1
|
Ishaq S, Shah IA, Lee SD, Wu BT. Effects of exercise training on nigrostriatal neuroprotection in Parkinson's disease: a systematic review. Front Neurosci 2025; 18:1464168. [PMID: 39844853 PMCID: PMC11752748 DOI: 10.3389/fnins.2024.1464168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Accepted: 12/20/2024] [Indexed: 01/24/2025] Open
Abstract
Introduction Parkinson's disease (PD) is characterized by progressive neurodegeneration within the nigrostriatum, leading to motor dysfunction. This systematic review aimed to summarize the effects of various exercise training regimens on protein or gene expression within the nigrostriatum and their role in neuroprotection and motor function improvement in animal models of Parkinson's disease (PD). Methods PubMed, EMBASE, and Web of Science were searched up to June 2024 and included sixteen studies that adhere to PRISMA guidelines and CAMARADES checklist scores ranging from 4 to 6 out of 10. Various exercise training regimens, administered 5 days per week for 6.5 weeks, were applied to MPTP, 6-OHDA, and PFF-α-synuclein-induced PD animal models. Results Exercise training was found to downregulate the inflammatory pathway by attenuating α-synuclein aggregation, inhibiting the TLR/MyD88/IκBα signaling cascade and NF-κB phosphorylation, and decreasing pro-inflammatory cytokines IL-1β and TNF-α while increasing anti-inflammatory cytokines IL-10 and TGF-β within the nigrostriatum. It also inhibited the ASC and NLRP3 inflammasome complex and reduced the BAX/ Bcl-2 ratio and caspase-1/3 proteins, thereby decreasing neuronal apoptosis in the nigrostriatum. Exercise training elevated the expression of Pro-BDNF, BDNF, GDNF, TrkB, and Erk1/2, providing neurotrophic support to dopaminergic neurons. Furthermore, it upregulated the dopaminergic signaling pathway by increasing the expression of TH, DAT, PSD-95, and synaptophysin in the nigrostriatum. Discussion The findings suggested that exercise training downregulated inflammatory and apoptotic pathways while upregulated BDNF/GDNF pathways and dopaminergic signaling within the nigrostriatum. These molecular changes contributed to neuroprotection, reduced dopaminergic neuron loss, and improved motor function in PD animal models. Systematic review registration CRD42024484537 https://www.crd.york.ac.uk/prospero/#recordDetails.
Collapse
Affiliation(s)
- Shahid Ishaq
- PhD Program in Healthcare Science, College of Healthcare Science, China Medical University, Taichung, Taiwan
| | - Iqbal Ali Shah
- PhD Program in Healthcare Science, College of Healthcare Science, China Medical University, Taichung, Taiwan
| | - Shin-Da Lee
- PhD Program in Healthcare Science, College of Healthcare Science, China Medical University, Taichung, Taiwan
- Department of Physical Therapy, China Medical University, Taichung, Taiwan
| | - Bor-Tsang Wu
- Department of Physical Therapy, China Medical University, Taichung, Taiwan
| |
Collapse
|
2
|
Moceri S, Bäuerle N, Habermeyer J, Ratz-Wirsching V, Harrer J, Distler J, Schulze-Krebs A, Timotius IK, Bluhm A, Hartlage-Rübsamen M, Roßner S, Winkler J, Xiang W, Hörsten SV. Young human alpha synuclein transgenic (BAC-SNCA) mice display sex- and gene-dose-dependent phenotypic disturbances. Behav Brain Res 2024; 460:114781. [PMID: 38043677 DOI: 10.1016/j.bbr.2023.114781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 11/10/2023] [Accepted: 11/20/2023] [Indexed: 12/05/2023]
Abstract
Parkinson's disease (PD) is a common neurodegenerative movement disorder, characterized by the loss of dopaminergic neurons in the substantia nigra pars compacta and the accumulation of aggregated alpha synuclein (aSyn). The disease often presents with early prodromal non-motor symptoms and later motor symptoms. Diagnosing PD based purely on motor symptoms is often too late for successful intervention, as a significant neuronal loss has already occurred. Furthermore, the lower prevalence of PD in females is not well understood, highlighting the need for a better understanding of the interaction between sex and aSyn, the crucial protein for PD pathogenesis. Here, we conducted a comprehensive phenotyping study in 1- to 5-month-old mice overexpressing human aSyn gene (SNCA) in a bacterial artificial chromosome (BAC-SNCA). We demonstrate a SNCA gene-dose-dependent increase of human aSyn and phosphorylated aSyn, as well as a decrease in tyrosine hydroxylase expression in BAC-SNCA mice, with more pronounced effects in male mice. Phosphorylated aSyn was already found in the dorsal motor nucleus of the vagus nerve of 2-month-old mice. This was time-wise associated with significant gait altrations in BAC-SNCA mice as early as 1 and 3 months of age using CatWalk gait analysis. Furthermore, anxiety-related behavioral tests revealed an increase in anxiety levels in male BAC-SNCA mice. Finally, 5-month-old male BAC-SNCA mice exhibited a SNCA gene-dose-dependent elevation in energy expenditure in automated home-cage monitoring. For the first time, these findings describe early-onset, sex- and gene-dose-dependent, aSyn-mediated disturbances in BAC-SNCA mice, providing a model for sex-differences, early-onset neuropathology, and prodromal symptoms of PD.
Collapse
Affiliation(s)
- Sandra Moceri
- Department of Experimental Therapy, Preclinical Experimental Center, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
| | - Natascha Bäuerle
- Department of Experimental Therapy, Preclinical Experimental Center, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
| | - Johanna Habermeyer
- Department of Experimental Therapy, Preclinical Experimental Center, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
| | - Veronika Ratz-Wirsching
- Department of Experimental Therapy, Preclinical Experimental Center, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
| | - Julia Harrer
- Department of Experimental Therapy, Preclinical Experimental Center, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
| | - Jörg Distler
- Department of Experimental Therapy, Preclinical Experimental Center, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
| | - Anja Schulze-Krebs
- Department of Experimental Therapy, Preclinical Experimental Center, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
| | - Ivanna K Timotius
- Department of Experimental Therapy, Preclinical Experimental Center, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany; Department of Electronic Engineering, Satya Wacana Christian University, 50711 Salatiga, Indonesia
| | - Alexandra Bluhm
- Paul-Flechsig-Institute for Brain Research, University of Leipzig, 04103 Leipzig, Germany
| | | | - Steffen Roßner
- Paul-Flechsig-Institute for Brain Research, University of Leipzig, 04103 Leipzig, Germany
| | - Jürgen Winkler
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
| | - Wei Xiang
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany.
| | - Stephan von Hörsten
- Department of Experimental Therapy, Preclinical Experimental Center, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany.
| |
Collapse
|
3
|
Liu Y, Huang Z, Zhang TX, Han B, Yang G, Jia D, Yang L, Liu Q, Lau AYL, Paul F, Verkhratsky A, Shi FD, Zhang C. Bruton's tyrosine kinase-bearing B cells and microglia in neuromyelitis optica spectrum disorder. J Neuroinflammation 2023; 20:309. [PMID: 38129902 PMCID: PMC10740299 DOI: 10.1186/s12974-023-02997-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 12/14/2023] [Indexed: 12/23/2023] Open
Abstract
BACKGROUND Neuromyelitis optica spectrum disorder (NMOSD) is an inflammatory autoimmune disease of the central nervous system that involves B-cell receptor signaling as well as astrocyte-microglia interaction, which both contribute to evolution of NMOSD lesions. MAIN BODY Through transcriptomic and flow cytometry analyses, we found that Bruton's tyrosine kinase (BTK), a crucial protein of B-cell receptor was upregulated both in the blood and cerebrospinal fluid of NMOSD patients. Blockade of BTK with zanubrutinib, a highly specific BTK inhibitor, mitigated the activation and maturation of B cells and reduced production of causal aquaporin-4 (AQP4) autoantibodies. In a mouse model of NMO, we found that both BTK and pBTK expression were significantly increased in microglia. Transmission electron microscope scan demonstrated that BTK inhibitor ameliorated demyelination, edema, and axonal injury in NMO mice. In the same mice colocalization of GFAP and Iba-1 immunofluorescence indicated a noticeable increase of astrocytes-microglia interaction, which was alleviated by zanubrutinib. The smart-seq analysis demonstrated that treatment with BTK inhibitor instigated microglial transcriptome changes including downregulation of chemokine-related genes and genes involved in the top 5 biological processes related to cell adhesion and migration, which are likely responsible for the reduced crosstalk of microglia and astrocytes. CONCLUSIONS Our results show that BTK activity is enhanced both in B cells and microglia and BTK inhibition contributes to the amelioration of NMOSD pathology. These data collectively reveal the mechanism of action of BTK inhibition and corroborate BTK as a viable therapeutic target.
