1
|
Radzimirski A, Croft M, Ireland N, Miller L, Newell-Caito J, Caito S. Dopaminergic- and Serotonergic-Dependent Behaviors Are Altered by Lanthanide Series Metals in Caenorhabditis elegans. TOXICS 2024; 12:754. [PMID: 39453174 PMCID: PMC11511074 DOI: 10.3390/toxics12100754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 10/07/2024] [Accepted: 10/15/2024] [Indexed: 10/26/2024]
Abstract
The lanthanide series elements are transition metals used as critical components of electronics, as well as rechargeable batteries, fertilizers, antimicrobials, contrast agents for medical imaging, and diesel fuel additives. With the surge in their utilization, lanthanide metals are being found more in our environment. However, little is known about the health effects associated with lanthanide exposure. Epidemiological studies as well as studies performed in rodents exposed to lanthanum (La) suggest neurological damage, learning and memory impairment, and disruption of neurotransmitter signaling, particularly in serotonin and dopamine pathways. Unfortunately, little is known about the neurological effects of heavier lanthanides. As dysfunctions of serotonergic and dopaminergic signaling are implicated in multiple neurological conditions, including Parkinson's disease, depression, generalized anxiety disorder, and post-traumatic stress disorder, it is of utmost importance to determine the effects of La and other lanthanides on these neurotransmitter systems. We therefore hypothesized that early-life exposure of light [La (III) or cerium (Ce (III))] or heavy [erbium (Er (III)) or ytterbium (Yb (III))] lanthanides in Caenorhabditis elegans could cause dysregulation of serotonergic and dopaminergic signaling upon adulthood. Serotonergic signaling was assessed by measuring pharyngeal pump rate, crawl-to-swim transition, as well as egg-laying behaviors. Dopaminergic signaling was assessed by measuring locomotor rate and egg-laying and swim-to-crawl transition behaviors. Treatment with La (III), Ce (III), Er (III), or Yb (III) caused deficits in serotonergic or dopaminergic signaling in all assays, suggesting both the heavy and light lanthanides disrupt these neurotransmitter systems. Concomitant with dysregulation of neurotransmission, all four lanthanides increased reactive oxygen species (ROS) generation and decreased glutathione and ATP levels. This suggests increased oxidative stress, which is a known modifier of neurotransmission. Altogether, our data suggest that both heavy and light lanthanide series elements disrupt serotonergic and dopaminergic signaling and may affect the development or pharmacological management of related neurological conditions.
Collapse
Affiliation(s)
- Anthony Radzimirski
- Department of Pharmaceutical Sciences, School of Pharmacy, Husson University, Bangor, ME 04401, USA
| | - Michael Croft
- Department of Molecular & Biomedical Sciences, University of Maine, Orono, ME 04469, USA (J.N.-C.)
| | - Nicholas Ireland
- Department of Pharmaceutical Sciences, School of Pharmacy, Husson University, Bangor, ME 04401, USA
| | - Lydia Miller
- Department of Pharmaceutical Sciences, School of Pharmacy, Husson University, Bangor, ME 04401, USA
| | - Jennifer Newell-Caito
- Department of Molecular & Biomedical Sciences, University of Maine, Orono, ME 04469, USA (J.N.-C.)
| | - Samuel Caito
- Department of Pharmaceutical Sciences, School of Pharmacy, Husson University, Bangor, ME 04401, USA
| |
Collapse
|
2
|
Ohgita T, Kono H, Morita I, Oyama H, Shimanouchi T, Kobayashi N, Saito H. Intramolecular interaction kinetically regulates fibril formation by human and mouse α-synuclein. Sci Rep 2023; 13:10885. [PMID: 37407638 DOI: 10.1038/s41598-023-38070-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 07/02/2023] [Indexed: 07/07/2023] Open
Abstract
Regulation of α-synuclein (αS) fibril formation is a potent therapeutic strategy for αS-related neurodegenerative disorders. αS, an intrinsically disordered 140-residue intraneural protein, comprises positively charged N-terminal, hydrophobic non-amyloid β component (NAC), and negatively charged C-terminal regions. Although mouse and human αS share 95% sequence identity, mouse αS forms amyloid fibrils faster than human αS. To evaluate the kinetic regulation of αS fibrillation, we examined the effects of mismatched residues in human and mouse αS on fibril formation and intramolecular interactions. Thioflavin T fluorescence assay using domain-swapped or C-terminal-truncated αS variants revealed that mouse αS exhibited higher nucleation and fibril elongation than human αS. In mouse αS, S87N substitution in the NAC region rather than A53T substitution is dominant for enhanced fibril formation. Fӧrester resonance energy transfer analysis demonstrated that the intramolecular interaction of the C-terminal region with the N-terminal and NAC regions observed in human αS is perturbed in mouse αS. In mouse αS, S87N substitution is responsible for the perturbed interaction. These results indicate that the interaction of the C-terminal region with the N-terminal and NAC regions suppresses αS fibril formation and that the human-to-mouse S87N substitution in the NAC region accelerates αS fibril formation by perturbing intramolecular interaction.
Collapse
Affiliation(s)
- Takashi Ohgita
- Laboratory of Biophysical Chemistry, Kyoto Pharmaceutical University, 5 Misasagi-Nakauchi-cho, Yamashina-ku, Kyoto, 607-8414, Japan.
