1
|
Lin YC, Zhang M, Chang YJ, Kuo TH. Comparisons of lifespan and stress resistance between sexes in Drosophila melanogaster. Heliyon 2023; 9:e18178. [PMID: 37576293 PMCID: PMC10415617 DOI: 10.1016/j.heliyon.2023.e18178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 07/06/2023] [Accepted: 07/10/2023] [Indexed: 08/15/2023] Open
Abstract
Animals exhibit different extents of sexual dimorphism in a variety of phenotypes. Sex differences in longevity, one of the most complex life history traits, have also been reported. Although lifespan regulation has been studied extensively in the fruit fly, Drosophila melanogaster, the sex differences in lifespan have not been consistent in previous studies. To explore this issue, we revisited this question by examining the lifespan and stress resistance of both sexes among 15 inbred strains. We first found positive correlations between males and females from the same strain in terms of lifespan and resistance to starvation and desiccation stress. Although the lifespan difference between male and female flies varied greatly depending on the strain, males across all strains collectively had a longer lifespan. In contrast, females showed better resistance to starvation and desiccation stress. We also observed greater variation in lifespan and resistance to starvation and desiccation stress in females. Unexpectedly, there was no notable correlation observed between lifespan and the three types of stress resistance in either males or females. Overall, our study provides new data regarding sexual dimorphism in fly lifespan and stress resistance; this information may promote the investigation of mechanisms underlying longevity in future research.
Collapse
Affiliation(s)
- Yu-Chiao Lin
- Institute of Systems Neuroscience, National Tsing Hua University, Hsinchu, Taiwan
| | - MingYang Zhang
- Institute of Systems Neuroscience, National Tsing Hua University, Hsinchu, Taiwan
| | | | - Tsung-Han Kuo
- Institute of Systems Neuroscience, National Tsing Hua University, Hsinchu, Taiwan
- Department of Life Science, National Tsing Hua University, Hsinchu 300, Taiwan
| |
Collapse
|
2
|
The Interplay between Gut Microbiota and Parkinson's Disease: Implications on Diagnosis and Treatment. Int J Mol Sci 2022; 23:ijms232012289. [PMID: 36293176 PMCID: PMC9603886 DOI: 10.3390/ijms232012289] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 10/05/2022] [Accepted: 10/12/2022] [Indexed: 11/05/2022] Open
Abstract
The bidirectional interaction between the gut microbiota (GM) and the Central Nervous System, the so-called gut microbiota brain axis (GMBA), deeply affects brain function and has an important impact on the development of neurodegenerative diseases. In Parkinson’s disease (PD), gastrointestinal symptoms often precede the onset of motor and non-motor manifestations, and alterations in the GM composition accompany disease pathogenesis. Several studies have been conducted to unravel the role of dysbiosis and intestinal permeability in PD onset and progression, but the therapeutic and diagnostic applications of GM modifying approaches remain to be fully elucidated. After a brief introduction on the involvement of GMBA in the disease, we present evidence for GM alterations and leaky gut in PD patients. According to these data, we then review the potential of GM-based signatures to serve as disease biomarkers and we highlight the emerging role of probiotics, prebiotics, antibiotics, dietary interventions, and fecal microbiota transplantation as supportive therapeutic approaches in PD. Finally, we analyze the mutual influence between commonly prescribed PD medications and gut-microbiota, and we offer insights on the involvement also of nasal and oral microbiota in PD pathology, thus providing a comprehensive and up-to-date overview on the role of microbial features in disease diagnosis and treatment.
Collapse
|
3
|
Lovejoy PC, Foley KE, Conti MM, Meadows SM, Bishop C, Fiumera AC. Genetic basis of susceptibility to low-dose paraquat and variation between the sexes in Drosophila melanogaster. Mol Ecol 2021; 30:2040-2053. [PMID: 33710693 DOI: 10.1111/mec.15878] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2019] [Accepted: 02/23/2021] [Indexed: 12/18/2022]
Abstract
Toxicant resistance is a complex trait, affected both by genetics and the environment. Like most complex traits, it can exhibit sexual dimorphism, yet sex is often overlooked as a factor in studies of toxicant resistance. Paraquat, one such toxicant, is a commonly used herbicide and is known to produce mitochondrial oxidative stress, decrease dopaminergic neurons and dopamine (DA) levels, and decrease motor ability. While the main effects of paraquat are well-characterized, less is known about the naturally occurring variation in paraquat susceptibility. The purpose of this study was to map the genes contributing to low-dose paraquat susceptibility in Drosophila melanogaster, and to determine if susceptibility differs between the sexes. One hundred of the Drosophila Genetic Reference Panel (DGRP) lines were scored for susceptibility via climbing ability and used in a genome-wide association study (GWAS). Variation in seventeen genes in females and thirty-five genes in males associated with paraquat susceptibility. Only two candidate genes overlapped between the sexes despite a significant positive correlation between male and female susceptibilities. Many associated polymorphisms had significant interactions with sex, with most having conditionally neutral effects. Conditional neutrality between the sexes probably stems from sex-biased expression which may result from partial resolution of sexual conflict. Candidate genes were verified with RNAi knockdowns, gene expression analyses, and DA quantification. Several of these genes are novel associations with paraquat susceptibility. This research highlights the importance of assessing both sexes when studying toxicant susceptibility.