Collapse
Affiliation(s)
- Ye Liu
- Department of Neurology and Institute of Neuroimmunology, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Zhenning Huang
- Department of Neurology and Institute of Neuroimmunology, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Tian-Xiang Zhang
- Department of Neurology and Institute of Neuroimmunology, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Bin Han
- Department of Neurology and Institute of Neuroimmunology, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Guili Yang
- Department of Neurology and Institute of Neuroimmunology, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Dongmei Jia
- Department of Neurology and Institute of Neuroimmunology, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
- Center of Neurological Diseases, China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Li Yang
- Department of Neurology and Institute of Neuroimmunology, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Qiang Liu
- Department of Neurology and Institute of Neuroimmunology, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Alexander Y L Lau
- Division of Neurology, Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Friedemann Paul
- Experimental and Clinical Research Center, Max Delbrueck Center for Molecular Medicine and Charité-Universitaetsmedizin Berlin, Berlin, Germany
- NeuroCure Clinical Research Center, Charité-Universitaetsmedizin Berlin, Berlin, Germany
| | - Alexei Verkhratsky
- Faculty of Biology, Health and Medicine, University of Manchester, Manchester, M13 9PL, UK
- Achucarro Centre for Neuroscience, IKERBASQUE, Basque Foundation for Science, 48011, Bilbao, Spain
- Department of Stem Cell Biology, State Research Institute Centre for Innovative Medicine, 01102, Vilnius, Lithuania
| | - Fu-Dong Shi
- Department of Neurology and Institute of Neuroimmunology, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
- Center of Neurological Diseases, China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Chao Zhang
- Department of Neurology and Institute of Neuroimmunology, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China.
- Center of Neurological Diseases, China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
4
|
Timotius IK, Roelofs RF, Richmond-Hacham B, Noldus LPJJ, von Hörsten S, Bikovski L. CatWalk XT gait parameters: a review of reported parameters in pre-clinical studies of multiple central nervous system and peripheral nervous system disease models. Front Behav Neurosci 2023; 17:1147784. [PMID: 37351154 PMCID: PMC10284348 DOI: 10.3389/fnbeh.2023.1147784] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 05/16/2023] [Indexed: 06/24/2023] Open
Abstract
Automated gait assessment tests are used in studies of disorders characterized by gait impairment. CatWalk XT is one of the first commercially available automated systems for analyzing the gait of rodents and is currently the most used system in peer-reviewed publications. This automated gait analysis system can generate a large number of gait parameters. However, this creates a new challenge in selecting relevant parameters that describe the changes within a particular disease model. Here, for the first time, we performed a multi-disorder review on published CatWalk XT data. We identify commonly reported CatWalk XT gait parameters derived from 91 peer-reviewed experimental studies in mice, covering six disorders of the central nervous system (CNS) and peripheral nervous system (PNS). The disorders modeled in mice were traumatic brain injury (TBI), stroke, sciatic nerve injury (SNI), spinal cord injury (SCI), Parkinson's disease (PD), and ataxia. Our review consisted of parameter selection, clustering, categorization, statistical evaluation, and data visualization. It suggests that certain gait parameters serve as potential indicators of gait dysfunction across multiple disease models, while others are specific to particular models. The findings also suggest that the more site-specific the injury is, the fewer parameters are reported to characterize its gait abnormalities. This study strives to present a clearly organized picture of gait parameters used in each one of the different mouse models, potentially helping novel CatWalk XT users to apply this information to similar or related mouse models they are working on.
Collapse
Affiliation(s)
- Ivanna K. Timotius
- Department of Electronics Engineering, Satya Wacana Christian University, Salatiga, Indonesia
- Department of Experimental Therapy, University Hospital Erlangen and Preclinical Experimental Animal Center, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | | | - Bar Richmond-Hacham
- Myers Neuro-Behavioral Core Facility, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv-Yafo, Israel
| | - Lucas P. J. J. Noldus
- Noldus Information Technology BV, Wageningen, Netherlands
- Donders Center for Neuroscience, Radboud University, Nijmegen, Netherlands
| | - Stephan von Hörsten
- Department of Experimental Therapy, University Hospital Erlangen and Preclinical Experimental Animal Center, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Lior Bikovski
- Myers Neuro-Behavioral Core Facility, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv-Yafo, Israel
- School of Behavioral Sciences, Netanya Academic College, Netanya, Israel
| |
Collapse
|
5
|
Shafiei A, Haghighi AH, Askari R, Keyhani A, Nabavizadeh MS, Asadi-Shekaari M. Effects of Moderate-Intensity Interval Training on Gene Expression and Antioxidant Status in the Hippocampus of Methamphetamine-Dependent Rats. Neurotox Res 2022; 40:1455-1463. [PMID: 35781220 DOI: 10.1007/s12640-022-00532-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 06/07/2022] [Accepted: 06/09/2022] [Indexed: 11/28/2022]
Abstract
Methamphetamine (METH) can cause neurotoxicity and increase the risk of neurodegenerative disorders such as Alzheimer's and Parkinson's diseases. This study aimed to investigate the effect of moderate-intensity interval training (MIIT) on gene expression and antioxidant status of the hippocampus of METH-dependent rats. Thirty-two male Wistar rats were randomly divided into four equal groups (n = 8): saline, METH, MIIT, and METH + MIIT. METH was injected intraperitoneally at 5 mg/kg for 21 days. The MIIT(interval running) was performed on the treadmill 5 days a week for 8 weeks. Morris water maze test was performed to measure learning and memory. Then, the hippocampal tissue was extracted to evaluate changes in gene expression and biochemical enzymes. The data were analyzed using one-way and two-way ANOVA methods at p < 0.05. The results showed that METH injection significantly reduced spatial memory and antioxidant enzymes and increased the expression of α-synuclein (α-syn), cyclin-dependent kinase 5 (CDK5), tau, and phosphorylated tau (p-tau) genes compared to the saline group. MIIT significantly increased spatial memory and antioxidant enzymes. However, it reduced α-syn, CDK5, tau, and p-tau expression. Thus, this study depicted that methamphetamine-dependent rats with memory deficits have lower antioxidant enzyme levels and higher expression of α-syn, CDK5, tau, and p-tau genes, and that an 8-week MIIT may have beneficial effects on the memory impairments as well as antioxidant status and gene expression in male rats.
