1
|
Chauhan H, Carruthers NJ, Stemmer PM, Schneider BL, Moszczynska A. Interactions of VMAT2 with CDCrel-1 and Parkin in Methamphetamine Neurotoxicity. Int J Mol Sci 2024; 25:13070. [PMID: 39684782 DOI: 10.3390/ijms252313070] [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: 08/25/2024] [Revised: 11/05/2024] [Accepted: 11/07/2024] [Indexed: 12/18/2024] Open
Abstract
In recent years, methamphetamine (METH) misuse in the US has been rapidly increasing, and there is no FDA-approved pharmacotherapy for METH use disorder (MUD). In addition to being dependent on the drug, people with MUD develop a variety of neurological problems related to the toxicity of this drug. A variety of molecular mechanisms underlying METH neurotoxicity has been identified, including the dysfunction of the neuroprotective protein parkin. However, it is not known whether parkin loss of function within striatal dopaminergic (DAergic) terminals translates into decreased DA storage capacity. This study examined the relationship between parkin, its substrate cell division cycle related-1 (CDCrel-1) associated with synaptic vesicles, and vesicular monoamine transporter-2 (VMAT2) responsible for packaging DA in an in vivo model of METH neurotoxicity. To assess the individual differences in response to METH's neurotoxic effects, a large group of male Sprague Dawley rats were treated with binge METH or saline and sacrificed 1 h or 24 h later. This study is the first to show that CDCrel-1 interacts with VMAT2 in the rat striatum and that binge METH can alter this interaction as well as the levels and subcellular localization of CDCrel-1. The proteomic analysis of VMAT-2-associated proteins revealed the upregulation of several proteins involved in the exocytosis/endocytosis cycle and responses to stress. The results suggest that DAergic neurons are engaged in counteracting METH-induced toxic effects, including attempts to increase endocytosis and autophagy at 1 h after the METH binge, with the responses varying widely between individual rats. Studying CDCrel-1, VMAT2, and other proteins in large groups of outbred rats can help define individual genetic and molecular differences in responses to METH neurotoxicity, which, in turn, may aid treating humans suffering from MUD and its neurological consequences.
Collapse
Affiliation(s)
- Heli Chauhan
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, 259 Mack Ave., Detroit, MI 48201, USA
| | - Nicholas J Carruthers
- Institute of Environmental Health Sciences and Proteomics Core Facility, 540 East Canfield Ave., Detroit, MI 48202, USA
- Bioinformatics Core, Michigan Medicine, University of Michigan, NCRC Building 14, 2800 Plymouth Rd., Ann Arbor, MI 48109, USA
| | - Paul M Stemmer
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, 259 Mack Ave., Detroit, MI 48201, USA
- Institute of Environmental Health Sciences and Proteomics Core Facility, 540 East Canfield Ave., Detroit, MI 48202, USA
| | - Bernard L Schneider
- Bertarelli Platform for Gene Therapy, École Polytechnique Fédérale de Lausanne, School of Life Sciences, Ch. Des Mines 9, CH-1202 Geneva, Switzerland
| | - Anna Moszczynska
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, 259 Mack Ave., Detroit, MI 48201, USA
| |
Collapse
|
2
|
Chauhan H, Carruthers N, Stemmer P, Schneider BP, Moszczynska A. Neurotoxic Methamphetamine Doses Alter CDCel-1 Levels and Its Interaction with Vesicular Monoamine Transporter-2 in Rat Striatum. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.21.604458. [PMID: 39091864 PMCID: PMC11291068 DOI: 10.1101/2024.07.21.604458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/04/2024]
Abstract
In recent years, methamphetamine METH misuse in the US has been rapidly increasing and there is no FDA-approved pharmacotherapy for METH use disorder (MUD). In addition to being dependent on the drug, people with MUD develop a variety of neurological problems related to the toxicity of this drug. A variety of molecular mechanisms underlying METH neurotoxicity has been identified, including dysfunction of the neuroprotective protein parkin. However, it is not known whether parkin loss of function within striatal dopaminergic (DAergic) terminals translates into a decrease in DA storage capacity. This study examined the relationship between parkin, its substrate cell division cycle related-1 (CDCrel-1), and vesicular monoamine transporter-2 (VMAT2) in METH neurotoxicity in male Sprague Dawley rats. To also assess individual differences in response to METH's neurotoxic effects, a large group of rats was treated with binge METH or saline and sacrificed 1h or 24h later. This study is the first to show that binge METH alters the levels and subcellular localization of CDCrel-1 and that CDCrel-1 interacts with VMAT2 and increases its levels at the plasma membrane. Furthermore, we found wide individual differences in the responses of measured indices to METH. Proteomic analysis of VMAT-2-associated proteins revealed upregulation of several proteins involved in the exocytosis/endocytosis cycle. The results suggest that at 1h after METH binge, DAergic neurons are engaged in counteracting METH-induced toxic effects, including oxidative stress- and hyperthermia-induced inhibition of synaptic vesicle cycling, with the responses varying between individual rats. Studying CDCrel-1, VMAT2, and other proteins in large groups of outbred rats can help define individual genetic and molecular differences in responses to METH neurotoxicity which, in turn, will aid treating humans suffering from METH use disorder and its neurological consequences.
Collapse
Affiliation(s)
- Heli Chauhan
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, 259 Mack Ave, Detroit, MI, USA 48201
| | - Nick Carruthers
- Institute of Environmental Health Sciences and Proteomics Core Facility, 540 East Canfield Ave., Detroit, MI 48202
| | - Paul Stemmer
- Institute of Environmental Health Sciences and Proteomics Core Facility, 540 East Canfield Ave., Detroit, MI 48202
| | - Bernard P. Schneider
- Brain Mind Institute École Polytechnique Fédérale de Lausanne School of Life Sciences, Ch. Des Mines, 9, CH-1202 Geneve, Switzerland
| | - Anna Moszczynska
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, 259 Mack Ave, Detroit, MI, USA 48201
| |
Collapse
|
3
|
Bhargava S, Kulkarni R, Dewangan B, Kulkarni N, Jiaswar C, Kumar K, Kumar A, Bodhe PR, Kumar H, Sahu B. Microtubule stabilising peptides: new paradigm towards management of neuronal disorders. RSC Med Chem 2023; 14:2192-2205. [PMID: 37974959 PMCID: PMC10650357 DOI: 10.1039/d3md00012e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 08/09/2023] [Indexed: 11/19/2023] Open
Abstract
Neuronal cells made of soma, axon, and dendrites are highly compartmentalized and possess a specialized transport system that can convey long-distance electrical signals for the cross-talk. The transport system is made up of microtubule (MT) polymers and MT-binding proteins. MTs play vital and diverse roles in various cellular processes. Therefore, defects and dysregulation of MTs and their binding proteins lead to many neurological disorders as exemplified by Parkinson's disease, Alzheimer's disease, amyotrophic lateral sclerosis, Huntington's disease, and many others. MT-stabilising agents (MSAs) altering the MT-associated protein connections have shown great potential for several neurodegenerative disorders. Peptides are an important class of molecules with high specificity, biocompatibility and are devoid of side effects. In the past, peptides have been explored in various neuronal disorders as therapeutics. Davunetide, a MT-stabilising octapeptide, has entered into phase II clinical trials for schizophrenia. Numerous examples of peptides emerging as MSAs reflect the emergence of a new paradigm for peptides which can be explored further as drug candidates for neuronal disorders. Although small molecule-based MSAs have been reviewed in the past, there is no systematic review in recent years focusing on peptides as MSAs apart from davunetide in 2013. Therefore, a systematic updated review on MT stabilising peptides may shed light on many hidden aspects and enable researchers to develop new therapies for diseases related to the CNS. In this review we have summarised the recent examples of peptides as MSAs.
Collapse
Affiliation(s)
- Shubhangi Bhargava
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research Ahmedabad India
| | - Riya Kulkarni
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research Ahmedabad India
| | - Bhaskar Dewangan
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research Ahmedabad India
| | - Neeraj Kulkarni
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research Ahmedabad India
| | - Chirag Jiaswar
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research Ahmedabad India
| | - Kunal Kumar
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research Ahmedabad India
| | - Amit Kumar
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research Ahmedabad India
| | - Praveen Reddy Bodhe
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research Ahmedabad India
| | - Hemant Kumar
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research Ahmedabad India
| | - Bichismita Sahu
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research Ahmedabad India
| |
Collapse
|
4
|
Kabir F, Atkinson R, Cook AL, Phipps AJ, King AE. The role of altered protein acetylation in neurodegenerative disease. Front Aging Neurosci 2023; 14:1025473. [PMID: 36688174 PMCID: PMC9845957 DOI: 10.3389/fnagi.2022.1025473] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 11/03/2022] [Indexed: 01/06/2023] Open
Abstract
Acetylation is a key post-translational modification (PTM) involved in the regulation of both histone and non-histone proteins. It controls cellular processes such as DNA transcription, RNA modifications, proteostasis, aging, autophagy, regulation of cytoskeletal structures, and metabolism. Acetylation is essential to maintain neuronal plasticity and therefore essential for memory and learning. Homeostasis of acetylation is maintained through the activities of histone acetyltransferases (HAT) and histone deacetylase (HDAC) enzymes, with alterations to these tightly regulated processes reported in several neurodegenerative diseases including Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD), and amyotrophic lateral sclerosis (ALS). Both hyperacetylation and hypoacetylation can impair neuronal physiological homeostasis and increase the accumulation of pathophysiological proteins such as tau, α-synuclein, and Huntingtin protein implicated in AD, PD, and HD, respectively. Additionally, dysregulation of acetylation is linked to impaired axonal transport, a key pathological mechanism in ALS. This review article will discuss the physiological roles of protein acetylation and examine the current literature that describes altered protein acetylation in neurodegenerative disorders.
Collapse
|
5
|
Kulcsarova K, Bang C, Berg D, Schaeffer E. Pesticides and the Microbiome-Gut-Brain Axis: Convergent Pathways in the Pathogenesis of Parkinson's Disease. JOURNAL OF PARKINSON'S DISEASE 2023; 13:1079-1106. [PMID: 37927277 PMCID: PMC10657696 DOI: 10.3233/jpd-230206] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 10/11/2023] [Indexed: 11/07/2023]
Abstract
The increasing global burden of Parkinson's disease (PD), termed the PD pandemic, is exceeding expectations related purely to population aging and is likely driven in part by lifestyle changes and environmental factors. Pesticides are well recognized risk factors for PD, supported by both epidemiological and experimental evidence, with multiple detrimental effects beyond dopaminergic neuron damage alone. The microbiome-gut-brain axis has gained much attention in recent years and is considered to be a significant contributor and driver of PD pathogenesis. In this narrative review, we first focus on how both pesticides and the microbiome may influence PD initiation and progression independently, describing pesticide-related central and peripheral neurotoxicity and microbiome-related local and systemic effects due to dysbiosis and microbial metabolites. We then depict the bidirectional interplay between pesticides and the microbiome in the context of PD, synthesizing current knowledge about pesticide-induced dysbiosis, microbiome-mediated alterations in pesticide availability, metabolism and toxicity, and complex systemic pesticide-microbiome-host interactions related to inflammatory and metabolic pathways, insulin resistance and other mechanisms. An overview of the unknowns follows, and the role of pesticide-microbiome interactions in the proposed body-/brain-first phenotypes of PD, the complexity of environmental exposures and gene-environment interactions is discussed. The final part deals with possible further steps for translation, consisting of recommendations on future pesticide use and research as well as an outline of promising preventive/therapeutic approaches targeted on strengthening or restoring a healthy gut microbiome, closing with a summary of current gaps and future perspectives in the field.
