1
|
Binda KH, Real CC, Simonsen MT, Grove EK, Bender D, Gjedde A, Brooks DJ, Landau AM. Acute transcutaneous auricular vagus nerve stimulation modulates presynaptic SV2A density in healthy rat brain: An in vivo microPET study. Psychophysiology 2024:e14709. [PMID: 39428713 DOI: 10.1111/psyp.14709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 10/01/2024] [Accepted: 10/04/2024] [Indexed: 10/22/2024]
Abstract
Vagus nerve stimulation (VNS) is the subject of exploration as an adjunct treatment for neurological disorders such as epilepsy, chronic migraine, pain, and depression. A non-invasive form of VNS is transcutaneous auricular VNS (taVNS). Combining animal models and positron emission tomography (PET) may lead to a better understanding of the elusive mechanisms of taVNS. We evaluated the acute effect of electrical stimulation of the left vagus nerve via the ear on brain synaptic vesicle glycoprotein 2A (SV2A) as a measure of presynaptic density and glucose metabolism in naïve rats. Female Sprague-Dawley rats were imaged with [11C]UCB-J (n = 11) or [18F]fluorodeoxyglucose ([18F]FDG) PET (n = 13) on two separate days, (1) at baseline, and (2) after acute unilateral left taVNS or sham stimulation (30 min). We calculated the regional volume of distribution (VT) for [11C]UCB-J and standard uptake values (SUV) for [18F]FDG. We observed regional reductions of [11C]UCB-J binding in response to taVNS ranging from 36% to 59%. The changes in taVNS compared to baseline were significantly larger than those induced by sham stimulation. The differences were observed bilaterally in the frontal cortex, striatum, and midbrain. The [18F]FDG PET uptake remained unchanged following acute taVNS or sham stimulation compared to baseline values. This proof-of-concept study shows for the first time that acute taVNS for 30 min can modulate in vivo synaptic SV2A density in cortical and subcortical regions of healthy rats. Preclinical disease models and PET ligands of different targets can be a powerful combination to assess the therapeutic potential of taVNS.
Collapse
Affiliation(s)
- Karina H Binda
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Caroline C Real
- Department of Nuclear Medicine and PET, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Mette T Simonsen
- Department of Nuclear Medicine and PET, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Ebbe K Grove
- Department of Nuclear Medicine and PET, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Dirk Bender
- Department of Nuclear Medicine and PET, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Albert Gjedde
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Neuroscience, University of Copenhagen, Copenhagen, Denmark
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada
| | - David J Brooks
- Department of Nuclear Medicine and PET, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Institute of Translational and Clinical Research, University of Newcastle upon Tyne, Newcastle Upon Tyne, UK
| | - Anne M Landau
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
2
|
Roodveldt C, Bernardino L, Oztop-Cakmak O, Dragic M, Fladmark KE, Ertan S, Aktas B, Pita C, Ciglar L, Garraux G, Williams-Gray C, Pacheco R, Romero-Ramos M. The immune system in Parkinson's disease: what we know so far. Brain 2024; 147:3306-3324. [PMID: 38833182 PMCID: PMC11449148 DOI: 10.1093/brain/awae177] [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: 02/20/2024] [Revised: 05/02/2024] [Accepted: 05/13/2024] [Indexed: 06/06/2024] Open
Abstract
Parkinson's disease is characterized neuropathologically by the degeneration of dopaminergic neurons in the ventral midbrain, the accumulation of α-synuclein (α-syn) aggregates in neurons and chronic neuroinflammation. In the past two decades, in vitro, ex vivo and in vivo studies have consistently shown the involvement of inflammatory responses mediated by microglia and astrocytes, which may be elicited by pathological α-syn or signals from affected neurons and other cell types, and are directly linked to neurodegeneration and disease development. Apart from the prominent immune alterations seen in the CNS, including the infiltration of T cells into the brain, more recent studies have demonstrated important changes in the peripheral immune profile within both the innate and adaptive compartments, particularly involving monocytes, CD4+ and CD8+ T cells. This review aims to integrate the consolidated understanding of immune-related processes underlying the pathogenesis of Parkinson's disease, focusing on both central and peripheral immune cells, neuron-glia crosstalk as well as the central-peripheral immune interaction during the development of Parkinson's disease. Our analysis seeks to provide a comprehensive view of the emerging knowledge of the mechanisms of immunity in Parkinson's disease and the implications of this for better understanding the overall pathogenesis of this disease.
Collapse
Affiliation(s)
- Cintia Roodveldt
- Centre for Molecular Biology and Regenerative Medicine-CABIMER, University of Seville-CSIC, Seville 41092, Spain
- Department of Medical Biochemistry, Molecular Biology and Immunology, Faculty of Medicine, University of Seville, Seville 41009, Spain
| | - Liliana Bernardino
- Health Sciences Research Center (CICS-UBI), Faculty of Health Sciences, University of Beira Interior, 6200-506, Covilhã, Portugal
| | - Ozgur Oztop-Cakmak
- Department of Neurology, Faculty of Medicine, Koç University, Istanbul 34010, Turkey
| | - Milorad Dragic
- Laboratory for Neurobiology, Department of General Physiology and Biophysics, Faculty of Biology, University of Belgrade, 11000 Belgrade, Serbia
- Department of Molecular Biology and Endocrinology, ‘VINČA’ Institute of Nuclear Sciences-National Institute of the Republic of Serbia, University of Belgrade, 11000 Belgrade, Serbia
| | - Kari E Fladmark
- Department of Biological Science, University of Bergen, 5006 Bergen, Norway
| | - Sibel Ertan
- Department of Neurology, Faculty of Medicine, Koç University, Istanbul 34010, Turkey
| | - Busra Aktas
- Department of Molecular Biology and Genetics, Burdur Mehmet Akif Ersoy University, Burdur 15200, Turkey
| | - Carlos Pita
- iNOVA4Health, NOVA Medical School, Faculdade de Ciências Médicas, NMS, FCM, Universidade Nova de Lisboa, 1169-056 Lisboa, Portugal
| | - Lucia Ciglar
- Center Health & Bioresources, Competence Unit Molecular Diagnostics, AIT Austrian Institute of Technology GmbH, 1210 Vienna, Austria
| | - Gaetan Garraux
- Movere Group, Faculty of Medicine, GIGA Institute, University of Liège, Liège 4000, Belgium
| | | | - Rodrigo Pacheco
- Laboratorio de Neuroinmunología, Centro Científico y Tecnológico de Excelencia Ciencia & Vida, Fundación Ciencia & Vida, Huechuraba 8580702, Santiago, Chile
- Facultad de Medicina y Ciencia, Universidad San Sebastián, Providencia 7510156, Santiago, Chile
| | - Marina Romero-Ramos
- Department of Biomedicine & The Danish Research Institute of Translational Neuroscience—DANDRITE, Aarhus University, DK-8000 Aarhus C, Denmark
| |
Collapse
|
3
|
He S, Ru Q, Chen L, Xu G, Wu Y. Advances in animal models of Parkinson's disease. Brain Res Bull 2024; 215:111024. [PMID: 38969066 DOI: 10.1016/j.brainresbull.2024.111024] [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: 05/12/2024] [Revised: 06/26/2024] [Accepted: 06/28/2024] [Indexed: 07/07/2024]
Abstract
Parkinson's disease is a complex neurodegenerative disease characterized by progressive movement impairments. Predominant symptoms encompass resting tremor, bradykinesia, limb rigidity, and postural instability. In addition, it also includes a series of non-motor symptoms such as sleep disorders, hyposmia, gastrointestinal dysfunction, autonomic dysfunction and cognitive impairment. Pathologically, the disease manifests through dopaminergic neuronal loss and the presence of Lewy bodies. At present, no significant breakthrough has been achieved in clinical Parkinson's disease treatment. Exploring treatment modalities necessitate the establishment of scientifically sound animal models. In recent years, researchers have focused on replicating the symptoms of human Parkinson's disease, resulting in the establishment of various experimental animal models primarily through drugs and transgenic methods to mimic relevant pathologies and identify more effective treatments. This review examines traditional neurotoxin and transgenic animal models as well as α-synuclein pre-formed fibrils models, non-human primate models and non-mammalian specie models. Additionally, it introduces emerging models, including models based on optogenetics, induced pluripotent stem cells, and gene editing, aiming to provide a reference for the utilization of experimental animal models and clinical research for researchers in this field.
Collapse
Affiliation(s)
- Sui He
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan 430056, China
| | - Qin Ru
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan 430056, China
| | - Lin Chen
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan 430056, China
| | - Guodong Xu
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan 430056, China
| | - Yuxiang Wu
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan 430056, China.
| |
Collapse
|
4
|
Nielsen J, Lauritsen J, Pedersen JN, Nowak JS, Bendtsen MK, Kleijwegt G, Lusser K, Pitarch LC, Moreno JV, Schneider MM, Krainer G, Goksøyr L, Khalifé P, Kaalund SS, Aznar S, Kjærgaard M, Sereikaité V, Strømgaard K, Knowles TPJ, Nielsen MA, Sander AF, Romero-Ramos M, Otzen DE. Molecular properties and diagnostic potential of monoclonal antibodies targeting cytotoxic α-synuclein oligomers. NPJ Parkinsons Dis 2024; 10:139. [PMID: 39075088 PMCID: PMC11286781 DOI: 10.1038/s41531-024-00747-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 07/04/2024] [Indexed: 07/31/2024] Open
Abstract
α-Synuclein (α-syn) accumulates as insoluble amyloid but also forms soluble α-syn oligomers (αSOs), thought to be even more cytotoxic than fibrils. To detect and block the unwanted activities of these αSOs, we have raised 30 monoclonal antibodies (mAbs) against different forms of αSOs, ranging from unmodified αSOs to species stabilized by lipid peroxidation products and polyphenols, αSOs formed by C-terminally truncated α-syn, and multivalent display of α-syn on capsid virus-like particles (cVLPs). While the mAbs generally show a preference for αSOs, they also bind fibrils, but to variable extents. Overall, we observe great diversity in the mAbs' relative affinities for monomers and αSOs, varied requirements for the C-terminal extension of α-syn, and only a modest effect on α-syn fibrillation. Several mAbs show several orders of magnitude preference for αSOs over monomers in in-solution studies, while the commercial antibody MJF14 only bound 10-fold more strongly to αSOs than monomeric α-syn. Gratifyingly, seven mAbs almost completely block αSO permeabilization of membrane vesicles. Five selected mAbs identified α-syn-related pathologies like Lewy bodies (LBs) and Lewy Neurites, as well as Glial Cytoplasmic Inclusions in postmortem brains from people diagnosed for PD, dementia with LBs or multiple system atrophy, although to different extents. Three mAbs were particularly useful for pathological evaluation of postmortem brain human tissue, including early stages of PD. Although there was no straightforward connection between the mAbs' biophysical and immunohistochemical properties, it is encouraging that this comprehensive collection of mAbs able to recognize different aggregated α-syn species in vitro also holds diagnostic potential.
Collapse
Affiliation(s)
- Janni Nielsen
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Aarhus C, Denmark
| | - Johanne Lauritsen
- DANDRITE & Department of Biomedicine, Aarhus University, Aarhus C, Denmark
| | - Jannik N Pedersen
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Aarhus C, Denmark
| | - Jan S Nowak
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Aarhus C, Denmark
| | - Malthe K Bendtsen
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Aarhus C, Denmark
| | - Giulia Kleijwegt
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Aarhus C, Denmark
| | - Kaija Lusser
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Aarhus C, Denmark
| | - Laia C Pitarch
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Aarhus C, Denmark
| | - Julián V Moreno
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Aarhus C, Denmark
| | | | - Georg Krainer
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, UK
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Louise Goksøyr
- Centre for Medical Parasitology at the Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Paul Khalifé
- Centre for Medical Parasitology at the Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Sanne Simone Kaalund
- Centre for Neuroscience and Stereology, Bispebjerg and Frederiksberg Hospital, Copenhagen, Denmark
| | - Susana Aznar
- Centre for Neuroscience and Stereology, Bispebjerg and Frederiksberg Hospital, Copenhagen, Denmark
| | - Magnus Kjærgaard
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus C, Denmark
| | - Vita Sereikaité
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Kristian Strømgaard
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Tuomas P J Knowles
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, UK
| | - Morten Agertoug Nielsen
- Centre for Medical Parasitology at the Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Adam F Sander
- Centre for Medical Parasitology at the Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | | | - Daniel E Otzen
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Aarhus C, Denmark.