Collapse
Affiliation(s)
- Ahad Shafiei
- Department of Exercise Physiology, Faculty of Sport Sciences, Hakim Sabzevari University, Sabzevar, Iran
| | - Amir Hossein Haghighi
- Department of Exercise Physiology, Faculty of Sport Sciences, Hakim Sabzevari University, Sabzevar, Iran
| | - Roya Askari
- Department of Exercise Physiology, Faculty of Sport Sciences, Hakim Sabzevari University, Sabzevar, Iran
| | - Alireza Keyhani
- Leishmaniasis Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | | | - Majid Asadi-Shekaari
- Neuroscience Research Center, Neuropharmacology Institute, Kerman University of Medical Sciences, Kerman, Iran.
| |
Collapse
|
6
|
Pieger K, Schmitt V, Gauer C, Gießl N, Prots I, Winner B, Winkler J, Brandstätter JH, Xiang W. Translocation of Distinct Alpha Synuclein Species from the Nucleus to Neuronal Processes during Neuronal Differentiation. Biomolecules 2022; 12:biom12081108. [PMID: 36009004 PMCID: PMC9406079 DOI: 10.3390/biom12081108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 08/09/2022] [Accepted: 08/10/2022] [Indexed: 11/16/2022] Open
Abstract
Alpha synuclein (aSyn) and its aggregation are crucial for the neurodegeneration of Parkinson’s disease (PD). aSyn was initially described in the nucleus and presynaptic nerve terminals. However, the biology of nuclear aSyn and the link of aSyn between subcellular compartments are less understood. Current knowledge suggests the existence of various aSyn species with distinct structural and biochemical properties. Here, we identified a C-terminal-targeting aSyn antibody (Nu-aSyn-C), which has a high immunoaffinity towards aSyn in the nucleus. Comparing the Nu-aSyn-C antibody to aSyn antibodies developed against phosphorylated or aggregated forms, we observed that nuclear aSyn differs from cytosolic aSyn by an increased phosphorylation and assembly level in proliferating cells. Employing Nu-aSyn-C, we characterized aSyn distribution during neuronal differentiation in midbrain dopaminergic neurons (mDANs) derived from human-induced pluripotent stem cells (hiPSCs) and Lund human mesencephalic cells, and in primary rat hippocampal neurons. We detected a specific translocation pattern of aSyn during neuronal differentiation from the nucleus to the soma and finally to neuronal processes. Interestingly, a remarkable shift of Nu-aSyn-C-positive species towards neurites was detected in hiPSC mDANs from a PD patient carrying aSyn gene duplication. Together, our results reveal distinct nuclear and cytosolic aSyn species that redistribute during neuronal differentiation—a process that is altered in PD-derived neurons.
Collapse
Affiliation(s)
- Katharina Pieger
- Department of Biology, Animal Physiology/Neurobiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91058 Erlangen, Germany
| | - Verena Schmitt
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Carina Gauer
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Nadja Gießl
- Department of Biology, Animal Physiology/Neurobiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91058 Erlangen, Germany
| | - Iryna Prots
- Department of Stem Cell Biology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Beate Winner
- Department of Stem Cell Biology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Jürgen Winkler
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Johann Helmut Brandstätter
- Department of Biology, Animal Physiology/Neurobiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91058 Erlangen, Germany
| | - Wei Xiang
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
- Correspondence: ; Tel.: +49-9131-85-44676
| |
Collapse
|
7
|
Alpha synuclein processing by MMP-3 - implications for synucleinopathies. Behav Brain Res 2022; 434:114020. [PMID: 35870616 DOI: 10.1016/j.bbr.2022.114020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 07/01/2022] [Accepted: 07/19/2022] [Indexed: 11/22/2022]
Abstract
α-Synuclein (aSyn) is a protein implicated in physiological functions such as neurotransmitter release at the synapse and the regulation of gene expression in the nucleus. In addition, pathological aSyn assemblies are characteristic for a class of protein aggregation disorders referred to as synucleinopathies, where aSyn aggregates appear as Lewy bodies and Lewy neurites. We recently discovered a novel post-translational pyroglutamate (pGlu) modification at Gln79 of N-truncated aSyn that promotes oligomer formation and neurotoxicity in human synucleinopathies. A priori, the appearance of pGlu79-aSyn in vivo involves a two-step process of free N-terminal Gln79 residue generation and subsequent cyclization of Gln79 into pGlu79. Prime candidate enzymes for these processes are matrix metalloproteinase-3 (MMP-3) and glutaminyl cyclase (QC). Here, we analyzed the expression of aSyn, MMP-3, QC and pGlu79-aSyn in brains of two transgenic mouse models for synucleinopathies (BAC-SNCA and ASO) by triple immunofluorescent labellings and confocal laser scanning microscopy. We report a co-localization of these proteins in brain structures typically affected by aSyn pathology, namely hippocampus in BAC-SNCA mice and substantia nigra in ASO mice. In addition, Western blot analyses revealed a high abundance of QC, MMP-3 and transgenic human aSyn in brain stem and thalamus but lower levels in cortex/hippocampus, whereas endogenous mouse aSyn was found to be most abundant in cortex/hippocampus, followed by thalamus and brain stem. During aging of ASO mice, we observed no differences between controls and transgenic mice in MMP-3 levels but higher QC content in thalamus of 6-month-old transgenic mice. Transgenic human aSyn abundance transiently increased and then showed decrease in oldest ASO mice analyzed. Immunohistochemistry revealed a successive increase in intraneuronal and extracellular formation of pGlu79-aSyn in substantia nigra during aging of ASO mice. Together, our data are supportive for a role of MMP-3 and QC in the generation of pGlu79-aSyn in brains affected by aSyn pathology.
Collapse
|
8
|
Dutta D, Paidi RK, Raha S, Roy A, Chandra S, Pahan K. Treadmill exercise reduces α-synuclein spreading via PPARα. Cell Rep 2022; 40:111058. [PMID: 35830804 PMCID: PMC9308946 DOI: 10.1016/j.celrep.2022.111058] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 12/29/2021] [Accepted: 06/15/2022] [Indexed: 11/30/2022] Open
Abstract
This study underlines the importance of treadmill exercise in reducing α-synuclein (α-syn) spreading in the A53T brain and protecting nigral dopaminergic neurons. Preformed α-syn fibril (PFF) seeding in the internal capsule of young A53T α-syn mice leads to increased spreading of α-syn to substantia nigra and motor cortex and concomitant loss of nigral dopaminergic neurons. However, regular treadmill exercise decreases α-syn spreading in the brain and protects nigral dopaminergic neurons in PFF-seeded mice. Accordingly, treadmill exercise also mitigates α-synucleinopathy in aged A53T mice. While investigating this mechanism, we have observed that treadmill exercise induces the activation of peroxisome proliferator-activated receptor α (PPARα) in the brain to stimulate lysosomal biogenesis via TFEB. Accordingly, treadmill exercise remains unable to stimulate TFEB and reduce α-synucleinopathy in A53T mice lacking PPARα, and fenofibrate, a prototype PPARα agonist, reduces α-synucleinopathy. These results delineate a beneficial function of treadmill exercise in reducing α-syn spreading in the brain via PPARα.
Collapse
Affiliation(s)
- Debashis Dutta
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL 60612, USA
| | - Ramesh Kumar Paidi
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL 60612, USA
| | - Sumita Raha
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL 60612, USA
| | - Avik Roy
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL 60612, USA; Division of Research and Development, Jesse Brown Veterans Affairs Medical Center, Chicago, IL, USA
| | - Sujyoti Chandra
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL 60612, USA
| | - Kalipada Pahan
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL 60612, USA; Division of Research and Development, Jesse Brown Veterans Affairs Medical Center, Chicago, IL, USA.
| |
Collapse
|
9
|
Cognition as a mediator for gait and balance impairments in GBA-related Parkinson's disease. NPJ Parkinsons Dis 2022; 8:78. [PMID: 35725575 PMCID: PMC9209443 DOI: 10.1038/s41531-022-00344-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 05/26/2022] [Indexed: 11/18/2022] Open
Abstract
The extent to which the heterogeneity of gait and balance problems in PD may be explained by genetic variation is unknown. Variants in the glucocerebrosidase (GBA) gene are the strongest known genetic risk factor for PD and are associated with greater motor and cognitive severity. However, the impact of GBA variants on comprehensive measures of gait and balance and their relationship to cognition remains unknown. We aimed to determine differences in gait and balance impairments in those with and without GBA variants (mutation carriers and E326K polymorphism) and explore direct and indirect effects of GBA status on gait, balance, and cognition. 332 participants, 43 of whom had GBA variants, were recruited. Participants completed a comprehensive, objective assessment of gait and standing balance using body-worn inertial sensors. Group differences in gait and balance between PD with and without GBA variants were assessed with linear regression, adjusting for age, gender, clinical testing site, disease duration, and apolipoprotein E (APOE) ɛ4 status. Structural equation modeling (SEM) explored direct relationships between GBA status and gait and balance and indirect relationships between GBA status and gait and balance via cognition. The GBA variant group had more impaired gait (pace and variability) and balance (sway area/jerk and sway velocity), than the non-GBA variant group. SEM demonstrated cognition as a mediator of GBA status on gait and balance. The close relationships among GBA, gait/balance, and cognition suggest potential for novel therapeutics to target the GBA pathway and/or cognition to improve mobility in PD GBA variants.