Collapse
Affiliation(s)
- Kristina Kulcsarova
- Department of Neurology, P. J. Safarik University, Kosice, Slovak Republic
- Department of Neurology, L. Pasteur University Hospital, Kosice, Slovak Republic
- Department of Clinical Neurosciences, University Scientific Park MEDIPARK, P. J. Safarik University, Kosice, Slovak Republic
| | - Corinna Bang
- Institute of Clinical Molecular Biology, Kiel University and University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Daniela Berg
- Department of Neurology, Kiel University and University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Eva Schaeffer
- Department of Neurology, Kiel University and University Medical Center Schleswig-Holstein, Kiel, Germany
| |
Collapse
|
6
|
Shi X, Jiang X, Chen C, Zhang Y, Sun X. The interconnections between the microtubules and mitochondrial networks in cardiocerebrovascular diseases: Implications for therapy. Pharmacol Res 2022; 184:106452. [PMID: 36116706 DOI: 10.1016/j.phrs.2022.106452] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 09/13/2022] [Accepted: 09/13/2022] [Indexed: 10/14/2022]
Abstract
Microtubules, a highly dynamic cytoskeleton, participate in many cellular activities including mechanical support, organelles interactions, and intracellular trafficking. Microtubule organization can be regulated by modification of tubulin subunits, microtubule-associated proteins (MAPs) or agents modulating microtubule assembly. Increasing studies demonstrate that microtubule disorganization correlates with various cardiocerebrovascular diseases including heart failure and ischemic stroke. Microtubules also mediate intracellular transport as well as intercellular transfer of mitochondria, a power house in cells which produce ATP for various physiological activities such as cardiac mechanical function. It is known to all that both microtubules and mitochondria participate in the progression of cancer and Parkinson's disease. However, the interconnections between the microtubules and mitochondrial networks in cardiocerebrovascular diseases remain unclear. In this paper, we will focus on the roles of microtubules in cardiocerebrovascular diseases, and discuss the interplay of mitochondria and microtubules in disease development and treatment. Elucidation of these issues might provide significant diagnostic value as well as potential targets for cardiocerebrovascular diseases.
Collapse
Affiliation(s)
- Xingjuan Shi
- School of Life Science and Technology, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, China.
| | - Xuan Jiang
- School of Life Science and Technology, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, China
| | - Congwei Chen
- School of Life Science and Technology, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, China
| | - Yu Zhang
- School of Life Science and Technology, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, China
| | - Xiaoou Sun
- Institute of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, China.
| |
Collapse
|
7
|
Zadeh-Haghighi H, Simon C. Radical pairs may play a role in microtubule reorganization. Sci Rep 2022; 12:6109. [PMID: 35414166 PMCID: PMC9005667 DOI: 10.1038/s41598-022-10068-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 04/01/2022] [Indexed: 12/14/2022] Open
Abstract
The exact mechanism behind general anesthesia remains an open question in neuroscience. It has been proposed that anesthetics selectively prevent consciousness and memory via acting on microtubules (MTs). It is known that the magnetic field modulates MT organization. A recent study shows that a radical pair model can explain the isotope effect in xenon-induced anesthesia and predicts magnetic field effects on anesthetic potency. Further, reactive oxygen species are also implicated in MT stability and anesthesia. Based on a simple radical pair mechanism model and a simple mathematical model of MT organization, we show that magnetic fields can modulate spin dynamics of naturally occurring radical pairs in MT. We propose that the spin dynamics influence a rate in the reaction cycle, which translates into a change in the MT density. We can reproduce magnetic field effects on the MT concentration that have been observed. Our model also predicts additional effects at slightly higher fields. Our model further predicts that the effect of zinc on the MT density exhibits isotopic dependence. The findings of this work make a connection between microtubule-based and radical pair-based quantum theories of consciousness.
Collapse
Affiliation(s)
- Hadi Zadeh-Haghighi
- Department of Physics and Astronomy, University of Calgary, Calgary, AB, T2N 1N4, Canada.
- Institute for Quantum Science and Technology, University of Calgary, Calgary, AB, T2N 1N4, Canada.
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, T2N 1N4, Canada.
| | - Christoph Simon
- Department of Physics and Astronomy, University of Calgary, Calgary, AB, T2N 1N4, Canada.
- Institute for Quantum Science and Technology, University of Calgary, Calgary, AB, T2N 1N4, Canada.
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, T2N 1N4, Canada.
| |
Collapse
|
8
|
Identifying the Effects of Reactive Oxygen Species on Mitochondrial Dynamics and Cytoskeleton Stability in Dictyostelium discoideum. Cells 2021; 10:cells10082147. [PMID: 34440916 PMCID: PMC8392674 DOI: 10.3390/cells10082147] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 08/12/2021] [Accepted: 08/16/2021] [Indexed: 12/14/2022] Open
Abstract
Defects in mitochondrial dynamics, fission, fusion, and motility have been implicated in the pathogenesis of multiple neurodegenerative diseases, including Parkinson's disease, Alzheimer's disease, Huntington's disease, and Charcot-Marie-Tooth disease. Another key feature of neurodegeneration is the increase in reactive oxygen species (ROS). Previous work has shown that the cytoskeleton, in particular the microtubules, and ROS generated by rotenone significantly regulate mitochondrial dynamics in Dictyostelium discoideum. The goal of this project is to study the effects of ROS on mitochondrial dynamics within our model organism D. discoideum to further understand the underlying issues that are the root of neurodegenerative diseases such as Alzheimer's disease and Parkinson's disease. We chose three likely ROS inducers, cumene hydroperoxide, hydroxylamine hydrochloride, and Antimycin A. Our work demonstrates that alteration of the microtubule cytoskeleton is not required to alter dynamics in response to ROS and there is no easy way to predict how mitochondrial dynamics will be altered based on which ROS generator is used. This research contributes to the better understanding of the cellular mechanisms that induce the pathogenesis of incurable neurodegenerative diseases with the hope that it will translate into developing new and more effective treatments for patients afflicted by them.
Collapse
|
9
|
Yang Y, Chen X, Feng Z, Cai X, Zhu X, Cao M, Yang L, Chen Y, Wang Y, Feng H. MEC17-induced α-tubulin acetylation restores mitochondrial transport function and alleviates axonal injury after intracerebral hemorrhage in mice. J Neurochem 2021; 160:51-63. [PMID: 34407220 DOI: 10.1111/jnc.15493] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 07/25/2021] [Accepted: 08/09/2021] [Indexed: 02/01/2023]
Abstract
Injury to long axonal projections is a central pathological feature at the early phase of intracerebral hemorrhage (ICH). It has been reported to contribute to persistent functional disability following ICH. However, the molecular mechanisms that drive axonal degeneration remain unclear. Autologous blood was injected into the striatum to mimic the pathology of ICH. Observed significant swollen axons with characteristic retraction bulbs were found around the striatal hematoma at 24 h after ICH. Electronic microscopic examination revealed highly disorganized microtubule and swollen mitochondria in the retraction bulbs. MEC17 is a specific α-tubulin acetyltransferase, ablation of acetylated α-tubulin in MEC17-/- mice aggravated axonal injury, axonal transport mitochondria dysfunction, and motor dysfunction. In contrast, treatment with tubastatin A (TubA), which promotes microtubule acetylation, significantly alleviated axonal injury and protected the integrity of the corticospinal tract and fine motor function after ICH. Moreover, results showed that 41% mitochondria were preferentially bundled to the acetylated α-tubulin in identifiable axons and dendrites in primary neurons. This impaired axonal transport of mitochondria in primary neurons of MEC17-/- mice. Given that opening of mitochondrial permeability transition pore (mPTP) induces mitochondrial dysfunction and impairs ATP supply thereby promoting axonal injury, we enhanced the availability of acetylated α-tubulin using TubA and inhibited mPTP opening with cyclosporin A. The results indicated that this combined treatment synergistically protected corticospinal tract integrity and promoted fine motor control recovery. These findings reveal key intracellular mechanisms that drive axonal degeneration after ICH and highlight the need to target multiple factors and respective regulatory mechanisms as an effective approach to prevent axonal degeneration and motor dysfunction after ICH.
Collapse
Affiliation(s)
- Yang Yang
- Department of Neurosurgery, The 904th Hospital of PLA, School of Medicine of Anhui Medical University, Wuxi, Jiangsu Province, China.,Department of Neurosurgery and State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University, Chongqing, China.,Chongqing Key Laboratory of Precision Neuromedicine and Neuroregenaration, Third Military Medical University (Army Medical University, Chongqing, China.,Wuxi Translational Medicine Center, Wuxi, Jiangsu, China
| | - Xuezhu Chen
- Department of Neurosurgery and State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University, Chongqing, China.,Chongqing Key Laboratory of Precision Neuromedicine and Neuroregenaration, Third Military Medical University (Army Medical University, Chongqing, China
| | - Zhizhong Feng
- Department of Neurosurgery, The 904th Hospital of PLA, School of Medicine of Anhui Medical University, Wuxi, Jiangsu Province, China
| | - Xianfeng Cai
- Department of Neurosurgery, The 904th Hospital of PLA, School of Medicine of Anhui Medical University, Wuxi, Jiangsu Province, China
| | - Xiaoming Zhu
- Department of Neurosurgery, The 904th Hospital of PLA, School of Medicine of Anhui Medical University, Wuxi, Jiangsu Province, China
| | - Ming Cao
- Department of Neurosurgery, The 904th Hospital of PLA, School of Medicine of Anhui Medical University, Wuxi, Jiangsu Province, China
| | - Likun Yang
- Department of Neurosurgery, The 904th Hospital of PLA, School of Medicine of Anhui Medical University, Wuxi, Jiangsu Province, China.,Wuxi Translational Medicine Center, Wuxi, Jiangsu, China
| | - Yujie Chen
- Department of Neurosurgery and State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University, Chongqing, China.,Chongqing Key Laboratory of Precision Neuromedicine and Neuroregenaration, Third Military Medical University (Army Medical University, Chongqing, China
| | - Yuhai Wang
- Department of Neurosurgery, The 904th Hospital of PLA, School of Medicine of Anhui Medical University, Wuxi, Jiangsu Province, China.,Wuxi Translational Medicine Center, Wuxi, Jiangsu, China
| | - Hua Feng
- Department of Neurosurgery and State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University, Chongqing, China.,Chongqing Key Laboratory of Precision Neuromedicine and Neuroregenaration, Third Military Medical University (Army Medical University, Chongqing, China
| |
Collapse
|
10
|
Sim TM, Tarini D, Dheen ST, Bay BH, Srinivasan DK. Nanoparticle-Based Technology Approaches to the Management of Neurological Disorders. Int J Mol Sci 2020; 21:E6070. [PMID: 32842530 PMCID: PMC7503838 DOI: 10.3390/ijms21176070] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Revised: 08/09/2020] [Accepted: 08/21/2020] [Indexed: 12/15/2022] Open
Abstract
Neurological disorders are the most devastating and challenging diseases associated with the central nervous system (CNS). The blood-brain barrier (BBB) maintains homeostasis of the brain and contributes towards the maintenance of a very delicate microenvironment, impairing the transport of many therapeutics into the CNS and making the management of common neurological disorders such as Alzheimer's disease (AD), Parkinson's disease (PD), cerebrovascular diseases (CVDs) and traumatic brain injury (TBI), exceptionally complicated. Nanoparticle (NP) technology offers a platform for the design of tissue-specific drug carrying systems owing to its versatile and modifiable nature. The prospect of being able to design NPs capable of successfully crossing the BBB, and maintaining a high drug bioavailability in neural parenchyma, has spurred much interest in the field of nanomedicine. NPs, which also come in an array of forms including polymeric NPs, solid lipid nanoparticles (SLNs), quantum dots and liposomes, have the flexibility of being conjugated with various macromolecules, such as surfactants to confer the physical or chemical property desired. These nanodelivery strategies represent potential novel and minimally invasive approaches to the treatment and diagnosis of these neurological disorders. Most of the strategies revolve around the ability of the NPs to cross the BBB via various influx mechanisms, such as adsorptive-mediated transcytosis (AMT) and receptor-mediated transcytosis (RMT), targeting specific biomarkers or lesions unique to that pathological condition, thereby ensuring high tissue-specific targeting and minimizing off-target side effects. In this article, insights into common neurological disorders and challenges of delivering CNS drugs due to the presence of BBB is provided, before an in-depth review of nanoparticle-based theranostic strategies.