| | - Hiroki Kono
- Laboratory of Biophysical Chemistry, Kyoto Pharmaceutical University, 5 Misasagi-Nakauchi-cho, Yamashina-ku, Kyoto, 607-8414, Japan
| | - Izumi Morita
- Department of Bioanalytical Chemistry, Kobe Pharmaceutical University, 4-19-1 Motoyama-Kitamachi, Higashinada-ku, Kobe, 658-8558, Japan
| | - Hiroyuki Oyama
- Department of Bioanalytical Chemistry, Kobe Pharmaceutical University, 4-19-1 Motoyama-Kitamachi, Higashinada-ku, Kobe, 658-8558, Japan
| | - Toshinori Shimanouchi
- Graduate School of Environmental and Life Science, Okayama University, Okayama, 700-8530, Japan
| | - Norihiro Kobayashi
- Department of Bioanalytical Chemistry, Kobe Pharmaceutical University, 4-19-1 Motoyama-Kitamachi, Higashinada-ku, Kobe, 658-8558, Japan
| | - Hiroyuki Saito
- Laboratory of Biophysical Chemistry, Kyoto Pharmaceutical University, 5 Misasagi-Nakauchi-cho, Yamashina-ku, Kyoto, 607-8414, Japan
| |
Collapse
|
3
|
Concha-Marambio L, Weber S, Farris CM, Dakna M, Lang E, Wicke T, Ma Y, Starke M, Ebentheuer J, Sixel-Döring F, Muntean ML, Schade S, Trenkwalder C, Soto C, Mollenhauer B. Accurate Detection of α-Synuclein Seeds in Cerebrospinal Fluid from Isolated Rapid Eye Movement Sleep Behavior Disorder and Patients with Parkinson's Disease in the DeNovo Parkinson (DeNoPa) Cohort. Mov Disord 2023; 38:567-578. [PMID: 36781413 PMCID: PMC10153075 DOI: 10.1002/mds.29329] [Citation(s) in RCA: 35] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 12/06/2022] [Accepted: 01/06/2023] [Indexed: 02/15/2023] Open
Abstract
BACKGROUND Misfolded α-synuclein (αSyn) aggregates (αSyn-seeds) in cerebrospinal fluid (CSF) are biomarkers for synucleinopathies such as Parkinson's disease (PD). αSyn-seeds have been detected in prodromal cases with isolated rapid eye movement sleep behavior disorder (iRBD). OBJECTIVES The objective of this study was to determine the accuracy of the αSyn-seed amplification assay (αS-SAA) in a comprehensively characterized cohort with a high proportion of PD and iRBD CSF samples collected at baseline. METHODS We used a high-throughput αS-SAA to analyze 233 blinded CSF samples from 206 participants of the DeNovo Parkinson Cohort (DeNoPa) (113 de novo PD, 64 healthy controls, 29 iRBD confirmed by video polysomnography). Results were compared with the final diagnosis, which was determined after up to 10 years of longitudinal clinical evaluations, including dopamine-transporter-single-photon emission computed tomography (DAT-SPECT) at baseline, CSF proteins, Movement Disorder Society-Unified Parkinson's Disease Rating Scale, and various cognitive and nonmotor scales. RESULTS αS-SAA detected αSyn-seeds in baseline PD-CSF with 98% accuracy. αSyn-seeds were detected in 93% of the iRBD cases. αS-SAA results showed higher agreement with the final than the initial diagnosis, as 14 patients were rediagnosed as non-αSyn aggregation disorder. For synucleinopathies, αS-SAA showed higher concordance with the final diagnosis than DAT-SPECT. Statistically significant correlations were found between assay parameters and disease progression. CONCLUSIONS Our results confirm αS-SAA accuracy at the first clinical evaluation when a definite diagnosis is most consequential. αS-SAA conditions reported here are highly sensitive, enabling the detection of αSyn-seeds in CSF from iRBD just months after the first symptoms, suggesting that αSyn-seeds are present in the very early prodromal phase of synucleinopathies. Therefore, αSyn-seeds are clear risk markers for synuclein-related disorders, but not for time of phenoconversion. © 2023 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
| | - Sandrina Weber
- Department of Neurology, University Medical Centre Goettingen, Robert-Koch Str. 40, 37073 Goettingen, Germany
- Paracelsus-Elena-Klinik, Klinikstrasse 16, 34119 Kassel, Germany
| | - Carly M. Farris
- R&D Unit, Amprion Inc., 11095 Flintkote Av., San Diego, California, 92121, USA
| | - Mohammed Dakna
- Department of Neurology, University Medical Centre Goettingen, Robert-Koch Str. 40, 37073 Goettingen, Germany
| | - Elisabeth Lang
- Paracelsus-Elena-Klinik, Klinikstrasse 16, 34119 Kassel, Germany
| | - Tamara Wicke
- Paracelsus-Elena-Klinik, Klinikstrasse 16, 34119 Kassel, Germany
| | - Yihua Ma
- R&D Unit, Amprion Inc., 11095 Flintkote Av., San Diego, California, 92121, USA
| | - Maritta Starke
- Paracelsus-Elena-Klinik, Klinikstrasse 16, 34119 Kassel, Germany
| | - Jens Ebentheuer
- Paracelsus-Elena-Klinik, Klinikstrasse 16, 34119 Kassel, Germany
| | - Friederike Sixel-Döring
- Paracelsus-Elena-Klinik, Klinikstrasse 16, 34119 Kassel, Germany
- Department of Neurology, Philipps University Marburg, Baldingerstraße 35043 Marburg, Germany
| | | | - Sebastian Schade
- Paracelsus-Elena-Klinik, Klinikstrasse 16, 34119 Kassel, Germany
| | - Claudia Trenkwalder
- Paracelsus-Elena-Klinik, Klinikstrasse 16, 34119 Kassel, Germany
- Department of Neurosurgery, University Medical Centre Goettingen, Robert-Koch Str. 40, 37073 Goettingen, Germany
| | - Claudio Soto
- R&D Unit, Amprion Inc., 11095 Flintkote Av., San Diego, California, 92121, USA
- Mitchell Center for Alzheimer’s Disease and Related Brain Disorders, University of Texas McGovern Medical School, Houston, TX, USA
| | - Brit Mollenhauer
- Department of Neurology, University Medical Centre Goettingen, Robert-Koch Str. 40, 37073 Goettingen, Germany
- Paracelsus-Elena-Klinik, Klinikstrasse 16, 34119 Kassel, Germany
| |
Collapse
|
4
|
Zhu C, Zhu J, Xiang Y, Bu XL, Jin WS, Wang YJ. A Conceptual Study on the Peripheral Clearance of Brain-Derived α-Synuclein in Humans. J Alzheimers Dis 2022; 90:1485-1492. [PMID: 36278352 DOI: 10.3233/jad-220742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND Abnormal intracellular expression and aggregation of α-synuclein (α-syn) is the histopathological hallmark of several neurodegenerative diseases especially Parkinson's disease. However, safe and efficient approaches to clear α-syn remain unavailable. OBJECTIVE This study aimed to investigate the process of peripheral catabolism of brain-derived α-syn. METHODS Thirty patients with atrioventricular reentrant tachycardia (AVRT) (left accessory pathways) who underwent radiofrequency catheter ablation (RFCA) were enrolled in this study. Blood was collected via catheters from superior vena cava (SVC), inferior vena cava (IVC) proximal to the hepatic vein (HV), the right femoral vein (FV), and femoral artery (FA) simultaneously during RFCA. Plasma α-syn levels of AVRT patients and soluble α-syn levels of the brain samples were measured using enzyme-linked immunosorbent assay kits. RESULTS The α-syn concentrations in different locations of veins were divided by time-matched arterial α-syn concentrations to generate the venous/arterial (V/A) ratio. The V/A ratio of α-syn from the SVC was 1.204 (1.069-1.339, 95% CI), while the V/A ratio of α-syn from IVC was 0.831 (0.734-0.928, 95% CI), suggesting that brain-derived α-syn in the arterial blood was physiologically cleared while going through the peripheral organs and tissues. And it was estimated that about half of brain soluble α-syn could efflux and be cleared in the periphery. Moreover, the glomerular filtration rate was found correlated with V-A difference (FA-ICV) (p = 0.0272). CONCLUSION Under physiological conditions, brain-derived α-syn could efflux into and be catabolized by the peripheral system. The kidney may play a potential role in the clearance of α-syn.