Collapse
|
10
|
Hartlage-Rübsamen M, Bluhm A, Moceri S, Machner L, Köppen J, Schenk M, Hilbrich I, Holzer M, Weidenfeller M, Richter F, Coras R, Serrano GE, Beach TG, Schilling S, von Hörsten S, Xiang W, Schulze A, Roßner S. A glutaminyl cyclase-catalyzed α-synuclein modification identified in human synucleinopathies. Acta Neuropathol 2021; 142:399-421. [PMID: 34309760 PMCID: PMC8357657 DOI: 10.1007/s00401-021-02349-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 07/13/2021] [Accepted: 07/13/2021] [Indexed: 12/22/2022]
Abstract
Parkinson’s disease (PD) is a progressive neurodegenerative disorder that is neuropathologically characterized by degeneration of dopaminergic neurons of the substantia nigra (SN) and formation of Lewy bodies and Lewy neurites composed of aggregated α-synuclein. Proteolysis of α-synuclein by matrix metalloproteinases was shown to facilitate its aggregation and to affect cell viability. One of the proteolysed fragments, Gln79-α-synuclein, possesses a glutamine residue at its N-terminus. We argue that glutaminyl cyclase (QC) may catalyze the pyroglutamate (pGlu)79-α-synuclein formation and, thereby, contribute to enhanced aggregation and compromised degradation of α-synuclein in human synucleinopathies. Here, the kinetic characteristics of Gln79-α-synuclein conversion into the pGlu-form by QC are shown using enzymatic assays and mass spectrometry. Thioflavin T assays and electron microscopy demonstrated a decreased potential of pGlu79-α-synuclein to form fibrils. However, size exclusion chromatography and cell viability assays revealed an increased propensity of pGlu79-α-synuclein to form oligomeric aggregates with high neurotoxicity. In brains of wild-type mice, QC and α-synuclein were co-expressed by dopaminergic SN neurons. Using a specific antibody against the pGlu-modified neo-epitope of α-synuclein, pGlu79-α-synuclein aggregates were detected in association with QC in brains of two transgenic mouse lines with human α-synuclein overexpression. In human brain samples of PD and dementia with Lewy body subjects, pGlu79-α-synuclein was shown to be present in SN neurons, in a number of Lewy bodies and in dystrophic neurites. Importantly, there was a spatial co-occurrence of pGlu79-α-synuclein with the enzyme QC in the human SN complex and a defined association of QC with neuropathological structures. We conclude that QC catalyzes the formation of oligomer-prone pGlu79-α-synuclein in human synucleinopathies, which may—in analogy to pGlu-Aβ peptides in Alzheimer’s disease—act as a seed for pathogenic protein aggregation.
Collapse
|
11
|
Clemen CS, Schmidt A, Winter L, Canneva F, Wittig I, Becker L, Coras R, Berwanger C, Hofmann A, Eggers B, Marcus K, Gailus-Durner V, Fuchs H, de Angelis MH, Krüger M, von Hörsten S, Eichinger L, Schröder R. N471D WASH complex subunit strumpellin knock-in mice display mild motor and cardiac abnormalities and BPTF and KLHL11 dysregulation in brain tissue. Neuropathol Appl Neurobiol 2021; 48:e12750. [PMID: 34312900 DOI: 10.1111/nan.12750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 07/12/2021] [Indexed: 11/30/2022]
Abstract
AIMS We investigated N471D WASH complex subunit strumpellin (Washc5) knock-in and Washc5 knock-out mice as models for hereditary spastic paraplegia type 8 (SPG8). METHODS We generated heterozygous and homozygous N471D Washc5 knock-in mice and subjected them to a comprehensive clinical, morphological and laboratory parameter screen, and gait analyses. Brain tissue was used for proteomic analysis. Furthermore, we generated heterozygous Washc5 knock-out mice. WASH complex subunit strumpellin expression was determined by qPCR and immunoblotting. RESULTS Homozygous N471D Washc5 knock-in mice showed mild dilated cardiomyopathy, decreased acoustic startle reactivity, thinner eye lenses, increased alkaline phosphatase and potassium levels and increased white blood cell counts. Gait analyses revealed multiple aberrations indicative of locomotor instability. Similarly, the clinical chemistry, haematology and gait parameters of heterozygous mice also deviated from the values expected for healthy animals, albeit to a lesser extent. Proteomic analysis of brain tissue depicted consistent upregulation of BPTF and downregulation of KLHL11 in heterozygous and homozygous knock-in mice. WASHC5-related protein interaction partners and complexes showed no change in abundancies. Heterozygous Washc5 knock-out mice showing normal WASHC5 levels could not be bred to homozygosity. CONCLUSIONS While biallelic ablation of Washc5 was prenatally lethal, expression of N471D mutated WASHC5 led to several mild clinical and laboratory parameter abnormalities, but not to a typical SPG8 phenotype. The consistent upregulation of BPTF and downregulation of KLHL11 suggest mechanistic links between the expression of N471D mutated WASHC5 and the roles of both proteins in neurodegeneration and protein quality control, respectively.