Collapse
Affiliation(s)
- Pamela C Lovejoy
- Department of Biological Sciences, Binghamton University, Binghamton, NY, USA.,Department of Biology, St. Joseph's College, Brooklyn, NY, USA
| | - Kate E Foley
- Department of Biological Sciences, Binghamton University, Binghamton, NY, USA
| | - Melissa M Conti
- Department of Psychology, Binghamton University, Binghamton, NY, USA
| | | | | | - Anthony C Fiumera
- Department of Biological Sciences, Binghamton University, Binghamton, NY, USA
| |
Collapse
|
4
|
Maitra U, Harding T, Liang Q, Ciesla L. GardeninA confers neuroprotection against environmental toxin in a Drosophila model of Parkinson's disease. Commun Biol 2021; 4:162. [PMID: 33547411 PMCID: PMC7864937 DOI: 10.1038/s42003-021-01685-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 01/11/2021] [Indexed: 02/08/2023] Open
Abstract
Parkinson’s disease is an age-associated neurodegenerative disorder characterized by the progressive loss of dopaminergic neurons from the midbrain. Epidemiological studies have implicated exposures to environmental toxins like the herbicide paraquat as major contributors to Parkinson’s disease etiology in both mammalian and invertebrate models. We have employed a paraquat-induced Parkinson’s disease model in Drosophila as an inexpensive in vivo platform to screen therapeutics from natural products. We have identified the polymethoxyflavonoid, GardeninA, with neuroprotective potential against paraquat-induced parkinsonian symptoms involving reduced survival, mobility defects, and loss of dopaminergic neurons. GardeninA-mediated neuroprotection is not solely dependent on its antioxidant activities but also involves modulation of the neuroinflammatory and cellular death responses. Furthermore, we have successfully shown GardeninA bioavailability in the fly heads after oral administration using ultra-performance liquid chromatography and mass spectrometry. Our findings reveal a molecular mechanistic insight into GardeninA-mediated neuroprotection against environmental toxin-induced Parkinson’s disease pathogenesis for novel therapeutic intervention. Maitra and colleagues identify the neuroprotective properties of GardeninA against environmental toxin-induced neurodegeneration in Drosophila. This study has the potential to influence future research into toxin-induced Parkinson’s disease pathogenesis.
Collapse
Affiliation(s)
- Urmila Maitra
- Department of Biological Sciences, University of Alabama, 2320 Science and Engineering Complex, Tuscaloosa, AL, 35487-0344, USA.
| | - Thomas Harding
- Department of Biological Sciences, University of Alabama, 2320 Science and Engineering Complex, Tuscaloosa, AL, 35487-0344, USA
| | - Qiaoli Liang
- Mass Spectrometry Facility, Department of Chemistry and Biochemistry, University of Alabama, 2004 Shelby Hall, Tuscaloosa, AL, 35487-0336, USA
| | - Lukasz Ciesla
- Department of Biological Sciences, University of Alabama, 2320 Science and Engineering Complex, Tuscaloosa, AL, 35487-0344, USA.
| |
Collapse
|
5
|
González-Lizárraga F, Ploper D, Ávila CL, Socías SB, Dos-Santos-Pereira M, Machín B, Del-Bel E, Michel PP, Pietrasanta LI, Raisman-Vozari R, Chehín R. CMT-3 targets different α-synuclein aggregates mitigating their toxic and inflammogenic effects. Sci Rep 2020; 10:20258. [PMID: 33219264 PMCID: PMC7679368 DOI: 10.1038/s41598-020-76927-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 11/03/2020] [Indexed: 12/22/2022] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder for which only symptomatic treatments are available. Repurposing drugs that target α-synuclein aggregation, considered one of the main drivers of PD progression, could accelerate the development of disease-modifying therapies. In this work, we focused on chemically modified tetracycline 3 (CMT-3), a derivative with reduced antibiotic activity that crosses the blood–brain barrier and is pharmacologically safe. We found that CMT-3 inhibited α-synuclein amyloid aggregation and led to the formation of non-toxic molecular species, unlike minocycline. Furthermore, CMT-3 disassembled preformed α-synuclein amyloid fibrils into smaller fragments that were unable to seed in subsequent aggregation reactions. Most interestingly, disaggregated species were non-toxic and less inflammogenic on brain microglial cells. Finally, we modelled the interactions between CMT-3 and α-synuclein aggregates by molecular simulations. In this way, we propose a mechanism for fibril disassembly. Our results place CMT-3 as a potential disease modifier for PD and possibly other synucleinopathies.
Collapse
Affiliation(s)
- Florencia González-Lizárraga
- Instituto de Investigación en Medicina Molecular y Celular Aplicada (IMMCA) (CONICET-UNT-SIPROSA), Pasaje Dorrego 1080, 4000, San Miguel de Tucumán, Argentina
| | - Diego Ploper
- Instituto de Investigación en Medicina Molecular y Celular Aplicada (IMMCA) (CONICET-UNT-SIPROSA), Pasaje Dorrego 1080, 4000, San Miguel de Tucumán, Argentina
| | - César L Ávila
- Instituto de Investigación en Medicina Molecular y Celular Aplicada (IMMCA) (CONICET-UNT-SIPROSA), Pasaje Dorrego 1080, 4000, San Miguel de Tucumán, Argentina
| | - Sergio B Socías
- Instituto de Investigación en Medicina Molecular y Celular Aplicada (IMMCA) (CONICET-UNT-SIPROSA), Pasaje Dorrego 1080, 4000, San Miguel de Tucumán, Argentina
| | | | - Belén Machín
- Instituto de Investigación en Medicina Molecular y Celular Aplicada (IMMCA) (CONICET-UNT-SIPROSA), Pasaje Dorrego 1080, 4000, San Miguel de Tucumán, Argentina
| | - Elaine Del-Bel
- Faculdade de Odontologia de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Patrick Pierre Michel
- Paris Brain Institute, Inserm U 1127, CNRS UMR 7225, Sorbonne Université UM75, Paris, France
| | - Lía I Pietrasanta
- Departamento de Física-Instituto de Física de Buenos Aires (IFIBA, UBA-CONICET) and Centro de Microscopías Avanzadas (CMA), Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, C1428EHA, Buenos Aires, Argentina
| | - Rita Raisman-Vozari
- Paris Brain Institute, Inserm U 1127, CNRS UMR 7225, Sorbonne Université UM75, Paris, France.
