of Anatomy, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
| | | | | | | | | |
Collapse
|
35
|
Redmond DE, McEntire CRS, Kingsbery JP, Leranth C, Elsworth JD, Bjugstad KB, Roth RH, Samulski RJ, Sladek JR. Comparison of fetal mesencephalic grafts, AAV-delivered GDNF, and both combined in an MPTP-induced nonhuman primate Parkinson's model. Mol Ther 2013; 21:2160-8. [PMID: 23913185 DOI: 10.1038/mt.2013.180] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2013] [Accepted: 07/20/2013] [Indexed: 02/08/2023] Open
Abstract
We combined viral vector delivery of human glial-derived neurotrophic factor (GDNF) with the grafting of dopamine (DA) precursor cells from fetal ventral mesencephalon (VM) to determine whether these strategies would improve the anti-Parkinson's effects in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-treated monkeys, an animal model for Parkinson's disease (PD). Both strategies have been reported as individually beneficial in animal models of PD, leading to clinical studies. GDNF delivery has also been reported to augment VM tissue implants, but no combined studies have been done in monkeys. Monkeys were treated with MPTP and placed into four balanced treatment groups receiving only recombinant adeno-associated virus serotype 5 (rAAV5)/hu-GDNF, only fetal DA precursor cells, both together, or a buffered saline solution (control). The combination of fetal precursors with rAAV5/hu-GDNF showed significantly higher striatal DA concentrations compared with the other treatments, but did not lead to greater functional improvement in this study. For the first time under identical conditions in primates, we show that all three treatments lead to improvement compared with control animals.
Collapse
Affiliation(s)
- D Eugene Redmond
- 1] Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut, USA [2] Department of Neurosurgery, Yale University School of Medicine, New Haven, Connecticut, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
The evaluation of AZ66, an optimized sigma receptor antagonist, against methamphetamine-induced dopaminergic neurotoxicity and memory impairment in mice. Int J Neuropsychopharmacol 2013; 16:1033-44. [PMID: 22932447 DOI: 10.1017/s1461145712000831] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
Sigma (σ) receptors have recently been identified as potential targets for the development of novel therapeutics aimed at mitigating the effects of methamphetamine. Particularly, σ receptors are believed to mitigate some of the neurotoxic effects of methamphetamine through modulation of dopamine, dopamine transporters and body temperature. Furthermore, recent evidence suggests that targeting σ receptors may prevent cognitive impairments produced by methamphetamine. In the present study, an optimized σ receptor antagonist, AZ66, was evaluated against methamphetamine-induced neurotoxicity and cognitive dysfunction. AZ66 was found to be highly selective for σ receptors compared to 64 other sites tested. Pretreatment of male, Swiss Webster mice with i.p. dosing of AZ66 significantly attenuated methamphetamine-induced striatal dopamine depletions, striatal dopamine transporter reductions and hyperthermia. Additionally, neurotoxic dosing with methamphetamine caused significant memory impairment in the object recognition test, which was attenuated when animals were pretreated with AZ66; similar trends were observed in the step-through passive avoidance test. Taken together, these results suggest that targeting σ receptors may provide neuroprotection against the neurotoxicity and cognitive impairments produced by methamphetamine.
Collapse
|
37
|
Loss of asymmetric spine synapses in prefrontal cortex of motor-asymptomatic, dopamine-depleted, cognitively impaired MPTP-treated monkeys. Int J Neuropsychopharmacol 2013; 16:905-12. [PMID: 22947206 PMCID: PMC3733504 DOI: 10.1017/s1461145712000892] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Parkinson's disease is usually characterized as a movement disorder; however, cognitive abilities that are dependent on the prefrontal cortex decline at an early stage of the disease in most patients. The changes that underlie cognitive deficits in Parkinson's disease are not well understood. We hypothesize that reduced dopamine signalling in the prefrontal cortex in Parkinson's disease is a harbinger of detrimental synaptic changes in pyramidal neurons in the prefrontal cortex, whose function is necessary for normal cognition. Our previous data showed that monkeys exposed to the neurotoxin, 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), but not exhibiting overt motor deficits (motor-asymptomatic), displayed cognitive deficits in prefrontal cortex-dependent tasks. The present results demonstrate that motor-asymptomatic MPTP-treated monkeys have a reduced dopamine concentration and a substantially lower number (50%) of asymmetric (excitatory) spine synapses in layer II/III, but not layer V, of the dorsolateral prefrontal cortex, compared to controls. In contrast, neither dopamine concentration nor asymmetric synapse number was altered in the entorhinal cortex of MPTP-treated monkeys. Together, these findings suggest that the number of asymmetric spine synapses on dendrites in the prefrontal cortex is dopamine-dependent and that the loss of synapses may be a morphological substrate of the cognitive deficits induced by a reduction in dopamine neurotransmission in this region.