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus C, Denmark.
| |
Collapse
|
5
|
An D, Xu Y. Environmental risk factors provoke new thinking for prevention and treatment of dementia with Lewy bodies. Heliyon 2024; 10:e30175. [PMID: 38707435 PMCID: PMC11068646 DOI: 10.1016/j.heliyon.2024.e30175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 04/09/2024] [Accepted: 04/22/2024] [Indexed: 05/07/2024] Open
Abstract
In recent years, environmental factors have received attention in the pathogenesis of neurodegenerative diseases. Other than genetic factors, the identification of environmental factors and modifiable risk factors may create opportunities to delay the onset or slow the progression of Lewy body disease. Researchers have made significant progress in understanding environmental and modifiable risk factors over the past 30 years. To date, despite the increasing number of articles assessing risk factors for Lewy body disease, few reviews have focused on their role in its onset. In this review, we reviewed the literature investigating the relationship between Lewy body disease and several environmental and other modifiable factors. We found that some air pollutants, exposure to some metals, and infection with some microorganisms may increase the risk of Lewy body disease. Coffee intake and the Mediterranean diet are protective factors. However, it is puzzling that low educational levels and smoking may have some protective effects. In addition, we proposed specific protocols for subsequent research directions on risk factors for neurodegenerative diseases and improved methods. By conducting additional case-control studies, we could explore the role of these factors in the etiopathogenesis of Lewy body disease, establishing a foundation for strategies aimed at preventing and reducing the onset and burden of the disease.
Collapse
Affiliation(s)
- Dinghao An
- Department of Neurology, Nanjing Drum Tower Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, China
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Yun Xu
- Department of Neurology, Nanjing Drum Tower Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, China
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- State Key Laboratory of Pharmaceutical Biotechnology and Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, China
- Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, China
- Nanjing Neurology Clinical Medical Center, Nanjing, China
| |
Collapse
|
6
|
Vaidya B, Gupta P, Biswas S, Laha JK, Roy I, Sharma SS. Effect of Clemizole on Alpha-Synuclein-Preformed Fibrils-Induced Parkinson's Disease Pathology: A Pharmacological Investigation. Neuromolecular Med 2024; 26:19. [PMID: 38703217 DOI: 10.1007/s12017-024-08785-2] [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: 01/29/2024] [Accepted: 04/02/2024] [Indexed: 05/06/2024]
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder associated with mitochondrial dysfunctions and oxidative stress. However, to date, therapeutics targeting these pathological events have not managed to translate from bench to bedside for clinical use. One of the major reasons for the lack of translational success has been the use of classical model systems that do not replicate the disease pathology and progression with the same degree of robustness. Therefore, we employed a more physiologically relevant model involving alpha-synuclein-preformed fibrils (PFF) exposure to SH-SY5Y cells and Sprague Dawley rats. We further explored the possible involvement of transient receptor potential canonical 5 (TRPC5) channels in PD-like pathology induced by these alpha-synuclein-preformed fibrils with emphasis on amelioration of oxidative stress and mitochondrial health. We observed that alpha-synuclein PFF exposure produced neurobehavioural deficits that were positively ameliorated after treatment with the TRPC5 inhibitor clemizole. Furthermore, Clemizole also reduced p-alpha-synuclein and diminished oxidative stress levels which resulted in overall improvements in mitochondrial biogenesis and functions. Finally, the results of the pharmacological modulation were further validated using siRNA-mediated knockdown of TRPC5 channels, which also decreased p-alpha-synuclein expression. Together, the results of this study could be superimposed in the future for exploring the beneficial effects of TRPC5 channel modulation for other neurodegenerative disorders and synucleopathies.
Collapse
Affiliation(s)
- Bhupesh Vaidya
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education, S.A.S. Nagar, Mohali, Punjab, 160062, India
| | - Pankaj Gupta
- Department of Pharmaceutical Technology (Process Chemistry), National Institute of Pharmaceutical Education and Research, S. A. S. Nagar, Mohali, Punjab, 160062, India
| | - Soumojit Biswas
- Department of Biotechnology, National Institute of Pharmaceutical Education, S.A.S. Nagar, Mohali, Punjab, 160062, India
| | - Joydev K Laha
- Department of Pharmaceutical Technology (Process Chemistry), National Institute of Pharmaceutical Education and Research, S. A. S. Nagar, Mohali, Punjab, 160062, India
| | - Ipsita Roy
- Department of Biotechnology, National Institute of Pharmaceutical Education, S.A.S. Nagar, Mohali, Punjab, 160062, India
| | - Shyam Sunder Sharma
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education, S.A.S. Nagar, Mohali, Punjab, 160062, India.
| |
Collapse
|
7
|
Centner A, Del Priore I, Chambers N, Cohen SR, Terry ML, Coyle M, Glinski J, Stoll AC, Patterson JR, Kemp CJ, Miller KM, Kubik M, Kuhn N, Luk KC, Sortwell CE, Bishop C. Deficits in basal and evoked striatal dopamine release following alpha-synuclein preformed fibril injection: An in vivo microdialysis study. Eur J Neurosci 2024; 59:1585-1603. [PMID: 38356120 DOI: 10.1111/ejn.16275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 01/22/2024] [Accepted: 01/26/2024] [Indexed: 02/16/2024]
Abstract
Parkinson's disease (PD) is characterized by the accumulation of misfolded alpha-synuclein (α-syn) protein, forming intraneuronal Lewy body (LB) inclusions. The α-syn preformed fibril (PFF) model of PD recapitulates α-syn aggregation, progressive nigrostriatal degeneration and motor dysfunction; however, little is known about the time course of PFF-induced alterations in basal and evoked dopamine (DA). In vivo microdialysis is well suited for identifying small changes in neurotransmitter levels over extended periods. In the present study, adult male Fischer 344 rats received unilateral, intrastriatal injections of either α-syn PFFs or phosphate-buffered saline (PBS). At 4 or 8 months post-injection (p.i.), animals underwent in vivo microdialysis to evaluate basal extracellular striatal DA and metabolite levels, local KCl-evoked striatal DA release and the effects of systemic levodopa (l-DOPA). Post-mortem analysis demonstrated equivalent PFF-induced reductions in tyrosine hydroxylase (TH) immunoreactive nigral neurons (~50%) and striatal TH (~20%) at both time points. Compared with reduction in striatal TH, reduction in striatal dopamine transporter (DAT) was more pronounced and progressed between the 4- and 8-month p.i. intervals (36% ➔ 46%). Significant PFF-induced deficits in basal and evoked striatal DA, as well as deficits in motor performance, were not observed until 8 months p.i. Responses to l-DOPA did not differ regardless of PBS or PFF treatment. These results suggest that basal and evoked striatal DA are maintained for several months following PFF injection, with loss of both associated with motor dysfunction. Our studies provide insight into the time course and magnitude of PFF-induced extracellular dopaminergic deficits in the striatum.
Collapse
Affiliation(s)
- Ashley Centner
- Department of Psychology, Binghamton University, Binghamton, New York, USA
| | | | - Nicole Chambers
- Department of Psychology, Binghamton University, Binghamton, New York, USA
| | - Sophie R Cohen
- Department of Psychology, Binghamton University, Binghamton, New York, USA
| | - Michelle L Terry
- Department of Psychology, Binghamton University, Binghamton, New York, USA
| | - Michael Coyle
- Department of Psychology, Binghamton University, Binghamton, New York, USA
| | - John Glinski
- Department of Psychology, Binghamton University, Binghamton, New York, USA
| | - Anna C Stoll
- Department of Translational Neuroscience, Michigan State University, Grand Rapids, Michigan, USA
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan, USA
| | - Joseph R Patterson
- Department of Translational Neuroscience, Michigan State University, Grand Rapids, Michigan, USA
| | - Christopher J Kemp
- Department of Translational Neuroscience, Michigan State University, Grand Rapids, Michigan, USA
| | - Kathryn M Miller
- Department of Translational Neuroscience, Michigan State University, Grand Rapids, Michigan, USA
| | - Michael Kubik
- Department of Translational Neuroscience, Michigan State University, Grand Rapids, Michigan, USA
| | - Nathan Kuhn
- Department of Translational Neuroscience, Michigan State University, Grand Rapids, Michigan, USA
| | - Kelvin C Luk
- Department of Pathology and Laboratory Medicine, Center for Neurodegenerative Disease Research, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Caryl E Sortwell
- Department of Translational Neuroscience, Michigan State University, Grand Rapids, Michigan, USA
| | - Christopher Bishop
- Department of Psychology, Binghamton University, Binghamton, New York, USA
| |
Collapse
|
8
|
Hansen ML, Ambjørn M, Harndahl MN, Benned-Jensen T, Fog K, Bjerregaard-Andersen K, Sotty F. Characterization of pSer129-αSyn Pathology and Neurofilament Light-Chain Release across In Vivo, Ex Vivo, and In Vitro Models of Pre-Formed-Fibril-Induced αSyn Aggregation. Cells 2024; 13:253. [PMID: 38334646 PMCID: PMC10854598 DOI: 10.3390/cells13030253] [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: 12/04/2023] [Revised: 01/18/2024] [Accepted: 01/26/2024] [Indexed: 02/10/2024] Open
Abstract
Protein aggregation is a predominant feature of many neurodegenerative diseases, including synucleinopathies, which are characterized by cellular inclusions containing α-Synuclein (αSyn) phosphorylated at serine 129 (pSer129). In the present study, we characterized the development of αSyn pre-formed fibril (PFF)-induced pSer129-αSyn pathology in F28tg mice overexpressing human wild-type αSyn, as well as in ex vivo organotypic cultures and in vitro primary cultures from the same mouse model. Concurrently, we collected cerebrospinal fluid (CSF) from mice and conditioned media from ex vivo and in vitro cultures and quantified the levels of neurofilament light chain (NFL), a biomarker of neurodegeneration. We found that the intra-striatal injection of PFFs induces the progressive spread of pSer129-αSyn pathology and microglial activation in vivo, as well as modest increases in NFL levels in the CSF. Similarly, PFF-induced αSyn pathology occurs progressively in ex vivo organotypic slice cultures and is accompanied by significant increases in NFL release into the media. Using in vitro primary hippocampal cultures, we further confirmed that pSer129-αSyn pathology and NFL release occur in a manner that correlates with the fibril dose and the level of the αSyn protein. Overall, we demonstrate that αSyn pathology is associated with NFL release across preclinical models of seeded αSyn aggregation and that the pharmacological inhibition of αSyn aggregation in vitro also significantly reduces NFL release.
Collapse
Affiliation(s)
- Maja L. Hansen
- Neuroscience, Molecular and Cellular Pharmacology, H. Lundbeck A/S, Valby, 2500 Copenhagen, Denmark; (M.L.H.); (M.A.); (K.F.)
| | - Malene Ambjørn
- Neuroscience, Molecular and Cellular Pharmacology, H. Lundbeck A/S, Valby, 2500 Copenhagen, Denmark; (M.L.H.); (M.A.); (K.F.)
| | - Mikkel N. Harndahl
- Biotherapeutic Discovery, H. Lundbeck A/S, Valby, 2500 Copenhagen, Denmark; (M.N.H.); (K.B.-A.)
| | - Tau Benned-Jensen
- Neuroscience, Molecular and Cellular Pharmacology, H. Lundbeck A/S, Valby, 2500 Copenhagen, Denmark; (M.L.H.); (M.A.); (K.F.)
| | - Karina Fog
- Neuroscience, Molecular and Cellular Pharmacology, H. Lundbeck A/S, Valby, 2500 Copenhagen, Denmark; (M.L.H.); (M.A.); (K.F.)
| | | | - Florence Sotty
- Neuroscience, Histology and Pathology Models, H. Lundbeck A/S, Valby, 2500 Copenhagen, Denmark
| |
Collapse
|
9
|
Patterson JR, Kochmanski J, Stoll AC, Kubik M, Kemp CJ, Duffy MF, Thompson K, Howe JW, Cole-Strauss A, Kuhn NC, Miller KM, Nelson S, Onyekpe CU, Beck JS, Counts SE, Bernstein AI, Steece-Collier K, Luk KC, Sortwell CE. Transcriptomic profiling of early synucleinopathy in rats induced with preformed fibrils. NPJ Parkinsons Dis 2024; 10:7. [PMID: 38172128 PMCID: PMC10764951 DOI: 10.1038/s41531-023-00620-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 12/08/2023] [Indexed: 01/05/2024] Open
Abstract
Examination of early phases of synucleinopathy when inclusions are present, but long before neurodegeneration occurs, is critical to both understanding disease progression and the development of disease modifying therapies. The rat alpha-synuclein (α-syn) preformed fibril (PFF) model induces synchronized synucleinopathy that recapitulates the pathological features of Parkinson's disease (PD) and can be used to study synucleinopathy progression. In this model, phosphorylated α-syn (pSyn) inclusion-containing neurons and reactive microglia (major histocompatibility complex-II immunoreactive) peak in the substantia nigra pars compacta (SNpc) months before appreciable neurodegeneration. However, it remains unclear which specific genes are driving these phenotypic changes. To identify transcriptional changes associated with early synucleinopathy, we used laser capture microdissection of the SNpc paired with RNA sequencing (RNASeq). Precision collection of the SNpc allowed for the assessment of differential transcript expression in the nigral dopamine neurons and proximal glia. Transcripts upregulated in early synucleinopathy were mainly associated with an immune response, whereas transcripts downregulated were associated with neurotransmission and the dopamine pathway. A subset of 29 transcripts associated with neurotransmission/vesicular release and the dopamine pathway were verified in a separate cohort of males and females to confirm reproducibility. Within this subset, fluorescent in situ hybridization (FISH) was used to localize decreases in the Syt1 and Slc6a3 transcripts to pSyn inclusion-containing neurons. Identification of transcriptional changes in early synucleinopathy provides insight into the molecular mechanisms driving neurodegeneration.