| | - Rosana Chehín
- Instituto de Investigación en Medicina Molecular y Celular Aplicada (IMMCA) (CONICET-UNT-SIPROSA), Pasaje Dorrego 1080, 4000, San Miguel de Tucumán, Argentina.
| |
Collapse
|
6
|
Chaudhuri A, Johnson R, Rakshit K, Bednářová A, Lackey K, Chakraborty SS, Krishnan N, Chaudhuri A. Exposure to Spectracide® causes behavioral deficits in Drosophila melanogaster: Insights from locomotor analysis and molecular modeling. CHEMOSPHERE 2020; 248:126037. [PMID: 32018111 DOI: 10.1016/j.chemosphere.2020.126037] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 01/24/2020] [Accepted: 01/25/2020] [Indexed: 06/10/2023]
Abstract
This study was focused on gaining insights into the mechanism by which the herbicide- Spectracide®, induces oxidative stress and alters behavior in Drosophila melanogaster. Exposure to Spectracide® (50%) significantly (p < 0.05) reduced the negative geotaxis response, jumping behavior and dampened locomotor activity rhythm in adult flies compared to non-exposed flies. Protein carbonyl levels indicative of oxidative damage increased significantly coupled with down-regulation of Sniffer gene expression encoding carbonyl reductase (CR) and its activity in Spectracide®-exposed flies. In silico modeling analysis revealed that the active ingredients of Spectracide® (atrazine, diquat dibromide, fluazifop-p-butyl, and dicamba) have significant binding affinity to the active site of CR enzyme, with atrazine having comparatively greater affinity. Our results suggest a mechanism by which ingredients in Spectracide® induce oxidative damage by competitive binding to the active site of a protective enzyme and impair its ability to prevent damage to proteins thereby leading to deficits in locomotor behavior in Drosophila.
Collapse
Affiliation(s)
- Ankur Chaudhuri
- Department of Microbiology, West Bengal State University, Barasat, Kolkata, 126, India
| | | | - Kuntol Rakshit
- Department of Physiology and Biomedical Engineering, Mayo Clinic School of Medicine, Mayo Clinic, Rochester, MN, USA
| | - Andrea Bednářová
- Department of Biochemistry, Molecular Biology, Entomology and Plant Pathology, Mississippi State University, MS, 39762, USA; Institute of Entomology, Biology Centre, Czech Academy of Sciences, Branišovská 31, 370 05, České Budĕjovice, Czech Republic
| | - Kimberly Lackey
- Department of Biological Sciences, University of Alabama, Tuscaloosa, AL, 35487, USA
| | | | - Natraj Krishnan
- Department of Biochemistry, Molecular Biology, Entomology and Plant Pathology, Mississippi State University, MS, 39762, USA.
| | - Anathbandhu Chaudhuri
- Biology Department, Stillman College, Tuscaloosa, AL, 35404, USA; Department of Biological Sciences, University of Alabama, Tuscaloosa, AL, 35487, USA.
| |
Collapse
|
7
|
Innate immune responses to paraquat exposure in a Drosophila model of Parkinson's disease. Sci Rep 2019; 9:12714. [PMID: 31481676 PMCID: PMC6722124 DOI: 10.1038/s41598-019-48977-6] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 08/13/2019] [Indexed: 12/25/2022] Open
Abstract
Parkinson’s disease (PD) is a progressive, neurodegenerative movement disorder characterized by the loss of dopaminergic (DA) neurons. Limited understanding of the early molecular pathways associated with the demise of DA neurons, including those of inflammatory exacerbation of neurodegeneration, is a major impediment to therapeutic development. Recent studies have implicated gene-environment interactions in PD susceptibility. We used transcriptomic profiling in a Drosophila PD model in response to paraquat (PQ)-induced oxidative stress to identify pre-symptomatic signatures of impending neuron dysfunction. Our RNAseq data analysis revealed extensive regulation of innate immune response genes following PQ ingestion. We found that PQ exposure leads to the activation of the NF-κB transcription factor, Relish, and the stress signaling factor JNK, encoded by the gene basket in Drosophila. Relish knockdown in the dopaminergic neurons confers PQ resistance and rescues mobility defects and DA neuron loss. Furthermore, PQ-induced toxicity is mediated through the immune deficiency signaling pathway. Surprisingly, the expression of Relish-dependent anti-microbial peptide (AMPs) genes is suppressed upon PQ exposure causing increased sensitivity to Gram-negative bacterial infection. This work provides a novel link between PQ exposure and innate immune system modulation underlying environmental toxin-induced neurodegeneration, thereby underscoring the role of the innate immune system in PD pathogenesis.