Collapse
Affiliation(s)
- Chi Zhu
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China.,Chongqing Key Laboratory of Ageing and Brain Diseases, Chongqing, China
| | - Jie Zhu
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China.,Chongqing Key Laboratory of Ageing and Brain Diseases, Chongqing, China
| | - Yang Xiang
- Department of Neurology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China.,Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, China
| | - Xian-Le Bu
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China.,Chongqing Key Laboratory of Ageing and Brain Diseases, Chongqing, China.,Institute of Brain and Intelligence, Third Military Medical University, Chongqing, China
| | - Wang-Sheng Jin
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China.,Chongqing Key Laboratory of Ageing and Brain Diseases, Chongqing, China.,Institute of Brain and Intelligence, Third Military Medical University, Chongqing, China
| | - Yan-Jiang Wang
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China.,Chongqing Key Laboratory of Ageing and Brain Diseases, Chongqing, China.,Institute of Brain and Intelligence, Third Military Medical University, Chongqing, China.,State Key Laboratory of Trauma, Burns and Combined Injury, Third Military Medical University, Chongqing, China.,Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
5
|
Kawahata I, Finkelstein DI, Fukunaga K. Pathogenic Impact of α-Synuclein Phosphorylation and Its Kinases in α-Synucleinopathies. Int J Mol Sci 2022; 23:ijms23116216. [PMID: 35682892 PMCID: PMC9181156 DOI: 10.3390/ijms23116216] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 05/29/2022] [Accepted: 05/31/2022] [Indexed: 12/30/2022] Open
Abstract
α-Synuclein is a protein with a molecular weight of 14.5 kDa and consists of 140 amino acids encoded by the SNCA gene. Missense mutations and gene duplications in the SNCA gene cause hereditary Parkinson’s disease. Highly phosphorylated and abnormally aggregated α-synuclein is a major component of Lewy bodies found in neuronal cells of patients with sporadic Parkinson’s disease, dementia with Lewy bodies, and glial cytoplasmic inclusion bodies in oligodendrocytes with multiple system atrophy. Aggregated α-synuclein is cytotoxic and plays a central role in the pathogenesis of the above-mentioned synucleinopathies. In a healthy brain, most α-synuclein is unphosphorylated; however, more than 90% of abnormally aggregated α-synuclein in Lewy bodies of patients with Parkinson’s disease is phosphorylated at Ser129, which is presumed to be of pathological significance. Several kinases catalyze Ser129 phosphorylation, but the role of phosphorylation enzymes in disease pathogenesis and their relationship to cellular toxicity from phosphorylation are not fully understood in α-synucleinopathy. Consequently, this review focuses on the pathogenic impact of α-synuclein phosphorylation and its kinases during the neurodegeneration process in α-synucleinopathy.
Collapse
Affiliation(s)
- Ichiro Kawahata
- Department of CNS Drug Innovation, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan
- Correspondence: (I.K.); (K.F.); Tel.: +81-22-795-6838 (I.K.); +81-22-795-6836 (K.F.); Fax: +81-22-795-6835 (I.K. & K.F.)
| | - David I. Finkelstein
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, VIC 3010, Australia;
| | - Kohji Fukunaga
- Department of CNS Drug Innovation, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan
- BRI Pharma Inc., Sendai 982-0804, Japan
- Correspondence: (I.K.); (K.F.); Tel.: +81-22-795-6838 (I.K.); +81-22-795-6836 (K.F.); Fax: +81-22-795-6835 (I.K. & K.F.)
| |
Collapse
|
6
|
Li ZF, Cui L, Jin MM, Hu DY, Hou XG, Liu SS, Zhang X, Zhu JH. A Matrigel-based 3D construct of SH-SY5Y cells models the α-synuclein pathologies of Parkinson's disease. Dis Model Mech 2022; 15:273997. [PMID: 35023548 PMCID: PMC8922027 DOI: 10.1242/dmm.049125] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 01/04/2022] [Indexed: 11/20/2022] Open
Abstract
Parkinson's disease (PD) is associated with α-synuclein-based Lewy body pathology, which has been difficult to observe in conventional two-dimensional (2D) cell culture and even in animal models. We herein aimed to develop a three-dimensional (3D) cellular model of PD to recapitulate the α-synuclein pathologies. All-trans-retinoic acid-differentiated human SH-SY5Y cells and Matrigel were optimized for 3D construction. The 3D cultured cells displayed higher tyrosine hydroxylase expression than 2D cells and improved dopaminergic-like phenotypes, as suggested by RNA-sequencing analyses. Multiple forms of α-synuclein, including monomer, and low- and high-molecular mass oligomers, were differentially present in the 2D and 3D cells, but mostly remained unchanged upon N-methyl-4-phenyl pyridine or rotenone treatment. Phosphorylated α-synuclein was accumulated, and detergent-insoluble α-synuclein fraction was observed, in the neurotoxin-treated 3D cells. Importantly, Lewy body-like inclusions were captured in the 3D system, including proteinase K-resistant α-synuclein aggregates, ubiquitin aggregation, and β-amyloid and β-sheet protein deposition. The study provides a unique and convenient 3D model of PD that recapitulates critical α-synuclein pathologies and should be useful in multiple PD-associated applications. Summary: This study provides a convenient 3D model of Parkinson's disease (PD), which recapitulates α-synuclein pathologies in human cells and could be used to investigate PD mechanisms and screen drugs.