Collapse
Affiliation(s)
- Tao Ming Sim
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore;
| | - Dinesh Tarini
- Government Kilpauk Medical College, The Tamilnadu Dr MGR Medical University, Chennai, Tamilnadu 600032, India;
| | - S. Thameem Dheen
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117594, Singapore; (S.T.D.); (B.H.B.)
| | - Boon Huat Bay
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117594, Singapore; (S.T.D.); (B.H.B.)
| | - Dinesh Kumar Srinivasan
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117594, Singapore; (S.T.D.); (B.H.B.)
| |
Collapse
|
11
|
Calogero AM, Mazzetti S, Pezzoli G, Cappelletti G. Neuronal microtubules and proteins linked to Parkinson's disease: a relevant interaction? Biol Chem 2020; 400:1099-1112. [PMID: 31256059 DOI: 10.1515/hsz-2019-0142] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 06/24/2019] [Indexed: 12/13/2022]
Abstract
Neuronal microtubules are key determinants of cell morphology, differentiation, migration and polarity, and contribute to intracellular trafficking along axons and dendrites. Microtubules are strictly regulated and alterations in their dynamics can lead to catastrophic effects in the neuron. Indeed, the importance of the microtubule cytoskeleton in many human diseases is emerging. Remarkably, a growing body of evidence indicates that microtubule defects could be linked to Parkinson's disease pathogenesis. Only a few of the causes of the progressive neuronal loss underlying this disorder have been identified. They include gene mutations and toxin exposure, but the trigger leading to neurodegeneration is still unknown. In this scenario, the evidence showing that mutated proteins in Parkinson's disease are involved in the regulation of the microtubule cytoskeleton is intriguing. Here, we focus on α-Synuclein, Parkin and Leucine-rich repeat kinase 2 (LRRK2), the three main proteins linked to the familial forms of the disease. The aim is to dissect their interaction with tubulin and microtubules in both physiological and pathological conditions, in which these proteins are overexpressed, mutated or absent. We highlight the relevance of such an interaction and suggest that these proteins could trigger neurodegeneration via defective regulation of the microtubule cytoskeleton.
Collapse
Affiliation(s)
- Alessandra M Calogero
- Department of Biosciences, Università degli Studi di Milano, via Celoria 26, I-20133 Milan, Italy
| | - Samanta Mazzetti
- Department of Biosciences, Università degli Studi di Milano, via Celoria 26, I-20133 Milan, Italy.,Fondazione Grigioni per il Morbo di Parkinson, via Zuretti 35, I-20135 Milan, Italy
| | - Gianni Pezzoli
- Fondazione Grigioni per il Morbo di Parkinson, via Zuretti 35, I-20135 Milan, Italy.,Parkinson Institute, ASST "G.Pini-CTO", via Bignami 1, I-20133 Milan, Italy
| | - Graziella Cappelletti
- Department of Biosciences, Università degli Studi di Milano, via Celoria 26, I-20133 Milan, Italy.,Center of Excellence on Neurodegenerative Diseases, Università degli Studi di Milano, via Balzaretti, I-20133 Milan, Italy
| |
Collapse
|
12
|
Li H, Jiang H, Zhang B, Feng J. Modeling Parkinson's Disease Using Patient-specific Induced Pluripotent Stem Cells. JOURNAL OF PARKINSONS DISEASE 2019; 8:479-493. [PMID: 30149462 PMCID: PMC6218140 DOI: 10.3233/jpd-181353] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Parkinson’s disease (PD) is the second most common neurodegenerative disorder. It is characterized by the degeneration of nigral dopaminergic (DA) neurons. While over 90% of cases are idiopathic, without a clear etiology, mutations in many genes have been linked to rare, familial forms of PD. It has been quite challenging to develop effective animal models of PD that capture salient features of PD. The discovery of induced pluripotent stem cells (iPSCs) makes it possible to generate patient-specific DA neurons to study PD. Here, we review the methods for the generation of iPSCs and discuss previous studies using iPSC-derived neurons from monogenic forms of PD. These investigations have revealed several converging pathways that intersect with the unique vulnerabilities of human nigral DA neurons. With the rapid development in stem cell biology, it is possible to generate patient-specific neurons that will be increasingly similar to those in the brain of the patient. Combined with the ability to edit the genome to generate isogenic iPSCs, the generation and analysis of patient-specific midbrain DA neurons will transform PD research by providing a valuable tool for mechanistic study and drug discovery.
Collapse
Affiliation(s)
- Hong Li
- Veterans Affairs Western New York Healthcare System, Buffalo, NY, USA.,Department of Physiology and Biophysics, State University of New York at Buffalo, Buffalo, NY, USA
| | - Houbo Jiang
- Veterans Affairs Western New York Healthcare System, Buffalo, NY, USA.,Department of Physiology and Biophysics, State University of New York at Buffalo, Buffalo, NY, USA
| | - Boyang Zhang
- Veterans Affairs Western New York Healthcare System, Buffalo, NY, USA.,Department of Physiology and Biophysics, State University of New York at Buffalo, Buffalo, NY, USA
| | - Jian Feng
- Veterans Affairs Western New York Healthcare System, Buffalo, NY, USA.,Department of Physiology and Biophysics, State University of New York at Buffalo, Buffalo, NY, USA
| |
Collapse
|
13
|
Chernivec E, Cooper J, Naylor K. Exploring the Effect of Rotenone-A Known Inducer of Parkinson's Disease-On Mitochondrial Dynamics in Dictyostelium discoideum. Cells 2018; 7:E201. [PMID: 30413037 PMCID: PMC6262481 DOI: 10.3390/cells7110201] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 10/31/2018] [Accepted: 11/06/2018] [Indexed: 02/07/2023] Open
Abstract
Current treatments for Parkinson's disease (PD) only alleviate symptoms doing little to inhibit the onset and progression of the disease, thus we must research the mechanism of Parkinson's. Rotenone is a known inducer of parkinsonian conditions in rats; we use rotenone to induce parkinsonian cellular conditions in Dictyostelium discoideum. In our model we primarily focus on mitochondrial dynamics. We found that rotenone disrupts the actin and microtubule cytoskeleton but mitochondrial morphology remains intact. Rotenone stimulates mitochondrial velocity while inhibiting mitochondrial fusion, increases reactive oxygen species (ROS) but has no effect on ATP levels. Antioxidants have been shown to decrease some PD symptoms thus we added ascorbic acid to our rotenone treated cells. Ascorbic acid administration suggests that rotenone effects may be specific to the disruption of the cytoskeleton rather than the increase in ROS. Our results imply that D. discoideum may be a valid cellular PD model and that the rotenone induced velocity increase and loss of fusion could prevent mitochondria from effectively providing energy and other mitochondrial products in high demand areas. The combination of these defects in mitochondrial dynamics and increased ROS could result in degeneration of neurons in PD.
Collapse
Affiliation(s)
- Ethan Chernivec
- Department of Biology, University of Central Arkansas, Conway, AR 72035, USA.
| | - Jacie Cooper
- Department of Biology, University of Central Arkansas, Conway, AR 72035, USA.
| | - Kari Naylor
- Department of Biology, University of Central Arkansas, Conway, AR 72035, USA.
| |
Collapse
|
14
|
Yang Y, Zhang K, Zhong J, Wang J, Yu Z, Lei X, Chen X, Quan Y, Xian J, Chen Y, Liu X, Feng H, Tan L. Stably maintained microtubules protect dopamine neurons and alleviate depression-like behavior after intracerebral hemorrhage. Sci Rep 2018; 8:12647. [PMID: 30140021 PMCID: PMC6107628 DOI: 10.1038/s41598-018-31056-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 08/08/2018] [Indexed: 11/25/2022] Open
Abstract
Mesolimbic dopamine (DA) system lesion plays a key role in the pathophysiology of depression, and our previous study demonstrated that reduced microtubule (MT) stability aggravated nigrostriatal pathway impairment after intracerebral hemorrhage (ICH). This study aimed to further investigate the occurrence regularity of depression-like behavior after ICH and determine whether maintaining MT stabilization could protect DA neurons in ventral tegmental area (VTA) and alleviate depression-like behavior after ICH. An intrastriatal injection of 20 μl of autologous blood or MT depolymerization reagent nocodazole (Noco) was used to mimic the pathology of ICH model in mice. The concentration of DA, number of DA neurons and acetylated α-tubulin (a marker for stable MT) in VTA were checked, and depression-related behavior tests were performed after ICH. A MT-stabilizing agent, epothilone B (EpoB), was administered to explore the effects of MT stabilization on DA neurons and depression-like behavior after ICH. The results showed that obvious depression-like behavior occurred at 7, 14, and 28 days (P < 0.01) after ICH. These time-points were related to significant decreases in the concentration of DA (P < 0.01) and number of DA neurons (P < 0.01) in VTA. Moreover, The decrease of acetylated α-tubulin expression after ICH and Noco injection contributed to DA neurons' impairment in VTA, and Noco injecton also aggravate ICH-induced depression-like behaviors and DA neurons' injury. Furthermore, EpoB treatment significantly ameliorated ICH and Noco-induced depression-like behaviors (P < 0.05) and increased the concentration of DA (P < 0.05) and number of DA neurons (P < 0.05) in VTA by increasing the level of acetylated α-tubulin. The results indicate that EpoB can protect DA neurons by enhancing MT stability, and alleviate post-ICH depressive behaviors. This MT-targeted therapeutic strategy shows promise as a bench-to-bedside translational method for treating depression after ICH.
Collapse
Affiliation(s)
- Yang Yang
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 29 Gaotanyan Street, 400038, China
| | - Kaiyuan Zhang
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 29 Gaotanyan Street, 400038, China
| | - Jun Zhong
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 29 Gaotanyan Street, 400038, China
| | - Ju Wang
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 29 Gaotanyan Street, 400038, China
| | - Zhongyuan Yu
- Battalion 3 of Cadet Brigade, Third Military Medical University (Army Medical University), Chongqing, 29 Gaotanyan Street, 400038, China
| | - Xuejiao Lei
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 29 Gaotanyan Street, 400038, China
| | - Xuezhu Chen
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 29 Gaotanyan Street, 400038, China
| | - Yulian Quan
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 29 Gaotanyan Street, 400038, China
| | - Jishu Xian
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 29 Gaotanyan Street, 400038, China
| | - Yujie Chen
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 29 Gaotanyan Street, 400038, China
| | - Xin Liu
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 29 Gaotanyan Street, 400038, China
| | - Hua Feng
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 29 Gaotanyan Street, 400038, China.
| | - Liang Tan
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 29 Gaotanyan Street, 400038, China.
| |
Collapse
|
15
|
Harris G, Eschment M, Orozco SP, McCaffery JM, Maclennan R, Severin D, Leist M, Kleensang A, Pamies D, Maertens A, Hogberg HT, Freeman D, Kirkwood A, Hartung T, Smirnova L. Toxicity, recovery, and resilience in a 3D dopaminergic neuronal in vitro model exposed to rotenone. Arch Toxicol 2018; 92:2587-2606. [PMID: 29955902 PMCID: PMC6063347 DOI: 10.1007/s00204-018-2250-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2018] [Accepted: 06/20/2018] [Indexed: 02/06/2023]
Abstract
To date, most in vitro toxicity testing has focused on acute effects of compounds at high concentrations. This testing strategy does not reflect real-life exposures, which might contribute to long-term disease outcome. We used a 3D-human dopaminergic in vitro LUHMES cell line model to determine whether effects of short-term rotenone exposure (100 nM, 24 h) are permanent or reversible. A decrease in complex I activity, ATP, mitochondrial diameter, and neurite outgrowth were observed acutely. After compound removal, complex I activity was still inhibited; however, ATP levels were increased, cells were electrically active and aggregates restored neurite outgrowth integrity and mitochondrial morphology. We identified significant transcriptomic changes after 24 h which were not present 7 days after wash-out. Our results suggest that testing short-term exposures in vitro may capture many acute effects which cells can overcome, missing adaptive processes, and long-term mechanisms. In addition, to study cellular resilience, cells were re-exposed to rotenone after wash-out and recovery period. Pre-exposed cells maintained higher metabolic activity than controls and presented a different expression pattern in genes previously shown to be altered by rotenone. NEF2L2, ATF4, and EAAC1 were downregulated upon single hit on day 14, but unchanged in pre-exposed aggregates. DAT and CASP3 were only altered after re-exposure to rotenone, while TYMS and MLF1IP were downregulated in both single-exposed and pre-exposed aggregates. In summary, our study shows that a human cell-based 3D model can be used to assess cellular adaptation, resilience, and long-term mechanisms relevant to neurodegenerative research.
