1
|
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
|
2
|
Borland H, Rasmussen I, Bjerregaard-Andersen K, Rasmussen M, Olsen A, Vilhardt F. α-synuclein build-up is alleviated via ESCRT-dependent endosomal degradation brought about by p38MAPK inhibition in cells expressing p25α. J Biol Chem 2022; 298:102531. [PMID: 36162505 PMCID: PMC9637583 DOI: 10.1016/j.jbc.2022.102531] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 08/24/2022] [Accepted: 09/04/2022] [Indexed: 11/24/2022] Open
Abstract
α-synucleinopathy is driven by an imbalance of synthesis and degradation of α-synuclein (αSyn), causing a build up of αSyn aggregates and post-translationally modified species, which not only interfere with normal cellular metabolism but also by their secretion propagates the disease. Therefore, a better understanding of αSyn degradation pathways is needed to address α-synucleinopathy. Here, we used the nerve growth factor–differentiated catecholaminergic PC12 neuronal cell line, which was conferred α-synucleinopathy by inducible expression of αSyn and tubulin polymerization-promoting protein p25α. p25α aggregates αSyn, and imposes a partial autophagosome–lysosome block to mimic aspects of lysosomal deficiency common in neurodegenerative disease. Under basal conditions, αSyn was degraded by multiple pathways but most prominently by macroautophagy and Nedd4/Ndfip1-mediated degradation. We found that expression of p25α induced strong p38MAPK activity. Remarkably, when opposed by inhibitor SB203580 or p38MAPK shRNA knockdown, endolysosomal localization and degradation of αSyn increased, and αSyn secretion and cytotoxicity decreased. This effect was specifically dependent on Hsc70 and the endosomal sorting complex required for transport machinery, but different from classical microautophagy, as the αSyn Hsc70 binding motif was unnecessary. Furthermore, in a primary neuronal (h)-αSyn seeding model, p38MAPK inhibition decreased pathological accumulation of phosphorylated serine-129-αSyn and cytotoxicity. In conclusion, p38MAPK inhibition shifts αSyn degradation from various forms of autophagy to an endosomal sorting complex required for transport–dependent uptake mechanism, resulting in increased αSyn turnover and cell viability in p25α-expressing cells. More generally, our results suggest that under conditions of autophagolysosomal malfunction, the uninterrupted endosomal pathway offers a possibility to achieve disease-associated protein degradation.
Collapse
Affiliation(s)
- Helena Borland
- Dept. of Cellular and Molecular Medicine, The Panum Institute, The Faculty of Health Sciences, University of Copenhagen, Copenhagen 2200N, Denmark; Dept. of Cell Biology, H. Lundbeck A/S, 2500 Valby, Denmark.
| | - Izabela Rasmussen
- Dept. of Cellular and Molecular Medicine, The Panum Institute, The Faculty of Health Sciences, University of Copenhagen, Copenhagen 2200N, Denmark.
| | | | - Michel Rasmussen
- Dept. of Cellular and Molecular Medicine, The Panum Institute, The Faculty of Health Sciences, University of Copenhagen, Copenhagen 2200N, Denmark.
| | - Anders Olsen
- Dept. of Chemistry and Bioscience, The Faculty of Engineering and Science, University of Aalborg, Fredrik Bajers Vej 7H, 9220 Aalborg, Denmark.
| | - Frederik Vilhardt
- Dept. of Cellular and Molecular Medicine, The Panum Institute, The Faculty of Health Sciences, University of Copenhagen, Copenhagen 2200N, Denmark.