Collapse
Affiliation(s)
- Zhao-Feng Li
- Institute of Nutrition and Diseases, Department of Preventive Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China.,Department of Neurology and Geriatrics, the Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Lei Cui
- Institute of Nutrition and Diseases, Department of Preventive Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Mi-Mi Jin
- Department of Neurology and Geriatrics, the Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Dong-Yan Hu
- Institute of Nutrition and Diseases, Department of Preventive Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xiao-Gang Hou
- Department of Neurology and Geriatrics, the Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Shu-Shu Liu
- Institute of Nutrition and Diseases, Department of Preventive Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xiong Zhang
- Department of Neurology and Geriatrics, the Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jian-Hong Zhu
- Institute of Nutrition and Diseases, Department of Preventive Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China.,Department of Neurology and Geriatrics, the Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
7
|
Qu M. The Neuroprotective Effect of Steroid Receptor Coactivator-Interacting Protein (SIP) in Astrocyte Model of 1-Methyl-4-Phenylpyridinium (MPP⁺)-Induced Parkinson's Disease. Med Sci Monit 2019; 25:5776-5784. [PMID: 31376345 PMCID: PMC6690214 DOI: 10.12659/msm.912106] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Background The purpose of this study was to investigate the role and mechanism of steroid receptor coactivator-interacting protein (SIP) in an astrocyte model of 1-methyl-4-phenylpyridinium (MPP+)-induced Parkinson’s disease. Material/Methods To perform our study, a Parkinson’s disease cell model was established by treating the rat glioblastoma cell line C6 with MPP+. SIP was overexpressed in C6 cells using SIP-plasmid. Cell viability and apoptosis were analyzed using MTT assay and flow cytometer respectively. Tumor necrosis factor (TNF)-α and interleukin (IL)-1β levels were detected using enzyme linked immunosorbent assay and quantitative reverse transcription PCR. Besides, lactate dehydrogenase (LDH) release, reactive oxygen species (ROS) production, and superoxide dismutase (SOD) enzyme activity were determined in the present study. For protein and mRNA detection, western blot assay, and qRT-PCR were performed respectively. Results SIP was decreased in MPP+-induced C6 cells. SIP overexpression relieved MPP+-induced cytotoxicity of C6 cells, displayed as increased cell viability and reduced cell apoptosis and reduced LDH release. Besides, SIP inhibited MPP+-induced inflammatory response and oxidative stress, evidenced by decreased levels of inflammatory factors (TNF-α and IL-1β), reduced ROS generation and enhanced SOD activity. Moreover, MPP+-induced nuclear factor-κB activation was inhibited by SIP overexpression. Conclusions SIP was downregulated in Parkinson’s disease and it played a protective role in the development Parkinson’s disease, thus may be a promising target for Parkinson’s disease treatment.
Collapse
Affiliation(s)
- Mingwei Qu
- Department of Neurology, Cangzhou Central Hospital, Cangzhou, Hebei, China (mainland)
| |
Collapse
|
8
|
Riederer P, Berg D, Casadei N, Cheng F, Classen J, Dresel C, Jost W, Krüger R, Müller T, Reichmann H, Rieß O, Storch A, Strobel S, van Eimeren T, Völker HU, Winkler J, Winklhofer KF, Wüllner U, Zunke F, Monoranu CM. α-Synuclein in Parkinson's disease: causal or bystander? J Neural Transm (Vienna) 2019; 126:815-840. [PMID: 31240402 DOI: 10.1007/s00702-019-02025-9] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 05/29/2019] [Indexed: 12/13/2022]
Abstract
Parkinson's disease (PD) comprises a spectrum of disorders with differing subtypes, the vast majority of which share Lewy bodies (LB) as a characteristic pathological hallmark. The process(es) underlying LB generation and its causal trigger molecules are not yet fully understood. α-Synuclein (α-syn) is a major component of LB and SNCA gene missense mutations or duplications/triplications are causal for rare hereditary forms of PD. As typical sporadic PD is associated with LB pathology, a factor of major importance is the study of the α-syn protein and its pathology. α-Syn pathology is, however, also evident in multiple system atrophy (MSA) and Lewy body disease (LBD), making it non-specific for PD. In addition, there is an overlap of these α-synucleinopathies with other protein-misfolding diseases. It has been proven that α-syn, phosphorylated tau protein (pτ), amyloid beta (Aβ) and other proteins show synergistic effects in the underlying pathogenic mechanisms. Multiple cell death mechanisms can induce pathological protein-cascades, but this can also be a reverse process. This holds true for the early phases of the disease process and especially for the progression of PD. In conclusion, while rare SNCA gene mutations are causal for a minority of familial PD patients, in sporadic PD (where common SNCA polymorphisms are the most consistent genetic risk factor across populations worldwide, accounting for 95% of PD patients) α-syn pathology is an important feature. Conversely, with regard to the etiopathogenesis of α-synucleinopathies PD, MSA and LBD, α-syn is rather a bystander contributing to multiple neurodegenerative processes, which overlap in their composition and individual strength. Therapeutic developments aiming to impact on α-syn pathology should take this fact into consideration.
Collapse
Affiliation(s)
- Peter Riederer
- Clinic and Policlinic for Psychiatry, Psychosomatics and Psychotherapy, University Hospital Würzburg, University of Würzburg, Margarete-Höppel-Platz 1, 97080, Würzburg, Germany. .,Department of Psychiatry, University of South Denmark, Odense, Denmark.
| | - Daniela Berg
- Department of Neurology, UKHS, Christian-Albrechts-Universität, Campus Kiel, Kiel, Germany
| | - Nicolas Casadei
- NGS Competence Center Tübingen, Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany
| | - Fubo Cheng
- NGS Competence Center Tübingen, Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany
| | - Joseph Classen
- Department of Neurology, University Hospital Leipzig, Leipzig, Germany
| | - Christian Dresel
- Department of Neurology, Center for Movement Disorders, Neuroimaging Center Mainz, Clinical Neurophysiology, Forschungszentrum Translationale Neurowissenschaften (FTN), Rhein-Main-Neuronetz, Mainz, Germany
| | | | - Rejko Krüger
- Clinical and Experimental Neuroscience, LCSB (Luxembourg Centre for Systems, Biomedicine), University of Luxembourg, Esch-sur-Alzette and Centre Hospitalier de Luxembourg (CHL), Luxembourg, Luxembourg.,National Center for Excellence in Research, Parkinson's disease (NCER-PD), Parkinson Research Clinic, Centre Hospitalier de Luxembourg, Luxembourg, Luxembourg
| | - Thomas Müller
- Department of Neurology, Alexianer St. Joseph Berlin-Weißensee, Berlin, Germany
| | - Heinz Reichmann
- Department of Neurology, University of Dresden, Dresden, Germany
| | - Olaf Rieß
- Institute of Medical Genetics and Applied Genomics, Tübingen, Germany
| | - Alexander Storch
- Department of Neurology, University of Rostock, Rostock, Germany.,German Centre for Neurodegenerative Diseases (DZNE) Rostock/Greifswald, Rostock, Germany
| | - Sabrina Strobel
- Department of Neuropathology, Institute of Pathology, University of Würzburg, Würzburg, Germany
| | - Thilo van Eimeren
- Department of Neurology, University Hospital of Cologne, Cologne, Germany
| | | | - Jürgen Winkler
- Department Kopfkliniken, Molekulare Neurologie, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Konstanze F Winklhofer
- Institute of Biochemistry and Pathobiochemistry, Ruhr-Universität Bochum, Bochum, Germany
| | - Ullrich Wüllner
- Department of Neurology, University of Bonn, German Center for Neurodegenerative Diseases (DZNE Bonn), Bonn, Germany
| | - Friederike Zunke
- Department of Biochemistry, Medical Faculty, University of Kiel, Kiel, Germany
| | - Camelia-Maria Monoranu
- Department of Neuropathology, Institute of Pathology, University of Würzburg, Würzburg, Germany
| |
Collapse
|
9
|
A secret that underlies Parkinson's disease: The damaging cycle. Neurochem Int 2019; 129:104484. [PMID: 31173779 DOI: 10.1016/j.neuint.2019.104484] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2019] [Revised: 05/30/2019] [Accepted: 06/03/2019] [Indexed: 01/21/2023]
Abstract
Parkinson's disease (PD) is a movement disorder, and its common characteristics include the loss of dopaminergic neurons and the accumulation of a special type of cytoplasmic inclusions called Lewy bodies in the substantia nigra pars compacta, which are more prevalent in the elderly. However, the pathophysiology of PD is still elusive. In this review, we summarized five common factors involved in PD, namely, (i) oxidative stress, (ii) mitochondrial dysfunction, (iii) inflammation, (iv) abnormal α-synuclein, and (v) endogenous neurotoxins, and proposed a hypothesis involving a damaging cycle. Oxidative stress-triggered aldehydes react with biogenic amines to produce endogenous neurotoxins. They cause mitochondrial dysfunction and the formation of inflammasomes, which induce the activation of neuroglial cells and the infiltration of T lymphocytes. The synergistic effect of these processes fosters chronic inflammation and α-synuclein aggregation and further exacerbates the impact of oxidative stress to establish a damaging cycle that eventually results in the degeneration of dopaminergic neurons. This damaging cycle provides an explanation of progressive neuronal death during the pathogenesis of PD and provides new potential targets beneficial for developing new drugs and approaches for clinical neuroprotection.