Collapse
Affiliation(s)
- Joseph R Patterson
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI, USA.
- Neuroscience Program, Michigan State University, East Lansing, MI, USA.
| | - Joseph Kochmanski
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI, USA
| | - Anna C Stoll
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI, USA
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, USA
| | - Michael Kubik
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI, USA
- Neuroscience Program, Michigan State University, East Lansing, MI, USA
| | - Christopher J Kemp
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI, USA
| | - Megan F Duffy
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI, USA
- Neuroscience Program, Michigan State University, East Lansing, MI, USA
| | - Kajene Thompson
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI, USA
| | - Jacob W Howe
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI, USA
- Neuroscience Program, Michigan State University, East Lansing, MI, USA
| | - Allyson Cole-Strauss
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI, USA
| | - Nathan C Kuhn
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI, USA
| | - Kathryn M Miller
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI, USA
- Neuroscience Program, Michigan State University, East Lansing, MI, USA
| | - Seth Nelson
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI, USA
| | - Christopher U Onyekpe
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI, USA
| | - John S Beck
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI, USA
| | - Scott E Counts
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI, USA
- Neuroscience Program, Michigan State University, East Lansing, MI, USA
| | - Alison I Bernstein
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI, USA
- Department of Pharmacology and Toxicology, Rutgers University, Piscataway, NJ, USA
- Environmental and Occupational Health Science Institute, Rutgers University, Piscataway, NJ, USA
| | - Kathy Steece-Collier
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI, USA
| | - Kelvin C Luk
- Center for Neurodegenerative Disease Research, Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Caryl E Sortwell
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI, USA
- Neuroscience Program, Michigan State University, East Lansing, MI, USA
| |
Collapse
|
10
|
Li Z, Dang Q, Wang P, Zhao F, Huang J, Wang C, Liu X, Min W. Food-Derived Peptides: Beneficial CNS Effects and Cross-BBB Transmission Strategies. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:20453-20478. [PMID: 38085598 DOI: 10.1021/acs.jafc.3c06518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2023]
Abstract
Food-derived peptides, as dietary supplements, have significant effects on promoting brain health and relieving central nervous system (CNS) diseases. However, the blood-brain barrier (BBB) greatly limits their in-brain bioavailability. Thus, overcoming the BBB to target the CNS is a major challenge for bioactive peptides in the prevention and treatment of CNS diseases. This review discusses improvement in the neuroprotective function of food-derived active peptides in CNS diseases, as well as the source of BBB penetrating peptides (BBB-shuttles) and the mechanism of transmembrane transport. Notably, this review also discusses various peptide modification methods to overcome the low permeability and stability of the BBB. Lipification, glycosylation, introduction of disulfide bonds, and cyclization are effective strategies for improving the penetration efficiency of peptides through the BBB. This review provides a new prospective for improving their neuroprotective function and developing treatments to delay or even prevent CNS diseases.
Collapse
Affiliation(s)
- Zehui Li
- College of Food and Health, Zhejiang A&F University, Hangzhou, Zhejiang 311300, P.R. China
- College of Food Science and Engineering, Jilin Agricultural University, ChangChun, Jilin 130118, P.R. China
| | - Qiao Dang
- College of Food Science and Engineering, Jilin Agricultural University, ChangChun, Jilin 130118, P.R. China
| | - Peng Wang
- College of Food and Health, Zhejiang A&F University, Hangzhou, Zhejiang 311300, P.R. China
| | - Fanrui Zhao
- College of Food and Health, Zhejiang A&F University, Hangzhou, Zhejiang 311300, P.R. China
- State Key Laboratory of Subtropical Silviculture, Zhejiang A&F University, Hangzhou 311300, P.R. China
| | - Jianqin Huang
- State Key Laboratory of Subtropical Silviculture, Zhejiang A&F University, Hangzhou 311300, P.R. China
| | - Chongchong Wang
- College of Food and Health, Zhejiang A&F University, Hangzhou, Zhejiang 311300, P.R. China
- State Key Laboratory of Subtropical Silviculture, Zhejiang A&F University, Hangzhou 311300, P.R. China
| | - Xingquan Liu
- College of Food and Health, Zhejiang A&F University, Hangzhou, Zhejiang 311300, P.R. China
| | - Weihong Min
- College of Food and Health, Zhejiang A&F University, Hangzhou, Zhejiang 311300, P.R. China
- State Key Laboratory of Subtropical Silviculture, Zhejiang A&F University, Hangzhou 311300, P.R. China
| |
Collapse
|
11
|
Ferreira SA, Li C, Klæstrup IH, Vitic Z, Rasmussen RK, Kirkegaard A, Toft GU, Betzer C, Svendsen P, Jensen PH, Luo Y, Etzerodt A, Moestrup SK, Romero-Ramos M. Sex-dimorphic neuroprotective effect of CD163 in an α-synuclein mouse model of Parkinson's disease. NPJ Parkinsons Dis 2023; 9:164. [PMID: 38092806 PMCID: PMC10719342 DOI: 10.1038/s41531-023-00606-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 11/24/2023] [Indexed: 12/17/2023] Open
Abstract
Alpha-synuclein (α-syn) aggregation and immune activation represent hallmark pathological events in Parkinson's disease (PD). The PD-associated immune response encompasses both brain and peripheral immune cells, although little is known about the immune proteins relevant for such a response. We propose that the upregulation of CD163 observed in blood monocytes and in the responsive microglia in PD patients is a protective mechanism in the disease. To investigate this, we used the PD model based on intrastriatal injections of murine α-syn pre-formed fibrils in CD163 knockout (KO) mice and wild-type littermates. CD163KO females revealed an impaired and differential early immune response to α-syn pathology as revealed by immunohistochemical and transcriptomic analysis. After 6 months, CD163KO females showed an exacerbated immune response and α-syn pathology, which ultimately led to dopaminergic neurodegeneration of greater magnitude. These findings support a sex-dimorphic neuroprotective role for CD163 during α-syn-induced neurodegeneration.
Collapse
Affiliation(s)
- Sara A Ferreira
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Danish Research Institute of Translational Neuroscience - DANDRITE, Aarhus University, Aarhus, Denmark
| | - Conghui Li
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Ida H Klæstrup
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Danish Research Institute of Translational Neuroscience - DANDRITE, Aarhus University, Aarhus, Denmark
| | - Zagorka Vitic
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Danish Research Institute of Translational Neuroscience - DANDRITE, Aarhus University, Aarhus, Denmark
| | | | - Asger Kirkegaard
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Danish Research Institute of Translational Neuroscience - DANDRITE, Aarhus University, Aarhus, Denmark
| | - Gitte U Toft
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Danish Research Institute of Translational Neuroscience - DANDRITE, Aarhus University, Aarhus, Denmark
| | - Cristine Betzer
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Danish Research Institute of Translational Neuroscience - DANDRITE, Aarhus University, Aarhus, Denmark
| | - Pia Svendsen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Poul H Jensen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Danish Research Institute of Translational Neuroscience - DANDRITE, Aarhus University, Aarhus, Denmark
| | - Yonglun Luo
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Danish Research Institute of Translational Neuroscience - DANDRITE, Aarhus University, Aarhus, Denmark
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark
| | - Anders Etzerodt
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Søren K Moestrup
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Marina Romero-Ramos
- Department of Biomedicine, Aarhus University, Aarhus, Denmark.
- Danish Research Institute of Translational Neuroscience - DANDRITE, Aarhus University, Aarhus, Denmark.
| |
Collapse
|
12
|
Wang X, Hu W, Qu L, Wang J, Wu A, Lo HH, Ng JPL, Tang Y, Yun X, Wu J, Wong VKW, Chung SK, Wang L, Luo W, Ji X, Law BYK. Tricin promoted ATG-7 dependent autophagic degradation of α-synuclein and dopamine release for improving cognitive and motor deficits in Parkinson's disease. Pharmacol Res 2023; 196:106874. [PMID: 37586619 DOI: 10.1016/j.phrs.2023.106874] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 07/27/2023] [Accepted: 07/28/2023] [Indexed: 08/18/2023]
Abstract
Tricin, a natural nontoxic flavonoid distributed in grasses and euphorbia plants, has been reported to scavenge free radicals, possess anti-inflammatory and antioxidative effects. However, its autophagic effect on Parkinson's disease (PD) has not been elucidated. By adopting cellular and C. elegans models of PD, the autophagic effect of tricin was identified based on the level of autophagy markers (LC3-II and p62). Besides, the pharmacological effects on neurotransmitters (dopamine), inflammatory cytokines (IFN γ, TNFα, MCP-1, IL-10, IL-6 and IL-17A), histology (hematoxylin & eosin and Nissl staining) and behavioural pathology (open-field test, hindlimb clasping, Y-maze, Morris water-maze and nest building test) were also confirmed in the A53T-α-synuclein transgenic PD mouse model. Further experiments demonstrated that tricin induced autophagic flux and lowered the level of α-synuclein through AMPK-p70s6K- and ATG7-dependent mechanism. Compared to the existing clinical PD drugs, tricin mitigated pathogenesis and symptoms of PD with no observable side effects. In summary, tricin is proposed as a potential adjuvant remedy or nutraceutical for the prevention and treatment of PD.
Collapse
Affiliation(s)
- Xingxia Wang
- Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau Special Administrative Region of China; Department of Neurology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Wei Hu
- Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau Special Administrative Region of China
| | - Liqun Qu
- Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau Special Administrative Region of China; Marine Traditional Chinese Medicine Research Center, Key Laboratory of Marine Traditional Chinese Medicine in Shandong Universities, Shandong Engineering and Technology Research Center on Omics of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Jian Wang
- Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau Special Administrative Region of China
| | - Anguo Wu
- Sichuan Key Medical Laboratory of New Drug Discovery and Drug ability Evaluation, Luzhou Key Laboratory of Activity Screening and Draggability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Hang Hong Lo
- Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau Special Administrative Region of China
| | - Jerome P L Ng
- Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau Special Administrative Region of China
| | - Yong Tang
- Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau Special Administrative Region of China; Sichuan Key Medical Laboratory of New Drug Discovery and Drug ability Evaluation, Luzhou Key Laboratory of Activity Screening and Draggability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Xiaoyun Yun
- Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau Special Administrative Region of China
| | - Jianhui Wu
- Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau Special Administrative Region of China
| | - Vincent Kam Wai Wong
- Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau Special Administrative Region of China
| | - Sookja Kim Chung
- Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau Special Administrative Region of China; Faculty of Medicine, Macau University of Science and Technology, Macau Special Administrative Region of China
| | - Linna Wang
- Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau Special Administrative Region of China
| | - Weidan Luo
- Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau Special Administrative Region of China
| | - Xiang Ji
- Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau Special Administrative Region of China
| | - Betty Yuen Kwan Law
- Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau Special Administrative Region of China.
| |
Collapse
|
13
|
Zhang S, Dauer K, Strohäker T, Tatenhorst L, Caldi Gomes L, Mayer S, Jung BC, Kim WS, Lee S, Becker S, Liesche‐Starnecker F, Zweckstetter M, Lingor P. Alpha-synuclein fibrils amplified from multiple system atrophy and Parkinson's disease patient brain spread after intracerebral injection into mouse brain. Brain Pathol 2023; 33:e13196. [PMID: 37485772 PMCID: PMC10467043 DOI: 10.1111/bpa.13196] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 07/07/2023] [Indexed: 07/25/2023] Open
Abstract
Parkinson's disease (PD), multiple system atrophy (MSA), and dementia with Lewy bodies (DLB) are neurodegenerative disorders with alpha-synuclein (α-syn) aggregation pathology. Different strains of α-syn with unique properties are suggested to cause distinct clinical and pathological manifestations resulting in PD, MSA, or DLB. To study individual α-syn spreading patterns, we injected α-syn fibrils amplified from brain homogenates of two MSA patients and two PD patients into the brains of C57BI6/J mice. Antibody staining against pS129-α-syn showed that α-syn fibrils amplified from the brain homogenates of the four different patients caused different levels of α-syn spreading. The strongest α-syn pathology was triggered by α-syn fibrils of one of the two MSA patients, followed by comparable pS129-α-syn induction by the second MSA and one PD patient material. Histological analysis using an antibody against Iba1 further showed that the formation of pS129-α-syn is associated with increased microglia activation. In contrast, no differences in dopaminergic neuron numbers or co-localization of α-syn in oligodendrocytes were observed between the different groups. Our data support the spreading of α-syn pathology in MSA, while at the same time pointing to spreading heterogeneity between different patients potentially driven by individual patient immanent factors.