| |
Collapse
|
3
|
Ahuja M, Kaidery NA, Dutta D, Attucks OC, Kazakov EH, Gazaryan I, Matsumoto M, Igarashi K, Sharma SM, Thomas B. Harnessing the Therapeutic Potential of the Nrf2/Bach1 Signaling Pathway in Parkinson's Disease. Antioxidants (Basel) 2022; 11:antiox11091780. [PMID: 36139853 PMCID: PMC9495572 DOI: 10.3390/antiox11091780] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 09/03/2022] [Accepted: 09/05/2022] [Indexed: 11/16/2022] Open
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative movement disorder characterized by a progressive loss of dopaminergic neurons in the substantia nigra pars compacta. Although a complex interplay of multiple environmental and genetic factors has been implicated, the etiology of neuronal death in PD remains unresolved. Various mechanisms of neuronal degeneration in PD have been proposed, including oxidative stress, mitochondrial dysfunction, neuroinflammation, α-synuclein proteostasis, disruption of calcium homeostasis, and other cell death pathways. While many drugs individually targeting these pathways have shown promise in preclinical PD models, this promise has not yet translated into neuroprotective therapies in human PD. This has consequently spurred efforts to identify alternative targets with multipronged therapeutic approaches. A promising therapeutic target that could modulate multiple etiological pathways involves drug-induced activation of a coordinated genetic program regulated by the transcription factor, nuclear factor E2-related factor 2 (Nrf2). Nrf2 regulates the transcription of over 250 genes, creating a multifaceted network that integrates cellular activities by expressing cytoprotective genes, promoting the resolution of inflammation, restoring redox and protein homeostasis, stimulating energy metabolism, and facilitating repair. However, FDA-approved electrophilic Nrf2 activators cause irreversible alkylation of cysteine residues in various cellular proteins resulting in side effects. We propose that the transcriptional repressor of BTB and CNC homology 1 (Bach1), which antagonizes Nrf2, could serve as a promising complementary target for the activation of both Nrf2-dependent and Nrf2-independent neuroprotective pathways. This review presents the current knowledge on the Nrf2/Bach1 signaling pathway, its role in various cellular processes, and the benefits of simultaneously inhibiting Bach1 and stabilizing Nrf2 using non-electrophilic small molecules as a novel therapeutic approach for PD.
Collapse
Affiliation(s)
- Manuj Ahuja
- Darby Children’s Research Institute, Medical University of South Carolina, Charleston, SC 29406, USA
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC 29406, USA
| | - Navneet Ammal Kaidery
- Darby Children’s Research Institute, Medical University of South Carolina, Charleston, SC 29406, USA
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC 29406, USA
| | - Debashis Dutta
- Darby Children’s Research Institute, Medical University of South Carolina, Charleston, SC 29406, USA
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC 29406, USA
| | | | | | - Irina Gazaryan
- Pace University, White Plains, NY 10601, USA
- Department of Chemical Enzymology, School of Chemistry, M.V. Lomonosov Moscow State University, 111401 Moscow, Russia
- Faculty of Biology and Biotechnologies, Higher School of Economics, 111401 Moscow, Russia
| | - Mitsuyo Matsumoto
- Department of Biochemistry, Graduate School of Medicine, Tohoku University, Sendai 980-8576, Japan
| | - Kazuhiko Igarashi
- Department of Biochemistry, Graduate School of Medicine, Tohoku University, Sendai 980-8576, Japan
| | - Sudarshana M. Sharma
- Department of Biochemistry & Molecular Biology, Medical University of South Carolina, Charleston, SC 29406, USA
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29406, USA
| | - Bobby Thomas
- Darby Children’s Research Institute, Medical University of South Carolina, Charleston, SC 29406, USA
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC 29406, USA