Collapse
|
8
|
Epigallocatechin-3-Gallate Protects and Prevents Paraquat-Induced Oxidative Stress and Neurodegeneration in Knockdown dj-1-β Drosophila melanogaster. Neurotox Res 2018; 34:401-416. [PMID: 29667128 DOI: 10.1007/s12640-018-9899-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 04/02/2018] [Accepted: 04/05/2018] [Indexed: 12/28/2022]
Abstract
Epigallocatechin-3-gallate (EGCG) is a polyhydroxyphenol constituent of green tea (e.g., Camellia sinensis) with known antioxidant properties. Due to these properties, others have proposed it as a potential therapeutic agent for the treatment of Parkinson's disease (PD). Previously, we demonstrated that EGCG prolonged the lifespan and locomotor activity in wild-type Canton-S flies exposed to the neurotoxicant paraquat (PQ), suggesting neuroprotective properties. Both gene mutations and environmental neurotoxicants (e.g., PQ) are factors involved in the development of PD. Thus, the first aim of this study was to create a suitable animal model of PD, which encompasses both of these factors. To create the model, we knocked down dj-1-β function specifically in the dopaminergic neurons to generate TH > dj-1-β-RNAi/+ Drosophila melanogaster flies. Next, we induced neurotoxicity in the transgenic flies with PQ. The second aim of this study was to validate the model by comparing the effects of vehicle, EGCG, and chemicals with known antioxidant and neuroprotective properties in vivo (e.g., propyl gallate and minocycline) on life-span, locomotor activity, lipid peroxidation, and neurodegeneration. The EGCG treatment provided protection and prevention from the PQ-induced reduction in the life-span and locomotor activity and from the PQ-induced increase in lipid peroxidation and neurodegeneration. These effects were augmented in the EGCG-treated flies when compared to the flies treated with either PG or MC. Altogether, these results suggest that the transgenic TH > dj-1-β-RNAi/+ flies treated with PQ serve as a suitable PD model for screening of potential therapeutic agents.
Collapse
|
9
|
Sanz FJ, Solana-Manrique C, Muñoz-Soriano V, Calap-Quintana P, Moltó MD, Paricio N. Identification of potential therapeutic compounds for Parkinson's disease using Drosophila and human cell models. Free Radic Biol Med 2017; 108:683-691. [PMID: 28455141 DOI: 10.1016/j.freeradbiomed.2017.04.364] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Revised: 04/11/2017] [Accepted: 04/17/2017] [Indexed: 12/15/2022]
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disorder after Alzheimer's disease. It is caused by a loss of dopaminergic neurons in the substantia nigra pars compacta, leading to a decrease in dopamine levels in the striatum and thus producing movement impairment. Major physiological causes of neurodegeneration in PD are oxidative stress (OS) and mitochondrial dysfunction; these pathophysiological changes can be caused by both genetic and environmental factors. Although most PD cases are sporadic, it has been shown that 5-10% of them are familial forms caused by mutations in certain genes. One of these genes is the DJ-1 oncogene, which is involved in an early-onset recessive PD form. Currently, PD is an incurable disease for which existing therapies are not sufficiently effective to counteract or delay the progression of the disease. Therefore, the discovery of alternative drugs for the treatment of PD is essential. In this study we used a Drosophila PD model to identify candidate compounds with therapeutic potential for this disease. These flies carry a loss-of-function mutation in the DJ-1β gene, the Drosophila ortholog of human DJ-1, and show locomotor defects reflected by a reduced climbing ability. A pilot modifier chemical screen was performed, and several candidate compounds were identified based on their ability to improve locomotor activity of PD model flies. We demonstrated that some of them were also able to reduce OS levels in these flies. To validate the compounds identified in the Drosophila screen, a human cell PD model was generated by knocking down DJ-1 function in SH-SY5Y neuroblastoma cells. Our results showed that some of the compounds were also able to increase the viability of the DJ-1-deficient cells subjected to OS, thus supporting the use of Drosophila for PD drug discovery. Interestingly, some of them have been previously proposed as alternative therapies for PD or tested in clinical trials and others are first suggested in this study as potential drugs for the treatment of this disease.
Collapse
Affiliation(s)
- Francisco José Sanz
- Departamento de Genética, Facultad CC Biológicas, Universidad de Valencia, 46100 Burjassot, Spain; Estructura de Recerca Interdisciplinar en Biotecnologia i Biomedicina (ERI BIOTECMED), Universidad de Valencia, 46100 Burjassot, Spain
| | - Cristina Solana-Manrique
- Departamento de Genética, Facultad CC Biológicas, Universidad de Valencia, 46100 Burjassot, Spain; Estructura de Recerca Interdisciplinar en Biotecnologia i Biomedicina (ERI BIOTECMED), Universidad de Valencia, 46100 Burjassot, Spain
| | - Verónica Muñoz-Soriano
- Departamento de Genética, Facultad CC Biológicas, Universidad de Valencia, 46100 Burjassot, Spain; Estructura de Recerca Interdisciplinar en Biotecnologia i Biomedicina (ERI BIOTECMED), Universidad de Valencia, 46100 Burjassot, Spain
| | - Pablo Calap-Quintana
- Departamento de Genética, Facultad CC Biológicas, Universidad de Valencia, 46100 Burjassot, Spain
| | - María Dolores Moltó
- Departamento de Genética, Facultad CC Biológicas, Universidad de Valencia, 46100 Burjassot, Spain; CIBERSAM, INCLIVA. Valencia, Spain
| | - Nuria Paricio
- Departamento de Genética, Facultad CC Biológicas, Universidad de Valencia, 46100 Burjassot, Spain; Estructura de Recerca Interdisciplinar en Biotecnologia i Biomedicina (ERI BIOTECMED), Universidad de Valencia, 46100 Burjassot, Spain.
| |
Collapse
|
10
|
Parashar A, Udayabanu M. Gut microbiota: Implications in Parkinson's disease. Parkinsonism Relat Disord 2017; 38:1-7. [PMID: 28202372 PMCID: PMC7108450 DOI: 10.1016/j.parkreldis.2017.02.002] [Citation(s) in RCA: 154] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Revised: 12/24/2016] [Accepted: 02/04/2017] [Indexed: 12/22/2022]
Abstract
Gut microbiota (GM) can influence various neurological outcomes, like cognition, learning, and memory. Commensal GM modulates brain development and behavior and has been implicated in several neurological disorders like Alzheimer's disease, multiple sclerosis, amyotrophic lateral sclerosis, anxiety, stress and much more. A recent study has shown that Parkinson's disease patients suffer from GM dysbiosis, but whether it is a cause or an effect is yet to be understood. In this review, we try to connect the dots between GM and PD pathology using direct and indirect evidence.