Collapse
Affiliation(s)
- Shuyu Zhang
- Clinical Department of Neurology, School of Medicine, University Hospital rechts der IsarTechnical University of MunichMunichGermany
| | - Karina Dauer
- Department of NeurologyUniversity Medical Center GöttingenGöttingenGermany
- Center for Biostructural Imaging of NeurodegenerationUniversity Medical Center GöttingenGöttingenGermany
| | - Timo Strohäker
- German Center for Neurodegenerative Diseases (DZNE)GöttingenGermany
| | - Lars Tatenhorst
- Department of NeurologyUniversity Medical Center GöttingenGöttingenGermany
- Center for Biostructural Imaging of NeurodegenerationUniversity Medical Center GöttingenGöttingenGermany
| | - Lucas Caldi Gomes
- Clinical Department of Neurology, School of Medicine, University Hospital rechts der IsarTechnical University of MunichMunichGermany
- Department of NeurologyUniversity Medical Center GöttingenGöttingenGermany
- Center for Biostructural Imaging of NeurodegenerationUniversity Medical Center GöttingenGöttingenGermany
| | - Simon Mayer
- Clinical Department of Neurology, School of Medicine, University Hospital rechts der IsarTechnical University of MunichMunichGermany
| | - Byung Chul Jung
- Department of Biomedical Sciences, Neuroscience Research Institute, Convergence Research Center for Dementia, College of MedicineSeoul National UniversitySeoulSouth Korea
| | - Woojin S. Kim
- Faculty of Medicine and Health, Brain and Mind Centre and School of Medical SciencesThe University of SydneySydneyNew South WalesAustralia
- School of Medical SciencesUniversity of New South Wales and Neuroscience Research AustraliaRandwickNew South WalesAustralia
| | - Seung‐Jae Lee
- Department of Biomedical Sciences, Neuroscience Research Institute, Convergence Research Center for Dementia, College of MedicineSeoul National UniversitySeoulSouth Korea
| | - Stefan Becker
- Department of NMR Based Structural BiologyMax Planck Institute for Multidisciplinary SciencesGöttingenGermany
| | - Friederike Liesche‐Starnecker
- Department of Neuropathology, Institute of Pathology, School of MedicineTechnical University MunichMunichGermany
- Department of Pathology and Molecular Diagnostics, Medical FacultyUniversity of AugsburgAugsburgGermany
| | - Markus Zweckstetter
- German Center for Neurodegenerative Diseases (DZNE)GöttingenGermany
- Department of NMR Based Structural BiologyMax Planck Institute for Multidisciplinary SciencesGöttingenGermany
| | - Paul Lingor
- Clinical Department of Neurology, School of Medicine, University Hospital rechts der IsarTechnical University of MunichMunichGermany
- Department of NeurologyUniversity Medical Center GöttingenGöttingenGermany
- Center for Biostructural Imaging of NeurodegenerationUniversity Medical Center GöttingenGöttingenGermany
| |
Collapse
|
14
|
Graves NJ, Gambin Y, Sierecki E. α-Synuclein Strains and Their Relevance to Parkinson's Disease, Multiple System Atrophy, and Dementia with Lewy Bodies. Int J Mol Sci 2023; 24:12134. [PMID: 37569510 PMCID: PMC10418915 DOI: 10.3390/ijms241512134] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 07/26/2023] [Accepted: 07/26/2023] [Indexed: 08/13/2023] Open
Abstract
Like many neurodegenerative diseases, Parkinson's disease (PD) is characterized by the formation of proteinaceous aggregates in brain cells. In PD, those proteinaceous aggregates are formed by the α-synuclein (αSyn) and are considered the trademark of this neurodegenerative disease. In addition to PD, αSyn pathological aggregation is also detected in atypical Parkinsonism, including Dementia with Lewy Bodies (DLB), Multiple System Atrophy (MSA), as well as neurodegeneration with brain iron accumulation, some cases of traumatic brain injuries, and variants of Alzheimer's disease. Collectively, these (and other) disorders are referred to as synucleinopathies, highlighting the relation between disease type and protein misfolding/aggregation. Despite these pathological relationships, however, synucleinopathies cover a wide range of pathologies, present with a multiplicity of symptoms, and arise from dysfunctions in different neuroanatomical regions and cell populations. Strikingly, αSyn deposition occurs in different types of cells, with oligodendrocytes being mainly affected in MSA, while aggregates are found in neurons in PD. If multiple factors contribute to the development of a pathology, especially in the cases of slow-developing neurodegenerative disorders, the common presence of αSyn aggregation, as both a marker and potential driver of disease, is puzzling. In this review, we will focus on comparing PD, DLB, and MSA, from symptomatology to molecular description, highlighting the role and contribution of αSyn aggregates in each disorder. We will particularly present recent evidence for the involvement of conformational strains of αSyn aggregates and discuss the reciprocal relationship between αSyn strains and the cellular milieu. Moreover, we will highlight the need for effective methodologies for the strainotyping of aggregates to ameliorate diagnosing capabilities and therapeutic treatments.
Collapse
Affiliation(s)
| | | | - Emma Sierecki
- EMBL Australia Node for Single Molecule Sciences and School of Biomedical Sciences, Faculty of Medicine, The University of New South Wales, Sydney, NSW 2052, Australia; (N.J.G.)
| |
Collapse
|
15
|
Calabresi P, Mechelli A, Natale G, Volpicelli-Daley L, Di Lazzaro G, Ghiglieri V. Alpha-synuclein in Parkinson's disease and other synucleinopathies: from overt neurodegeneration back to early synaptic dysfunction. Cell Death Dis 2023; 14:176. [PMID: 36859484 PMCID: PMC9977911 DOI: 10.1038/s41419-023-05672-9] [Citation(s) in RCA: 131] [Impact Index Per Article: 131.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 01/18/2023] [Accepted: 02/09/2023] [Indexed: 03/03/2023]
Abstract
Although the discovery of the critical role of α-synuclein (α-syn) in the pathogenesis of Parkinson's disease (PD) is now twenty-five years old, it still represents a milestone in PD research. Abnormal forms of α-syn trigger selective and progressive neuronal death through mitochondrial impairment, lysosomal dysfunction, and alteration of calcium homeostasis not only in PD but also in other α-syn-related neurodegenerative disorders such as dementia with Lewy bodies, multiple system atrophy, pure autonomic failure, and REM sleep behavior disorder. Furthermore, α-syn-dependent early synaptic and plastic alterations and the underlying mechanisms preceding overt neurodegeneration have attracted great interest. In particular, the presence of early inflammation in experimental models and PD patients, occurring before deposition and spreading of α-syn, suggests a mechanistic link between inflammation and synaptic dysfunction. The knowledge of these early mechanisms is of seminal importance to support the research on reliable biomarkers to precociously identify the disease and possible disease-modifying therapies targeting α-syn. In this review, we will discuss these critical issues, providing a state of the art of the role of this protein in early PD and other synucleinopathies.
Collapse
Affiliation(s)
- Paolo Calabresi
- Sezione di Neurologia, Dipartimento di Neuroscienze, Facoltà di Medicina e Chirurgia, Università Cattolica del Sacro Cuore, Rome, 00168, Italy. .,Neurologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, 00168, Italy.
| | - Alessandro Mechelli
- Dipartimento di Scienze Mediche e Chirurgiche, Istituto di Neurologia, Università "Magna Graecia", Catanzaro, Italy
| | - Giuseppina Natale
- Sezione di Neurologia, Dipartimento di Neuroscienze, Facoltà di Medicina e Chirurgia, Università Cattolica del Sacro Cuore, Rome, 00168, Italy
| | - Laura Volpicelli-Daley
- Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Giulia Di Lazzaro
- Neurologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, 00168, Italy
| | - Veronica Ghiglieri
- Neurologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, 00168, Italy.,Università Telematica San Raffaele, Rome, 00166, Italy
| |
Collapse
|
16
|
Abstract
The midbrain dopamine (mDA) system is composed of molecularly and functionally distinct neuron subtypes that mediate specific behaviours and are linked to various brain diseases. Considerable progress has been made in identifying mDA neuron subtypes, and recent work has begun to unveil how these neuronal subtypes develop and organize into functional brain structures. This progress is important for further understanding the disparate physiological functions of mDA neurons and their selective vulnerability in disease, and will ultimately accelerate therapy development. This Review discusses recent advances in our understanding of molecularly defined mDA neuron subtypes and their circuits, ranging from early developmental events, such as neuron migration and axon guidance, to their wiring and function, and future implications for therapeutic strategies.
Collapse
|
17
|
Ni R. PET imaging in animal models of Parkinson's disease. Behav Brain Res 2023; 438:114174. [PMID: 36283568 DOI: 10.1016/j.bbr.2022.114174] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Revised: 09/20/2022] [Accepted: 09/27/2022] [Indexed: 12/05/2022]
Abstract
Alpha-synucleinopathies, such as Parkinson's disease, dementia with Lewy bodies and multiple system atrophy, are characterized by aberrant accumulation of alpha-synuclein and synaptic dysfunction leading to motor and cognitive deficits. Animal models of alpha-synucleinopathy have greatly facilitated the mechanistic understanding of the disease and the development of therapeutics. Various transgenic, alpha-synuclein fibril-injected, and toxin-injected animal models of Parkinson's disease and multiple system atrophy that recapitulate the disease pathology have been developed and widely used. Recent advances in positron emission tomography have allowed the noninvasive visualization of molecular alterations, underpinning behavioral dysfunctions in the brains of animal models and the longitudinal monitoring of treatment effects. Imaging studies in these disease animal models have employed multi-tracer PET designs to reveal dopaminergic deficits together with other molecular alterations. This review focuses on the development of new positron emission tomography tracers and studies of alpha-synuclein, synaptic vesicle glycoprotein 2A neurotransmitter receptor deficits such as dopaminergic receptor, dopaminergic transporter, serotonergic receptor, vesicular monoamine transporter 2, hypometabolism, neuroinflammation, mitochondrial dysfunction and leucine rich repeat kinase 2 in animal models of Parkinson's disease. The outstanding challenges and emerging applications are outlined, such as investigating the gut-brain-axis by using positron emission tomography in animal models, and provide a future outlook.
Collapse
Affiliation(s)
- Ruiqing Ni
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland; Institute for Biomedical Engineering, University of Zurich & ETH Zurich, Zurich, Switzerland.
| |
Collapse
|
18
|
Zheng C, Toyonaga T, Chen B, Nicholson L, Mennie W, Liu M, Spurrier J, Deluca K, Strittmatter SM, Carson RE, Huang Y, Cai Z. Decreased synaptic vesicle glycoprotein 2A binding in a rodent model of familial Alzheimer's disease detected by [ 18F]SDM-16. Front Neurol 2023; 14:1045644. [PMID: 36846134 PMCID: PMC9945093 DOI: 10.3389/fneur.2023.1045644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 01/17/2023] [Indexed: 02/11/2023] Open
Abstract
Introduction Synapse loss is one of the hallmarks of Alzheimer's disease (AD) and is associated with cognitive decline. In this study, we tested [18F]SDM-16, a novel metabolically stable SV2A PET imaging probe, in the transgenic APPswe/PS1dE9 (APP/PS1) mouse model of AD and age-matched wild-type (WT) mice at 12 months of age. Methods Based on previous preclinical PET imaging studies using [11C]UCB-J and [18F]SynVesT-1 in the same strain animals, we used the simplified reference tissue model (SRTM), with brain stem as the pseudo reference region to calculate distribution volume ratios (DVRs). Results To simplify and streamline the quantitative analysis, we compared the standardized uptake value ratios (SUVRs) from different imaging windows to DVRs and found that the averaged SUVRs from 60-90 min post-injection (p.i.) are most consistent with the DVRs. Thus, we used averaged SUVRs from 60-90 min for group comparisons and found statistically significant differences in the tracer uptake in different brain regions, e.g., hippocampus (p = 0.001), striatum (p = 0.002), thalamus (p = 0.003), and cingulate cortex (p = 0.0003). Conclusions In conclusion, [18F]SDM-16 was used to detect decreased SV2A levels in the brain of APP/PS1 AD mouse model at one year old. Our data suggest that [18F]SDM-16 has similar statistical power in detecting the synapse loss in APP/PS1 mice as [11C]UCB-J and [18F]SynVesT-1, albeit later imaging window (60-90 min p.i.) is needed when SUVR is used as a surrogate for DVR for [18F]SDM-16 due to its slower brain kinetics.