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29406, USA
- Department of Drug Discovery, Medical University of South Carolina, Charleston, SC 29406, USA
- Correspondence:
| |
Collapse
|
4
|
Reimer L, Haikal C, Gram H, Theologidis V, Kovacs G, Ruesink H, Baun A, Nielsen J, Otzen DE, Li JY, Jensen PH. Low dose DMSO treatment induces oligomerization and accelerates aggregation of α-synuclein. Sci Rep 2022; 12:3737. [PMID: 35260646 PMCID: PMC8904838 DOI: 10.1038/s41598-022-07706-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 02/10/2022] [Indexed: 01/05/2023] Open
Abstract
Dimethyl sulfoxide (DMSO) is a highly utilized small molecule that serves many purposes in scientific research. DMSO offers unique polar, aprotic and amphiphilic features, which makes it an ideal solvent for a wide variety of both polar and nonpolar molecules. Furthermore, DMSO is often used as a cryoprotectant in cell-based research. However, recent reports suggest that DMSO, even at low concentration, might interfere with important cellular processes, and cause macromolecular changes to proteins where a shift from α-helical to β-sheet structure can be observed. To investigate how DMSO might influence current research, we assessed biochemical and cellular impacts of DMSO treatment on the structure of the aggregation-prone protein α-synuclein, which plays a central role in the etiology of Parkinson’s disease, and other brain-related disorders, collectively termed the synucleinopathies. Here, we found that addition of DMSO increased the particle-size of α-synuclein, and accelerated the formation of seeding-potent fibrils in a dose-dependent manner. These fibrils made in the presence of DMSO were indistinguishable from fibrils made in pure PBS, when assessed by proteolytic digestion, cytotoxic profile and their ability to seed cellular aggregation of α-synuclein. Moreover, as evident through binding to the MJFR-14-6-4-2 antibody, which preferentially recognizes aggregated forms of α-synuclein, and a bimolecular fluorescence complementation assay, cells exposed to DMSO experienced increased aggregation of α-synuclein. However, no observable α-synuclein abnormalities nor differences in neuronal survival were detected after oral DMSO-treatment in either C57BL/6- or α-synuclein transgenic F28 mice. In summary, we demonstrate that low concentrations of DMSO makes α-synuclein susceptible to undergo aggregation both in vitro and in cells. This may affect experimental outcomes when studying α-synuclein in the presence of DMSO, and should call for careful consideration when such experiments are planned.
Collapse
Affiliation(s)
- Lasse Reimer
- Danish Research Institute of Translational Neuroscience - DANDRITE, Aarhus University, Aarhus C, Denmark. .,Department of Biomedicine, Aarhus University, Aarhus C, Denmark.
| | - Caroline Haikal
- Neural Plasticity and Repair Unit, Wallenberg Neuroscience Center, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Hjalte Gram
- Danish Research Institute of Translational Neuroscience - DANDRITE, Aarhus University, Aarhus C, Denmark.,Department of Biomedicine, Aarhus University, Aarhus C, Denmark
| | - Vasileios Theologidis
- Danish Research Institute of Translational Neuroscience - DANDRITE, Aarhus University, Aarhus C, Denmark.,Department of Biomedicine, Aarhus University, Aarhus C, Denmark
| | - Gergo Kovacs
- Danish Research Institute of Translational Neuroscience - DANDRITE, Aarhus University, Aarhus C, Denmark.,Department of Biomedicine, Aarhus University, Aarhus C, Denmark
| | - Harm Ruesink
- Danish Research Institute of Translational Neuroscience - DANDRITE, Aarhus University, Aarhus C, Denmark.,Department of Biomedicine, Aarhus University, Aarhus C, Denmark
| | - Andreas Baun
- Danish Research Institute of Translational Neuroscience - DANDRITE, Aarhus University, Aarhus C, Denmark.,Department of Biomedicine, Aarhus University, Aarhus C, Denmark
| | - Janni Nielsen
- Interdisciplinary Nanoscience Center - iNANO, Aarhus University, Aarhus C, Denmark
| | - Daniel Erik Otzen