Collapse
Affiliation(s)
- Georgina Harris
- Center for Alternatives to Animal Testing (CAAT), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Melanie Eschment
- Center for Alternatives to Animal Testing (CAAT) Europe, Department of Biology, University of Konstanz, Konstanz, Germany
| | - Sebastian Perez Orozco
- The Integrated Imaging Center, Department of Biology, Engineering in Oncology Center and The Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, USA
| | - J Michael McCaffery
- The Integrated Imaging Center, Department of Biology, Engineering in Oncology Center and The Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, USA
| | | | - Daniel Severin
- The Mind/Brain Institute, Johns Hopkins University, Baltimore, MD, USA
| | - Marcel Leist
- Center for Alternatives to Animal Testing (CAAT) Europe, Department of Biology, University of Konstanz, Konstanz, Germany
| | - Andre Kleensang
- Center for Alternatives to Animal Testing (CAAT), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - David Pamies
- Center for Alternatives to Animal Testing (CAAT), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Alexandra Maertens
- Center for Alternatives to Animal Testing (CAAT), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Helena T Hogberg
- Center for Alternatives to Animal Testing (CAAT), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Dana Freeman
- Department of Environmental Health and Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Alfredo Kirkwood
- The Mind/Brain Institute, Johns Hopkins University, Baltimore, MD, USA.,Department of Neuroscience, Johns Hopkins University, Baltimore, MD, USA
| | - Thomas Hartung
- Center for Alternatives to Animal Testing (CAAT), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA.,Center for Alternatives to Animal Testing (CAAT) Europe, Department of Biology, University of Konstanz, Konstanz, Germany
| | - Lena Smirnova
- Center for Alternatives to Animal Testing (CAAT), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA.
| |
Collapse
|
16
|
Stephenson SEM, Aumann TD, Taylor JM, Riseley JR, Li R, Mann JR, Tomas D, Lockhart PJ. Generation and characterisation of a parkin-Pacrg knockout mouse line and a Pacrg knockout mouse line. Sci Rep 2018; 8:7528. [PMID: 29760428 PMCID: PMC5951884 DOI: 10.1038/s41598-018-25766-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Accepted: 04/25/2018] [Indexed: 11/24/2022] Open
Abstract
Mutations in PARK2 (parkin) can result in Parkinson's disease (PD). Parkin shares a bidirectional promoter with parkin coregulated gene (PACRG) and the transcriptional start sites are separated by only ~200 bp. Bidirectionally regulated genes have been shown to function in common biological pathways. Mice lacking parkin have largely failed to recapitulate the dopaminergic neuronal loss and movement impairments seen in individuals with parkin-mediated PD. We aimed to investigate the function of PACRG and test the hypothesis that parkin and PACRG function in a common pathway by generating and characterizing two novel knockout mouse lines harbouring loss of both parkin and Pacrg or Pacrg alone. Successful modification of the targeted allele was confirmed at the genomic, transcriptional and steady state protein levels for both genes. At 18-20 months of age, there were no significant differences in the behaviour of parental and mutant lines when assessed by openfield, rotarod and balance beam. Subsequent neuropathological examination suggested there was no gross abnormality of the dopaminergic system in the substantia nigra and no significant difference in the number of dopaminergic neurons in either knockout model compared to wildtype mice.
Collapse
Affiliation(s)
- Sarah E M Stephenson
- Bruce Lefroy Centre for Genetic Health Research, Murdoch Children's Research Institute, Flemington Road, Parkville, Victoria, Australia
- Department of Paediatrics, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, Victoria, Australia
| | - Timothy D Aumann
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia
| | - Juliet M Taylor
- Department of Pharmacology and Therapeutics, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, Victoria, Australia
| | - Jessica R Riseley
- Bruce Lefroy Centre for Genetic Health Research, Murdoch Children's Research Institute, Flemington Road, Parkville, Victoria, Australia
| | - Ruili Li
- Department of Paediatrics, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, Victoria, Australia
- Surgical Research, Murdoch Children's Research Institute, Flemington Road, Parkville, Victoria, Australia
| | - Jeffrey R Mann
- Monash Genome Modification Platform, Monash University, Clayton, Victoria, Australia
| | - Doris Tomas
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia
| | - Paul J Lockhart
- Bruce Lefroy Centre for Genetic Health Research, Murdoch Children's Research Institute, Flemington Road, Parkville, Victoria, Australia.
- Department of Paediatrics, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, Victoria, Australia.
| |
Collapse
|
17
|
Yang Y, Zhang X, Ge H, Liu W, Sun E, Ma Y, Zhao H, Li R, Chen W, Yuan J, Chen Q, Chen Y, Liu X, Zhang JH, Hu R, Fan X, Feng H. Epothilone B Benefits Nigrostriatal Pathway Recovery by Promoting Microtubule Stabilization After Intracerebral Hemorrhage. J Am Heart Assoc 2018; 7:JAHA.117.007626. [PMID: 29348323 PMCID: PMC5850167 DOI: 10.1161/jaha.117.007626] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Background Many previous clinical studies have demonstrated that the nigrostriatal pathway, which plays a vital role in movement adjustment, is significantly impaired after stroke, according to medical imaging and autopsies. However, the basic pathomorphological changes have been poorly investigated to date. This study was designed to explore the pathomorphological changes, mechanism, and therapeutic method of nigrostriatal impairment after intracerebral hemorrhage (ICH). Methods and Results Intrastriatal injection of autologous blood or microtubule depolymerization reagent nocodazole was performed to mimic the pathology of ICH in C57/BL6 mice. Immunofluorescence, Western blotting, electron microscopy, functional behavioral tests, and anterograde and retrograde neural circuit tracking techniques were used in these mice. The data showed that the number of dopamine neurons and the dopamine concentration were severely decreased and that fine motor function was impaired after ICH. Microtubule depolymerization was the main contributor to the loss of dopamine neurons and to motor function deficits after ICH, as was also proven by intrastriatal injection of nocodazole. Moreover, administration of the microtubule stabilizer epothilone B (1.5 mg/kg) improved the integrity of the nigrostriatal pathway neural circuit, increased the number of dopamine neurons (4598±896 versus 3125±355; P=0.034) and the dopamine concentration (4.28±0.99 versus 3.08±0.75 ng/mg; P=0.041), and enhanced fine motor functional recovery associated with increased acetylated α‐tubulin expression to maintain microtubule stabilization after ICH. Conclusions Our results clarified the pathomorphological changes of the nigrostriatal pathway after ICH and found that epothilone B helped alleviate nigrostriatal pathway injury after ICH, associated with promoting α‐tubulin acetylation to maintain microtubule stabilization, thus facilitating motor recovery.
Collapse
Affiliation(s)
- Yang Yang
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Xuan Zhang
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Hongfei Ge
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Wei Liu
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Eryi Sun
- Neurosurgery Department of Guizhou, Medical University Affiliated Hospital, Guiyang, Guizhou, China
| | - Yuanyuan Ma
- Department of Basic Nursing, School of Nursing, Third Military Medical University, Chongqing, China
| | - Hengli Zhao
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Rongwei Li
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Weixiang Chen
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Jichao Yuan
- Department of Neurology, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Qianwei Chen
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Yujie Chen
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Xin Liu
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - John H Zhang
- Department of Anesthesiology, Neurosurgery and Physiology, Loma Linda University, Loma Linda, CA
| | - Rong Hu
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Xiaotang Fan
- Department of Developmental Neuropsychology, School of Psychology, Third Military Medical University, Chongqing, China
| | - Hua Feng
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, Chongqing, China
| |
Collapse
|
18
|
Chuang YH, Paul KC, Bronstein JM, Bordelon Y, Horvath S, Ritz B. Parkinson's disease is associated with DNA methylation levels in human blood and saliva. Genome Med 2017; 9:76. [PMID: 28851441 PMCID: PMC5576382 DOI: 10.1186/s13073-017-0466-5] [Citation(s) in RCA: 110] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Accepted: 08/08/2017] [Indexed: 12/21/2022] Open
Abstract
Background Several articles suggest that DNA methylation levels in blood relate to Parkinson’s disease (PD) but there is a need for a large-scale study that involves suitable population based controls. The purposes of the study were: (1) to study whether PD status is associated with DNA methylation levels in blood/saliva; (2) to study whether observed associations relate to blood cell types; and (3) to characterize genome-wide significant markers (“CpGs”) and clusters of CpGs (co-methylation modules) in terms of biological pathways. Methods In a population-based case control study of PD, we studied blood samples from 335 PD cases and 237 controls and saliva samples from another 128 cases and 131 controls. DNA methylation data were generated from over 486,000 CpGs using the Illumina Infinium array. We identified modules of CpGs (clusters) using weighted correlation network analysis (WGCNA). Results Our cross-sectional analysis of blood identified 82 genome-wide significant CpGs (including cg02489202 in LARS2 p = 8.3 × 10–11 and cg04772575 in ABCB9 p = 4.3 × 10–10). Three out of six PD related co-methylation modules in blood were significantly enriched with immune system related genes. Our analysis of saliva identified five significant CpGs. PD-related CpGs are located near genes that relate to mitochondrial function, neuronal projection, cytoskeleton organization, systemic immune response, and iron handling. Conclusions This study demonstrates that: (1) PD status has a profound association with DNA methylation levels in blood and saliva; and (2) the most significant PD-related changes reflect changes in blood cell composition. Overall, this study highlights the role of the immune system in PD etiology but future research will need to address the causal structure of these relationships. Electronic supplementary material The online version of this article (doi:10.1186/s13073-017-0466-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yu-Hsuan Chuang
- Department of Epidemiology, Fielding School of Public Health, University of California Los Angeles, 650 Charles E. Young Drive, Box 951772, Los Angeles, CA, 90095-1772, USA
| | - Kimberly C Paul
- Department of Epidemiology, Fielding School of Public Health, University of California Los Angeles, 650 Charles E. Young Drive, Box 951772, Los Angeles, CA, 90095-1772, USA
| | - Jeff M Bronstein
- Department of Neurology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Yvette Bordelon
- Department of Neurology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Steve Horvath
- Department of Human Genetics, David Geffen School of Medicine, University of California Los Angeles, 695 Charles E. Young Drive South, Box 708822, Los Angeles, CA, 90095-7088, USA. .,Department of Biostatistics, Fielding School of Public Health, University of California Los Angeles, Los Angeles, CA, 90095, USA.
| | - Beate Ritz
- Department of Epidemiology, Fielding School of Public Health, University of California Los Angeles, 650 Charles E. Young Drive, Box 951772, Los Angeles, CA, 90095-1772, USA. .,Department of Neurology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, 90095, USA. .,Department of Environmental Health, Fielding School of Public Health, University of California Los Angeles, Los Angeles, CA, 90095, USA.
| |
Collapse
|
19
|
Moffat JJ, Ka M, Jung EM, Smith AL, Kim WY. The role of MACF1 in nervous system development and maintenance. Semin Cell Dev Biol 2017; 69:9-17. [PMID: 28579452 DOI: 10.1016/j.semcdb.2017.05.020] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Revised: 05/12/2017] [Accepted: 05/29/2017] [Indexed: 12/14/2022]
Abstract
Microtubule-actin crosslinking factor 1 (MACF1), also known as actin crosslinking factor 7 (ACF7), is essential for proper modulation of actin and microtubule cytoskeletal networks. Most MACF1 isoforms are expressed broadly in the body, but some are exclusively found in the nervous system. Consequentially, MACF1 is integrally involved in multiple neural processes during development and in adulthood, including neurite outgrowth and neuronal migration. Furthermore, MACF1 participates in several signaling pathways, including the Wnt/β-catenin and GSK-3 signaling pathways, which regulate key cellular processes, such as proliferation and cell migration. Genetic mutation or dysregulation of the MACF1 gene has been associated with neurodevelopmental and neurodegenerative diseases, specifically schizophrenia and Parkinson's disease. MACF1 may also play a part in neuromuscular disorders and have a neuroprotective role in the optic nerve. In this review, the authors seek to synthesize recent findings relating to the roles of MACF1 within the nervous system and explore potential novel functions of MACF1 not yet examined.