Collapse
Affiliation(s)
- Christoph S Clemen
- Institute of Aerospace Medicine, German Aerospace Center, Cologne, Germany.,Center for Physiology and Pathophysiology, Institute of Vegetative Physiology, Medical Faculty, University of Cologne, Cologne, Germany.,Center for Biochemistry, Institute of Biochemistry I, Medical Faculty, University of Cologne, Cologne, Germany
| | - Andreas Schmidt
- Center for Molecular Medicine and Excellence Cluster "Cellular Stress Responses in Aging-Associated Diseases" (CECAD), University of Cologne, Cologne, Germany
| | - Lilli Winter
- Institute of Neuropathology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany.,Neuromuscular Research Department, Center for Anatomy and Cell Biology, Medical University of Vienna, Vienna, Austria
| | - Fabio Canneva
- Experimental Therapy, University Hospital Erlangen and Preclinical Experimental Center, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Ilka Wittig
- Functional Proteomics, Medical School, Goethe University, Frankfurt, Germany
| | - Lore Becker
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Roland Coras
- Institute of Neuropathology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Carolin Berwanger
- Institute of Aerospace Medicine, German Aerospace Center, Cologne, Germany
| | | | - Andreas Hofmann
- Faculty of Veterinary and Agricultural Sciences, University of Melbourne, Parkville, Victoria, Australia
| | - Britta Eggers
- Medical Proteome Center, Medical Faculty, and Medical Proteome Analysis, Center for Protein Diagnostics (PRODI), Ruhr-University Bochum, Bochum, Germany
| | - Katrin Marcus
- Medical Proteome Center, Medical Faculty, and Medical Proteome Analysis, Center for Protein Diagnostics (PRODI), Ruhr-University Bochum, Bochum, Germany
| | - Valerie Gailus-Durner
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Helmut Fuchs
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Martin Hrabe de Angelis
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany.,TUM School of Life Sciences (SoLS), Technical University of Munich, Freising, Germany.,German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Marcus Krüger
- Center for Molecular Medicine and Excellence Cluster "Cellular Stress Responses in Aging-Associated Diseases" (CECAD), University of Cologne, Cologne, Germany
| | - Stephan von Hörsten
- Experimental Therapy, University Hospital Erlangen and Preclinical Experimental Center, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Ludwig Eichinger
- Center for Biochemistry, Institute of Biochemistry I, Medical Faculty, University of Cologne, Cologne, Germany
| | - Rolf Schröder
- Institute of Neuropathology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
12
|
Brown JM, Baker LS, Seroogy KB, Genter MB. Intranasal Carnosine Mitigates α-Synuclein Pathology and Motor Dysfunction in the Thy1-aSyn Mouse Model of Parkinson's Disease. ACS Chem Neurosci 2021; 12:2347-2359. [PMID: 34138535 PMCID: PMC9996643 DOI: 10.1021/acschemneuro.1c00096] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Parkinson's disease (PD) is a debilitating neurodegenerative disorder. Early symptoms include motor dysfunction and impaired olfaction. Toxic aggregation of α-synuclein (aSyn) in the olfactory bulb (OB) and substantia nigra pars compacta (SNpc) is a hallmark of PD neuropathology. Intranasal (IN) carnosine (2 mg/d for 8 weeks) was previously demonstrated to improve motor behavior and mitochondrial function in Thy1-aSyn mice, a model of PD. The present studies evaluated the efficacy of IN carnosine at a higher dose in slowing progression of motor deficits and aSyn accumulation in Thy1-aSyn mice. After baseline neurobehavioral assessments, IN carnosine was administered (0.0, 2.0, or 4.0 mg/day) to wild-type and Thy1-aSyn mice for 8 weeks. Olfactory and motor behavioral measurements were repeated prior to end point tissue collection. Brain sections were immunostained for aSyn and tyrosine hydroxylase (TH). Immunopositive cells were counted using design-based stereology in the SNpc and OB mitral cell layer (MCL). Behavioral assessments revealed a dose-dependent improvement in motor function with increasing carnosine dose. Thy1-aSyn mice treated with 2.0 or 4.0 mg/d IN carnosine exhibited fewer aSyn-positive (aSyn(+)) cell bodies in the SNpc compared to vehicle-treated mice. Moreover, the number of aSyn(+) cell bodies in carnosine-treated Thy1-aSyn mice was reduced to vehicle-treated wild-type levels in the SNpc. Carnosine treatment did not affect the number of aSyn(+) cell bodies in the OB-MCL or the number of TH(+) cells in the SNpc. In summary, intranasal carnosine treatment decreased aSyn accumulation in the SNpc, which may underlie its mitigation of motor deficits in the Thy1-aSyn mice.
Collapse
Affiliation(s)
- Josephine M Brown
- Department of Environmental and Public Health Sciences, College of Medicine, University of Cincinnati, 160 Panzeca Way, Cincinnati, Ohio 45267-0056, United States
| | - Lauren S Baker
- Department of Environmental and Public Health Sciences, College of Medicine, University of Cincinnati, 160 Panzeca Way, Cincinnati, Ohio 45267-0056, United States
| | - Kim B Seroogy
- Department of Neurology and Rehabilitation Medicine, College of Medicine, University of Cincinnati, Cincinnati, Ohio 45267-0536, United States
| | - Mary Beth Genter
- Department of Environmental and Public Health Sciences, College of Medicine, University of Cincinnati, 160 Panzeca Way, Cincinnati, Ohio 45267-0056, United States
| |
Collapse
|
13
|
Schmidt MY, Chamoli M, Lithgow GJ, Andersen JK. Swimming exercise reduces native ⍺-synuclein protein species in a transgenic C. elegans model of Parkinson's disease. MICROPUBLICATION BIOLOGY 2021; 2021. [PMID: 34222835 PMCID: PMC8243224 DOI: 10.17912/micropub.biology.000413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Exercise has been historically recommended to prevent many disease conditions. Intense exercise in particular, has been shown to be beneficial for Parkinson's disease (PD) - stopping and even reversing symptoms in some patients. Recent research in mammalian animal models of Parkinson's have shown that exercise affects ⍺-synuclein aggregate species, considered to be a hallmark of PD. However, the exact changes in native ⍺-synuclein protein species after exercise and the downstream effects of exercise upon the health of the animals remains unclear. Recently, it was shown that swimming constitutes a form of exercise in C. elegans worms that confers a protective effect in several worm models of tau and Huntington protein neurodegeneration. Here we show that a period of swimming exercise (Ex) - 15-20 mins - dramatically reduces several native human ⍺-synuclein protein species in the NL5901 C. elegans worm model of Parkinson's. Exercise on Day 1 of adulthood was found to improve motor function measured by the thrashing rate of worms on Day 2 and Day 4 when compared to both control (untreated) and food restricted (FR) worms. Moreover, exercised worms show smaller ⍺-synuclein::YFP puncta than food restricted worms. Here we show that exercise reduces native human ⍺-synuclein levels independent of food restriction in C. elegans.
Collapse
Affiliation(s)
- Minna Y Schmidt
- The Buck Institute for Research on Aging.,The University of Southern California, Leonard Davis School of Gerontology
| | | | - Gordon J Lithgow
- The Buck Institute for Research on Aging.,The University of Southern California, Leonard Davis School of Gerontology
| | - Julie K Andersen
- The Buck Institute for Research on Aging.,The University of Southern California, Leonard Davis School of Gerontology
| |
Collapse
|
14
|
Mészáros L, Riemenschneider MJ, Gassner H, Marxreiter F, von Hörsten S, Hoffmann A, Winkler J. Human alpha-synuclein overexpressing MBP29 mice mimic functional and structural hallmarks of the cerebellar subtype of multiple system atrophy. Acta Neuropathol Commun 2021; 9:68. [PMID: 33853667 PMCID: PMC8048356 DOI: 10.1186/s40478-021-01166-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 03/19/2021] [Indexed: 12/13/2022] Open
Abstract
Multiple system atrophy (MSA) is a rare, but fatal atypical parkinsonian disorder. The prototypical pathological hallmark are oligodendroglial cytoplasmic inclusions (GCIs) containing alpha-synuclein (α-syn). Currently, two MSA phenotypes are classified: the parkinsonian (MSA-P) and the cerebellar subtype (MSA-C), clinically characterized by predominant parkinsonism or cerebellar ataxia, respectively. Previous studies have shown that the transgenic MSA mouse model overexpressing human α-syn controlled by the oligodendroglial myelin basic protein (MBP) promoter (MBP29-hα-syn mice) mirrors crucial characteristics of the MSA-P subtype. However, it remains elusive, whether this model recapitulates important features of the MSA-C-related phenotype. First, we examined MSA-C-associated cerebellar pathology using human post-mortem tissue of MSA-C patients and controls. We observed the prototypical GCI pathology and a preserved number of oligodendrocytes in the cerebellar white matter (cbw) accompanied by severe myelin deficit, microgliosis, and a profound loss of Purkinje cells. Secondly, we phenotypically characterized MBP29-hα-syn mice using a dual approach: structural analysis of the hindbrain and functional assessment of gait. Matching the neuropathological features of MSA-C, GCI pathology within the cbw of MBP29-hα-syn mice was accompanied by a severe myelin deficit despite an increased number of oligodendrocytes and a high number of myeloid cells even at an early disease stage. Intriguingly, MBP29-hα-syn mice developed a significant loss of Purkinje cells at a more advanced disease stage. Catwalk XT gait analysis revealed decreased walking speed, increased stride length and width between hind paws. In addition, less dual diagonal support was observed toward more dual lateral and three paw support. Taken together, this wide-based and unsteady gait reflects cerebellar ataxia presumably linked to the cerebellar pathology in MBP29-hα-syn mice. In conclusion, the present study strongly supports the notion that the MBP29-hα-syn mouse model mimics important characteristics of the MSA-C subtype providing a powerful preclinical tool for evaluating future interventional strategies.