- Interdisciplinary Nanoscience Center - iNANO, Aarhus University, Aarhus C, Denmark
| | - Jia-Yi Li
- Neural Plasticity and Repair Unit, Wallenberg Neuroscience Center, Department of Experimental Medical Science, Lund University, Lund, Sweden.,Institute of Health Sciences, China Medical University, 110112, Shenyang, People's Republic of China
| | - Poul Henning Jensen
- Danish Research Institute of Translational Neuroscience - DANDRITE, Aarhus University, Aarhus C, Denmark.,Department of Biomedicine, Aarhus University, Aarhus C, Denmark
| |
Collapse
|
5
|
Pathogenic LRRK2 requires secondary factors to induce cellular toxicity. Biosci Rep 2021; 40:226517. [PMID: 32975566 PMCID: PMC7560525 DOI: 10.1042/bsr20202225] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 09/08/2020] [Accepted: 09/24/2020] [Indexed: 12/02/2022] Open
Abstract
Pathogenic mutations in the leucine-rich repeat kinase 2 (LRRK2) gene belong to the most common genetic causes of inherited Parkinson’s disease (PD) and variations in its locus increase the risk to develop sporadic PD. Extensive research efforts aimed at understanding how changes in the LRRK2 function result in molecular alterations that ultimately lead to PD. Cellular LRRK2-based models revealed several potential pathophysiological mechanisms including apoptotic cell death, LRRK2 protein accumulation and deficits in neurite outgrowth. However, highly variable outcomes between different cellular models have been reported. Here, we have investigated the effect of different experimental conditions, such as the use of different tags and gene transfer methods, in various cellular LRRK2 models. Readouts included cell death, sensitivity to oxidative stress, LRRK2 relocalization, α-synuclein aggregation and neurite outgrowth in cell culture, as well as neurite maintenance in vivo. We show that overexpression levels and/or the tag fused to LRRK2 affect the relocalization of LRRK2 to filamentous and skein-like structures. We found that overexpression of LRRK2 per se is not sufficient to induce cellular toxicity or to affect α-synuclein-induced toxicity and aggregate formation. Finally, neurite outgrowth/retraction experiments in cell lines and in vivo revealed that secondary, yet unknown, factors are required for the pathogenic LRRK2 effects on neurite length. Our findings stress the importance of technical and biological factors in LRRK2-induced cellular phenotypes and hence imply that conclusions based on these types of LRRK2-based assays should be interpreted with caution.
Collapse
|
6
|
Wallings RL, Herrick MK, Tansey MG. LRRK2 at the Interface Between Peripheral and Central Immune Function in Parkinson's. Front Neurosci 2020; 14:443. [PMID: 32508566 PMCID: PMC7253584 DOI: 10.3389/fnins.2020.00443] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Accepted: 04/09/2020] [Indexed: 12/20/2022] Open
Abstract
It is becoming increasingly accepted that there is an interplay between the peripheral immune response and neuroinflammation in the pathophysiology of Parkinson's disease (PD). Mutations in the leucine-rich-repeat kinase 2 (LRRK2) gene are associated with familial and sporadic cases of PD but are also found in immune-related disorders, such as inflammatory bowel disease (IBD) and leprosy. Furthermore, LRRK2 has been associated with bacterial infections such as Mycobacterium tuberculosis and Salmonella typhimurium. Recent evidence suggests a role of LRRK2 in the regulation of the immune system and modulation of inflammatory responses, at a systemic level, with LRRK2 functionally implicated in both the immune system of the central nervous system (CNS) and the periphery. It has therefore been suggested that peripheral immune signaling may play an important role in the regulation of neurodegeneration in LRRK2 as well as non-LRRK2-associated PD. This review will discuss the current evidence for this hypothesis and will provide compelling rationale for placing LRRK2 at the interface between peripheral immune responses and neuroinflammation.