Collapse
|
10
|
Naja mossambica mossambica Cobra Cardiotoxin Targets Mitochondria to Disrupt Mitochondrial Membrane Structure and Function. Toxins (Basel) 2019; 11:toxins11030152. [PMID: 30857180 PMCID: PMC6468758 DOI: 10.3390/toxins11030152] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 03/01/2019] [Accepted: 03/05/2019] [Indexed: 01/07/2023] Open
Abstract
Cobra venom cardiotoxins (CVCs) can translocate to mitochondria to promote apoptosis by eliciting mitochondrial dysfunction. However, the molecular mechanism(s) by which CVCs are selectively targeted to the mitochondrion to disrupt mitochondrial function remains to be elucidated. By studying cardiotoxin from Naja mossambica mossambica cobra (cardiotoxin VII4), a basic three-fingered S-type cardiotoxin, we hypothesized that cardiotoxin VII4 binds to cardiolipin (CL) in mitochondria to alter mitochondrial structure/function and promote neurotoxicity. By performing confocal analysis, we observed that red-fluorescently tagged cardiotoxin rapidly translocates to mitochondria in mouse primary cortical neurons and in human SH-SY5Y neuroblastoma cells to promote aberrant mitochondrial fragmentation, a decline in oxidative phosphorylation, and decreased energy production. In addition, by employing electron paramagnetic resonance (EPR) and protein nuclear magnetic resonance (1H-NMR) spectroscopy and phosphorescence quenching of erythrosine in model membranes, our compiled biophysical data show that cardiotoxin VII4 binds to anionic CL, but not to zwitterionic phosphatidylcholine (PC), to increase the permeability and formation of non-bilayer structures in CL-enriched membranes that biochemically mimic the outer and inner mitochondrial membranes. Finally, molecular dynamics simulations and in silico docking studies identified CL binding sites in cardiotoxin VII4 and revealed a molecular mechanism by which cardiotoxin VII4 interacts with CL and PC to bind and penetrate mitochondrial membranes.
Collapse
|
11
|
Bonito-Oliva A, Schedin-Weiss S, Younesi SS, Tiiman A, Adura C, Paknejad N, Brendel M, Romin Y, Parchem RJ, Graff C, Vukojević V, Tjernberg LO, Terenius L, Winblad B, Sakmar TP, Graham WV. Conformation-specific antibodies against multiple amyloid protofibril species from a single amyloid immunogen. J Cell Mol Med 2019; 23:2103-2114. [PMID: 30663210 PMCID: PMC6378190 DOI: 10.1111/jcmm.14119] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 11/28/2018] [Accepted: 12/07/2018] [Indexed: 12/01/2022] Open
Abstract
We engineered and employed a chaperone‐like amyloid‐binding protein Nucleobindin 1 (NUCB1) to stabilize human islet amyloid polypeptide (hIAPP) protofibrils for use as immunogen in mice. We obtained multiple monoclonal antibody (mAb) clones that were reactive against hIAPP protofibrils. A secondary screen was carried out to identify clones that cross‐reacted with amyloid beta‐peptide (Aβ42) protofibrils, but not with Aβ40 monomers. These mAbs were further characterized in several in vitro assays, in immunohistological studies of a mouse model of Alzheimer's disease (AD) and in AD patient brain tissue. We show that mAbs obtained by immunizing mice with the NUCB1‐hIAPP complex cross‐react with Aβ42, specifically targeting protofibrils and inhibiting their further aggregation. In line with conformation‐specific binding, the mAbs appear to react with an intracellular antigen in diseased tissue, but not with amyloid plaques. We hypothesize that the mAbs we describe here recognize a secondary or quaternary structural epitope that is common to multiple amyloid protofibrils. In summary, we report a method to create mAbs that are conformation‐sensitive and sequence‐independent and can target more than one type of protofibril species.
Collapse
Affiliation(s)
- Alessandra Bonito-Oliva
- Laboratory of Chemical Biology & Signal Transduction, The Rockefeller University, New York City, New York
| | - Sophia Schedin-Weiss
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet, Solna, Sweden
| | - Shahab S Younesi
- Department of Neuroscience, Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, Houston, Texas
| | - Ann Tiiman
- Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Carolina Adura
- High Throughput and Spectroscopy Resource Center, The Rockefeller University, New York City, New York
| | - Navid Paknejad
- Molecular Cytology Core Facility, Memorial Sloan-Kettering Cancer Center, New York City, New York
| | - Matt Brendel
- Molecular Cytology Core Facility, Memorial Sloan-Kettering Cancer Center, New York City, New York
| | - Yevgeniy Romin
- Molecular Cytology Core Facility, Memorial Sloan-Kettering Cancer Center, New York City, New York
| | - Ronald J Parchem
- Department of Neuroscience, Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, Houston, Texas
| | - Caroline Graff
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet, Solna, Sweden.,Theme Aging, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Vladana Vukojević
- Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Lars O Tjernberg
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet, Solna, Sweden
| | - Lars Terenius
- Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Bengt Winblad
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet, Solna, Sweden
| | - Thomas P Sakmar
- Laboratory of Chemical Biology & Signal Transduction, The Rockefeller University, New York City, New York.,Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet, Solna, Sweden
| | - W Vallen Graham
- Laboratory of Chemical Biology & Signal Transduction, The Rockefeller University, New York City, New York
| |
Collapse
|
12
|
Investigation of α-Synuclein Amyloid Fibrils Using the Fluorescent Probe Thioflavin T. Int J Mol Sci 2018; 19:ijms19092486. [PMID: 30142878 PMCID: PMC6163839 DOI: 10.3390/ijms19092486] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Revised: 08/08/2018] [Accepted: 08/20/2018] [Indexed: 11/29/2022] Open
Abstract
In this work, α-synuclein amyloid fibrils—the formation of which is a biomarker of Parkinson’s disease—were investigated using the fluorescent probe thioflavin T (ThT). The experimental conditions of protein fibrillogenesis were chosen so that a sufficient number of continuous measurements could be performed to characterize and analyze all stages of this process. The reproducibility of fibrillogenesis and the structure of the obtained aggregates (which is a critical point for further investigation) were proven using a wide range of physical-chemical methods. For the determination of ThT-α-synuclein amyloid fibril binding parameters, the sample and reference solutions were prepared using equilibrium microdialysis. By utilizing absorption spectroscopy of these solutions, the ThT-fibrils binding mode with a binding constant of about 104 M−1 and stoichiometry of ThT per protein molecule of about 1:8 was observed. Fluorescence spectroscopy of the same solutions with the subsequent correction of the recorded fluorescence intensity on the primary inner filter effect allowed us to determine another mode of ThT binding to fibrils, with a binding constant of about 106 M−1 and stoichiometry of about 1:2500. Analysis of the photophysical characteristics of the dye molecules bound to the sites of different binding modes allowed us to assume the possible localization of these sites. The obtained differences in the ThT binding parameters to the amyloid fibrils formed from α-synuclein and other amyloidogenic proteins, as well as in the photophysical characteristics of the bound dye, confirmed the hypothesis of amyloid fibril polymorphism.