Collapse
Affiliation(s)
- Chao Zheng
- Department of Radiology and Biomedical Imaging, PET Center, Yale School of Medicine, New Haven, CT, United States
| | - Takuya Toyonaga
- Department of Radiology and Biomedical Imaging, PET Center, Yale School of Medicine, New Haven, CT, United States
| | - Baosheng Chen
- Department of Radiology and Biomedical Imaging, PET Center, Yale School of Medicine, New Haven, CT, United States
| | - LaShae Nicholson
- Program in Cellular Neuroscience, Neurodegeneration, and Repair, Departments of Neuroscience and Neurology, Yale University School of Medicine, New Haven, CT, United States
| | - William Mennie
- Department of Radiology and Biomedical Imaging, PET Center, Yale School of Medicine, New Haven, CT, United States
| | - Michael Liu
- Department of Radiology and Biomedical Imaging, PET Center, Yale School of Medicine, New Haven, CT, United States
| | - Joshua Spurrier
- Program in Cellular Neuroscience, Neurodegeneration, and Repair, Departments of Neuroscience and Neurology, Yale University School of Medicine, New Haven, CT, United States
| | - Kristin Deluca
- Program in Cellular Neuroscience, Neurodegeneration, and Repair, Departments of Neuroscience and Neurology, Yale University School of Medicine, New Haven, CT, United States
| | - Stephen M. Strittmatter
- Program in Cellular Neuroscience, Neurodegeneration, and Repair, Departments of Neuroscience and Neurology, Yale University School of Medicine, New Haven, CT, United States
| | - Richard E. Carson
- Department of Radiology and Biomedical Imaging, PET Center, Yale School of Medicine, New Haven, CT, United States
| | - Yiyun Huang
- Department of Radiology and Biomedical Imaging, PET Center, Yale School of Medicine, New Haven, CT, United States
| | - Zhengxin Cai
- Department of Radiology and Biomedical Imaging, PET Center, Yale School of Medicine, New Haven, CT, United States
| |
Collapse
|
19
|
The Interplay between α-Synuclein and Microglia in α-Synucleinopathies. Int J Mol Sci 2023; 24:ijms24032477. [PMID: 36768798 PMCID: PMC9916729 DOI: 10.3390/ijms24032477] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 01/19/2023] [Accepted: 01/24/2023] [Indexed: 02/01/2023] Open
Abstract
Synucleinopathies are a set of devastating neurodegenerative diseases that share a pathologic accumulation of the protein α-synuclein (α-syn). This accumulation causes neuronal death resulting in irreversible dementia, deteriorating motor symptoms, and devastating cognitive decline. While the etiology of these conditions remains largely unknown, microglia, the resident immune cells of the central nervous system (CNS), have been consistently implicated in the pathogenesis of synucleinopathies. Microglia are generally believed to be neuroprotective in the early stages of α-syn accumulation and contribute to further neurodegeneration in chronic disease states. While the molecular mechanisms by which microglia achieve this role are still being investigated, here we highlight the major findings to date. In this review, we describe how structural varieties of inherently disordered α-syn result in varied microglial receptor-mediated interactions. We also summarize which microglial receptors enable cellular recognition and uptake of α-syn. Lastly, we review the downstream effects of α-syn processing within microglia, including spread to other brain regions resulting in neuroinflammation and neurodegeneration in chronic disease states. Understanding the mechanism of microglial interactions with α-syn is vital to conceptualizing molecular targets for novel therapeutic interventions. In addition, given the significant diversity in the pathophysiology of synucleinopathies, such molecular interactions are vital in gauging all potential pathways of neurodegeneration in the disease state.
Collapse
|
20
|
Feng L, Lo H, You H, Wu W, Cheng X, Xin J, Ye Z, Chen X, Pan X. Loss of cannabinoid receptor 2 promotes α-Synuclein-induced microglial synaptic pruning in nucleus accumbens by modulating the pCREB-c-Fos signaling pathway and complement system. Exp Neurol 2023; 359:114230. [PMID: 36162511 DOI: 10.1016/j.expneurol.2022.114230] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 08/31/2022] [Accepted: 09/19/2022] [Indexed: 12/30/2022]
Abstract
The disruption of nucleus accumbens (NAc) function impacts mood and learning behavior in α-Synucleinopathy, in which microglial synaptic pruning plays a pivotal role in modulating the neuropathologic progression. Available literature documents that in microglia, the activation of cannabinoid receptor 2 (CB2R) decreases inflammation, but it remains obscured regarding the roles of CB2R in microglia-mediated synaptic pruning in the NAc during the neuropathological progression of α-Synucleinopathy. We adopted the fibrillar α-Synuclein (α-Syn) treatment to characterize the effect of genetic CB2R deletion on microglial function and the signaling pathway. CB2R knockout (CB2-/-) mice and wild-type (CB2+/+) mice were divided into the α-Syn or saline treatment groups. Biochemical and microscopy approaches, including immunofluorescence, real-time PCR, and western blotting, were employed to assess the changes in homeostasis of synaptic pruning in NAc under the α-Syn-induced microglia. Moreover, the underlying mechanisms of CB2R on α-Syn induced microglial activity was assessed in vitro. After the injection of α-Syn into the NAc, distinct microglial morphological changes and M1 phenotype transformation were observed between CB2-/- and CB2+/+ mice. Meanwhile, after the α-Syn treatment, CB2-/- mice showed an increased upregulation of CD68 protein and IL-1β mRNA but decreased brain-derived neurotrophic factor (BDNF) and TGF-β mRNA compared with CB2+/+ mice. Additionally, CB2-/- microglia after the treatment showed a highly enriched complement 3a receptor (C3aR) producing excessive pruning of cholinergic synapses but less engulfment of dopaminergic synapses. Mechanistically, the loss of CB2R function in the α-Syn stimulation triggered c-Fos activation in microglia, but not in neurons. Further inhibition of microglial CB2R functions under α-Syn stimulation activated the phosphorylated cAMP-response element-binding protein (pCREB)-c-Fos, which was closely related to the C3aR upregulation. Our results reveal a critical and mechanistic role of CB2R in altering the microglial function and its value in the homeostasis of synaptic circuits in the NAc under the α-Syn pathology.
Collapse
Affiliation(s)
- Linjuan Feng
- Department of Neurology, Center for Cognitive Neurology, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou 350001, China; Fujian Institute of Geriatrics, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou 350001, China; Institute of Clinical Neurology, Fujian Medical University, 29 Xinquan Road, Fuzhou 350001, China; Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, 88 Jiaotong Road, Fuzhou 350001, China
| | - Hsuan Lo
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Hanlin You
- Department of Neurology, Center for Cognitive Neurology, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou 350001, China; Fujian Institute of Geriatrics, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou 350001, China; Institute of Clinical Neurology, Fujian Medical University, 29 Xinquan Road, Fuzhou 350001, China; Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, 88 Jiaotong Road, Fuzhou 350001, China
| | - Wei Wu
- Key Laboratory of Brain Aging and Neurodegenerative Diseases, Fujian Medical University, 1 Xueyuan Road, Fuzhou 350001, China
| | - Xiaojuan Cheng
- Department of Neurology, Center for Cognitive Neurology, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou 350001, China; Fujian Institute of Geriatrics, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou 350001, China; Institute of Clinical Neurology, Fujian Medical University, 29 Xinquan Road, Fuzhou 350001, China; Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, 88 Jiaotong Road, Fuzhou 350001, China
| | - Jiawei Xin
- Department of Neurology, Center for Cognitive Neurology, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou 350001, China; Fujian Institute of Geriatrics, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou 350001, China; Institute of Clinical Neurology, Fujian Medical University, 29 Xinquan Road, Fuzhou 350001, China
| | - Zucheng Ye
- Key Laboratory of Brain Aging and Neurodegenerative Diseases, Fujian Medical University, 1 Xueyuan Road, Fuzhou 350001, China
| | - Xiaochun Chen
- Department of Neurology, Center for Cognitive Neurology, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou 350001, China; Institute of Clinical Neurology, Fujian Medical University, 29 Xinquan Road, Fuzhou 350001, China; Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, 88 Jiaotong Road, Fuzhou 350001, China
| | - Xiaodong Pan
- Department of Neurology, Center for Cognitive Neurology, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou 350001, China; Fujian Institute of Geriatrics, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou 350001, China; Institute of Clinical Neurology, Fujian Medical University, 29 Xinquan Road, Fuzhou 350001, China; Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, 88 Jiaotong Road, Fuzhou 350001, China; Key Laboratory of Brain Aging and Neurodegenerative Diseases, Fujian Medical University, 1 Xueyuan Road, Fuzhou 350001, China.
| |
Collapse
|
21
|
Real CC, Binda KH, Thomsen MB, Lillethorup TP, Brooks DJ, Landau AM. Selecting the Best Animal Model of Parkinson's Disease for Your Research Purpose: Insight from in vivo PET Imaging Studies. Curr Neuropharmacol 2023; 21:1241-1272. [PMID: 36797611 PMCID: PMC10286593 DOI: 10.2174/1570159x21666230216101659] [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: 08/17/2022] [Revised: 09/08/2022] [Accepted: 09/08/2022] [Indexed: 02/18/2023] Open
Abstract
Parkinson's disease (PD) is a debilitating neurodegenerative multisystem disorder leading to motor and non-motor symptoms in millions of individuals. Despite intense research, there is still no cure, and early disease biomarkers are lacking. Animal models of PD have been inspired by basic elements of its pathogenesis, such as dopamine dysfunction, alpha-synuclein accumulation, neuroinflammation and disruption of protein degradation, and these have been crucial for a deeper understanding of the mechanisms of pathology, the identification of biomarkers, and evaluation of novel therapies. Imaging biomarkers are non-invasive tools to assess disease progression and response to therapies; their discovery and validation have been an active field of translational research. Here, we highlight different considerations of animal models of PD that can be applied to future research, in terms of their suitability to answer different research questions. We provide the reader with important considerations of the best choice of model to use based on the disease features of each model, including issues related to different species. In addition, positron emission tomography studies conducted in PD animal models in the last 5 years are presented. With a variety of different species, interventions and genetic information, the choice of the most appropriate model to answer research questions can be daunting, especially since no single model recapitulates all aspects of this complex disorder. Appropriate animal models in conjunction with in vivo molecular imaging tools, if selected properly, can be a powerful combination for the assessment of novel therapies and developing tools for early diagnosis.
Collapse
Affiliation(s)
- Caroline Cristiano Real
- Department of Nuclear Medicine and PET Center, Aarhus University Hospital, Aarhus, Denmark
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Karina Henrique Binda
- Department of Nuclear Medicine and PET Center, Aarhus University Hospital, Aarhus, Denmark
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Majken Borup Thomsen
- Department of Nuclear Medicine and PET Center, Aarhus University Hospital, Aarhus, Denmark
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Thea Pinholt Lillethorup
- Department of Nuclear Medicine and PET Center, Aarhus University Hospital, Aarhus, Denmark
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - David James Brooks
- Department of Nuclear Medicine and PET Center, Aarhus University Hospital, Aarhus, Denmark
- Institute of Translational and Clinical Research, University of Newcastle, Upon Tyne, UK
| | - Anne Marlene Landau
- Department of Nuclear Medicine and PET Center, Aarhus University Hospital, Aarhus, Denmark
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
22
|
Bai X, Guo T, Chen J, Guan X, Zhou C, Wu J, Liu X, Wu H, Wen J, Gu L, Gao T, Xuan M, Huang P, Zhang B, Xu X, Zhang M. Microstructural but not macrostructural cortical degeneration occurs in Parkinson’s disease with mild cognitive impairment. NPJ Parkinsons Dis 2022; 8:151. [DOI: 10.1038/s41531-022-00416-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 10/14/2022] [Indexed: 11/11/2022] Open
Abstract
AbstractThis study aimed to investigate the cortical microstructural/macrostructural degenerative patterns in Parkinson’s disease (PD) patients with mild cognitive impairment (MCI). Overall, 38 PD patients with normal cognition (PD-NC), 38 PD-MCI, and 32 healthy controls (HC) were included. PD-MCI was diagnosed according to the MDS Task Force level II criteria. Cortical microstructural alterations were evaluated with Neurite Orientation Dispersion and Density Imaging. Cortical thickness analyses were derived from T1-weighted imaging using the FreeSurfer software. For cortical microstructural analyses, compared with HC, PD-NC showed lower orientation dispersion index (ODI) in bilateral cingulate and paracingulate gyri, supplementary motor area, right paracentral lobule, and precuneus (PFWE < 0.05); while PD-MCI showed lower ODI in widespread regions covering bilateral frontal, parietal, occipital, and right temporal areas and lower neurite density index in left frontal area, left cingulate, and paracingulate gyri (PFWE < 0.05). Furthermore, compared with PD-NC, PD-MCI showed reduced ODI in right frontal area and bilateral caudate nuclei (voxel P < 0.01 and cluster >100 voxels) and the ODI values were associated with the Montreal Cognitive Assessment scores (r = 0.440, P < 0.001) and the memory performance (r = 0.333, P = 0.004) in the PD patients. However, for cortical thickness analyses, there was no difference in the between-group comparisons. In conclusion, cortical microstructural alterations may precede macrostructural changes in PD-MCI. This study provides insightful evidence for the degenerative patterns in PD-MCI and contributes to our understanding of the latent biological basis of cortical neurite changes for early cognitive impairment in PD.