Collapse
Affiliation(s)
- Rebecca L. Wallings
- Department of Neuroscience and Center for Translational Research in Neurodegenerative Disease, University of Florida College of Medicine, Gainesville, FL, United States
| | - Mary K. Herrick
- Department of Neuroscience and Center for Translational Research in Neurodegenerative Disease, University of Florida College of Medicine, Gainesville, FL, United States
- Laney Graduate School, Emory University, Atlanta, GA, United States
| | - Malú Gámez Tansey
- Department of Neuroscience and Center for Translational Research in Neurodegenerative Disease, University of Florida College of Medicine, Gainesville, FL, United States
| |
Collapse
|
7
|
Labonne JDJ, Driessen TM, Harris ME, Kong IK, Brakta S, Theisen J, Sangare M, Layman LC, Kim CH, Lim J, Kim HG. Comparative Genomic Mapping Implicates LRRK2 for Intellectual Disability and Autism at 12q12, and HDHD1, as Well as PNPLA4, for X-Linked Intellectual Disability at Xp22.31. J Clin Med 2020; 9:jcm9010274. [PMID: 31963867 PMCID: PMC7019335 DOI: 10.3390/jcm9010274] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 11/28/2019] [Accepted: 12/06/2019] [Indexed: 01/01/2023] Open
Abstract
We report a genomic and phenotypic delineation for two chromosome regions with candidate genes for syndromic intellectual disability at 12q12 and Xp22.31, segregating independently in one family with four affected members. Fine mapping of three affected members, along with six unreported small informative CNVs, narrowed down the candidate chromosomal interval to one gene LRRK2 at 12q12. Expression studies revealed high levels of LRRK2 transcripts in the whole human brain, cerebral cortex and hippocampus. RT-qPCR assays revealed that LRRK2 transcripts were dramatically reduced in our microdeletion patient DGDP289A compared to his healthy grandfather with no deletion. The decreased expression of LRRK2 may affect protein–protein interactions between LRRK2 and its binding partners, of which eight have previously been linked to intellectual disability. These findings corroborate with a role for LRRK2 in cognitive development, and, thus, we propose that intellectual disability and autism, displayed in the 12q12 microdeletions, are likely caused by LRRK2. Using another affected member, DGDP289B, with a microdeletion at Xp22.31, in this family, we performed the genomic and clinical delineation with six published and nine unreported cases. We propose HDHD1 and PNPLA4 for X-linked intellectual disability in this region, since their high transcript levels in the human brain substantiate their role in intellectual functioning.
Collapse
Affiliation(s)
- Jonathan D. J. Labonne
- Section of Reproductive Endocrinology, Infertility & Genetics, Department of Obstetrics & Gynecology, Augusta University, Augusta, GA 30912, USA (M.E.H.); (S.B.); (J.T.); (L.C.L.)
| | - Terri M. Driessen
- Department of Genetics, Yale University, New Haven, CT 06510, USA; (T.M.D.); (J.L.)
| | - Marvin E. Harris
- Section of Reproductive Endocrinology, Infertility & Genetics, Department of Obstetrics & Gynecology, Augusta University, Augusta, GA 30912, USA (M.E.H.); (S.B.); (J.T.); (L.C.L.)
| | - Il-Keun Kong
- Department of Animal Science, Division of Applied Life Science (BK21plus), Institute of Agriculture and Life Science, Gyeongsang National University, Jinju 52828, Korea;
| | - Soumia Brakta
- Section of Reproductive Endocrinology, Infertility & Genetics, Department of Obstetrics & Gynecology, Augusta University, Augusta, GA 30912, USA (M.E.H.); (S.B.); (J.T.); (L.C.L.)
| | - John Theisen
- Section of Reproductive Endocrinology, Infertility & Genetics, Department of Obstetrics & Gynecology, Augusta University, Augusta, GA 30912, USA (M.E.H.); (S.B.); (J.T.); (L.C.L.)
| | - Modibo Sangare
- Faculty of Medicine and Odontostomatology (FMOS), University of Sciences, Techniques and Technologies of Bamako (USTTB), Bamako, Mali;
| | - Lawrence C. Layman
- Section of Reproductive Endocrinology, Infertility & Genetics, Department of Obstetrics & Gynecology, Augusta University, Augusta, GA 30912, USA (M.E.H.); (S.B.); (J.T.); (L.C.L.)
- Department of Neuroscience and Regenerative Medicine, Augusta University, Augusta, GA 30912, USA
| | - Cheol-Hee Kim
- Department of Biology, Chungnam National University, Daejeon 34134, Korea;
| | - Janghoo Lim
- Department of Genetics, Yale University, New Haven, CT 06510, USA; (T.M.D.); (J.L.)
- Department of Neuroscience, Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale Stem Cell Center, Yale University, New Haven, CT 06510, USA
| | - Hyung-Goo Kim
- Section of Reproductive Endocrinology, Infertility & Genetics, Department of Obstetrics & Gynecology, Augusta University, Augusta, GA 30912, USA (M.E.H.); (S.B.); (J.T.); (L.C.L.)