Collapse
|
38
|
Elsworth JD, Redmond DE, Roth RH. Coordinated expression of dopamine transporter and vesicular monoamine transporter in the primate striatum during development. Synapse 2013; 67:580-5. [PMID: 23468413 DOI: 10.1002/syn.21662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2013] [Revised: 02/04/2013] [Accepted: 02/24/2013] [Indexed: 11/06/2022]
Abstract
Several addictive or neurotoxic drugs are dependent on the dopamine transporter (DAT) and/or vesicular monoamine transporter (VMAT2) to exert their detrimental effects on dopamine neurons. For example, methamphetamine and MPTP are substrates for both DAT and VMAT2, with the ratio of DAT to VMAT2 in striatum being a determinant of the degree of toxicity inflicted by these drugs on dopamine neurons. Thus, the susceptibility of dopamine neurons to agents whose pharmacology involves DAT and VMAT2 may vary during development if the ontogeny of DAT and VMAT2 differs, and this is relevant as exposure of dopamine neurons to toxic agents during development is hypothesized to underlie some neurological or psychiatric disorders. However, the relative expression of DAT and VMAT2 has not been studied in either primate or nonprimate fetal brain, and this was addressed in the present study by measuring the binding of specific radioligands of DAT and VMAT2 to striatal membranes from nonhuman primates at mid-gestation, late-gestation, and the postnatal and adult periods. Dopamine concentration was also determined in striatal tissue from the same brains. These data indicate that in striatum of primates, unlike rodents, there is a sharp increase in DAT and VMAT2 expression after mid-gestation, with adult levels being attained at the time of birth. In addition, this study demonstrated that there is a coordinated expression of DAT and VMAT2 from the time of mid-gestation to adulthood.
Collapse
Affiliation(s)
- John D Elsworth
- Neuropsychopharmacology Research Laboratory, Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut 06511, USA.
| | | | | |
Collapse
|
39
|
Elsworth JD, Jentsch JD, Vandevoort CA, Roth RH, Redmond DE, Leranth C. Prenatal exposure to bisphenol A impacts midbrain dopamine neurons and hippocampal spine synapses in non-human primates. Neurotoxicology 2013; 35:113-20. [PMID: 23337607 DOI: 10.1016/j.neuro.2013.01.001] [Citation(s) in RCA: 98] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2012] [Revised: 12/29/2012] [Accepted: 01/03/2013] [Indexed: 11/29/2022]
Abstract
Prevalent use of bisphenol-A (BPA) in the manufacture of resins, plastics and paper products has led to frequent exposure of most people to this endocrine disruptor. Some rodent studies have suggested that BPA can exert detrimental effects on brain development. However as rodent models cannot be relied on to predict consequences of human exposure to BPA during development, it is important to investigate the effects of BPA on non-human primate brain development. Previous research suggests that BPA preferentially targets dopamine neurons in ventral mesencephalon and glutamatergic neurons in hippocampus, so the present work examined the susceptibility of these systems to low dose BPA exposure at the fetal and juvenile stages of development in non-human primates. Exposure of pregnant rhesus monkeys to relatively low levels of BPA during the final 2 months of gestation, induced abnormalities in fetal ventral mesencephalon and hippocampus. Specifically, light microscopy revealed a decrease in tyrosine hydroxylase-expressing (dopamine) neurons in the midbrain of BPA-exposed fetuses and electron microscopy identified a reduction in spine synapses in the CA1 region of hippocampus. In contrast, administration of BPA to juvenile vervet monkeys (14-18 months of age) was without effect on these indices, or on dopamine and serotonin concentrations in striatum and prefrontal cortex, or on performance of a cognitive task that tests working memory capacity. These data indicate that BPA exerts an age-dependent detrimental impact on primate brain development, at blood levels within the range measured in humans having only environmental contact with BPA.
Collapse
Affiliation(s)
- John D Elsworth
- Department of Psychiatry, Yale University, School of Medicine, New Haven, CT, USA.
| | | | | | | | | | | |
Collapse
|
40
|
Neural development, a risky period. Exp Neurol 2012; 237:43-5. [DOI: 10.1016/j.expneurol.2012.05.024] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2012] [Revised: 05/24/2012] [Accepted: 05/31/2012] [Indexed: 12/13/2022]
|
41
|
Yadav S, Dixit A, Agrawal S, Singh A, Srivastava G, Singh AK, Srivastava PK, Prakash O, Singh MP. Rodent models and contemporary molecular techniques: notable feats yet incomplete explanations of Parkinson's disease pathogenesis. Mol Neurobiol 2012; 46:495-512. [PMID: 22736079 DOI: 10.1007/s12035-012-8291-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2012] [Accepted: 06/13/2012] [Indexed: 12/20/2022]
Abstract
Rodent models and molecular tools, mainly omics and RNA interference, have been rigorously used to decode the intangible etiology and pathogenesis of Parkinson's disease (PD). Although convention of contemporary molecular techniques and multiple rodent models paved imperative leads in deciphering the role of putative causative factors and sequential events leading to PD, complete and clear-cut mechanisms of pathogenesis are still hard to pin down. The current article reviews the implications and pros and cons of rodent models and molecular tools in understanding the molecular and cellular bases of PD pathogenesis based on the existing literature. Probable rationales for short of comprehensive leads and future possibilities in spite of the extensive applications of molecular tools and rodent models have also been discussed.