Collapse
|
23
|
Leveraging the preformed fibril model to distinguish between alpha-synuclein inclusion- and nigrostriatal degeneration-associated immunogenicity. Neurobiol Dis 2022; 171:105804. [PMID: 35764290 PMCID: PMC9803935 DOI: 10.1016/j.nbd.2022.105804] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 05/04/2022] [Accepted: 06/22/2022] [Indexed: 01/03/2023] Open
Abstract
Neuroinflammation has become a well-accepted pathologic hallmark of Parkinson's disease (PD). However, it remains unclear whether inflammation, triggered by α-syn aggregation and/or degeneration, contributes to the progression of the disease. Studies examining neuroinflammation in PD are unable to distinguish between Lewy body-associated inflammation and degeneration-associated inflammation, as both pathologies are present simultaneously. Intrastriatal and intranigral injections of alpha-synuclein (α-syn) preformed fibrils (PFFs) results in two distinct pathologic phases: Phase 1: The accumulation and peak formation of α-syn inclusions in nigrostriatal system and, Phase 2: Protracted dopaminergic neuron degeneration. In this review we summarize the current understanding of neuroinflammation in the α-syn PFF model, leveraging the distinct Phase 1 aggregation phase and Phase 2 degeneration phase to guide our interpretations. Studies consistently demonstrate an association between pathologic α-syn aggregation in the substantia nigra (SN) and activation of the innate immune system. Further, major histocompatibility complex-II (MHC-II) antigen presentation is proportionate to inclusion load. The α-syn aggregation phase is also associated with peripheral and adaptive immune cell infiltration to the SN. These findings suggest that α-syn like aggregates are immunogenic and thus have the potential to contribute to the degenerative process. Studies examining neuroinflammation during the neurodegenerative phase reveal elevated innate, adaptive, and peripheral immune cell markers, however limitations of single time point experimental design hinder interpretations as to whether this neuroinflammation preceded, or was triggered by, nigral degeneration. Longitudinal studies across both the aggregation and degeneration phases of the model suggest that microglial activation (MHC-II) is greater in magnitude during the aggregation phase that precedes degeneration. Overall, the consistency between neuroinflammatory markers in the parkinsonian brain and in the α-syn PFF model, combined with the distinct aggregation and degenerative phases, establishes the utility of this model platform to yield insights into pathologic events that contribute to neuroinflammation and disease progression in PD.
Collapse
|
24
|
Sossi V, Patterson JR, McCormick S, Kemp CJ, Miller KM, Stoll AC, Kuhn N, Kubik M, Kochmanski J, Luk KC, Sortwell CE. Dopaminergic Positron Emission Tomography Imaging in the Alpha-Synuclein Preformed Fibril Model Reveals Similarities to Early Parkinson's Disease. Mov Disord 2022; 37:1739-1748. [PMID: 35524682 PMCID: PMC9391270 DOI: 10.1002/mds.29051] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 02/24/2022] [Accepted: 04/18/2022] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND Positron emission tomography (PET) imaging in early Parkinson's disease (PD) subjects reveals that increased dopamine (DA) turnover and reduced dopamine transporter (DAT) density precede decreases in DA synthesis and storage. The rat α-synuclein preformed fibril (α-syn PFF) model provides a platform to investigate DA dynamics during multiple stages of α-syn inclusion-triggered nigrostriatal degeneration. OBJECTIVES We investigated multiple aspects of in vivo dopaminergic deficits longitudinally and similarities to human PD using translational PET imaging readouts. METHODS Longitudinal imaging was performed every 2 months in PFF and control rats for 7 months. [18 F]-Fluoro-3,4-dihydroxyphenyl-L-alanine (FDOPA) imaging was performed to investigate DA synthesis and storage (Kocc ) and DA turnover, estimated by its inverse, the effective distribution volume ratio (EDVR). 11 C-Methylphenidate (MP) was used to estimate DAT density (BPND ). RESULTS Early DA turnover increases and DAT binding decreases were observed in the ipsilateral striatum of PFF rats, progressing longitudinally. EDVR decreased 26%, 38%, and 47%, and BPND decreased 36%, 50%, and 65% at the 2-, 4-, and 6-month time points, respectively, compared to ipsilateral control striatum. In contrast, deficits in DA synthesis and storage were not observed in the ipsilateral striatum of PFF rats compared to control injections and were relatively preserved up to 6 months (Kocc decreased 20% at 6 months). CONCLUSIONS The relative preservation of DA synthesis and storage compared to robust progressive deficits in DAT density and increases in DA turnover in the rat α-syn PFF model display remarkable face validity to dopaminergic alterations in human PD. © 2022 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Vesna Sossi
- Department of Physics and AstronomyUniversity of British ColumbiaVancouverBritish ColumbiaCanada
| | - Joseph R. Patterson
- Department of Translational NeuroscienceMichigan State UniversityGrand RapidsMichiganUSA
| | - Siobhan McCormick
- Pacific Parkinson's Research CentreUniversity of British ColumbiaVancouverBritish ColumbiaCanada
| | - Christopher J. Kemp
- Department of Translational NeuroscienceMichigan State UniversityGrand RapidsMichiganUSA
| | - Kathryn M. Miller
- Department of Translational NeuroscienceMichigan State UniversityGrand RapidsMichiganUSA
| | - Anna C. Stoll
- Department of Translational NeuroscienceMichigan State UniversityGrand RapidsMichiganUSA
| | - Nathan Kuhn
- Department of Translational NeuroscienceMichigan State UniversityGrand RapidsMichiganUSA
| | - Michael Kubik
- Department of Translational NeuroscienceMichigan State UniversityGrand RapidsMichiganUSA
| | - Joseph Kochmanski
- Department of Translational NeuroscienceMichigan State UniversityGrand RapidsMichiganUSA
| | - Kelvin C. Luk
- Department of Pathology and Laboratory MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Caryl E. Sortwell
- Department of Translational NeuroscienceMichigan State UniversityGrand RapidsMichiganUSA
| |
Collapse
|
25
|
Rossi R, Arjmand S, Bærentzen SL, Gjedde A, Landau AM. Synaptic Vesicle Glycoprotein 2A: Features and Functions. Front Neurosci 2022; 16:864514. [PMID: 35573314 PMCID: PMC9096842 DOI: 10.3389/fnins.2022.864514] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 04/05/2022] [Indexed: 01/05/2023] Open
Abstract
In recent years, the field of neuroimaging dramatically moved forward by means of the expeditious development of specific radioligands of novel targets. Among these targets, the synaptic vesicle glycoprotein 2A (SV2A) is a transmembrane protein of synaptic vesicles, present in all synaptic terminals, irrespective of neurotransmitter content. It is involved in key functions of neurons, focused on the regulation of neurotransmitter release. The ubiquitous expression in gray matter regions of the brain is the basis of its candidacy as a marker of synaptic density. Following the development of molecules derived from the structure of the anti-epileptic drug levetiracetam, which selectively binds to SV2A, several radiolabeled markers have been synthetized to allow the study of SV2A distribution with positron emission tomography (PET). These radioligands permit the evaluation of in vivo changes of SV2A distribution held to be a potential measure of synaptic density in physiological and pathological conditions. The use of SV2A as a biomarker of synaptic density raises important questions. Despite numerous studies over the last decades, the biological function and the expressional properties of SV2A remain poorly understood. Some functions of SV2A were claimed, but have not been fully elucidated. While the expression of SV2A is ubiquitous, stronger associations between SV2A and Υ amino butyric acid (GABA)-ergic rather than glutamatergic synapses were observed in some brain structures. A further issue is the unclear interaction between SV2A and its tracers, which reflects a need to clarify what really is detected with neuroimaging tools. Here, we summarize the current knowledge of the SV2A protein and we discuss uncertain aspects of SV2A biology and physiology. As SV2A expression is ubiquitous, but likely more strongly related to a certain type of neurotransmission in particular circumstances, a more extensive knowledge of the protein would greatly facilitate the analysis and interpretation of neuroimaging results by allowing the evaluation not only of an increase or decrease of the protein level, but also of the type of neurotransmission involved.
Collapse
Affiliation(s)
- Rachele Rossi
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.,Department of Nuclear Medicine and PET Center, Aarhus University Hospital, Aarhus, Denmark
| | - Shokouh Arjmand
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Simone Larsen Bærentzen
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.,Department of Nuclear Medicine and PET Center, Aarhus University Hospital, Aarhus, Denmark
| | - Albert Gjedde
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.,Department of Neuroscience, University of Copenhagen, Copenhagen, Denmark.,Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| | - Anne M Landau
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.,Department of Nuclear Medicine and PET Center, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
26
|
Lillethorup TP, Noer O, Alstrup AKO, Real CC, Stokholm K, Thomsen MB, Zaer H, Orlowski D, Mikkelsen TW, Glud AN, Nielsen EHT, Schacht AC, Winterdahl M, Brooks DJ, Sørensen JCH, Landau AM. Spontaneous Partial Recovery of Striatal Dopaminergic Uptake Despite Nigral Cell Loss in Asymptomatic MPTP-Lesioned Female Minipigs. Neurotoxicology 2022; 91:166-176. [DOI: 10.1016/j.neuro.2022.05.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 04/06/2022] [Accepted: 05/09/2022] [Indexed: 11/16/2022]
|
27
|
Changes in CD163+, CD11b+, and CCR2+ peripheral monocytes relate to Parkinson's disease and cognition. Brain Behav Immun 2022; 101:182-193. [PMID: 35026420 DOI: 10.1016/j.bbi.2022.01.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 01/04/2022] [Accepted: 01/07/2022] [Indexed: 12/15/2022] Open
Abstract
Alpha-synuclein pathology is associated with immune activation and neurodegeneration in Parkinson's disease. The immune activation involves not only microglia but also peripheral immune cells, such as mononuclear phagocytes found in blood and infiltrated in the brain. Understanding peripheral immune involvement is essential for developing immunomodulatory treatment. Therefore, we aimed to study circulating mononuclear phagocytes in early- and late-stage Parkinson's disease, defined by disease duration of less or more than five years, respectively, and analyze their association with clinical phenotypes. We performed a cross-sectional multi-color flow cytometry study on 78 sex-balanced individuals with sporadic Parkinson's disease, 28 controls, and longitudinal samples from seven patients and one control. Cell frequencies and surface marker expressions on natural killer cells, monocyte subtypes, and dendritic cells were compared between groups and correlated with standardized clinical scores. We found elevated frequencies and surface levels of migration- (CCR2, CD11b) and phagocytic- (CD163) markers, particularly on classical and intermediate monocytes in early Parkinson's disease. HLA-DR expression was increased in advanced stages of the disease, whereas TLR4 expression was decreased in women with Parkinson's Disease. The disease-associated immune changes of CCR2 and CD11b correlated with worse cognition. Increased TLR2 expression was related to worse motor symptoms. In conclusion, our data highlights the TLR2 relevance in the symptomatic motor presentation of the disease and a role for peripheral CD163+ and migration-competent monocytes in Parkinson's disease cognitive defects. Our study suggests that the peripheral immune system is dynamically altered in Parkinson's disease stages and directly related to both symptoms and the sex bias of the disease.
Collapse
|
28
|
α-Synuclein Overexpression Increases Dopamine D2/3 Receptor Binding and Immune Activation in a Model of Early Parkinson’s Disease. Biomedicines 2021; 9:biomedicines9121876. [PMID: 34944691 PMCID: PMC8698691 DOI: 10.3390/biomedicines9121876] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 12/01/2021] [Accepted: 12/07/2021] [Indexed: 12/14/2022] Open
Abstract
Progressive degeneration of dopaminergic neurons, immune activation, and α-synuclein pathology characterize Parkinson’s disease (PD). We previously reported that unilateral intranigral injection of recombinant adeno-associated viral (rAAV) vectors encoding wild-type human α-synuclein produced a rat model of early PD with dopamine terminal dysfunction. Here we tested the hypothesis that decreases in dopamine result in increased postsynaptic dopamine D2/D3 receptor expression, neuroinflammation, and reduced synaptic vesicle glycoprotein 2A (SV2A) density. Rats were injected with rAAV encoding α-synuclein or green fluorescent protein and subjected to non-pharmacological motor tests, before euthanization at 12 weeks post-injection. We performed: (1) in situ hybridization of nigral tyrosine hydroxylase mRNA, (2) HPLC of striatal dopamine content, and (3) autoradiography with [3H]raclopride, [3H]DTBZ, [3H]GBR12935, [3H]PK11195, and [3H]UCB-J to measure binding at D2/3 receptors, vesicular monoamine transporter 2, dopamine transporters, mitochondrial translocator protein, and SV2A, respectively. rAAV-α-synuclein induced motor asymmetry and reduced tyrosine hydroxylase mRNA and dopamine content in ipsilateral brain regions. This was paralleled by elevated ipsilateral postsynaptic dopamine D2/3 receptor expression and immune activation, with no changes to synaptic SV2A density. In conclusion, α-synuclein overexpression results in dopaminergic degeneration that induced compensatory increases in D2/3 binding and immune activation, recapitulating many of the pathological characteristics of PD.