Collapse
Affiliation(s)
- Arun Parashar
- Jaypee University of Information Technology, Waknaghat, District- Solan, Himachal Pradesh, PIN-173234, India
| | - Malairaman Udayabanu
- Jaypee University of Information Technology, Waknaghat, District- Solan, Himachal Pradesh, PIN-173234, India.
| |
Collapse
|
11
|
Ortega-Arellano HF, Jimenez-Del-Rio M, Velez-Pardo C. Minocycline protects, rescues and prevents knockdown transgenic parkin Drosophila against paraquat/iron toxicity: Implications for autosomic recessive juvenile parkinsonism. Neurotoxicology 2017; 60:42-53. [PMID: 28284907 DOI: 10.1016/j.neuro.2017.03.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Revised: 02/03/2017] [Accepted: 03/06/2017] [Indexed: 01/03/2023]
Abstract
Autosomal recessive Juvenile Parkinsonism (AR-JP) is a chronic, progressive neurodegenerative disorder caused by mutation in the PARKIN gene, and invariably associated with dopaminergic (DAergic) neuronal loss and brain iron accumulation. Since current medical therapy is symptomatic and lacks significant disease-modifying effects, other treatment approaches are urgently needed it. In the present work, we investigate the role of minocycline (MC) in paraquat (PQ)/iron-induced neurotoxicity in the Drosophila TH>parkin-RNAi/+ (w[*]; UAS-parkin-RNAi; TH-GAL4) fly and have shown the following: (i) MC increased life span and restored the locomotor activity of knockdown (KD) transgenic parkin flies in comparison with the control (vehicle) group; (ii) MC at low (0.1 and 0.3mM) and middle (0.5mM) concentrations protected, rescued and prevented KD parkin Drosophila against PQ toxicity. However, MC at high (1mM) concentration aggravated the toxic effect of PQ; (iii) MC protected and rescued DAergic neurons against the PQ toxic effect according to tyrosine hydroxylase (TH)>green-fluorescent protein (GFP) reporter protein microscopy and anti-TH Western blotting analysis; (iv) MC protected DAergic neurons against PQ/iron toxicity; (v) MC significantly abridged lipid peroxidation (LPO) in the protection, rescue and prevention treatment in TH>parkin-RNAi/+ flies against PQ or iron alone or combined (PQ/iron)-induced neuronal oxidative stress (OS). Our results suggest that MC exerts neuroprotection against PQ/iron-induced OS in DAergic neurons most probably by the scavenging activity of reactive oxygen species (ROS), and by chelating iron. Therefore, MC might be a potential therapeutic drug to delay, revert, or prevent AR-JP.
Collapse
Affiliation(s)
- Hector Flavio Ortega-Arellano
- Neuroscience Research Group, Medical Research Institute, Faculty of Medicine, University of Antioquia (UdeA), Calle 70 No. 52-21, and Calle 62 # 52-59, Building 1, Room 412, SIU, Medellin, Colombia
| | - Marlene Jimenez-Del-Rio
- Neuroscience Research Group, Medical Research Institute, Faculty of Medicine, University of Antioquia (UdeA), Calle 70 No. 52-21, and Calle 62 # 52-59, Building 1, Room 412, SIU, Medellin, Colombia.
| | - Carlos Velez-Pardo
- Neuroscience Research Group, Medical Research Institute, Faculty of Medicine, University of Antioquia (UdeA), Calle 70 No. 52-21, and Calle 62 # 52-59, Building 1, Room 412, SIU, Medellin, Colombia.
| |
Collapse
|
12
|
Phom L, Achumi B, Alone DP, Muralidhara, Yenisetti SC. Curcumin's neuroprotective efficacy in Drosophila model of idiopathic Parkinson's disease is phase specific: implication of its therapeutic effectiveness. Rejuvenation Res 2015; 17:481-9. [PMID: 25238331 DOI: 10.1089/rej.2014.1591] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Selective degeneration of dopaminergic neurons in the substantia nigra underlies the basic motor impairments of Parkinson's disease (PD). Curcumin has been used for centuries in traditional medicines in India. Our aim is to understand the efficacy of genotropic drug curcumin as a neuroprotective agent in PD. Analysis of different developmental stages in model organisms revealed that they are characterized by different patterns of gene expression which is similar to that of developmental stages of human. Genotropic drugs would be effective only during those life cycle stages for which their target molecules are available. Hence there exists a possibility that targets of genotropic compounds such as curcumin may not be present in all life stages. However, no reports are available in PD models illustrating the efficacy of curcumin in later phases of adult life. This is important because this is the period during which late-onset disorders such as idiopathic PD set in. To understand this paradigm, we tested the protective efficacy of curcumin in different growth stages (early, late health stage, and transition phase) in adult Drosophila flies. Results showed that it can rescue the motor defects during early stages of life but is ineffective at later phases. This observation was substantiated with the finding that curcumin treatment could replenish depleted brain dopamine levels in the PD model only during early stages of life cycle, clearly suggesting its limitation as a therapeutic agent in late-onset neurodegenerative disorders such as PD.