Collapse
|
13
|
Zondler L, Kostka M, Garidel P, Heinzelmann U, Hengerer B, Mayer B, Weishaupt JH, Gillardon F, Danzer KM. Proteasome impairment by α-synuclein. PLoS One 2017; 12:e0184040. [PMID: 28945746 PMCID: PMC5612461 DOI: 10.1371/journal.pone.0184040] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Accepted: 08/16/2017] [Indexed: 11/28/2022] Open
Abstract
Parkinson’s disease (PD) is the second most prevalent neurodegenerative disorder worldwide and characterized by the loss of dopaminergic neurons in the patients’ midbrains. Both the presence of the protein α-synuclein in intracellular protein aggregates in surviving neurons and the genetic linking of the α-synuclein encoding gene point towards a major role of α-synuclein in PD etiology. The exact pathogenic mechanisms of PD development are not entirely described to date, neither is the specific role of α-synuclein in this context. Previous studies indicate that one aspect of α-synuclein-related cellular toxicity might be direct proteasome impairment. The 20/26S proteasomal machinery is an important instrument of intracellular protein degradation. Thus, direct proteasome impairment by α-synuclein might explain or at least contribute to the formation of intracellular protein aggregates. Therefore this study investigates direct proteasomal impairment by α-synuclein both in vitro using recombinant α-synuclein and isolated proteasomes as well as in living cells. Our experiments demonstrate that the impairment of proteasome activity by α-synuclein is highly dependent upon the cellular background and origin. We show that recombinant α-synuclein oligomers and fibrils scarcely affect 20S proteasome function in vitro, neither does transient α-synuclein expression in U2OS ps 2042 (Ubi(G76V)-GFP) cells. However, stable expression of both wild-type and mutant α-synuclein in dopaminergic SH-SY5Y and PC12 cells results in a prominent impairment of the chymotrypsin-like 20S/26S proteasomal protein cleavage. Thus, our results support the idea that α-synuclein in a specific cellular environment, potentially present in dopaminergic cells, cannot be processed by the proteasome and thus contributes to a selective vulnerability of dopaminergic cells to α-synuclein pathology.
Collapse
Affiliation(s)
- Lisa Zondler
- Neurology Department, Ulm University, Ulm, Germany
| | - Marcus Kostka
- Boehringer Ingelheim Pharma GmbH Co.KG, Biberach an der Riß, Germany
| | - Patrick Garidel
- Boehringer Ingelheim Pharma GmbH Co.KG, Biberach an der Riß, Germany
| | - Udo Heinzelmann
- Boehringer Ingelheim Pharma GmbH Co.KG, Biberach an der Riß, Germany
| | - Bastian Hengerer
- Boehringer Ingelheim Pharma GmbH Co.KG, Biberach an der Riß, Germany
| | - Benjamin Mayer
- Institute for Epidemiology and Medical Biometry, Ulm University, Ulm, Germany
| | | | - Frank Gillardon
- Boehringer Ingelheim Pharma GmbH Co.KG, Biberach an der Riß, Germany
| | | |
Collapse
|
14
|
Eastwood TA, Baker K, Brooker HR, Frank S, Mulvihill DP. An enhanced recombinant amino-terminal acetylation system and novel in vivo high-throughput screen for molecules affecting α-synuclein oligomerisation. FEBS Lett 2017; 591:833-841. [PMID: 28214355 PMCID: PMC5396276 DOI: 10.1002/1873-3468.12597] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Revised: 02/13/2017] [Accepted: 02/14/2017] [Indexed: 01/19/2023]
Abstract
Amino‐terminal acetylation is a ubiquitous protein modification affecting the majority of eukaryote proteins to regulate stability and function. We describe an optimised recombinant expression system for rapid production of amino terminal‐acetylated proteins within bacteria. We go on to describe the system's use in a fluorescence based in vivo assay for use in the high‐throughput screen to identify drugs that impact amino‐terminal acetylation‐dependent oligomerisation. These new tools and protocols will allow researchers to enhance routine recombinant protein production and identify new molecules for use in research and clinical applications.
Collapse
Affiliation(s)
| | - Karen Baker
- School of Biosciences, University of Kent, Canterbury, UK
| | | | - Stefanie Frank
- School of Biosciences, University of Kent, Canterbury, UK
| | | |
Collapse
|
15
|
Sierecki E, Giles N, Bowden Q, Polinkovsky ME, Steinbeck J, Arrioti N, Rahman D, Bhumkar A, Nicovich PR, Ross I, Parton RG, Böcking T, Gambin Y. Nanomolar oligomerization and selective co-aggregation of α-synuclein pathogenic mutants revealed by single-molecule fluorescence. Sci Rep 2016; 6:37630. [PMID: 27892477 PMCID: PMC5385372 DOI: 10.1038/srep37630] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Accepted: 10/28/2016] [Indexed: 02/08/2023] Open
Abstract
Protein aggregation is a hallmark of many neurodegenerative diseases, notably Alzheimer's and Parkinson's disease. Parkinson's disease is characterized by the presence of Lewy bodies, abnormal aggregates mainly composed of α-synuclein. Moreover, cases of familial Parkinson's disease have been linked to mutations in α-synuclein. In this study, we compared the behavior of wild-type (WT) α-synuclein and five of its pathological mutants (A30P, E46K, H50Q, G51D and A53T). To this end, single-molecule fluorescence detection was coupled to cell-free protein expression to measure precisely the oligomerization of proteins without purification, denaturation or labelling steps. In these conditions, we could detect the formation of oligomeric and pre-fibrillar species at very short time scale and low micromolar concentrations. The pathogenic mutants surprisingly segregated into two classes: one group forming large aggregates and fibrils while the other tending to form mostly oligomers. Strikingly, co-expression experiments reveal that members from the different groups do not generally interact with each other, both at the fibril and monomer levels. Together, this data paints a completely different picture of α-synuclein aggregation, with two possible pathways leading to the development of fibrils.