Collapse
Affiliation(s)
- Jeffrey J Moffat
- Department of Developmental Neuroscience, Munroe-Meyer Institute, University of Nebraska Medical Center, Omaha, NE, USA.
| | - Minhan Ka
- Department of Developmental Neuroscience, Munroe-Meyer Institute, University of Nebraska Medical Center, Omaha, NE, USA.
| | - Eui-Man Jung
- Department of Developmental Neuroscience, Munroe-Meyer Institute, University of Nebraska Medical Center, Omaha, NE, USA.
| | - Amanda L Smith
- Department of Developmental Neuroscience, Munroe-Meyer Institute, University of Nebraska Medical Center, Omaha, NE, USA.
| | - Woo-Yang Kim
- Department of Developmental Neuroscience, Munroe-Meyer Institute, University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
20
|
Louis ED, Hernandez N, Chen KP, Naranjo KV, Park J, Clark LN, Ottman R. Action Tremor Asymmetry Profile Does Not Aggregate in Families with Essential Tremor. Front Neurol 2017; 8:148. [PMID: 28469594 PMCID: PMC5395646 DOI: 10.3389/fneur.2017.00148] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Accepted: 03/31/2017] [Indexed: 12/18/2022] Open
Abstract
Background Action tremor is the hallmark feature of essential tremor (ET). While the tremor typically is mildly asymmetric, in some patients, it is markedly asymmetric. There are few data on factors that influence this asymmetry. ET is also a highly familial disease. Whether the tremor asymmetry profile (i.e., differential expression of tremor in each arm in a given patient) is similar across family members is not known. The alternative possibility is that this feature is not heritable. There are no published data addressing this issue. The aim of this study was to determine whether the extent of action tremor asymmetry ran in ET families. Methods ET probands and relatives were enrolled in a genetic study at Yale and Columbia Universities. An in-person evaluation included a videotaped neurological examination, including a detailed assessment of tremors. A senior movement disorders neurologist reviewed all videotaped examinations, and the severity of postural and kinetic arm tremors was rated on 12 examination items using a reliable rating scale. The tremor asymmetry index = right arm tremor score − left arm tremor score. We used a bivariate linear regression model to assess the predictors of the tremor asymmetry index in relatives; this model used the tremor asymmetry index in the proband as a primary predictor of interest. In an analysis of variance (ANOVA), we tested for heterogeneity across families in the tremor asymmetry index (i.e., to see whether there was a significant family effect). Results There were 187 enrollees (59 probands, 128 affected relatives). In a bivariate linear regression model, the tremor asymmetry index in the proband was not a predictor of the tremor asymmetry index in their relatives (p = 0.66). In an ANOVA, family grouping did not explain a significant proportion of the total variance in the tremor asymmetry index (p = 0.56). Conclusion Tremor asymmetry did not aggregate in families with ET. Therefore, this does not seem to be a disease feature that is heritable. These data will provide added value to the clinical dialog, giving patients one more piece of information about the way the disease manifests within families.
Collapse
Affiliation(s)
- Elan D Louis
- Division of Movement Disorders, Department of Neurology, Yale School of Medicine, Yale University, New Haven, CT, USA.,Department of Chronic Disease Epidemiology, Yale School of Public Health, Yale University, New Haven, CT, USA.,Center for Neuroepidemiology and Clinical Neurological Research, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Nora Hernandez
- Division of Movement Disorders, Department of Neurology, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Karen P Chen
- Division of Movement Disorders, Department of Neurology, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Kelly V Naranjo
- Division of Movement Disorders, Department of Neurology, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Jemin Park
- Division of Movement Disorders, Department of Neurology, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Lorraine N Clark
- College of Physicians and Surgeons, Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY, USA.,Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY, USA
| | - Ruth Ottman
- G.H. Sergievsky Center, College of Physicians and Surgeons, Columbia University, New York, NY, USA.,Department of Neurology, College of Physicians and Surgeons, Columbia University, New York, NY, USA.,Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, NY, USA.,Division of Epidemiology, New York State Psychiatric Institute, New York, NY, USA
| |
Collapse
|
21
|
Kweon JH, Kim S, Lee SB. The cellular basis of dendrite pathology in neurodegenerative diseases. BMB Rep 2017; 50:5-11. [PMID: 27502014 PMCID: PMC5319658 DOI: 10.5483/bmbrep.2017.50.1.131] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Indexed: 01/30/2023] Open
Abstract
One of the characteristics of the neurons that distinguishes them from other cells is their complex and polarized structure consisting of dendrites, cell body, and axon. The complexity and diversity of dendrites are particularly well recognized, and accumulating evidences suggest that the alterations in the dendrite structure are associated with many neurodegenerative diseases. Given the importance of the proper dendritic structures for neuronal functions, the dendrite pathology appears to have crucial contribution to the pathogenesis of neurodegenerative diseases. Nonetheless, the cellular and molecular basis of dendritic changes in the neurodegenerative diseases remains largely elusive. Previous studies in normal condition have revealed that several cellular components, such as local cytoskeletal structures and organelles located locally in dendrites, play crucial roles in dendrite growth. By reviewing what has been unveiled to date regarding dendrite growth in terms of these local cellular components, we aim to provide an insight to categorize the potential cellular basis that can be applied to the dendrite pathology manifested in many neurodegenerative diseases. [BMB Reports 2017; 50(1): 5-11].
Collapse
Affiliation(s)
- Jung Hyun Kweon
- Department of Brain & Cognitive Sciences, DGIST, Daegu 42988, Korea
| | - Sunhong Kim
- Disease Target Structure Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141; Department of Biomolecular Science, University of Science and Technology, Daejeon 34141, Korea
| | - Sung Bae Lee
- Department of Brain & Cognitive Sciences, DGIST, Daegu 42988, Korea
| |
Collapse
|
22
|
Liu B, Lv C, Zhang J, Liu Y, Sun J, Cheng X, Mao W, Ma Y, Li S. Effects of eldepryl on glial cell proliferation and activation in the substantia nigra and striatum in a rat model of Parkinson’s disease. Neurol Res 2017; 39:459-467. [PMID: 28276259 DOI: 10.1080/01616412.2017.1297911] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Affiliation(s)
- Bin Liu
- First Department of Neurology, Hospital Affiliated to North China University of Science and Technology, Tangshan, China
| | - Chaonan Lv
- First Department of Neurology, Hospital Affiliated to North China University of Science and Technology, Tangshan, China
| | - Jinxia Zhang
- First Department of Neurology, Hospital Affiliated to North China University of Science and Technology, Tangshan, China
| | - Ying Liu
- First Department of Neurology, Hospital Affiliated to North China University of Science and Technology, Tangshan, China
| | - Jing Sun
- First Department of Neurology, Hospital Affiliated to North China University of Science and Technology, Tangshan, China
| | - Xiaohua Cheng
- First Department of Neurology, Hospital Affiliated to North China University of Science and Technology, Tangshan, China
| | - Wenjing Mao
- First Department of Neurology, Hospital Affiliated to North China University of Science and Technology, Tangshan, China
| | - Yuanyuan Ma
- First Department of Neurology, Hospital Affiliated to North China University of Science and Technology, Tangshan, China
| | - Shiying Li
- First Department of Neurology, Hospital Affiliated to North China University of Science and Technology, Tangshan, China
| |
Collapse
|
23
|
Antunes Dos Santos A, Appel Hort M, Culbreth M, López-Granero C, Farina M, Rocha JBT, Aschner M. Methylmercury and brain development: A review of recent literature. J Trace Elem Med Biol 2016; 38:99-107. [PMID: 26987277 PMCID: PMC5011031 DOI: 10.1016/j.jtemb.2016.03.001] [Citation(s) in RCA: 112] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2016] [Accepted: 03/02/2016] [Indexed: 02/02/2023]
Abstract
Methylmercury (MeHg) is a potent environmental pollutant, which elicits significant toxicity in humans. The central nervous system (CNS) is the primary target of toxicity, and is particularly vulnerable during development. Maternal exposure to MeHg via consumption of fish and seafood can have irreversible effects on the neurobehavioral development of children, even in the absence of symptoms in the mother. It is well documented that developmental MeHg exposure may lead to neurological alterations, including cognitive and motor dysfunction. The neurotoxic effects of MeHg on the developing brain have been extensively studied. The mechanism of toxicity, however, is not fully understood. No single process can explain the multitude of effects observed in MeHg-induced neurotoxicity. This review summarizes the most current knowledge on the effects of MeHg during nervous system development considering both, in vitro and in vivo experimental models. Considerable attention was directed towards the role of glutamate and calcium dyshomeostasis, mitochondrial dysfunction, as well as the effects of MeHg on cytoskeletal components/regulators.
Collapse
Affiliation(s)
| | - Mariana Appel Hort
- Institute of Biological Sciences, Federal University of Rio Grande, Campus Carreiros, Rio Grande do Sul, Brazil
| | - Megan Culbreth
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Caridad López-Granero
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Marcelo Farina
- Department of Biochemistry, Federal University of Santa Catarina, Florianopolis, Santa Catarina, Brazil
| | - Joao B T Rocha
- Department of Biochemistry, Federal University of Santa Maria, Santa Maria, Rio Grande do Sul, Brazil
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
24
|
Effects of Microtubule Stabilization by Epothilone B Depend on the Type and Age of Neurons. Neural Plast 2016; 2016:5056418. [PMID: 27872763 PMCID: PMC5107872 DOI: 10.1155/2016/5056418] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Revised: 09/07/2016] [Accepted: 09/21/2016] [Indexed: 01/06/2023] Open
Abstract
Several studies have demonstrated the therapeutic potential of applying microtubule- (MT-) stabilizing agents (MSAs) that cross the blood-brain barrier to promote axon regeneration and prevent axonal dystrophy in rodent models of spinal cord injury and neurodegenerative diseases. Paradoxically, administration of MSAs, which have been widely prescribed to treat malignancies, is well known to cause debilitating peripheral neuropathy and axon degeneration. Despite the growing interest of applying MSAs to treat the injured or degenerating central nervous system (CNS), consequences of MSA exposure to neurons in the central and peripheral nervous system (PNS) have not been thoroughly investigated. Here, we have examined and compared the effects of a brain-penetrant MSA, epothilone B, on cortical and sensory neurons in culture and show that epothilone B exhibits both beneficial and detrimental effects, depending on not only the concentration of drug but also the type and age of a neuron, as seen in clinical settings. Therefore, to exploit MSAs to their full benefit and minimize unwanted side effects, it is important to understand the properties of neuronal MTs and strategies should be devised to deliver minimal effective concentration directly to the site where needed.
Collapse
|
25
|
Microtubule Destabilization Paves the Way to Parkinson's Disease. Mol Neurobiol 2016; 54:6762-6774. [PMID: 27757833 DOI: 10.1007/s12035-016-0188-5] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Accepted: 09/30/2016] [Indexed: 01/20/2023]
Abstract
Microtubules are dynamic structures normally associated to the cell division, during which they form the mitotic spindle, as well as to the initial phases of specification and polarization of various cell types, including neurons. Although microtubules could have a role in the death of many cells and tissues, the microtubule-based degenerative mechanisms have been poorly investigated; nevertheless, during the last two decades, many clues have been accumulated suggesting the importance of the microtubule system during neurodegeneration. Thus, the aim of this review is to analyse how the changes of the microtubule cytoskeleton, in terms of organization and dynamics, as well as the failure of the microtubule-dependent neuronal processes, as axonal transport, may play a pivotal role in the chain of events leading to Parkinson's disease. Last but not least, since disease-modifying or neuroprotective strategies are a clinical priority in Parkinson's disease, we will also present the hints about the concrete possibility of a microtubule-targeted therapy, which would have the potentiality to block the running degenerative events and to prompt the regeneration of the lost tissues.
Collapse
|
26
|
eSNPO: An eQTL-based SNP Ontology and SNP functional enrichment analysis platform. Sci Rep 2016; 6:30595. [PMID: 27470167 PMCID: PMC4965794 DOI: 10.1038/srep30595] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Accepted: 07/04/2016] [Indexed: 02/06/2023] Open
Abstract
Genome-wide association studies (GWASs) have mined many common genetic variants associated with human complex traits like diseases. After that, the functional annotation and enrichment analysis of significant SNPs are important tasks. Classic methods are always based on physical positions of SNPs and genes. Expression quantitative trait loci (eQTLs) are genomic loci that contribute to variation in gene expression levels and have been proven efficient to connect SNPs and genes. In this work, we integrated the eQTL data and Gene Ontology (GO), constructed associations between SNPs and GO terms, then performed functional enrichment analysis. Finally, we constructed an eQTL-based SNP Ontology and SNP functional enrichment analysis platform. Taking Parkinson Disease (PD) as an example, the proposed platform and method are efficient. We believe eSNPO will be a useful resource for SNP functional annotation and enrichment analysis after we have got significant disease related SNPs.