Collapse
Affiliation(s)
- Limamanen Phom
- 1 Drosophila Neurobiology Laboratory, Department of Zoology, Nagaland University (Central) , Lumami, Nagaland, India
| | | | | | | | | |
Collapse
|
13
|
Lisiecka DM, Suckling J, Barnes TRE, Chaudhry IB, Dazzan P, Husain N, Jones PB, Joyce EM, Lawrie SM, Upthegrove R, Deakin B. The benefit of minocycline on negative symptoms in early-phase psychosis in addition to standard care - extent and mechanism (BeneMin): study protocol for a randomised controlled trial. Trials 2015; 16:71. [PMID: 25886254 PMCID: PMC4351843 DOI: 10.1186/s13063-015-0580-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Accepted: 01/22/2015] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Negative symptoms of psychosis do not respond to the traditional therapy with first- or second-generation antipsychotics and are among main causes of a decrease in quality of life observed in individuals suffering from the disorder. Minocycline, a broad-spectrum tetracyclic antibiotic displaying neuroprotective properties has been suggested as a new potential therapy for negative symptoms. In the two previous clinical trials comparing minocycline and placebo, both added to the standard care, patients receiving minocycline showed increased reduction in negative symptoms. Three routes to neuroprotection by minocycline have been identified: neuroprotection against grey matter loss, anti-inflammatory action and stabilisation of glutamate receptors. However, it is not yet certain what the extent of the benefit of minocycline in psychosis is and what its mechanism is. We present a protocol for a multi-centre double-blind randomised placebo-controlled clinical trial entitled The Benefit of Minocycline on Negative Symptoms of Psychosis: Extent and Mechanism (BeneMin). METHODS After providing informed consent, 226 participants in the early phase of psychosis will be randomised to receive either 100 mg modified-release capsules of minocycline or similar capsules with placebo for 12 months in addition to standard care. The participants will be tested for outcome variables before and after the intervention period. The extent of benefit will be tested via clinical outcome measures, namely the Positive and Negative Syndrome Scale score, social and cognitive functioning scores, antipsychotic medication dose equivalent and level of weight gain. The mechanism of action of minocycline will be tested via blood screening for circulating cytokines and magnetic resonance imaging with three-dimensional T1-weighted rapid gradient-echo, proton density T2-weighted dual echo and T2*-weighted gradient echo planar imaging with N-back task and resting state. Eight research centres in UK and 15 National Health Service Trusts and Health Boards will be involved in recruiting participants, performing the study and analysing the data. DISCUSSION The BeneMin trial can inform as to whether in minocycline we have found a new and effective therapy against negative symptoms of psychosis. The European Union Clinical Trial Register: EudraCT 2010-022463-35 with the registration finalised in July 2011. The recruitment in the trial started in January 2013 with the first patient recruited in March 2013.
Collapse
Affiliation(s)
- Danuta M Lisiecka
- Department of Psychiatry, Behavioural and Clinical Neuroscience Institute, University of Cambridge, Cambridge, UK.
- Department of Psychiatry, Brain Mapping Unit, University of Cambridge, Herchel Smith Building for Brain and Mind Sciences, Robinson Way, Cambridge, CB2 0SZ, UK.
| | - John Suckling
- Department of Psychiatry, Behavioural and Clinical Neuroscience Institute, University of Cambridge, Cambridge, UK.
- Department of Psychiatry, Brain Mapping Unit, University of Cambridge, Herchel Smith Building for Brain and Mind Sciences, Robinson Way, Cambridge, CB2 0SZ, UK.
- Cambridge and Peterborough NHS Foundation Trust, Cambridge, UK.
| | - Thomas R E Barnes
- Department of Medicine, Centre for Mental Health, Faculty of Medicine, Imperial College, London, UK.
- West London Mental Health NHS Trust, London, UK.
| | - Imran B Chaudhry
- Institute of Brain, Behaviour and Mental Health, Clinical and Cognitive Neurosciences, University of Manchester, Manchester, UK.
- Lancashire Care Early Intervention Service, Accrington, UK.
| | - Paola Dazzan
- Department of Psychosis Studies, Institute of Psychiatry, King's College, London, UK.
| | - Nusrat Husain
- Institute of Brain, Behaviour and Mental Health, Clinical and Cognitive Neurosciences, University of Manchester, Manchester, UK.
| | - Peter B Jones
- Cambridge and Peterborough NHS Foundation Trust, Cambridge, UK.
- Department of Psychiatry, University of Cambridge, Cambridge, UK.
| | - Eileen M Joyce
- Institute of Neurology, University College London, London, UK.
| | - Stephen M Lawrie
- Division of Psychiatry, University of Edinburgh, Royal Edinburgh Hospital, Edinburgh, UK.
| | - Rachel Upthegrove
- School of Clinical and Experimental Medicine, University of Birmingham, Birmingham, UK.
- Early Intervention Service, Birmingham and Solihull Mental Health NHS Foundation Trust, Birmingham, UK.
| | - Bill Deakin
- Institute of Brain, Behaviour and Mental Health, Clinical and Cognitive Neurosciences, University of Manchester, Manchester, UK.
- Manchester Mental Health and Social Care Trust, Manchester, UK.
| |
Collapse
|
14
|
Wang X, Wang ML, Lu XY, Zhang P, Yu HG, Hu YK. Glucagon-like peptide-1 suppresses palmitic acid induced L02 cell apoptosis through JNK pathway. Shijie Huaren Xiaohua Zazhi 2015; 23:16-21. [DOI: 10.11569/wcjd.v23.i1.16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the potential effects of glucagon-like peptide-1 (GLP-1) on palmitic acid (PA) induced apoptosis of hepatic L02 cells and the underlying mechanism.
METHODS: L02 cells were stimulated with different levels (0.125, 0.250 and 0.500 mmol/L) of PA for different durations (12, 24 and 48 h) in the presence or absence of GLP-1. Cell Counting Kit-8 (CCK-8) assay was used to analyze the inhibitory effects on growth of L02 cells, and terminal-deoxynucleoitidyl transferase mediated nick end labeling (TUNEL) assay was used to observe the apoptotic rate of the cells. The expression of c-Jun N-terminal kinase (JNK), c-Jun, p-JNK and p-c-Jun proteins was detected by Western blot assay.