Collapse
Affiliation(s)
- Emma Sierecki
- EMBL Australia Node in Single Molecule Science, The University of New South Wales, Sydney NSW 2032 Australia
| | - Nichole Giles
- EMBL Australia Node in Single Molecule Science, The University of New South Wales, Sydney NSW 2032 Australia
| | - Quill Bowden
- EMBL Australia Node in Single Molecule Science, The University of New South Wales, Sydney NSW 2032 Australia
| | - Mark E. Polinkovsky
- EMBL Australia Node in Single Molecule Science, The University of New South Wales, Sydney NSW 2032 Australia
| | - Janina Steinbeck
- Institute for Molecular Bioscience, The University of Queensland, St Lucia QLD 4072, Australia
| | - Nicholas Arrioti
- Institute for Molecular Bioscience, The University of Queensland, St Lucia QLD 4072, Australia
| | - Diya Rahman
- Institute for Molecular Bioscience, The University of Queensland, St Lucia QLD 4072, Australia
| | - Akshay Bhumkar
- EMBL Australia Node in Single Molecule Science, The University of New South Wales, Sydney NSW 2032 Australia
| | - Philip R. Nicovich
- EMBL Australia Node in Single Molecule Science, The University of New South Wales, Sydney NSW 2032 Australia
| | - Ian Ross
- Institute for Molecular Bioscience, The University of Queensland, St Lucia QLD 4072, Australia
| | - Robert G. Parton
- Institute for Molecular Bioscience, The University of Queensland, St Lucia QLD 4072, Australia
| | - Till Böcking
- EMBL Australia Node in Single Molecule Science, The University of New South Wales, Sydney NSW 2032 Australia
| | - Yann Gambin
- EMBL Australia Node in Single Molecule Science, The University of New South Wales, Sydney NSW 2032 Australia
| |
Collapse
|
16
|
Subcellular Parkinson's Disease-Specific Alpha-Synuclein Species Show Altered Behavior in Neurodegeneration. Mol Neurobiol 2016; 54:7639-7655. [PMID: 27837450 DOI: 10.1007/s12035-016-0266-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Accepted: 10/26/2016] [Indexed: 01/29/2023]
Abstract
Parkinson's disease and other synucleinopathies are characterized by the presence of intra-neuronal protein aggregates enriched in the presynaptic protein α-synuclein. α-synuclein is considered an intrinsically disordered 14 kDa monomer, and although poorly understood, its transition to higher-order multimeric species may play central roles in healthy neurons and during Parkinson's disease pathogenesis. In this study, we demonstrate that α-synuclein exists as defined, subcellular-specific species that change characteristics in response to oxidative stress in neuroblastoma cells and in response to Parkinson's disease pathogenesis in human cerebellum and frontal cortex. We further show that the phosphorylation patterns of different α-synuclein species are subcellular specific and dependent on the oxidative environment. Using high-performance liquid chromatography and mass spectrometry, we identify a Parkinson's disease enriched, cytosolic ~36-kDa α-synuclein species which can be recapitulated in Parkinson's disease model neuroblastoma cells. The characterization of subcellular-specific α-synuclein features in neurodegeneration will allow for the identification of neurotoxic α-synuclein species, which represent prime targets to reduce α-synuclein pathogenicity.
Collapse
|
17
|
Qiao C, Yin N, Gu HY, Zhu JL, Ding JH, Lu M, Hu G. Atp13a2 Deficiency Aggravates Astrocyte-Mediated Neuroinflammation via NLRP3 Inflammasome Activation. CNS Neurosci Ther 2016; 22:451-60. [PMID: 26848562 DOI: 10.1111/cns.12514] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Revised: 12/21/2015] [Accepted: 12/22/2015] [Indexed: 12/24/2022] Open
Abstract
AIM Atp13a2 (Park9) gene encodes a transmembrane lysosomal P5-type ATPase (ATP13A2), and its missense or truncation mutations leads to lysosomal dysfunction and consequently results in neuronal death in the pathogenesis of Parkinson's disease (PD). Nevertheless, the roles of ATP13A2 in the biological features of astrocytes, especially in the regulation of PD-related neuroinflammation, have not been investigated. METHODS We cultured primary neurons and astrocytes from mouse midbrain to investigate the mechanisms for astrocyte ATP13A2-regulated lysosomal function and neuroinflammation following 1-methyl-4-phenylpyridinium (MPP(+) ) treatment. RESULTS We found that astrocytes expressed considerable levels of ATP13A2 and deficiency of ATP13A2 in astrocyte-induced intense inflammation, which exacerbated dopaminergic neuron damage after exposure to MPP(+) . Notably, lack of ATP13A2 increased lysosomal membrane permeabilization and cathepsin B release, which in turn exacerbated activation of nod-like receptor protein 3 (NLRP3) inflammasome to produce excess IL-1β from astrocytes. Furthermore, overexpression of ATP13A2 reversed MPP(+) -induced cathepsin B release and NLRP3 inflammasome activation in astrocytes. CONCLUSIONS Our results have revealed a novel role of ATP13A2 in modulating astrocyte-mediated neuroinflammation via NLRP3 inflammasome activation, thus bringing to light of a direct link between astrocyte lysosome and neuroinflammation in the pathological model of PD.