- Neurological Disorders Research Center, Qatar Biomedical Research Institute, Hamad Bin Khalifa University, Doha P.O. Box 34110, Qatar
- Correspondence:
| |
Collapse
|
8
|
LRRK2, alpha-synuclein, and tau: partners in crime or unfortunate bystanders? Biochem Soc Trans 2019; 47:827-838. [PMID: 31085616 DOI: 10.1042/bst20180466] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 04/18/2019] [Accepted: 04/23/2019] [Indexed: 12/11/2022]
Abstract
The identification of genetic forms of Parkinson's disease (PD) has tremendously expanded our understanding of the players and mechanisms involved. Mutations in the genes encoding for alpha-synuclein (aSyn), LRRK2, and tau have been associated with familial and sporadic forms of the disease. aSyn is the major component of Lewy bodies and Lewy neurites, which are pathognomonic protein inclusions in PD. Hyperphosphorylated tau protein accumulates in neurofibrillary tangles in the brains of Alzheimer's disease patients but is also seen in the brains of PD patients. LRRK2 is a complex multi-domain protein with kinase and GTPase enzymatic activity. Since aSyn and tau are phosphoproteins, we review the possible interplay between the three proteins. Understanding the interplay between LRRK2, aSyn and tau is extremely important, as this may enable the identification of novel targets and pathways for therapeutic intervention.
Collapse
|
9
|
Kluss JH, Mamais A, Cookson MR. LRRK2 links genetic and sporadic Parkinson's disease. Biochem Soc Trans 2019; 47:651-661. [PMID: 30837320 PMCID: PMC6563926 DOI: 10.1042/bst20180462] [Citation(s) in RCA: 134] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 02/05/2019] [Accepted: 02/07/2019] [Indexed: 12/21/2022]
Abstract
The past two decades in research has revealed the importance of leucine-rich repeat kinase 2 (LRRK2) in both monogenic and sporadic forms of Parkinson's disease (PD). In families, mutations in LRRK2 can cause PD with age-dependent but variable penetrance and genome-wide association studies have found variants of the gene that are risk factors for sporadic PD. Functional studies have suggested that the common mechanism that links all disease-associated variants is that they increase LRRK2 kinase activity, albeit in different ways. Here, we will discuss the roles of LRRK2 in areas of inflammation and vesicular trafficking in the context of monogenic and sporadic PD. We will also provide a hypothetical model that links inflammation and vesicular trafficking together in an effort to outline how these pathways might interact and eventually lead to neuronal cell death. We will also highlight the translational potential of LRRK2-specific kinase inhibitors for the treatment of PD.
Collapse
Affiliation(s)
- Jillian H Kluss
- Cell Biology and Gene Expression Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bldg. 35, 35 Convent Drive, Bethesda, MD 20892-3707, U.S.A
| | - Adamantios Mamais
- Cell Biology and Gene Expression Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bldg. 35, 35 Convent Drive, Bethesda, MD 20892-3707, U.S.A
| | - Mark R Cookson
- Cell Biology and Gene Expression Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bldg. 35, 35 Convent Drive, Bethesda, MD 20892-3707, U.S.A.
| |
Collapse
|
10
|
Benson DL, Huntley GW. Are we listening to everything the PARK genes are telling us? J Comp Neurol 2019; 527:1527-1540. [PMID: 30680728 DOI: 10.1002/cne.24642] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 12/30/2018] [Accepted: 01/02/2019] [Indexed: 12/17/2022]
Abstract
The cardinal motor symptoms that define Parkinson's disease (PD) clinically have been recognized for over 200 years. That these symptoms arise following the loss of dopamine neurons in the substantia nigra has been known for the last 50. These long-established facts have fueled a broadly held expectation that degenerating dopaminergic neurons alone hold the key to understanding and curing PD. This prevalent expectation is at odds with the observation that many nonmotor symptoms, including depression and cognitive inflexibility among others, can appear years earlier than the overt dopaminergic neuron degeneration that drives motor abnormalities and are not improved by levodopa treatment. Thus, preserving or rescuing dopamine neuron health and function is of paramount importance, but this alone fails to capture the underlying neurobiology of earlier-appearing nonmotor symptoms. Insight into the complete landscape of disease-related abnormalities and the context in which they arise can be gleaned from a more comprehensive consideration of the PARK genes that are known to cause PD. Here, we make the case that a full incorporation of research showing when and where PARK genes are expressed as well as the impact of gene mutation on function throughout life, in tandem with research studying how dopaminergic neuron degeneration begins, is essential for a full understanding of the multi-dimensional etiology of PD. A broad view may also reveal something about long-term adjustments cells and systems make in response to gene mutation and help to identify mechanisms conferring the resilience or susceptibility of some cells and systems over others.