Collapse
|
29
|
Raval NR, Gudmundsen F, Juhl M, Andersen IV, Speth N, Videbæk A, Petersen IN, Mikkelsen JD, Fisher PM, Herth MM, Plavén-Sigray P, Knudsen GM, Palner M. Synaptic Density and Neuronal Metabolic Function Measured by Positron Emission Tomography in the Unilateral 6-OHDA Rat Model of Parkinson's Disease. Front Synaptic Neurosci 2021; 13:715811. [PMID: 34867258 PMCID: PMC8636601 DOI: 10.3389/fnsyn.2021.715811] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 10/06/2021] [Indexed: 11/30/2022] Open
Abstract
Parkinson’s disease (PD) is caused by progressive neurodegeneration and characterised by motor dysfunction. Neurodegeneration of dopaminergic neurons also causes aberrations within the cortico-striato-thalamo-cortical (CSTC) circuit, which has been hypothesised to lead to non-motor symptoms such as depression. Individuals with PD have both lower synaptic density and changes in neuronal metabolic function in the basal ganglia, as measured using [11C]UCB-J and [18F]FDG positron emission tomography (PET), respectively. However, the two radioligands have not been directly compared in the same PD subject or in neurodegeneration animal models. Here, we investigate [11C]UCB-J binding and [18F]FDG uptake in the CSTC circuit following a unilateral dopaminergic lesion in rats and compare it to sham lesioned rats. Rats received either a unilateral injection of 6-hydroxydopamine (6-OHDA) or saline in the medial forebrain bundle and rostral substantia nigra (n = 4/group). After 3 weeks, all rats underwent two PET scans using [18F]FDG, followed by [11C]UCB-J on a separate day. [18F]FDG uptake and [11C]UCB-J binding were both lower in the ipsilateral striatal regions compared to the contralateral regions. Using [11C]UCB-J, we could detect an 8.7% decrease in the ipsilateral ventral midbrain, compared to a 2.9% decrease in ventral midbrain using [18F]FDG. Differential changes between hemispheres for [11C]UCB-J and [18F]FDG outcomes were also evident in the CSTC circuit’s cortical regions, especially in the orbitofrontal cortex and medial prefrontal cortex where higher synaptic density yet lower neuronal metabolic function was observed, following lesioning. In conclusion, [11C]UCB-J and [18F]FDG PET can detect divergent changes following a dopaminergic lesion in rats, especially in cortical regions that are not directly affected by the neurotoxin. These results suggest that combined [11C]UCB-J and [18F]FDG scans could yield a better picture of the heterogeneous cerebral changes in neurodegenerative disorders.
Collapse
Affiliation(s)
- Nakul Ravi Raval
- Neurobiology Research Unit, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark.,Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Frederik Gudmundsen
- Neurobiology Research Unit, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Morten Juhl
- Cardiology Stem Cell Centre, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Ida Vang Andersen
- Neurobiology Research Unit, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark.,Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Nikolaj Speth
- Neurobiology Research Unit, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Annesofie Videbæk
- Neurobiology Research Unit, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Ida Nymann Petersen
- Department of Clinical Physiology, Nuclear Medicine & PET, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Jens D Mikkelsen
- Neurobiology Research Unit, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark.,Department of Neuroscience, University of Copenhagen, Copenhagen, Denmark
| | | | - Matthias Manfred Herth
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Department of Clinical Physiology, Nuclear Medicine & PET, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Pontus Plavén-Sigray
- Neurobiology Research Unit, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Gitte Moos Knudsen
- Neurobiology Research Unit, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark.,Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Mikael Palner
- Neurobiology Research Unit, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark.,Clinical Physiology and Nuclear Medicine, Department of Clinical Research, University of Southern Denmark, Odense, Denmark.,Department of Nuclear Medicine, Odense University Hospital, Odense, Denmark
| |
Collapse
|
30
|
Yan JN, Zhang HY, Li JR, Chen Y, Jiang YC, Shen JB, Ke KF, Gu XS. Schwann cells differentiated from skin-derived precursors provide neuroprotection via autophagy inhibition in a cellular model of Parkinson's disease. Neural Regen Res 2021; 17:1357-1363. [PMID: 34782582 PMCID: PMC8643066 DOI: 10.4103/1673-5374.327353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Autophagy has been shown to play an important role in Parkinson’s disease. We hypothesized that skin-derived precursor cells exhibit neuroprotective effects in Parkinson’s disease through affecting autophagy. In this study, 6-hydroxydopamine-damaged SH-SY5Y cells were pretreated with a culture medium containing skin-derived precursors differentiated into Schwann cells (SKP-SCs). The results showed that the SKP-SC culture medium remarkably enhanced the activity of SH-SY5Y cells damaged by 6-hydroxydopamine, reduced excessive autophagy, increased tyrosine hydroxylase expression, reduced α-synuclein expression, reduced the autophagosome number, and activated the PI3K/AKT/mTOR pathway. Autophagy activator rapamycin inhibited the effects of SKP-SCs, and autophagy inhibitor 3-methyladenine had the opposite effect. These findings confirm that SKP-SCs modulate the PI3K/AKT/mTOR pathway to inhibit autophagy, thereby exhibiting a neuroprotective effect in a cellular model of Parkinson’s disease. This study was approved by the Animal Ethics Committee of Laboratory Animal Center of Nantong University (approval No. S20181009-205) on October 9, 2018.
Collapse
Affiliation(s)
- Jia-Nan Yan
- Department of Neurology, Affiliated Hospital of Nantong University; Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China
| | - Hai-Ying Zhang
- Department of Neurology, Affiliated Hospital of Nantong University; Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China
| | - Jun-Rui Li
- Department of Clinical Medicine, The First Clinical Medical College of Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Ying Chen
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong; Department of Neurology and Suzhou Clinical Research Center of Neurological Disease, The Second Afflicted Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Yong-Cheng Jiang
- Department of Neurology, Affiliated Hospital of Nantong University; Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China
| | - Jia-Bing Shen
- Department of Neurology, Affiliated Hospital of Nantong University; Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China
| | - Kai-Fu Ke
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China
| | - Xiao-Su Gu
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China
| |
Collapse
|
31
|
Iyer V, Venkiteswaran K, Savaliya S, Lieu CA, Handly E, Gilmour TP, Kunselman AR, Subramanian T. The cross-hemispheric nigrostriatal pathway prevents the expression of levodopa-induced dyskinesias. Neurobiol Dis 2021; 159:105491. [PMID: 34461264 PMCID: PMC8597404 DOI: 10.1016/j.nbd.2021.105491] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 08/17/2021] [Accepted: 08/25/2021] [Indexed: 11/29/2022] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative movement disorder that is routinely treated with levodopa. Unfortunately, long-term dopamine replacement therapy using levodopa leads to levodopa-induced dyskinesias (LID), a significant and disabling side-effect. Clinical findings indicate that LID typically only occurs following the progression of PD motor symptoms from the unilateral (Hoehn and Yahr (HY) Stage I) to the bilateral stage (HY Stage II). This suggests the presence of some compensatory interhemispheric mechanisms that delay the occurrence of LID. We therefore investigated the role of interhemispheric connections of the nigrostriatal pathway on LID expression in a rat model of PD. The striatum of one hemisphere of rats was first injected with a retrograde tracer to label the ipsi- and cross-hemispheric nigrostriatal pathways. Rats were then split into groups and unilaterally lesioned in the striatum or medial forebrain bundle of the tracer-injected hemisphere to induce varying levels of hemiparkinsonism. Finally, rats were treated with levodopa and tested for the expression of LID. Distinct subsets emerged from rats that underwent the same lesioning paradigm based on LID. Strikingly, non-dyskinetic rats had significant sparing of their cross-hemispheric nigrostriatal pathway projecting from the unlesioned hemisphere. In contrast, dyskinetic rats only had a small proportion of this cross-hemispheric nigrostriatal pathway survive lesioning. Crucially, both non-dyskinetic and dyskinetic rats had nearly identical levels of ipsi-hemispheric nigrostriatal pathway survival and parkinsonian motor deficits. Our data suggest that the survival of the cross-hemispheric nigrostriatal pathway plays a crucial role in preventing the expression of LID and represents a potentially novel target to halt the progression of this devastating side-effect of a common anti-PD therapeutic.
Collapse
Affiliation(s)
- Vishakh Iyer
- Department of Neurology and Neural and Behavioral Sciences, The Pennsylvania State University College of Medicine, Hershey, PA, United States of America
| | - Kala Venkiteswaran
- Department of Neurology and Neural and Behavioral Sciences, The Pennsylvania State University College of Medicine, Hershey, PA, United States of America
| | - Sandip Savaliya
- Department of Neurosurgery, The Pennsylvania State University College of Medicine, Hershey, PA, United States of America
| | - Christopher A Lieu
- Department of Neurology and Neural and Behavioral Sciences, The Pennsylvania State University College of Medicine, Hershey, PA, United States of America
| | - Erin Handly
- Department of Neurology and Neural and Behavioral Sciences, The Pennsylvania State University College of Medicine, Hershey, PA, United States of America
| | - Timothy P Gilmour
- Department of Electrical Engineering, John Brown University, Siloam Springs, AR, United States of America
| | - Allen R Kunselman
- Department of Public Health Sciences, The Pennsylvania State University College of Medicine, Hershey, PA, United States of America
| | - Thyagarajan Subramanian
- Department of Neurology and Neural and Behavioral Sciences, The Pennsylvania State University College of Medicine, Hershey, PA, United States of America.
| |
Collapse
|
32
|
Li Y, Xia Y, Yin S, Wan F, Hu J, Kou L, Sun Y, Wu J, Zhou Q, Huang J, Xiong N, Wang T. Targeting Microglial α-Synuclein/TLRs/NF-kappaB/NLRP3 Inflammasome Axis in Parkinson's Disease. Front Immunol 2021; 12:719807. [PMID: 34691027 PMCID: PMC8531525 DOI: 10.3389/fimmu.2021.719807] [Citation(s) in RCA: 76] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Accepted: 09/22/2021] [Indexed: 01/04/2023] Open
Abstract
According to emerging studies, the excessive activation of microglia and the subsequent release of pro-inflammatory cytokines play important roles in the pathogenesis and progression of Parkinson's disease (PD). However, the exact mechanisms governing chronic neuroinflammation remain elusive. Findings demonstrate an elevated level of NLRP3 inflammasome in activated microglia in the substantia nigra of PD patients. Activated NLRP3 inflammasome aggravates the pathology and accelerates the progression of neurodegenerative diseases. Abnormal protein aggregation of α-synuclein (α-syn), a pathologically relevant protein of PD, were reported to activate the NLRP3 inflammasome of microglia through interaction with toll-like receptors (TLRs). This eventually releases pro-inflammatory cytokines through the translocation of nuclear factor kappa-B (NF-κB) and causes an impairment of mitochondria, thus damaging the dopaminergic neurons. Currently, therapeutic drugs for PD are primarily aimed at providing relief from its clinical symptoms, and there are no well-established strategies to halt or reverse this disease. In this review, we aimed to update existing knowledge on the role of the α-syn/TLRs/NF-κB/NLRP3 inflammasome axis and microglial activation in PD. In addition, this review summarizes recent progress on the α-syn/TLRs/NF-κB/NLRP3 inflammasome axis of microglia as a potential target for PD treatment by inhibiting microglial activation.
Collapse
Affiliation(s)
- Yunna Li
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yun Xia
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Sijia Yin
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fang Wan
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Junjie Hu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Liang Kou
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yadi Sun
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiawei Wu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qiulu Zhou
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jinsha Huang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Nian Xiong
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tao Wang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
33
|
Polinski NK. A Summary of Phenotypes Observed in the In Vivo Rodent Alpha-Synuclein Preformed Fibril Model. JOURNAL OF PARKINSONS DISEASE 2021; 11:1555-1567. [PMID: 34486988 PMCID: PMC8609716 DOI: 10.3233/jpd-212847] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The use of wildtype recombinant alpha-synuclein preformed fibrils (aSyn PFFs) to induce endogenous alpha-synuclein to form pathological phosphorylation and trigger neurodegeneration is a popular model for studying Parkinson's disease (PD) biology and testing therapeutic strategies. The strengths of this model lie in its ability to recapitulate the phosphorylation/aggregation of aSyn and nigrostriatal degeneration seen in PD, as well as its suitability for studying the progressive nature of PD and the spread of aSyn pathology. Although the model is commonly used and has been adopted by many labs, variability in observed phenotypes exists. Here we provide summaries of the study design and reported phenotypes from published reports characterizing the aSyn PFF in vivo model in rodents following injection into the brain, gut, muscle, vein, peritoneum, and eye. These summaries are designed to facilitate an introduction to the use of aSyn PFFs to generate a rodent model of PD-highlighting phenotypes observed in papers that set out to thoroughly characterize the model. This information will hopefully improve the understanding of this model and clarify when the aSyn PFF model may be an appropriate choice for one's research.