RESULTS: The growth of the L02 cells was significantly inhibited by PA in vitro, and PA induced the expression of p-JNK and p-c-Jun. GLP-1 suppressed the activation of JNK and c-Jun induced by PA.
CONCLUSION: PA can inhibit the proliferation of L02 cells and induce cell apoptosis. The JNK signaling pathway is probably involved in the mechanism of PA induced apoptosis.
Collapse
|
15
|
Signaling pathways involved in 1-octen-3-ol-mediated neurotoxicity in Drosophila melanogaster: implication in Parkinson’s disease. Neurotox Res 2014; 25:183-91. [PMID: 23959949 DOI: 10.1007/s12640-013-9418-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2013] [Revised: 08/06/2013] [Accepted: 08/06/2013] [Indexed: 01/20/2023]
Abstract
Previously, we have pioneered Drosophila melanogaster as a reductionist model to show that 1-octen-3-ol, a musty-smelling volatile compound emitted by fungi and other organisms, causes loss of dopaminergic neurons and Parkinson’s disease-like symptoms in flies. Using our in vivo Drosophila system, the modulatory roles of important signaling pathways—JNK, Akt and the caspase-3-dependent apoptotic pathway were investigated in the context of 1-octen-3-ol-induced dopamine neurotoxicity. When heterozygous flies carrying mutant alleles for these proteins were exposed to 0.5 ppm of 1-octen-3-ol, they had shorter survival times than wild-type Drosophila. The overexpressed levels of wild-type JNK and Akt, (UAS-bsk and UAS-Akt) with TH-GAL4 and elav-GAL4 drivers improved the survival duration of exposed flies compared with controls. Thus, we found that Akt and JNK both protect against loss of dopamine activity associated with 1-octen-3-ol exposure, indicating the pro-survival role of these signaling pathways. Further, 1-octen-3-ol exposure was associated with activation of caspase 3, a hallmark for apoptosis.
Collapse
|
16
|
Minocycline increases the activity of superoxide dismutase and reduces the concentration of nitric oxide, hydrogen peroxide and mitochondrial malondialdehyde in manganese treated Drosophila melanogaster. Neurochem Res 2014; 39:1270-8. [PMID: 24756376 DOI: 10.1007/s11064-014-1309-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2014] [Revised: 04/02/2014] [Accepted: 04/11/2014] [Indexed: 10/25/2022]
Abstract
The toxicity caused by high concentrations of manganese (Mn) could be due to a production of free radicals. Minocycline is an effective antioxidant with a high potential to capture free radicals. We investigated the effect of minocycline in the activities of superoxide dismutase (SOD) and catalase, and in the concentrations of nitric oxide (NO), hydrogen peroxide (H2O2) and mitochondrial malondialdehyde (MDA) in manganese-treated Drosophila melanogaster. Five groups of flies were used: (1) control: not treated; (2) continuously treated with minocycline (0.05 mM); (3) treated with 30 mM Mn for 6 days and then no additional treatment; (4) continuously treated with Mn; (5) treated only with Mn for 6 days and then treated with minocycline; (6) simultaneously treated with Mn and minocycline. On the 6th day, Mn treatment caused 50% mortality; in the surviving flies increased levels of MDA (67.93%), NO (11.04%), H2O2 (14.62%) and SOD and catalase activity (165.34 and 71.43%, respectively) were detected. All the flies continuously treated with Mn died by the 21st day. On day 40, MDA levels were decreased in groups two, three and five (43.04, 29.67, and 34.72% respectively), as well as NO in group two (29.21%) and H2O2 in groups two and five (53.94% and 78.69%, respectively), while in group three the concentration of H2O2 was increased (408.25%). In conclusion, Mn exerted a pro-oxidant effect on the 6th day as shown by the increased levels of oxidative markers. Minocycline extended the lifespan, increased the activity of SOD and reduced the levels of NO, H2O2 and mitochondrial MDA.
Collapse
|
17
|
Ortega-Arellano HF, Jimenez-Del-Rio M, Velez-Pardo C. Dmp53, basket and drICE gene knockdown and polyphenol gallic acid increase life span and locomotor activity in a Drosophila Parkinson's disease model. Genet Mol Biol 2013; 36:608-15. [PMID: 24385865 PMCID: PMC3873193 DOI: 10.1590/s1415-47572013000400020] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2013] [Accepted: 10/08/2013] [Indexed: 12/22/2022] Open
Abstract
Understanding the mechanism(s) by which dopaminergic (DAergic) neurons are eroded in Parkinson's disease (PD) is critical for effective therapeutic strategies. By using the binary tyrosine hydroxylase (TH)-Gal4/UAS-X RNAi Drosophila melanogaster system, we report that Dmp53, basket and drICE gene knockdown in dopaminergic neurons prolong life span (p < 0.05; log-rank test) and locomotor activity (p < 0.05; χ(2) test) in D. melanogaster lines chronically exposed to (1 mM) paraquat (PQ, oxidative stress (OS) generator) compared to untreated transgenic fly lines. Likewise, knockdown flies displayed higher climbing performance than control flies. Amazingly, gallic acid (GA) significantly protected DAergic neurons, ameliorated life span, and climbing abilities in knockdown fly lines treated with PQ compared to flies treated with PQ only. Therefore, silencing specific gene(s) involved in neuronal death might constitute an excellent tool to study the response of DAergic neurons to OS stimuli. We propose that a therapy with antioxidants and selectively "switching off" death genes in DAergic neurons could provide a means for pre-clinical PD individuals to significantly ameliorate their disease condition.