Collapse
Affiliation(s)
- Lisa Mészáros
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054, Erlangen, Germany
| | | | - Heiko Gassner
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054, Erlangen, Germany
| | - Franz Marxreiter
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054, Erlangen, Germany
| | - Stephan von Hörsten
- Experimental Therapy, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054, Erlangen, Germany
| | - Alana Hoffmann
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054, Erlangen, Germany.
| | - Jürgen Winkler
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054, Erlangen, Germany.
| |
Collapse
|
15
|
Chaprov K, Rezvykh A, Funikov S, Ivanova TA, Lysikova EA, Deykin AV, Kukharsky MS, Yu Aksinenko A, Bachurin SO, Ninkina N, Buchman VL. A bioisostere of Dimebon/Latrepirdine delays the onset and slows the progression of pathology in FUS transgenic mice. CNS Neurosci Ther 2021; 27:765-775. [PMID: 33754495 PMCID: PMC8193697 DOI: 10.1111/cns.13637] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 03/04/2021] [Accepted: 03/07/2021] [Indexed: 12/13/2022] Open
Abstract
Aims To assess effects of DF402, a bioisostere of Dimebon/Latrepirdine, on the disease progression in the transgenic model of amyotrophic lateral sclerosis (ALS) caused by expression of pathogenic truncated form of human FUS protein. Methods Mice received DF402 from the age of 42 days and the onset of clinical signs, the disease duration and animal lifespan were monitored for experimental and control animals, and multiple parameters of their gait were assessed throughout the pre‐symptomatic stage using CatWalk system followed by a bioinformatic analysis. RNA‐seq was used to compare the spinal cord transcriptomes of wild‐type, untreated, and DF402‐treated FUS transgenic mice. Results DF402 delays the onset and slows the progression of pathology. We developed a CatWalk analysis protocol that allows detection of gait changes in FUS transgenic mice and the effect of DF402 on their gait already at early pre‐symptomatic stage. At this stage, a limited number of genes significantly change expression in transgenic mice and for 60% of these genes, DF402 treatment causes the reversion of the expression pattern. Conclusion DF402 slows down the disease progression in the mouse model of ALS, which is consistent with previously reported neuroprotective properties of Dimebon and its other bioisosteres. These results suggest that these structures can be considered as lead compounds for further optimization to obtain novel medicines that might be used as components of complex ALS therapy.
Collapse
Affiliation(s)
- Kirill Chaprov
- Institute of Physiologically Active Compounds, Russian Academy of Science, Chernogolovka, Russia
| | - Alexander Rezvykh
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia.,Moscow Institute of Physics and Technology, Dolgoprudny, Russia
| | - Sergei Funikov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Tamara A Ivanova
- Institute of Physiologically Active Compounds, Russian Academy of Science, Chernogolovka, Russia
| | - Ekaterina A Lysikova
- Institute of Physiologically Active Compounds, Russian Academy of Science, Chernogolovka, Russia
| | - Alexei V Deykin
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Institute of Gene Biology, Russian Academy of Sciences, Moscow, Russia.,Laboratory of Genome Editing for Veterinary and Biomedicine, Belgorod State National Research University, Belgorod region, Russia
| | - Michail S Kukharsky
- Institute of Physiologically Active Compounds, Russian Academy of Science, Chernogolovka, Russia.,Pirogov Russian National Research Medical University, Moscow, Russia.,School of Biosciences, Cardiff University, Cardiff, UK
| | - Alexey Yu Aksinenko
- Institute of Physiologically Active Compounds, Russian Academy of Science, Chernogolovka, Russia
| | - Sergey O Bachurin
- Institute of Physiologically Active Compounds, Russian Academy of Science, Chernogolovka, Russia
| | - Natalia Ninkina
- Institute of Physiologically Active Compounds, Russian Academy of Science, Chernogolovka, Russia.,School of Biosciences, Cardiff University, Cardiff, UK
| | - Vladimir L Buchman
- Institute of Physiologically Active Compounds, Russian Academy of Science, Chernogolovka, Russia.,School of Biosciences, Cardiff University, Cardiff, UK
| |
Collapse
|
16
|
Chaprov KD, Teterina EV, Roman AY, Ivanova TA, Goloborshcheva VV, Kucheryanu VG, Morozov SG, Lysikova EA, Lytkina OA, Koroleva IV, Popova NI, Antohin AI, Ovchinnikov RK, Kukharsky MS. Comparative Analysis of MPTP Neurotoxicity in Mice with a Constitutive Knockout of the α-Synuclein Gene. Mol Biol 2021. [DOI: 10.1134/s0026893321010039] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
17
|
Mahalakshmi B, Maurya N, Lee SD, Bharath Kumar V. Possible Neuroprotective Mechanisms of Physical Exercise in Neurodegeneration. Int J Mol Sci 2020; 21:ijms21165895. [PMID: 32824367 PMCID: PMC7460620 DOI: 10.3390/ijms21165895] [Citation(s) in RCA: 161] [Impact Index Per Article: 32.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 08/13/2020] [Accepted: 08/15/2020] [Indexed: 12/22/2022] Open
Abstract
Physical exercise (PE) improves physical performance, mental status, general health, and well-being. It does so by affecting many mechanisms at the cellular and molecular level. PE is beneficial for people suffering from neuro-degenerative diseases because it improves the production of neurotrophic factors, neurotransmitters, and hormones. PE promotes neuronal survival and neuroplasticity and also optimizes neuroendocrine and physiological responses to psychosocial and physical stress. PE sensitizes the parasympathetic nervous system (PNS), Autonomic Nervous System (ANS) and central nervous system (CNS) by promoting many processes such as synaptic plasticity, neurogenesis, angiogenesis, and autophagy. Overall, it carries out many protective and preventive activities such as improvements in memory, cognition, sleep and mood; growth of new blood vessels in nervous system; and the reduction of stress, anxiety, neuro-inflammation, and insulin resistance. In the present work, the protective effects of PE were overviewed. Suitable examples from the current research work in this context are also given in the article.
Collapse
Affiliation(s)
- B. Mahalakshmi
- Institute of Research and Development, Duy Tan University, Da Nang 550000, Vietnam;
| | - Nancy Maurya
- Department of Botany, Government Science College, Pandhurna, Chhindwara, Madhya Pradesh 480334, India;
| | - Shin-Da Lee
- Department of Physical Therapy, Asia University, Taichung 41354, Taiwan
- Department of Physical Therapy Graduate Institute of Rehabilitation Science, China Medical University, Taichung 40402, Taiwan
- Correspondence: (S.-D.L.); (V.B.K.); Tel.: +886-4-22053366 (ext. 7300) (S.-D.L.); +886-4-2332-3456 (ext. 6352 or 6353) (V.B.K.); Fax: 886-4-22065051 (S.-D.L.); +886-4-23305834 (V.B.K.)
| | - V. Bharath Kumar
- Department of Medical Laboratory Science and Biotechnology, Asia University, Taichung 41354, Taiwan
- Correspondence: (S.-D.L.); (V.B.K.); Tel.: +886-4-22053366 (ext. 7300) (S.-D.L.); +886-4-2332-3456 (ext. 6352 or 6353) (V.B.K.); Fax: 886-4-22065051 (S.-D.L.); +886-4-23305834 (V.B.K.)