Collapse
Affiliation(s)
- Nicole K Polinski
- The Michael J. Fox Foundation for Parkinson'sResearch, New York, NY, USA
| |
Collapse
|
34
|
Ferreira N, Gram H, Sorrentino ZA, Gregersen E, Schmidt SI, Reimer L, Betzer C, Perez-Gozalbo C, Beltoja M, Nagaraj M, Wang J, Nowak JS, Dong M, Willén K, Cholak E, Bjerregaard-Andersen K, Mendez N, Rabadia P, Shahnawaz M, Soto C, Otzen DE, Akbey Ü, Meyer M, Giasson BI, Romero-Ramos M, Jensen PH. Multiple system atrophy-associated oligodendroglial protein p25α stimulates formation of novel α-synuclein strain with enhanced neurodegenerative potential. Acta Neuropathol 2021; 142:87-115. [PMID: 33978813 PMCID: PMC8217051 DOI: 10.1007/s00401-021-02316-0] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 04/20/2021] [Accepted: 04/21/2021] [Indexed: 01/08/2023]
Abstract
Pathology consisting of intracellular aggregates of alpha-Synuclein (α-Syn) spread through the nervous system in a variety of neurodegenerative disorders including Parkinson's disease, dementia with Lewy bodies, and multiple system atrophy. The discovery of structurally distinct α-Syn polymorphs, so-called strains, supports a hypothesis where strain-specific structures are templated into aggregates formed by native α-Syn. These distinct strains are hypothesised to dictate the spreading of pathology in the tissue and the cellular impact of the aggregates, thereby contributing to the variety of clinical phenotypes. Here, we present evidence of a novel α-Syn strain induced by the multiple system atrophy-associated oligodendroglial protein p25α. Using an array of biophysical, biochemical, cellular, and in vivo analyses, we demonstrate that compared to α-Syn alone, a substoichiometric concentration of p25α redirects α-Syn aggregation into a unique α-Syn/p25α strain with a different structure and enhanced in vivo prodegenerative properties. The α-Syn/p25α strain induced larger inclusions in human dopaminergic neurons. In vivo, intramuscular injection of preformed fibrils (PFF) of the α-Syn/p25α strain compared to α-Syn PFF resulted in a shortened life span and a distinct anatomical distribution of inclusion pathology in the brain of a human A53T transgenic (line M83) mouse. Investigation of α-Syn aggregates in brain stem extracts of end-stage mice demonstrated that the more aggressive phenotype of the α-Syn/p25α strain was associated with an increased load of α-Syn aggregates based on a Förster resonance energy transfer immunoassay and a reduced α-Syn aggregate seeding activity based on a protein misfolding cyclic amplification assay. When injected unilaterally into the striata of wild-type mice, the α-Syn/p25α strain resulted in a more-pronounced motoric phenotype than α-Syn PFF and exhibited a "tropism" for nigro-striatal neurons compared to α-Syn PFF. Overall, our data support a hypothesis whereby oligodendroglial p25α is responsible for generating a highly prodegenerative α-Syn strain in multiple system atrophy.
Collapse
|
35
|
Binda KH, Lillethorup TP, Real CC, Bærentzen SL, Nielsen MN, Orlowski D, Brooks DJ, Chacur M, Landau AM. Exercise protects synaptic density in a rat model of Parkinson's disease. Exp Neurol 2021; 342:113741. [PMID: 33965411 DOI: 10.1016/j.expneurol.2021.113741] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 05/04/2021] [Indexed: 01/09/2023]
Abstract
BACKGROUND Parkinson's disease (PD) is characterized by Lewy body and neurite pathology associated with dopamine terminal dysfunction. Clinically, it is associated with motor slowing, rigidity, and tremor. Postural instability and pain are also features. Physical exercise benefits PD patients - possibly by promoting neuroplasticity including synaptic regeneration. OBJECTIVES In a parkinsonian rat model, we test the hypotheses that exercise: (a) increases synaptic density and reduces neuroinflammation and (b) lowers the nociceptive threshold by increasing μ-opioid receptor expression. METHODS Brain autoradiography was performed on rats unilaterally injected with either 6-hydroxydopamine (6-OHDA) or saline and subjected to treadmill exercise over 5 weeks. [3H]UCB-J was used to measure synaptic vesicle glycoprotein 2A (SV2A) density. Dopamine D2/3 receptor and μ-opioid receptor availability were assessed with [3H]Raclopride and [3H]DAMGO, respectively, while neuroinflammation was detected with the 18kDA translocator protein (TSPO) marker [3H]PK11195. The nociceptive threshold was determined prior to and throughout the exercise protocol. RESULTS We confirmed a dopaminegic deficit with increased striatal [3H]Raclopride D2/3 receptor availability and reduced nigral tyrosine hydroxylase immunoreactivity in the ipsilateral hemisphere of all 6-OHDA-injected rats. Sedentary rats lesioned with 6-OHDA showed significant reduction of ipsilateral striatal and substantia nigra [3H]UCB-J binding while [3H]PK11195 showed increased ipsilateral striatal neuroinflammation. Lesioned rats who exercised had higher levels of ipsilateral striatal [3H]UCB-J binding and lower levels of neuroinflammation compared to sedentary lesioned rats. Striatal 6-OHDA injections reduced thalamic μ-opioid receptor availability but subsequent exercise restored binding. Exercise also raised thalamic and hippocampal SV2A synaptic density in 6-OHDA lesioned rats, accompanied by a rise in nociceptive threshold. CONCLUSION These data suggest that treadmill exercise protects nigral and striatal synaptic integrity in a rat lesion model of PD - possibly by promoting compensatory mechanisms. Exercise was also associated with reduced neuroinflammation post lesioning and altered opioid transmission resulting in an increased nociceptive threshold.
Collapse
Affiliation(s)
- K H Binda
- Translational Neuropsychiatry Unit, Aarhus University, Universitetsbyen 13, Building 2b, Aarhus C 8000, Denmark; Department of Nuclear Medicine and PET, Aarhus University and Hospital, Palle Juul-Jensens Boulevard 165, J109, Aarhus N 8200, Denmark; Laboratory of Functional Neuroanatomy of Pain, Departamento de Anatomia, Universidade de Sao Paulo, Sao Paulo, SP, Brazil.
| | - T P Lillethorup
- Department of Nuclear Medicine and PET, Aarhus University and Hospital, Palle Juul-Jensens Boulevard 165, J109, Aarhus N 8200, Denmark.
| | - C C Real
- Translational Neuropsychiatry Unit, Aarhus University, Universitetsbyen 13, Building 2b, Aarhus C 8000, Denmark; Department of Nuclear Medicine and PET, Aarhus University and Hospital, Palle Juul-Jensens Boulevard 165, J109, Aarhus N 8200, Denmark; Laboratory of Nuclear Medicine (LIM 43), Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, SP, Brazil.
| | - S L Bærentzen
- Translational Neuropsychiatry Unit, Aarhus University, Universitetsbyen 13, Building 2b, Aarhus C 8000, Denmark; Department of Nuclear Medicine and PET, Aarhus University and Hospital, Palle Juul-Jensens Boulevard 165, J109, Aarhus N 8200, Denmark.
| | - M N Nielsen
- Translational Neuropsychiatry Unit, Aarhus University, Universitetsbyen 13, Building 2b, Aarhus C 8000, Denmark.
| | - D Orlowski
- Center for Experimental Neuroscience (CENSE), Department of Clinical Medicine, Aarhus University and Department of Neurosurgery, Aarhus University Hospital, Aarhus N, 8200, Denmark.
| | - D J Brooks
- Department of Nuclear Medicine and PET, Aarhus University and Hospital, Palle Juul-Jensens Boulevard 165, J109, Aarhus N 8200, Denmark; Institute for Translational and Clinical Research, Newcastle University, Newcastle upon Tyne NE4 5PL, UK.
| | - M Chacur
- Laboratory of Functional Neuroanatomy of Pain, Departamento de Anatomia, Universidade de Sao Paulo, Sao Paulo, SP, Brazil.
| | - A M Landau
- Translational Neuropsychiatry Unit, Aarhus University, Universitetsbyen 13, Building 2b, Aarhus C 8000, Denmark; Department of Nuclear Medicine and PET, Aarhus University and Hospital, Palle Juul-Jensens Boulevard 165, J109, Aarhus N 8200, Denmark.
| |
Collapse
|
36
|
Cui H, Wang W, Zheng X, Xia D, Liu H, Qin C, Tian H, Teng J. Decreased AQP4 Expression Aggravates ɑ-Synuclein Pathology in Parkinson's Disease Mice, Possibly via Impaired Glymphatic Clearance. J Mol Neurosci 2021; 71:2500-2513. [PMID: 33772424 DOI: 10.1007/s12031-021-01836-4] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Accepted: 03/19/2021] [Indexed: 12/13/2022]
Abstract
The pathological hallmarks of Parkinson's disease (PD), a neurodegenerative disorder, are the selective loss of dopamine neurons in the substantia nigra pars compacta (SNpc) and the presence of α-synuclein (α-syn) aggregates in the form of Lewy bodies/Lewy neurites (LBs/LNs) in neurons. Recent studies have indicated that aquaporin 4 (AQP4), as a predominant water channel protein in the brain, is involved in the progression of Parkinson's disease (PD). However, it remains unclear whether AQP4 expression affects α-syn pathology in Parkinson's disease. In this study, we established a progressive PD model by subjecting AQP4 null (AQP4+/-) mice to bilateral intrastriatal injection of α-syn preformed fibrils (PFFs) and investigated the effect of decreased AQP4 expression on the development of PD. We found that decreased expression of AQP4 accelerated pathologic deposition of α-syn and facilitated the loss of dopamine neurons and behavioral disorders. Draining of macromolecules from the brain via the glymphatic pathway was slowed due to decreased AQP4 expression. Taken together, these findings indicate that decreased AQP4 expression may aggravate PD-like pathology, possibly via impairment of the glymphatic pathway.
Collapse
Affiliation(s)
- Huili Cui
- Department of Neurology, Institute of Parkinson and Movement Disorder, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Wenkang Wang
- Department of Orthopedics, Graduate School of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China
| | - Xinhui Zheng
- Department of Orthopedics, Graduate School of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China
| | - Danhao Xia
- Department of Neurology, Institute of Parkinson and Movement Disorder, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Han Liu
- Department of Neurology, Institute of Parkinson and Movement Disorder, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Chi Qin
- Department of Neurology, Institute of Parkinson and Movement Disorder, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Haiyan Tian
- Department of Neurology, Institute of Parkinson and Movement Disorder, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Junfang Teng
- Department of Neurology, Institute of Parkinson and Movement Disorder, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
| |
Collapse
|
37
|
Gcwensa NZ, Russell DL, Cowell RM, Volpicelli-Daley LA. Molecular Mechanisms Underlying Synaptic and Axon Degeneration in Parkinson's Disease. Front Cell Neurosci 2021; 15:626128. [PMID: 33737866 PMCID: PMC7960781 DOI: 10.3389/fncel.2021.626128] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 02/05/2021] [Indexed: 01/13/2023] Open
Abstract
Parkinson’s disease (PD) is a progressive neurodegenerative disease that impairs movement as well as causing multiple other symptoms such as autonomic dysfunction, rapid eye movement (REM) sleep behavior disorder, hyposmia, and cognitive changes. Loss of dopamine neurons in the substantia nigra pars compacta (SNc) and loss of dopamine terminals in the striatum contribute to characteristic motor features. Although therapies ease the symptoms of PD, there are no treatments to slow its progression. Accumulating evidence suggests that synaptic impairments and axonal degeneration precede neuronal cell body loss. Early synaptic changes may be a target to prevent disease onset and slow progression. Imaging of PD patients with radioligands, post-mortem pathologic studies in sporadic PD patients, and animal models of PD demonstrate abnormalities in presynaptic terminals as well as postsynaptic dendritic spines. Dopaminergic and excitatory synapses are substantially reduced in PD, and whether other neuronal subtypes show synaptic defects remains relatively unexplored. Genetic studies implicate several genes that play a role at the synapse, providing additional support for synaptic dysfunction in PD. In this review article we: (1) provide evidence for synaptic defects occurring in PD before neuron death; (2) describe the main genes implicated in PD that could contribute to synapse dysfunction; and (3) show correlations between the expression of Snca mRNA and mouse homologs of PD GWAS genes demonstrating selective enrichment of Snca and synaptic genes in dopaminergic, excitatory and cholinergic neurons. Altogether, these findings highlight the need for novel therapeutics targeting the synapse and suggest that future studies should explore the roles for PD-implicated genes across multiple neuron types and circuits.
Collapse
Affiliation(s)
- Nolwazi Z Gcwensa
- Department of Neurobiology, Center for Neurodegeneration and Experimental Therapeutics, Civitan International Research Center, Birmingham, AL, United States
| | - Drèson L Russell
- Department of Neurobiology, Center for Neurodegeneration and Experimental Therapeutics, Civitan International Research Center, Birmingham, AL, United States
| | - Rita M Cowell
- Department of Neuroscience, Southern Research, Birmingham, AL, United States
| | - Laura A Volpicelli-Daley
- Department of Neurobiology, Center for Neurodegeneration and Experimental Therapeutics, Civitan International Research Center, Birmingham, AL, United States
| |
Collapse
|