Collapse
Affiliation(s)
- Deanna L Benson
- Department of Neuroscience, Friedman Brain Institute and the Graduate School of Biomedical Sciences Icahn School of Medicine at Mount Sinai, New York, New York
| | - George W Huntley
- Department of Neuroscience, Friedman Brain Institute and the Graduate School of Biomedical Sciences Icahn School of Medicine at Mount Sinai, New York, New York
| |
Collapse
|
11
|
Zhao HT, John N, Delic V, Ikeda-Lee K, Kim A, Weihofen A, Swayze EE, Kordasiewicz HB, West AB, Volpicelli-Daley LA. LRRK2 Antisense Oligonucleotides Ameliorate α-Synuclein Inclusion Formation in a Parkinson's Disease Mouse Model. MOLECULAR THERAPY. NUCLEIC ACIDS 2017; 8:508-519. [PMID: 28918051 PMCID: PMC5573879 DOI: 10.1016/j.omtn.2017.08.002] [Citation(s) in RCA: 148] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Revised: 08/07/2017] [Accepted: 08/07/2017] [Indexed: 01/30/2023]
Abstract
No treatments exist to slow or halt Parkinson's disease (PD) progression; however, inhibition of leucine-rich repeat kinase 2 (LRRK2) activity represents one of the most promising therapeutic strategies. Genetic ablation and pharmacological LRRK2 inhibition have demonstrated promise in blocking α-synuclein (α-syn) pathology. However, LRRK2 kinase inhibitors may reduce LRRK2 activity in several tissues and induce systemic phenotypes in the kidney and lung that are undesirable. Here, we test whether antisense oligonucleotides (ASOs) provide an alternative therapeutic strategy, as they can be restricted to the CNS and provide a stable, long-lasting reduction of protein throughout the brain. Administration of LRRK2 ASOs to the brain reduces LRRK2 protein levels and fibril-induced α-syn inclusions. Mice exposed to α-syn fibrils treated with LRRK2 ASOs show more tyrosine hydroxylase (TH)-positive neurons compared to control mice. Furthermore, intracerebral injection of LRRK2 ASOs avoids unwanted phenotypes associated with loss of LRRK2 expression in the periphery. This study further demonstrates that a reduction of endogenous levels of normal LRRK2 reduces the formation of α-syn inclusions. Importantly, this study points toward LRRK2 ASOs as a potential therapeutic strategy for preventing PD-associated pathology and phenotypes without causing potential adverse side effects in peripheral tissues associated with LRRK2 inhibition.
Collapse
Affiliation(s)
| | - Neena John
- Center for Neurodegeneration and Experimental Therapeutics, University of Alabama, Birmingham, AL 35294, USA
| | - Vedad Delic
- Center for Neurodegeneration and Experimental Therapeutics, University of Alabama, Birmingham, AL 35294, USA
| | | | - Aneeza Kim
- Ionis Pharmaceuticals, Inc., Carlsbad, CA 92010, USA
| | | | - Eric E Swayze
- Ionis Pharmaceuticals, Inc., Carlsbad, CA 92010, USA
| | | | - Andrew B West
- Center for Neurodegeneration and Experimental Therapeutics, University of Alabama, Birmingham, AL 35294, USA
| | - Laura A Volpicelli-Daley
- Center for Neurodegeneration and Experimental Therapeutics, University of Alabama, Birmingham, AL 35294, USA.