Collapse
Affiliation(s)
- Chen Qiao
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Nuo Yin
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Huan-Yu Gu
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jia-Lei Zhu
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jian-Hua Ding
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Ming Lu
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Gang Hu
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, Jiangsu, China.,Biomedical Functional Materials Collaborative Innovation Center, College of Chemistry and Materials Science, Nanjing Normal University, Nanjing, Jiangsu, China
| |
Collapse
|
18
|
Ghio S, Kamp F, Cauchi R, Giese A, Vassallo N. Interaction of α-synuclein with biomembranes in Parkinson's disease--role of cardiolipin. Prog Lipid Res 2015; 61:73-82. [PMID: 26703192 DOI: 10.1016/j.plipres.2015.10.005] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Revised: 10/14/2015] [Accepted: 10/30/2015] [Indexed: 12/15/2022]
Abstract
One of the key molecular events underlying the pathogenesis of Parkinson's disease (PD) is the aberrant misfolding and aggregation of the α-synuclein (αS) protein into higher-order oligomers that play a key role in neuronal dysfunction and degeneration. A wealth of experimental data supports the hypothesis that the neurotoxicity of αS oligomers is intrinsically linked with their ability to interact with, and disrupt, biological membranes; especially those membranes having negatively-charged surfaces and/or lipid packing defects. Consequences of αS-lipid interaction include increased membrane tension, permeation by pore formation, membrane lysis and/or leakage due to the extraction of lipids from the bilayer. Moreover, we assert that the interaction of αS with a liquid-disordering phospholipid uniquely enriched in mitochondrial membranes, namely cardiolipin (1,3-diphosphatidyl-sn-glycerol, CL), helps target the αS oligomeric complexes intracellularly to mitochondria. Binding mediated by CL may thus represent an important pathomechanism by which cytosolic αS could physically associate with mitochondrial membranes and disrupt their integrity. Impaired mitochondrial function culminates in a cellular bioenergetic crisis and apoptotic death. To conclude, we advocate the accelerated discovery of new drugs targeting this pathway in order to restore mitochondrial function in PD.
Collapse
Affiliation(s)
- Stephanie Ghio
- Dept. of Physiology and Biochemistry, University of Malta, Msida, Malta
| | - Frits Kamp
- Biomedical Center (BMC), Biochemistry, Ludwig-Maximilians-University & DZNE, 81377 Munich, Germany
| | - Ruben Cauchi
- Dept. of Physiology and Biochemistry, University of Malta, Msida, Malta
| | - Armin Giese
- Zentrum für Neuropathologie und Prionforschung, Ludwig-Maximilians-University, 81377 Munich, Germany
| | - Neville Vassallo
- Dept. of Physiology and Biochemistry, University of Malta, Msida, Malta.
| |
Collapse
|
19
|
Comparison between α-synuclein wild-type and A53T mutation in a progressive Parkinson's disease model. Biochem Biophys Res Commun 2015; 464:988-993. [PMID: 26192120 DOI: 10.1016/j.bbrc.2015.07.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 07/01/2015] [Indexed: 11/21/2022]
Abstract
BACKGROUND AND AIMS Vector based over-expression of α-synuclein is a newly developed method to establish animal Parkinson's disease (PD) model. In this paper, we inject the rat brain with recombinant adeno-associated virus (rAAV) to express α-synuclein wild-type and A53T mutation, and compared the degeneration of dopaminergic neurons between them. METHOD AND RESULTS The rAAV vectors were injected into the substantia nigra pars compacta (SNpc) of rat brain. In different time point, immunohistochemistry was used to detect the expression of α-synuclein. The expression level was lower in the 3rd and 6th week and increased from the 9th week. α-synuclein spread around the neurons in SNpc in the 12th week. The loss of dopaminergic neurons was increasing along the expression of α-synuclein, and damage extent was more serious in the A53T group than the WT group. In the A53T group, there were more insoluble inclusions can be detected, and the phosphorylation of α-synuclein was also higher. CONLUSION The result of comparison between the two types of α-synuclein showed that A53T mutated α-synuclein was more effective to establish PD model, and the model based A53T mutated α-synuclein was a suitable model to early-onset PD.
Collapse
|
20
|
MINAMI AKARI, NAKANISHI ATSUKO, MATSUDA SATORU, KITAGISHI YASUKO, OGURA YASUNORI. Function of α-synuclein and PINK1 in Lewy body dementia (Review). Int J Mol Med 2014; 35:3-9. [DOI: 10.3892/ijmm.2014.1980] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2014] [Accepted: 10/16/2014] [Indexed: 11/06/2022] Open
|
21
|
Zondler L, Miller-Fleming L, Repici M, Gonçalves S, Tenreiro S, Rosado-Ramos R, Betzer C, Straatman KR, Jensen PH, Giorgini F, Outeiro TF. DJ-1 interactions with α-synuclein attenuate aggregation and cellular toxicity in models of Parkinson's disease. Cell Death Dis 2014; 5:e1350. [PMID: 25058424 PMCID: PMC4123098 DOI: 10.1038/cddis.2014.307] [Citation(s) in RCA: 129] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2014] [Revised: 06/11/2014] [Accepted: 06/13/2014] [Indexed: 11/09/2022]
Abstract
Parkinson's disease (PD) is a devastating neurodegenerative disorder characterized by the loss of neurons in the substantia nigra pars compacta and the presence of Lewy bodies in surviving neurons. These intracellular protein inclusions are primarily composed of misfolded α-synuclein (aSyn), which has also been genetically linked to familial and sporadic forms of PD. DJ-1 is a small ubiquitously expressed protein implicated in several pathways associated with PD pathogenesis. Although mutations in the gene encoding DJ-1 lead to familial early-onset PD, the exact mechanisms responsible for its role in PD pathogenesis are still elusive. Previous work has found that DJ-1--which has protein chaperone-like activity--modulates aSyn aggregation. Here, we investigated possible physical interactions between aSyn and DJ-1 and any consequent functional and pathological relevance. We found that DJ-1 interacts directly with aSyn monomers and oligomers in vitro, and that this also occurs in living cells. Notably, several PD-causing mutations in DJ-1 constrain this interaction. In addition, we found that overexpression of DJ-1 reduces aSyn dimerization, whereas mutant forms of DJ-1 impair this process. Finally, we found that human DJ-1 as well as yeast orthologs of DJ-1 reversed aSyn-dependent cellular toxicity in Saccharomyces cerevisiae. Taken together, these data suggest that direct interactions between DJ-1 and aSyn constitute the basis for a neuroprotective mechanism and that familial mutations in DJ-1 may contribute to PD by disrupting these interactions.
Collapse
Affiliation(s)
- L Zondler
- Department of NeuroDegeneration and Restorative Research, University Medical Center Göttingen, Göttingen, Germany
| | - L Miller-Fleming
- 1] Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, Lisboa, Portugal [2] Department of Genetics, University of Leicester, Leicester LE1 7RH, UK
| | - M Repici
- Department of Genetics, University of Leicester, Leicester LE1 7RH, UK
| | - S Gonçalves
- Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, Lisboa, Portugal
| | - S Tenreiro
- Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, Lisboa, Portugal
| | - R Rosado-Ramos
- Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, Lisboa, Portugal
| | - C Betzer
- Danish Research Institute of Translational Neuroscience - Dandrite, Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - K R Straatman
- Centre for Core Biotechnology Services, University of Leicester, Leicester LE1 7RH, UK
| | - P H Jensen
- Danish Research Institute of Translational Neuroscience - Dandrite, Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - F Giorgini
- Department of Genetics, University of Leicester, Leicester LE1 7RH, UK
| | - T F Outeiro
- 1] Department of NeuroDegeneration and Restorative Research, University Medical Center Göttingen, Göttingen, Germany [2] Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, Lisboa, Portugal
| |
Collapse
|