Collapse
|
27
|
Genetic Variants of Microtubule Actin Cross-linking Factor 1 (MACF1) Confer Risk for Parkinson’s Disease. Mol Neurobiol 2016; 54:2878-2888. [DOI: 10.1007/s12035-016-9861-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Accepted: 03/17/2016] [Indexed: 01/12/2023]
|
28
|
Penazzi L, Bakota L, Brandt R. Microtubule Dynamics in Neuronal Development, Plasticity, and Neurodegeneration. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2015; 321:89-169. [PMID: 26811287 DOI: 10.1016/bs.ircmb.2015.09.004] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Neurons are the basic information-processing units of the nervous system. In fulfilling their task, they establish a structural polarity with an axon that can be over a meter long and dendrites with a complex arbor, which can harbor ten-thousands of spines. Microtubules and their associated proteins play important roles during the development of neuronal morphology, the plasticity of neurons, and neurodegenerative processes. They are dynamic structures, which can quickly adapt to changes in the environment and establish a structural scaffold with high local variations in composition and stability. This review presents a comprehensive overview about the role of microtubules and their dynamic behavior during the formation and maturation of processes and spines in the healthy brain, during aging and under neurodegenerative conditions. The review ends with a discussion of microtubule-targeted therapies as a perspective for the supportive treatment of neurodegenerative disorders.
Collapse
Affiliation(s)
- Lorène Penazzi
- Department of Neurobiology, University of Osnabrück, Osnabrück, Germany
| | - Lidia Bakota
- Department of Neurobiology, University of Osnabrück, Osnabrück, Germany
| | - Roland Brandt
- Department of Neurobiology, University of Osnabrück, Osnabrück, Germany
| |
Collapse
|
29
|
Autophagy-related protein expression in the substantia nigra and eldepryl intervention in rat models of Parkinson׳s disease. Brain Res 2015; 1625:180-8. [DOI: 10.1016/j.brainres.2015.08.033] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Revised: 08/18/2015] [Accepted: 08/25/2015] [Indexed: 01/09/2023]
|
30
|
Ren Y, Jiang H, Hu Z, Fan K, Wang J, Janoschka S, Wang X, Ge S, Feng J. Parkin mutations reduce the complexity of neuronal processes in iPSC-derived human neurons. Stem Cells 2015; 33:68-78. [PMID: 25332110 DOI: 10.1002/stem.1854] [Citation(s) in RCA: 83] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2014] [Accepted: 08/24/2014] [Indexed: 12/13/2022]
Abstract
Parkinson's disease (PD) is characterized by the degeneration of nigral dopaminergic (DA) neurons and non-DA neurons in many parts of the brain. Mutations of parkin, an E3 ubiquitin ligase that strongly binds to microtubules, are the most frequent cause of recessively inherited PD. The lack of robust PD phenotype in parkin knockout mice suggests a unique vulnerability of human neurons to parkin mutations. Here, we show that the complexity of neuronal processes as measured by total neurite length, number of terminals, number of branch points, and Sholl analysis was greatly reduced in induced pluripotent stem cell (iPSC)-derived TH(+) or TH(-) neurons from PD patients with parkin mutations. Consistent with these, microtubule stability was significantly decreased by parkin mutations in iPSC-derived neurons. Overexpression of parkin, but not its PD-linked mutant nor green fluorescent protein, restored the complexity of neuronal processes and the stability of microtubules. Consistent with these, the microtubule-depolymerizing agent colchicine mimicked the effect of parkin mutations by decreasing neurite length and complexity in control neurons while the microtubule-stabilizing drug taxol mimicked the effect of parkin overexpression by enhancing the morphology of parkin-deficient neurons. The results suggest that parkin maintains the morphological complexity of human neurons by stabilizing microtubules.
Collapse
Affiliation(s)
- Yong Ren
- Department of Physiology and Biophysics, State University of New York at Buffalo, Buffalo, New York, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Behaviour and prefrontal protein differences in C57BL/6N and 129 X1/SvJ mice. Brain Res Bull 2015; 116:16-24. [PMID: 26003851 DOI: 10.1016/j.brainresbull.2015.05.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Revised: 05/11/2015] [Accepted: 05/12/2015] [Indexed: 01/24/2023]
Abstract
Experimental animals provide valuable opportunities to establish aetiological mechanisms and test new treatments for neurodevelopmental psychiatric conditions. However, it is increasingly appreciated that inter-strain differences cannot be neglected in the experimental design. In addition, the importance of including females in preclinical - but also clinical - research is now recognised. Here, we compared behaviour and prefrontal protein differences in male and female C57BL/6N and 129X1/SvJ mice as both are commonly used experimental rodents. Relative to 129X1/SvJ mice, both sexes of C57BL/6N mice had weaker sensorimotor gating, measured in the prepulse inhibition (PPI) of startle paradigm, and were more sensitive to amphetamine challenge in the open field. The pattern of protein expression in the prefrontal cortex of C57BL6N mice was also clearly distinct from 129X1/SvJ mice. Proteins differentially expressed were those associated with oxidative metabolism, receptor protein signalling, cell communication and signal transduction and energy pathways. We suggest that the C57BL/6N mouse may usefully proxy features of the neurodevelopmental disorders and could have application in pre-translational screening of new therapeutic approaches. The 129X1/SvJ strain in contrast, might be better suited to experimental studies of causal risk factors expected to lower PPI and increase amphetamine sensitivity.
Collapse
|
32
|
Capurro A, Bodea LG, Schaefer P, Luthi-Carter R, Perreau VM. Computational deconvolution of genome wide expression data from Parkinson's and Huntington's disease brain tissues using population-specific expression analysis. Front Neurosci 2015; 8:441. [PMID: 25620908 PMCID: PMC4288238 DOI: 10.3389/fnins.2014.00441] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Accepted: 12/15/2014] [Indexed: 01/09/2023] Open
Abstract
The characterization of molecular changes in diseased tissues gives insight into pathophysiological mechanisms and is important for therapeutic development. Genome-wide gene expression analysis has proven valuable for identifying biological processes in neurodegenerative diseases using post mortem human brain tissue and numerous datasets are publically available. However, many studies utilize heterogeneous tissue samples consisting of multiple cell types, all of which contribute to global gene expression values, confounding biological interpretation of the data. In particular, changes in numbers of neuronal and glial cells occurring in neurodegeneration confound transcriptomic analyses, particularly in human brain tissues where sample availability and controls are limited. To identify cell specific gene expression changes in neurodegenerative disease, we have applied our recently published computational deconvolution method, population specific expression analysis (PSEA). PSEA estimates cell-type-specific expression values using reference expression measures, which in the case of brain tissue comprises mRNAs with cell-type-specific expression in neurons, astrocytes, oligodendrocytes and microglia. As an exercise in PSEA implementation and hypothesis development regarding neurodegenerative diseases, we applied PSEA to Parkinson's and Huntington's disease (PD, HD) datasets. Genes identified as differentially expressed in substantia nigra pars compacta neurons by PSEA were validated using external laser capture microdissection data. Network analysis and Annotation Clustering (DAVID) identified molecular processes implicated by differential gene expression in specific cell types. The results of these analyses provided new insights into the implementation of PSEA in brain tissues and additional refinement of molecular signatures in human HD and PD.
Collapse
Affiliation(s)
- Alberto Capurro
- Department of Cell Physiology and Pharmacology, University of Leicester Leicester, UK
| | - Liviu-Gabriel Bodea
- Neural Regeneration Unit, Institute of Reconstructive Neurobiology, University of Bonn Bonn, Germany ; Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland St Lucia, QLD, Australia
| | | | - Ruth Luthi-Carter
- Department of Cell Physiology and Pharmacology, University of Leicester Leicester, UK
| | - Victoria M Perreau
- The Bioinformatics Core and The Synaptic Neurobiology Laboratory, The Florey Institute of Neuroscience and Mental Health Parkville, VIC, Australia
| |
Collapse
|
33
|
Fang W, Gao G, Zhao H, Xia Y, Guo X, Li N, Li Y, Yang Y, Chen L, Wang Q, Li L. Role of the Akt/GSK-3β/CRMP-2 pathway in axon degeneration of dopaminergic neurons resulting from MPP+ toxicity. Brain Res 2014; 1602:9-19. [PMID: 25150591 DOI: 10.1016/j.brainres.2014.08.030] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2014] [Revised: 08/12/2014] [Accepted: 08/13/2014] [Indexed: 12/26/2022]
Abstract
Parkinson׳s disease (PD) is the most common neurodegenerative disease of the basal ganglia. Earlier reports suggest that the main pathological change in PD is due to apoptosis of dopaminergic neuronal soma in the substantia nigra (SN). The therapies for PD are also largely focused on the prevention of degeneration of the neuronal soma. However, these treatments can only provide temporary relief by delaying the progression of the disease and are therefore unable to prevent the long term neurodegeneration process. This limitation of the existing therapeutic treatment indicates that there may be other causes that either occur earlier or are independent of apoptosis of neuronal soma. Previous studies have shown that axon degeneration may play an important role in PD, and that this may occur at an early stage of the disease. Thus, preventing axon degeneration may be a potential new approach for therapeutic treatment for PD and future therapies can be useful if emphasis is given on the mechanisms of axon degeneration. It has been recognized that microtubule disassembly leads to axon degeneration because the depolymerized microtubules are more likely to be degraded. Previous studies have shown that glycogen synthase kinase-3β (GSK-3β)/collapsin response mediator protein 2 (CRMP-2) signaling pathway could be regulated by Akt for axonal-dendritic polarity. CRMP-2 is critical for specifying axon/dendrite fate possibly by promoting neurite elongation via microtubule assembly. However, whether Akt could regulate GSK-3β/CRMP-2 pathway and the possible effects of this regulation is unclear in dopaminergic axon degeneration induced by 1-methyl-4-phenylpyridiniumion (MPP+). In this study, we observe the degeneration of axon and neuronal soma by scanning electron microscope and tyrosine hydroxylase staining (TH) using a PD model in dopaminergic neurons in vitro. In addition to this, we detect the expression of total and phosphorylated form of Akt, GSK-3β and CRMP-2, as well as the axonal injury marker amyloid precursor protein (APP). From our studies, we observe that axon degeneration is a characteristic feature in the cascade of events that follow when neurons are induced by MPP+. This degeneration process occurs earlier in case of PD and is more severe than the degeneration of the neuronal soma and Akt/ GSK-3β/CRMP-2 pathway is involved in this process.