Collapse
Affiliation(s)
- Sharawan Yadav
- CSIR-Indian Institute of Toxicology Research, Lucknow-226 001, Uttar Pradesh, India
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Morrow BA, Roth RH, Redmond DE, Diano S, Elsworth JD. Susceptibility to a parkinsonian toxin varies during primate development. Exp Neurol 2012; 235:273-81. [PMID: 22366325 DOI: 10.1016/j.expneurol.2012.02.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2011] [Revised: 01/18/2012] [Accepted: 02/06/2012] [Indexed: 12/21/2022]
Abstract
Symptoms of Parkinson's disease typically emerge later in life when loss of nigrostriatal dopamine neuron function exceeds the threshold of compensatory mechanisms in the basal ganglia. Although nigrostriatal dopamine neurons are lost during aging, in Parkinson's disease other detrimental factors must play a role to produce greater than normal loss of these neurons. Early development has been hypothesized to be a potentially vulnerable period when environmental or genetic abnormalities may compromise central dopamine neurons. This study uses a specific parkinsonian neurotoxin, 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), to probe the relative vulnerability of nigrostriatal dopamine neurons at different stages of primate development. Measures of dopamine, homovanillic acid, 1-methyl-pyridinium concentrations and tyrosine hydroxylase immunoreactive neurons indicated that at mid-gestation dopamine neurons are relatively vulnerable to MPTP, whereas later in development or in the young primate these neurons are resistant to the neurotoxin. These studies highlight a potentially greater risk to the fetus of exposure during mid-gestation to environmental agents that cause oxidative stress. In addition, the data suggest that uncoupling protein-2 may be a target for retarding the progressive loss of nigrostriatal dopamine neurons that occurs in Parkinson's disease and aging.
Collapse
Affiliation(s)
- B A Morrow
- Neuropsychopharmacology Research Laboratory, Department of Psychiatry, Yale University School of Medicine, New Haven, CT 06511, USA
| | | | | | | | | |
Collapse
|
43
|
Kanthasamy A, Jin H, Anantharam V, Sondarva G, Rangasamy V, Rana A, Kanthasamy A. Emerging neurotoxic mechanisms in environmental factors-induced neurodegeneration. Neurotoxicology 2012; 33:833-7. [PMID: 22342404 DOI: 10.1016/j.neuro.2012.01.011] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2011] [Revised: 01/19/2012] [Accepted: 01/23/2012] [Indexed: 12/17/2022]
Abstract
Exposure to environmental neurotoxic metals, pesticides and other chemicals is increasingly recognized as a key risk factor in the pathogenesis of chronic neurodegenerative disorders such as Parkinson's and Alzheimer's diseases. Oxidative stress and apoptosis have been actively investigated as neurotoxic mechanisms over the past two decades, resulting in a greater understanding of neurotoxic processes. Nevertheless, emerging evidence indicates that epigenetic changes, protein aggregation and autophagy are important cellular and molecular correlates of neurodegenerative diseases resulting from chronic neurotoxic chemical exposure. During the Joint Conference of the 13th International Neurotoxicology Association and the 11th International Symposium on Neurobehavioral Methods and Effects in Occupational and Environmental Health, the recent progress made toward understanding epigenetic mechanisms, protein aggregation, autophagy, and deregulated kinase activation following neurotoxic chemical exposure and the relevance to neurodegenerative conditions were one of the themes of the symposium. Dr. Anumantha G. Kanthasamy described the role of acetylation of histones and non-histone proteins in neurotoxicant-induced neurodegenerative processes in the nigral dopaminergic neuronal system. Dr. Arthi Kanthasamy illustrated the role of autophagy as a key determinant in cell death events during neurotoxic insults. Dr. Ajay Rana provided evidence for posttranslational modification of α-synuclein protein by the Mixed Linage Kinase (MLK) group of kinases to initiate protein aggregation in cell culture and animal models of Parkinson's disease. These presentations outlined emerging cutting edge mechanisms that might set the stage for future mechanistic investigations into new frontiers of molecular neurotoxicology. This report summarizes the views of symposium participants, with emphasis on future directions for study of environmentally and occupationally linked chronic neurodegenerative diseases.
Collapse
Affiliation(s)
- Anumantha Kanthasamy
- Parkinson's Disorder Research Laboratory, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA 50011, USA.
| | | | | | | | | | | | | |
Collapse
|