| |
Collapse
|
18
|
Húngaro TGR, Freitas-Lima LC, Gregnani MF, Perilhão MS, Alves-Silva T, Arruda AC, Barrera-Chimal J, Estrela GR, Araújo RC. Physical Exercise Exacerbates Acute Kidney Injury Induced by LPS via Toll-Like Receptor 4. Front Physiol 2020; 11:768. [PMID: 32765291 PMCID: PMC7380174 DOI: 10.3389/fphys.2020.00768] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 06/11/2020] [Indexed: 12/15/2022] Open
Abstract
Introduction: Lipopolysaccharide (LPS) is a systemic response-triggering endotoxin, which has the kidney as one of its first targets, thus causing acute injuries to this organ. Physical exercise is capable of promoting physiological alterations and modulating inflammatory responses in the infectious process through multiple parameters, including the toll-like receptor (TLR)-4 pathway, which is the main LPS signaling in sepsis. Additionally, previous studies have shown that physical exercise can be both a protector factor and an aggravating factor for some kidney diseases. This study aims at analyzing whether physical exercise before the induction of LPS endotoxemia can protect kidneys from acute kidney injury. Methods: C57BL/6J male mice, 12 weeks old, were distributed into four groups: (1) sedentary (control, N = 7); (2) sedentary + LPS (N = 7); (3) trained (N = 7); and (4) trained + LPS (N = 7). In the training groups, the animals exercised 5×/week in a treadmill, 60 min/day, for 4 weeks (60% of max. velocity). Sepsis was induced in the training group by the application of a single dose of LPS (5 mg/kg i.p.). Sedentary animals received LPS on the same day, and the non-LPS groups received a saline solution instead. All animals were euthanized 24 h after the administration of LPS or saline. Results: The groups receiving LPS presented a significant increase in serum urea (p < 0.0001) and creatinine (p < 0.001) concentration and renal gene expression of inflammatory markers, such as tumor necrosis factor alpha and interleukin-6, as well as TLRs. In addition, LPS promoted a decrease in reduced glutathione. Compared to the sedentary + LPS group, trained + LPS showed overexpression of a gene related to kidney injury (NGAL, p < 0.01) and the protein levels of LPS receptor TLR-4 (p < 0.01). Trained + LPS animals showed an expansion of the tubulointerstitial space in the kidney (p < 0.05) and a decrease in the gene expression of hepatic AOAH (p < 0.01), an enzyme involved in LPS clearance. Conclusion: In contrast to our hypothesis, training was unable to mitigate the renal inflammatory response caused by LPS. On the contrary, it seems to enhance injury by accentuating endotoxin-induced TLR-4 signaling. This effect could be partly due to the modulation of a hepatic enzyme that detoxifies LPS.
Collapse
Affiliation(s)
- Talita Guerreiro Rodrigues Húngaro
- Laboratório de Genética e Metabolismo do Exercício, Programa de Nefrologia, Departamento de Biofísica, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Leandro Ceotto Freitas-Lima
- Laboratório de Genética e Metabolismo do Exercício, Programa de Biologia Molecular, Departamento de Biofísica, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Marcos Fernandes Gregnani
- Laboratório de Genética e Metabolismo do Exercício, Programa de Biologia Molecular, Departamento de Biofísica, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Mauro Sérgio Perilhão
- Laboratório de Genética e Metabolismo do Exercício, Programa de Nefrologia, Departamento de Biofísica, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Thaís Alves-Silva
- Laboratório de Genética e Metabolismo do Exercício, Programa de Biologia Molecular, Departamento de Biofísica, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Adriano Cleis Arruda
- Laboratório de Genética e Metabolismo do Exercício, Programa de Nefrologia, Departamento de Biofísica, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Jonatan Barrera-Chimal
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico.,Unidad de Investigación UNAM-INC, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico
| | - Gabriel Rufino Estrela
- Departamento de Oncologia Clínica e Experimental, Disciplina de Hematologia e Hematoterapia, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Ronaldo Carvalho Araújo
- Laboratório de Genética e Metabolismo do Exercício, Programa de Nefrologia, Departamento de Biofísica, Universidade Federal de São Paulo, São Paulo, Brazil.,Laboratório de Genética e Metabolismo do Exercício, Programa de Biologia Molecular, Departamento de Biofísica, Universidade Federal de São Paulo, São Paulo, Brazil
| |
Collapse
|
19
|
Kwok A, Rosas S, Bateman TA, Livingston E, Smith TL, Moore J, Zawieja DC, Hampton T, Mao XW, Delp MD, Willey JS. Altered rodent gait characteristics after ~35 days in orbit aboard the International Space Station. LIFE SCIENCES IN SPACE RESEARCH 2020; 24:9-17. [PMID: 31987483 DOI: 10.1016/j.lssr.2019.10.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 10/23/2019] [Accepted: 10/29/2019] [Indexed: 06/10/2023]
Abstract
The long-term adaptations to microgravity and other spaceflight challenges within the confines of a spacecraft, and readaptations to weight-bearing upon reaching a destination, are unclear. While post-flight gait change in astronauts have been well documented and reflect multi-system deficits, no data from rodents have been collected. Thus, the purpose of this study was to evaluate gait changes in response to spaceflight. A prospective collection of gait data was collected on 3 groups of mice: those who spent~35 days in orbit (FLIGHT) aboard the International Space Station (ISS); a ground-based control with the same habitat conditions as ISS (Ground Control; GC); and a vivarium control with typical rodent housing conditions (VIV). Pre-flight and post-flight gait measurements were conducted utilizing an optimized and portable gait analysis system (DigiGait, Mouse Specifics, Inc). The total data acquisition time for gait patterns of FLIGHT and control mice was 1.5-5 min/mouse, allowing all 20 mice per group to be assessed in less than an hour. Patterns of longitudinal gait changes were observed in the hind limbs and the forelimbs of the FLIGHT mice after ~35 days in orbit; few differences were observed in gait characteristics within the GC and VIV controls from the initial to the final gait assessment, and between groups. For FLIGHT mice, 12 out of 18 of the evaluated gait characteristics in the hind limbs were significantly changed, including: stride width variability; stride length and variance; stride, swing, and stance duration; paw angle and area at peak stance; and step angle, among others. Gait characteristics that decreased included stride frequency, and others. Moreover, numerous forelimb gait characteristics in the FLIGHT mice were changed at post-flight measures relative to pre-flight. This rapid DigiGait gait measurement tool and customized spaceflight protocol is useful for providing preliminary insight into how spaceflight could affect multiple systems in rodents in which deficits are reflected by altered gait characteristics.
Collapse
Affiliation(s)
- Andy Kwok
- Department of Radiation Oncology, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Samuel Rosas
- Department of Radiation Oncology, Wake Forest School of Medicine, Winston-Salem, NC, United States; Department of Orthopaedic Surgery, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Ted A Bateman
- Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Eric Livingston
- Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Thomas L Smith
- Department of Orthopaedic Surgery, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Joseph Moore
- Department of Radiation Oncology, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - David C Zawieja
- Department of Medical Physiology, Texas A&M University, College Station, TX, United States
| | - Tom Hampton
- Mouse Specifics, Framingham, MA, United States
| | - Xiao W Mao
- Department of Basic Sciences, Division of Biomedical Engineering Sciences (BMES), Loma Linda University School of Medicine and Medical Center, Loma Linda, CA, United States
| | - Michael D Delp
- Department of Nutrition, Food and Exercise Sciences, Florida State University, Tallahassee, FL, United States
| | - Jeffrey S Willey
- Department of Radiation Oncology, Wake Forest School of Medicine, Winston-Salem, NC, United States.