Collapse
Affiliation(s)
- Wei Fang
- Department of Neurosurgery, Tangdu Hospital, Fourth Military Medical University, Xi׳an City 710038, PR China
| | - Guodong Gao
- Department of Neurosurgery, Tangdu Hospital, Fourth Military Medical University, Xi׳an City 710038, PR China
| | - Haikang Zhao
- The second Affiliated Hospital,Xi׳an Medical University, Xi׳an City 710038, PR China
| | - Yi Xia
- Department of Neurosurgery, Tangdu Hospital, Fourth Military Medical University, Xi׳an City 710038, PR China
| | - Xiaodong Guo
- Department of Neurosurgery, 153rd Hospital of PLA, Zhengzhou 450042, PR China
| | - Nan Li
- Department of Neurosurgery, Tangdu Hospital, Fourth Military Medical University, Xi׳an City 710038, PR China
| | - Yuqian Li
- Department of Neurosurgery, Tangdu Hospital, Fourth Military Medical University, Xi׳an City 710038, PR China
| | - Yang Yang
- Department of Neurosurgery, Tangdu Hospital, Fourth Military Medical University, Xi׳an City 710038, PR China
| | - Lei Chen
- Department of Neurosurgery, Tangdu Hospital, Fourth Military Medical University, Xi׳an City 710038, PR China
| | - Qiang Wang
- Department of Neurosurgery, Tangdu Hospital, Fourth Military Medical University, Xi׳an City 710038, PR China
| | - Lihong Li
- Department of Neurosurgery, Tangdu Hospital, Fourth Military Medical University, Xi׳an City 710038, PR China.
| |
Collapse
|
34
|
Zou T, Tang X, Huang Z, Xu N, Hu Z. The Pael-R gene does not mediate the changes in rotenone-induced Parkinson's disease model cells. Neural Regen Res 2014; 9:402-6. [PMID: 25206827 PMCID: PMC4146201 DOI: 10.4103/1673-5374.128245] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/19/2014] [Indexed: 11/04/2022] Open
Abstract
In this study, we established cell models for Parkinson's disease using rotenone. An RNA interference vector targeting Parkin-associated endothelin receptor-like receptor (Pael-R) was transfected into the model cells. The results of reverse-transcription polymerase chain reaction and western blot analysis showed that Pael-R expression was decreased after RNA interference compared with the control group (no treatment) and the model group (rotenone treatment), while the rate of apoptosis and survival of dopaminergic cells did not differ significantly between groups, as detected by flow cytometry and an MTT assay. These experimental findings indicate that the Pael-R gene has no role in the changes in rotenone-induced Parkinson's disease model cells.
Collapse
Affiliation(s)
- Ting Zou
- Department of Neurology, Second Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Xiangqi Tang
- Department of Neurology, Second Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Zhiling Huang
- Department of Neurology, Second Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Niangui Xu
- Department of Neurology, Second Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Zhiping Hu
- Department of Neurology, Second Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| |
Collapse
|
35
|
|
36
|
Sanders LH, Timothy Greenamyre J. Oxidative damage to macromolecules in human Parkinson disease and the rotenone model. Free Radic Biol Med 2013; 62:111-120. [PMID: 23328732 PMCID: PMC3677955 DOI: 10.1016/j.freeradbiomed.2013.01.003] [Citation(s) in RCA: 425] [Impact Index Per Article: 35.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2012] [Revised: 01/08/2013] [Accepted: 01/08/2013] [Indexed: 11/25/2022]
Abstract
Parkinson disease (PD), the most common neurodegenerative movement disorder, is associated with selective degeneration of nigrostriatal dopamine neurons. Although the underlying mechanisms contributing to neurodegeneration in PD seem to be multifactorial, mitochondrial impairment and oxidative stress are widely considered to be central to many forms of the disease. Whether oxidative stress is a cause or a consequence of dopaminergic death, there is substantial evidence for oxidative stress both in human PD patients and in animal models of PD, especially using rotenone, a complex I inhibitor. There are many indices of oxidative stress, but this review covers the recent evidence for oxidative damage to nucleic acids, lipids, and proteins in both the brain and the peripheral tissues in human PD and in the rotenone model. Limitations of the existing literature and future perspectives are discussed. Understanding how each particular macromolecule is damaged by oxidative stress and the interplay of secondary damage to other biomolecules may help us design better targets for the treatment of PD.
Collapse
Affiliation(s)
- Laurie H Sanders
- Pittsburgh Institute for Neurodegenerative Diseases, Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - J Timothy Greenamyre
- Pittsburgh Institute for Neurodegenerative Diseases, Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15260, USA.
| |
Collapse
|
37
|
DJ-1 deficiency perturbs microtubule dynamics and impairs striatal neurite outgrowth. Neurobiol Aging 2013; 34:489-98. [DOI: 10.1016/j.neurobiolaging.2012.04.008] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2012] [Revised: 04/13/2012] [Accepted: 04/16/2012] [Indexed: 11/21/2022]
|
38
|
Xu F, Farkas S, Kortbeek S, Zhang FX, Chen L, Zamponi GW, Syed NI. Mercury-induced toxicity of rat cortical neurons is mediated through N-Methyl-D-Aspartate receptors. Mol Brain 2012; 5:30. [PMID: 22980357 PMCID: PMC3462706 DOI: 10.1186/1756-6606-5-30] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2012] [Accepted: 09/11/2012] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Mercury is a well-known neurotoxin implicated in a wide range of neurological or psychiatric disorders including autism spectrum disorders, Alzheimer's disease, Parkinson's disease, epilepsy, depression, mood disorders and tremor. Mercury-induced neuronal degeneration is thought to invoke glutamate-mediated excitotoxicity, however, the underlying mechanisms remain poorly understood. Here, we examine the effects of various mercury concentrations (including pathological levels present in human plasma or cerebrospinal fluid) on cultured, rat cortical neurons. RESULTS We found that inorganic mercuric chloride (HgCl₂--at 0.025 to 25 μM) not only caused neuronal degeneration but also perturbed neuronal excitability. Whole-cell patch-clamp recordings of pyramidal neurons revealed that HgCl₂ not only enhanced the amplitude and frequency of synaptic, inward currents, but also increased spontaneous synaptic potentials followed by sustained membrane depolarization. HgCl₂ also triggered sustained, 2-5 fold rises in intracellular calcium concentration ([Ca²⁺]i). The observed increases in neuronal activity and [Ca²⁺]i were substantially reduced by the application of MK 801, a non-competitive antagonist of N-Methyl-D-Aspartate (NMDA) receptors. Importantly, our study further shows that a pre incubation or co-application of MK 801 prevents HgCl₂-induced reduction of cell viability and a disruption of β-tubulin. CONCLUSIONS Collectively, our data show that HgCl₂-induced toxic effects on central neurons are triggered by an over-activation of NMDA receptors, leading to cytoskeleton instability.
Collapse
Affiliation(s)
- Fenglian Xu
- Department of Cell Biology & Anatomy, University of Calgary, Alberta, Canada.
| | | | | | | | | | | | | |
Collapse
|
39
|
Byers B, Cord B, Nguyen HN, Schüle B, Fenno L, Lee PC, Deisseroth K, Langston JW, Pera RR, Palmer TD. SNCA triplication Parkinson's patient's iPSC-derived DA neurons accumulate α-synuclein and are susceptible to oxidative stress. PLoS One 2011; 6:e26159. [PMID: 22110584 PMCID: PMC3217921 DOI: 10.1371/journal.pone.0026159] [Citation(s) in RCA: 237] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2011] [Accepted: 09/21/2011] [Indexed: 12/21/2022] Open
Abstract
Parkinson's disease (PD) is an incurable age-related neurodegenerative disorder affecting both the central and peripheral nervous systems. Although common, the etiology of PD remains poorly understood. Genetic studies infer that the disease results from a complex interaction between genetics and environment and there is growing evidence that PD may represent a constellation of diseases with overlapping yet distinct underlying mechanisms. Novel clinical approaches will require a better understanding of the mechanisms at work within an individual as well as methods to identify the specific array of mechanisms that have contributed to the disease. Induced pluripotent stem cell (iPSC) strategies provide an opportunity to directly study the affected neuronal subtypes in a given patient. Here we report the generation of iPSC-derived midbrain dopaminergic neurons from a patient with a triplication in the α-synuclein gene (SNCA). We observed that the iPSCs readily differentiated into functional neurons. Importantly, the PD-affected line exhibited disease-related phenotypes in culture: accumulation of α-synuclein, inherent overexpression of markers of oxidative stress, and sensitivity to peroxide induced oxidative stress. These findings show that the dominantly-acting PD mutation is intrinsically capable of perturbing normal cell function in culture and confirm that these features reflect, at least in part, a cell autonomous disease process that is independent of exposure to the entire complexity of the diseased brain.
Collapse
Affiliation(s)
- Blake Byers
- Department of Bioengineering, Stanford University, Stanford, California, United States of America
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, California, United States of America
| | - Branden Cord
- Department of Obstetrics and Gynecology, Stanford University, Stanford, California, United States of America
| | - Ha Nam Nguyen
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, California, United States of America
| | - Birgitt Schüle
- Parkinson's Institute and Clinical Center, Sunnyvale, California, United States of America
| | - Lief Fenno
- Department of Neuroscience, Stanford University, Stanford, California, United States of America
| | - Patrick C. Lee
- Department of Obstetrics and Gynecology, Stanford University, Stanford, California, United States of America
| | - Karl Deisseroth
- Department of Bioengineering, Stanford University, Stanford, California, United States of America
- Department of Neuroscience, Stanford University, Stanford, California, United States of America
- Howard Hughes Medical Institute, Stanford University, Stanford, California, United States of America
- Department of Psychiatry and Behavioral Sciences, Stanford, California, United States of America
| | - J. William Langston
- Parkinson's Institute and Clinical Center, Sunnyvale, California, United States of America
| | - Renee Reijo Pera
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, California, United States of America
- Department of Obstetrics and Gynecology, Stanford University, Stanford, California, United States of America
- * E-mail: (RRP); (TDP)
| | - Theo D. Palmer
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, California, United States of America
- Department of Neurosurgery, Stanford University, Stanford, California, United States of America
- * E-mail: (RRP); (TDP)
| |
Collapse
|
40
|
Modulation of microtubule dynamics by a TIR domain protein from the intracellular pathogen Brucella melitensis. Biochem J 2011; 439:79-83. [PMID: 21692747 DOI: 10.1042/bj20110577] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
TIR (Toll/interleukin-1 receptor) domain-containing proteins play a crucial role in innate immunity in eukaryotes. Brucella is a highly infectious intracellular bacterium that encodes a TIR domain protein (TcpB) to subvert host innate immune responses to establish a beneficial niche for pathogenesis. TcpB inhibits NF-κB (nuclear factor κB) activation and pro-inflammatory cytokine secretions mediated by TLR (Toll-like receptor) 2 and TLR4. In the present study, we have demonstrated that TcpB modulates microtubule dynamics by acting as a stabilization factor. TcpB increased the rate of nucleation as well as the polymerization phases of microtubule formation in a similar manner to paclitaxel. TcpB could efficiently inhibit nocodazole- or cold-induced microtubule disassembly. Microtubule stabilization by TcpB is attributed to the BB-loop region of the TIR domain, and a point mutation affected the microtubule stabilization as well as the TLR-suppression properties of TcpB.
Collapse
|
41
|
|
42
|
Larsen NJ, Ambrosi G, Mullett SJ, Berman SB, Hinkle DA. DJ-1 knock-down impairs astrocyte mitochondrial function. Neuroscience 2011; 196:251-64. [PMID: 21907265 DOI: 10.1016/j.neuroscience.2011.08.016] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2011] [Accepted: 08/05/2011] [Indexed: 12/21/2022]
Abstract
Mitochondrial dysfunction has long been implicated in the pathogenesis of Parkinson's disease (PD). PD brain tissues show evidence for mitochondrial respiratory chain Complex I deficiency. Pharmacological inhibitors of Complex I, such as rotenone, cause experimental parkinsonism. The cytoprotective protein DJ-1, whose deletion is sufficient to cause genetic PD, is also known to have mitochondria-stabilizing properties. We have previously shown that DJ-1 is over-expressed in PD astrocytes, and that DJ-1 deficiency impairs the capacity of astrocytes to protect co-cultured neurons against rotenone. Since DJ-1 modulated, astrocyte-mediated neuroprotection against rotenone may depend upon proper astrocytic mitochondrial functioning, we hypothesized that DJ-1 deficiency would impair astrocyte mitochondrial motility, fission/fusion dynamics, membrane potential maintenance, and respiration, both at baseline and as an enhancement of rotenone-induced mitochondrial dysfunction. In astrocyte-enriched cultures, we observed that DJ-1 knock-down reduced mitochondrial motility primarily in the cellular processes of both untreated and rotenone treated cells. In these same cultures, DJ-1 knock-down did not appreciably affect mitochondrial fission, fusion, or respiration, but did enhance rotenone-induced reductions in the mitochondrial membrane potential. In neuron-astrocyte co-cultures, astrocytic DJ-1 knock-down reduced astrocyte process mitochondrial motility in untreated cells, but this effect was not maintained in the presence of rotenone. In the same co-cultures, astrocytic DJ-1 knock-down significantly reduced mitochondrial fusion in the astrocyte cell bodies, but not the processes, under the same conditions of rotenone treatment in which DJ-1 deficiency is known to impair astrocyte-mediated neuroprotection. Our studies therefore demonstrated the following new findings: (i) DJ-1 deficiency can impair astrocyte mitochondrial physiology at multiple levels, (ii) astrocyte mitochondrial dynamics vary with sub-cellular region, and (iii) the physical presence of neurons can affect astrocyte mitochondrial behavior.