Collapse
Affiliation(s)
- Hector Flavio Ortega-Arellano
- Neuroscience Research Group, Medical Research Institute, Faculty of Medicine, University of Antioquia, Medellin, Colombia
| | - Marlene Jimenez-Del-Rio
- Neuroscience Research Group, Medical Research Institute, Faculty of Medicine, University of Antioquia, Medellin, Colombia
| | - Carlos Velez-Pardo
- Neuroscience Research Group, Medical Research Institute, Faculty of Medicine, University of Antioquia, Medellin, Colombia
| |
Collapse
|
18
|
Jahromi SR, Haddadi M, Shivanandappa T, Ramesh SR. Neuroprotective effect of Decalepis hamiltonii in paraquat-induced neurotoxicity in Drosophila melanogaster: biochemical and behavioral evidences. Neurochem Res 2013; 38:2616-24. [PMID: 24173775 DOI: 10.1007/s11064-013-1179-9] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2013] [Revised: 10/08/2013] [Accepted: 10/10/2013] [Indexed: 01/24/2023]
Abstract
In this paper, we have demonstrated for the first time, the antioxidant and neuroprotective effects of Decalepis hamiltonii (Dh) root extract against paraquat (PQ)-induced oxidative stress and neurotoxicity in Drosophila melanogaster. Exposure of adult D. melanogaster (Oregon K) to PQ induced oxidative stress as evidenced by glutathione depletion, lipid peroxidation and enhanced activities of antioxidant enzymes such as catalase, superoxide dismutase as well as elevated levels of acetylcholine esterase. Pretreatment of flies by feeding with Dh extract (0.1, 0.5 %) for 14 days boosted the activities of antioxidant enzymes and prevented the PQ-induced oxidative stress. Dietary feeding of Dh extract prior to PQ exposure showed a lower incidence of mortality and enhanced motor activities of flies in a negative geotaxis assay; both suggesting the neuroprotective potential of Dh. Based on the results, we contemplate that the roots of Dh might prevent and ameliorate the human diseases caused by oxidative stress. The neuroprotective action of Dh can be attributed to the antioxidant constituents while the precise mechanism of its action needs further investigations.
Collapse
Affiliation(s)
- Samaneh Reiszadeh Jahromi
- Department of Studies in Zoology, University of Mysore, Manasagangotri, Mysore, 570006, Karnataka, India
| | | | | | | |
Collapse
|
19
|
Zheng HF, Yang YP, Hu LF, Wang MX, Wang F, Cao LD, Li D, Mao CJ, Xiong KP, Wang JD, Liu CF. Autophagic impairment contributes to systemic inflammation-induced dopaminergic neuron loss in the midbrain. PLoS One 2013; 8:e70472. [PMID: 23936437 PMCID: PMC3735600 DOI: 10.1371/journal.pone.0070472] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2013] [Accepted: 06/19/2013] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Neuroinflammation plays an important role in the pathogenesis of Parkinson's disease (PD), inducing and accelerating dopaminergic (DA) neuron loss. Autophagy, a critical mechanism for clearing misfolded or aggregated proteins such as α-synuclein (α-SYN), may affect DA neuron survival in the midbrain. However, whether autophagy contributes to neuroinflammation-induced toxicity in DA neurons remains unknown. RESULTS Intraperitoneal injection of lipopolysaccharide (LPS, 5 mg/kg) into young (3-month-old) and aged (16-month-old) male C57BL/6J mice was observed to cause persistent neuroinflammation that was associated with a delayed and progressive loss of DA neurons and accumulation of α-SYN in the midbrain. The autophagic substrate-p62 (SQSTM1) persistently increased, whereas LC3-II and HDAC6 exhibited early increases followed by a decline. In vitro studies further demonstrated that TNF-α induced cell death in PC12 cells. Moreover, a sublethal dose of TNF-α (50 ng/ml) increased the expression of LC3-II, p62, and α-SYN, implying that TNF-α triggered autophagic impairment in cells. CONCLUSION Neuroinflammation may cause autophagic impairment, which could in turn result in DA neuron degeneration in midbrain.
Collapse
Affiliation(s)
- Hui-Fen Zheng
- Department of Neurology, Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Institute of Neuroscience, Soochow University, Suzhou, Jiangsu, China
- Department of Neurology, Yixing People’s Hospital, Yixing, Jiangsu, China
| | - Ya-Ping Yang
- Department of Neurology, Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Institute of Neuroscience, Soochow University, Suzhou, Jiangsu, China
| | - Li-Fang Hu
- Department of Neurology, Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Institute of Neuroscience, Soochow University, Suzhou, Jiangsu, China
| | - Mei-Xia Wang
- Department of Neurology, Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Institute of Neuroscience, Soochow University, Suzhou, Jiangsu, China
| | - Fen Wang
- Department of Neurology, Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Institute of Neuroscience, Soochow University, Suzhou, Jiangsu, China
| | - Li-Dan Cao
- Department of Neurology, Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Institute of Neuroscience, Soochow University, Suzhou, Jiangsu, China
| | - Da Li
- Department of Neurology, Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Institute of Neuroscience, Soochow University, Suzhou, Jiangsu, China
| | - Cheng-Jie Mao
- Department of Neurology, Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Institute of Neuroscience, Soochow University, Suzhou, Jiangsu, China
| | - Kang-Ping Xiong
- Department of Neurology, Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Institute of Neuroscience, Soochow University, Suzhou, Jiangsu, China
| | - Jian-Da Wang
- Department of Neurology, Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Institute of Neuroscience, Soochow University, Suzhou, Jiangsu, China
| | - Chun-Feng Liu
- Department of Neurology, Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Institute of Neuroscience, Soochow University, Suzhou, Jiangsu, China
- * E-mail:
| |
Collapse
|