| |
Collapse
|
20
|
Timotius IK, Canneva F, Minakaki G, Moceri S, Plank AC, Casadei N, Riess O, Winkler J, Klucken J, Eskofier B, von Hörsten S. Systematic data analysis and data mining in CatWalk gait analysis by heat mapping exemplified in rodent models for neurodegenerative diseases. J Neurosci Methods 2019; 326:108367. [PMID: 31351096 DOI: 10.1016/j.jneumeth.2019.108367] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 07/12/2019] [Accepted: 07/15/2019] [Indexed: 12/15/2022]
Abstract
BACKGROUND Motor impairment appears as a characteristic symptom of several diseases and injuries. Therefore, tests for analyzing motor dysfunction are widely applied across preclinical models and disease stages. Among those, gait analysis tests are commonly used, but they generate a huge number of gait parameters. Thus, complications in data analysis and reporting raise, which often leads to premature parameter selection. NEW METHODS In order to avoid arbitrary parameter selection, we present here a systematic initial data analysis by utilizing heat-maps for data reporting. We exemplified this approach within an intervention study, as well as applied it to two longitudinal studies in rodent models related to Parkinson's disease (PD) and Huntington disease (HD). RESULTS The systematic initial data analysis (IDA) is feasible for exploring gait parameters, both in experimental and longitudinal studies. The resulting heat maps provided a visualization of gait parameters within a single chart, highlighting important clusters of differences. COMPARISON WITH EXISTING METHOD Often, premature parameter selection is practiced, lacking comprehensiveness. Researchers often use multiple separated graphs on distinct gait parameters for reporting. Additionally, negative results are often not reported. CONCLUSIONS Heat mapping utilized in initial data analysis is advantageous for reporting clustered gait parameter differences in one single chart and improves data mining.
Collapse
Affiliation(s)
- Ivanna K Timotius
- Machine Learning and Data Analytics Lab, Dept. of Computer Science, Faculty of Engineering, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Germany; Dept. of Electronics Engineering, Satya Wacana Christian University, Salatiga, Indonesia
| | - Fabio Canneva
- Dept. Experimental Therapy, University Hospital Erlangen (UKEr) and Preclinical Experimental Animal Center, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Germany
| | - Georgia Minakaki
- Dept. of Molecular Neurology, University Hospital Erlangen, University of Erlangen-Nürnberg (FAU), Germany
| | - Sandra Moceri
- Dept. Experimental Therapy, University Hospital Erlangen (UKEr) and Preclinical Experimental Animal Center, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Germany
| | - Anne-Christine Plank
- Dept. Experimental Therapy, University Hospital Erlangen (UKEr) and Preclinical Experimental Animal Center, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Germany
| | - Nicolas Casadei
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Germany
| | - Olaf Riess
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Germany
| | - Jürgen Winkler
- Dept. of Molecular Neurology, University Hospital Erlangen, University of Erlangen-Nürnberg (FAU), Germany
| | - Jochen Klucken
- Dept. of Molecular Neurology, University Hospital Erlangen, University of Erlangen-Nürnberg (FAU), Germany
| | - Bjoern Eskofier
- Machine Learning and Data Analytics Lab, Dept. of Computer Science, Faculty of Engineering, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Germany
| | - Stephan von Hörsten
- Dept. Experimental Therapy, University Hospital Erlangen (UKEr) and Preclinical Experimental Animal Center, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Germany.
| |
Collapse
|
21
|
Wassouf Z, Schulze-Hentrich JM. Alpha-synuclein at the nexus of genes and environment: the impact of environmental enrichment and stress on brain health and disease. J Neurochem 2019; 150:591-604. [PMID: 31165472 PMCID: PMC6771760 DOI: 10.1111/jnc.14787] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 05/24/2019] [Accepted: 05/29/2019] [Indexed: 12/16/2022]
Abstract
Accumulation of alpha‐synuclein protein aggregates is the hallmark neuropathologic feature of synucleinopathies such as Parkinson’s disease. Rare point mutations and multiplications in SNCA, the gene encoding alpha‐synuclein, as well as other genetic alterations are linked to familial Parkinson’s disease cases with high penetrance and hence constitute major genetic risk factors for Parkinson’s disease. However, the preponderance of cases seems sporadic, most likely based on a complex interplay between genetic predispositions, aging processes and environmental influences. Deciphering the impact of these environmental factors and their interactions with the individual genetic background in humans is challenging and often requires large cohorts, complicated study designs, and longitudinal set‐ups. In contrast, rodent models offer an ideal system to study the influence of individual environmental aspects under controlled genetic background and standardized conditions. In this review, we highlight findings from studies examining effects of environmental enrichment mimicking stimulation of the brain by its physical and social surroundings as well as of environmental stressors on brain health in the context of Parkinson’s disease. We discuss possible internal molecular transducers of such environmental cues in Parkinson’s disease rodent models and emphasize their potential in developing novel avenues to much‐needed therapies for this still incurable disease. ![]()
This article is part of the Special Issue “Synuclein”
Collapse
Affiliation(s)
- Zinah Wassouf
- German Center for Neurodegenerative Diseases, Göttingen, Germany.,Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany
| | | |
Collapse
|
22
|
Chuang CS, Chang JC, Soong BW, Chuang SF, Lin TT, Cheng WL, Orr HT, Liu CS. Treadmill training increases the motor activity and neuron survival of the cerebellum in a mouse model of spinocerebellar ataxia type 1. Kaohsiung J Med Sci 2019; 35:679-685. [PMID: 31271500 DOI: 10.1002/kjm2.12106] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Accepted: 06/14/2019] [Indexed: 12/19/2022] Open
Abstract
Spinocerebellar ataxia (SCA) type 1 (SCA1) is a rare autosomal dominant disorder that is characterized by worsening of disordered coordination, ataxia of the trunk, and other neurological symptoms. Physical activity improves both mobility and the daily living activities of patients with SCA. Intervention with daily regular treadmill exercise may slow the deterioration of cerebellar neurons in SCA1. Therefore, the signal changes and performance of cerebellar neurons after exercise in SCA1 was investigated in this study. We employed a transgenic mouse model of SCA1, generated by amplifying the cytosine-adenine-guanine trinucleotide repeat expansions, and the mice underwent 1 month of moderate daily treadmill exercise for 1 hour. The rotarod test revealed that the motor function of the SCA1 mice that underwent training was superior to that of the control SCA1 mice, which did not undergo training. Moreover, the cerebellar pathology revealed preserved Purkinje neurons stained by carbindin with an increase of the neuronal Per Arnt Sim domain protein 4, a key regulation in the structural and functional plasticity of neurons, in the excised SCA1 mice relative to the controls. The mechanism was related to an increase of phosphorylation of ribosomal protein S6, a downstream target of the mammalian target of rapamycin pathway, but not to autophagy activation. This study determined that regular treadmill exercise may play a crucial role in the viable support of cerebellar neurons in SCA1.
Collapse
Affiliation(s)
- Chieh-Sen Chuang
- Department of Neurology, Changhua Christian Hospital, Changhua, Taiwan.,Vascular and Genomic Center, Changhua Christian Hospital, Changhua, Taiwan
| | - Jui-Chih Chang
- Vascular and Genomic Center, Changhua Christian Hospital, Changhua, Taiwan
| | - Bing-Wen Soong
- Department of Neurology, Shuang Ho Hospital, Taipei Medical University, Taipei, Taiwan.,Institute of Neuroscience, Taipei Medical University, Taipei, Taiwan
| | - Sheng-Fei Chuang
- Vascular and Genomic Center, Changhua Christian Hospital, Changhua, Taiwan
| | - Ta-Tsung Lin
- Vascular and Genomic Center, Changhua Christian Hospital, Changhua, Taiwan
| | - Wen-Ling Cheng
- Vascular and Genomic Center, Changhua Christian Hospital, Changhua, Taiwan
| | - Harry T Orr
- Department of Laboratory Medicine and Pathology, Minnesota Supercomputing Institute, University of Minnesota, Minneapolis, Minnesota
| | - Chin-San Liu
- Department of Neurology, Changhua Christian Hospital, Changhua, Taiwan.,Vascular and Genomic Center, Changhua Christian Hospital, Changhua, Taiwan.,School of Chinese Medicine, Graduate Institute of Integrated Medicine, Research Center for Chinese Medicine and Acupuncture, China Medical University, Taichung, Taiwan
| |
Collapse
|