Collapse
Affiliation(s)
- N J Larsen
- Department of Neurology, Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | | | | | | | | |
Collapse
|
43
|
Parisiadou L, Cai H. LRRK2 function on actin and microtubule dynamics in Parkinson disease. Commun Integr Biol 2011; 3:396-400. [PMID: 21057624 DOI: 10.4161/cib.3.5.12286] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2010] [Accepted: 05/05/2010] [Indexed: 01/11/2023] Open
Abstract
The mutations in the LRRK2 gene cause clinically typical, late-onset Parkinson disease, strengthening the idea that the familial forms of the disease represent an important tool for the study of the idiopathic forms. Despite the great effort to describe and functionally characterize the LRRK2 gene product, its physiological role remains elusive. In this article, we will discuss along with other references, our recent findings that assigned a critical role of LRRK2 protein on cytosleketal dynamics and how this direction could provide a valuable platform to further appreciate the mechanism underlying LRRK2-mediated pathophysiology of the disease.
Collapse
Affiliation(s)
- Loukia Parisiadou
- Unit of Transgenesis; Laboratory of Neurogenetics; National Institute on Aging; National Institutes of Health; Bethesda, MD USA
| | | |
Collapse
|
44
|
Choi WS, Palmiter RD, Xia Z. Loss of mitochondrial complex I activity potentiates dopamine neuron death induced by microtubule dysfunction in a Parkinson's disease model. ACTA ACUST UNITED AC 2011; 192:873-82. [PMID: 21383081 PMCID: PMC3051820 DOI: 10.1083/jcb.201009132] [Citation(s) in RCA: 134] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The combination of microtubule depolymerization and the accumulation of cytosolic dopamine and reactive oxygen species selectively affects survival of dopaminergic neurons. Mitochondrial complex I dysfunction is regarded as underlying dopamine neuron death in Parkinson’s disease models. However, inactivation of the Ndufs4 gene, which compromises complex I activity, does not affect the survival of dopamine neurons in culture or in the substantia nigra pars compacta of 5-wk-old mice. Treatment with piericidin A, a complex I inhibitor, does not induce selective dopamine neuron death in either Ndufs4+/+ or Ndufs4−/− mesencephalic cultures. In contrast, rotenone, another complex I inhibitor, causes selective toxicity to dopamine neurons, and Ndufs4 inactivation potentiates this toxicity. We identify microtubule depolymerization and the accumulation of cytosolic dopamine and reactive oxygen species as alternative mechanisms underlying rotenone-induced dopamine neuron death. Enhanced rotenone toxicity to dopamine neurons from Ndufs4 knockout mice may involve enhanced dopamine synthesis caused by the accumulation of nicotinamide adenine dinucleotide reduced. Our results suggest that the combination of disrupting microtubule dynamics and inhibiting complex I, either by mutations or exposure to toxicants, may be a risk factor for Parkinson’s disease.
Collapse
Affiliation(s)
- Won-Seok Choi
- Department of Environmental and Occupational Health Sciences, Graduate Program in Neurobiology and Behavior, University of Washington, Seattle, WA 98195, USA
| | | | | |
Collapse
|
45
|
Gardan-Salmon D, Dixon JM, Lonergan SM, Selsby JT. Proteomic assessment of the acute phase of dystrophin deficiency in mdx mice. Eur J Appl Physiol 2011; 111:2763-73. [PMID: 21409400 DOI: 10.1007/s00421-011-1906-3] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2010] [Accepted: 03/03/2011] [Indexed: 11/27/2022]
Abstract
Duchenne muscular dystrophy (DMD) is caused by the absence of a functional dystrophin protein and is modeled by the mdx mouse. The mdx mouse suffers an early necrotic bout in the hind limb muscles lasting from approximately 4 to 7 weeks. The purpose of this investigation was to determine the extent to which dystrophin deficiency changed the proteome very early in the disease process. In order to accomplish this, proteins from gastrocnemius from 6-week-old C57 (n = 6) and mdx (n = 6) mice were labeled with fluorescent dye and subjected to two-dimensional differential in-gel electrophoresis (2D-DIGE). Resulting differentially expressed spots were excised and protein identity determined via MALDI-TOF followed by database searching using MASCOT. Proteins of the immediate energy system and glycolysis were generally down-regulated in mdx mice compared to C57 mice. Conversely, expression of proteins involved in the Kreb's cycle and electron transport chain were increased in dystrophin-deficient muscle compared to control. Expression of cytoskeletal components, including tubulins, vimentin, and collagen, were increased in mdx mice compared to C57 mice. Importantly, these changes are occurring at only 6 weeks of age and are caused by acute dystrophin deficiency rather than more chronic injury. These data may provide insight regarding early pathologic changes occurring in dystrophin-deficient skeletal muscle.
Collapse
Affiliation(s)
- D Gardan-Salmon
- Department of Animal Science, Iowa State University, 2356 Kildee Hall, Ames, IA 50011, USA
| | | | | | | |
Collapse
|
46
|
Mullett SJ, Hinkle DA. DJ-1 deficiency in astrocytes selectively enhances mitochondrial Complex I inhibitor-induced neurotoxicity. J Neurochem 2011; 117:375-87. [PMID: 21219333 DOI: 10.1111/j.1471-4159.2011.07175.x] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Parkinson's disease (PD) brains show evidence of mitochondrial respiratory Complex I deficiency, oxidative stress, and neuronal death. Complex I-inhibiting neurotoxins, such as the pesticide rotenone, cause neuronal death and parkinsonism in animal models. We have previously shown that DJ-1 over-expression in astrocytes augments their capacity to protect neurons against rotenone, that DJ-1 knock-down impairs astrocyte-mediated neuroprotection against rotenone, and that each process involves astrocyte-released factors. To further investigate the mechanism behind these findings, we developed a high-throughput, plate-based bioassay that can be used to assess how genetic manipulations in astrocytes affect their ability to protect co-cultured neurons. We used this bioassay to show that DJ-1 deficiency-induced impairments in astrocyte-mediated neuroprotection occur solely in the presence of pesticides that inhibit Complex I (rotenone, pyridaben, fenazaquin, and fenpyroximate); not with agents that inhibit Complexes II-V, that primarily induce oxidative stress, or that inhibit the proteasome. This is a potentially PD-relevant finding because pesticide exposure is epidemiologically-linked with an increased risk for PD. Further investigations into our model suggested that astrocytic GSH and heme oxygenase-1 antioxidant systems are not central to the neuroprotective mechanism.
Collapse
Affiliation(s)
- Steven J Mullett
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15260, USA
| | | |
Collapse
|
47
|
Esteves AR, Arduíno DM, Silva DFF, Oliveira CR, Cardoso SM. Mitochondrial Dysfunction: The Road to Alpha-Synuclein Oligomerization in PD. PARKINSON'S DISEASE 2011; 2011:693761. [PMID: 21318163 PMCID: PMC3026982 DOI: 10.4061/2011/693761] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/20/2010] [Revised: 12/21/2010] [Accepted: 12/27/2010] [Indexed: 12/21/2022]
Abstract
While the etiology of Parkinson's disease remains largely elusive, there is accumulating evidence suggesting that mitochondrial dysfunction occurs prior to the onset of symptoms in Parkinson's disease. Mitochondria are remarkably primed to play a vital role in neuronal cell survival since they are key regulators of energy metabolism (as ATP producers), of intracellular calcium homeostasis, of NAD(+)/NADH ratio, and of endogenous reactive oxygen species production and programmed cell death. In this paper, we focus on mitochondrial dysfunction-mediated alpha-synuclein aggregation. We highlight some of the findings that provide proof of evidence for a mitochondrial metabolism control in Parkinson's disease, namely, mitochondrial regulation of microtubule-dependent cellular traffic and autophagic lysosomal pathway. The knowledge that microtubule alterations may lead to autophagic deficiency and may compromise the cellular degradation mechanisms that culminate in the progressive accumulation of aberrant protein aggregates shields new insights to the way we address Parkinson's disease. In line with this knowledge, an innovative window for new therapeutic strategies aimed to restore microtubule network may be unlocked.
Collapse
Affiliation(s)
- A. R. Esteves
- Centro de Neurociências e Biologia Celular, Universidade de Coimbra, 3004 Coimbra, Portugal
| | - D. M. Arduíno
- Centro de Neurociências e Biologia Celular, Universidade de Coimbra, 3004 Coimbra, Portugal
| | - D. F. F. Silva
- Centro de Neurociências e Biologia Celular, Universidade de Coimbra, 3004 Coimbra, Portugal
| | - C. R. Oliveira
- Centro de Neurociências e Biologia Celular, Universidade de Coimbra, 3004 Coimbra, Portugal
- Faculdade de Medicina, Universidade de Coimbra, 3000 Coimbra, Portugal
| | - S. M. Cardoso
- Centro de Neurociências e Biologia Celular, Universidade de Coimbra, 3004 Coimbra, Portugal
- Faculdade de Medicina, Universidade de Coimbra, 3000 Coimbra, Portugal
| |
Collapse
|
48
|
|
49
|
Cartelli D, Ronchi C, Maggioni MG, Rodighiero S, Giavini E, Cappelletti G. Microtubule dysfunction precedes transport impairment and mitochondria damage in MPP+ -induced neurodegeneration. J Neurochem 2010; 115:247-58. [PMID: 20649848 DOI: 10.1111/j.1471-4159.2010.06924.x] [Citation(s) in RCA: 95] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Dysfunction of the microtubule (MT) system is an emerging theme in the pathogenesis of Parkinson's disease. This study was designed to investigate the putative role of MT dysfunction in dopaminergic neuron death induced by the neurotoxin 1-methyl-4-phenylpiridinium (MPP(+)). In nerve growth factor-differentiated PC12 cells, we have analyzed post-translational modifications of tubulin known to be associated with differently dynamic MTs and show that MPP(+) causes a selective loss of dynamic MTs and a concomitant enrichment of stable MTs. Through a direct live cell imaging approach, we show a significant reduction of MT dynamics following exposure to MPP(+) and a reorientation of MTs. Furthermore, these alterations precede the impairment of intracellular transport as revealed by changes in mitochondria movements along neurites and their accumulation into varicosities. We have also analyzed activation of caspase 3 and mitochondrial injury, well-known alterations induced by MPP(+), and found that they are noticeable only when MT dysfunction is already established. These data provide the first evidence that axonal transport impairment and mitochondrial damage might be a consequence of MT dysfunction in MPP(+) -induced neurodegeneration, lending support to the concept that alterations of MT organization and dynamics could play a pivotal role in neuronal death in Parkinson's disease.
Collapse
Affiliation(s)
- Daniele Cartelli
- Dipartimento di Biologia, Università degli Studi di Milano, Milan, Italy
| | | | | | | | | | | |
Collapse
|
50
|
The Drosophila vesicular monoamine transporter reduces pesticide-induced loss of dopaminergic neurons. Neurobiol Dis 2010; 40:102-12. [PMID: 20472063 DOI: 10.1016/j.nbd.2010.05.008] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2010] [Revised: 05/04/2010] [Accepted: 05/06/2010] [Indexed: 12/21/2022] Open
Abstract
Dopamine is cytotoxic and may play a role in the development of Parkinson's disease. However, its interaction with environmental risk factors such as pesticides remains poorly understood. The vesicular monoamine transporter (VMAT) regulates intracellular dopamine content, and we have tested the neuroprotective effects of VMAT in vivo using the model organism Drosophila melanogaster. We find that Drosophila VMAT (dVMAT) mutants contain fewer dopaminergic neurons than wild type, consistent with a developmental effect, and that dopaminergic cell loss in the mutant is exacerbated by the pesticides rotenone and paraquat. Overexpression of DVMAT protein does not increase the survival of animals exposed to rotenone, but blocks the loss of dopaminergic neurons caused by this pesticide. These results are the first to demonstrate an interaction between a VMAT and pesticides in vivo, and provide an important model to investigate the mechanisms by which pesticides and cellular DA may interact to kill dopaminergic cells.
Collapse
|