| |
Collapse
|
12
|
Animal models of α-synucleinopathy for Parkinson disease drug development. Nat Rev Neurosci 2017; 18:515-529. [DOI: 10.1038/nrn.2017.75] [Citation(s) in RCA: 126] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
13
|
Andersson R, Galter D, Papadia D, Fisahn A. Histamine induces KCNQ channel-dependent gamma oscillations in rat hippocampus via activation of the H1 receptor. Neuropharmacology 2017; 118:13-25. [PMID: 28274820 DOI: 10.1016/j.neuropharm.2017.03.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Revised: 03/01/2017] [Accepted: 03/02/2017] [Indexed: 11/30/2022]
Abstract
Histamine is an aminergic neurotransmitter, which regulates wakefulness, arousal and attention in the central nervous system. Histamine receptors have been the target of efforts to develop pro-cognitive drugs to treat disorders such as Alzheimer's disease and schizophrenia. Cognitive functions including attention are closely associated with gamma oscillations, a rhythmical electrical activity pattern in the 30-80 Hz range, which depends on the synchronized activity of excitatory pyramidal cells and inhibitory fast-spiking interneurons. We set out to explore whether histamine has a role in promoting gamma oscillations in the hippocampus. Using in-situ hybridization we demonstrate that histamine receptor subtypes 1, 2 and 3 are expressed in stratum pyramidale of area CA3 in rats. We show that both pyramidal cells and fast-spiking interneurons depolarize and increase action potential firing in response to histamine in vitro. The activation of histamine receptors generates dose-dependent, transient gamma oscillations in area CA3 of the hippocampus - the locus of the gamma rhythm generator. We also demonstrate that this histamine effect is independent of muscarinic receptors. Using specific antagonists we provide evidence that histamine gamma rhythmogenesis specifically depends on the H1 receptor. Histamine also depolarized both pyramidal cells and fast-spiking interneurons and increased membrane resistance in pyramidal cells. The increased membrane resistance is potentially mediated by the inhibition of potassium channels because application of the KCNQ channel opener ICA110381 abolished the oscillations. Taken together our data demonstrate a novel and physiological mechanism for generating gamma oscillations in hippocampus and suggest a role for KCNQ channels in this cognition-relevant brain activity.
Collapse
Affiliation(s)
- Richard Andersson
- Neuronal Oscillations Laboratory, Neurogeriatrics Division, Center for Alzheimer Research, Dept. of Neurobiology, Care Sciences and Society, Karolinska Institutet, 14186 Stockholm, Sweden
| | - Dagmar Galter
- Dept. of Neuroscience, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Daniela Papadia
- Neuronal Oscillations Laboratory, Neurogeriatrics Division, Center for Alzheimer Research, Dept. of Neurobiology, Care Sciences and Society, Karolinska Institutet, 14186 Stockholm, Sweden
| | - André Fisahn
- Neuronal Oscillations Laboratory, Neurogeriatrics Division, Center for Alzheimer Research, Dept. of Neurobiology, Care Sciences and Society, Karolinska Institutet, 14186 Stockholm, Sweden.
| |
Collapse
|
14
|
Chumarina M, Azevedo C, Bigarreau J, Vignon C, Kim KS, Li JY, Roybon L. Derivation of mouse embryonic stem cell lines from tyrosine hydroxylase reporter mice crossed with a human SNCA transgenic mouse model of Parkinson's disease. Stem Cell Res 2016; 19:17-20. [PMID: 28413000 PMCID: PMC5626016 DOI: 10.1016/j.scr.2016.12.026] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2016] [Accepted: 12/16/2016] [Indexed: 10/25/2022] Open
Abstract
Mouse embryonic stem cell (mESC) lines were derived by crossing heterozygous transgenic (tg) mice expressing green fluorescent protein (GFP) under the control of the rat tyrosine hydroxylase (TH) promoter, with homozygous alpha-synuclein (aSYN) mice expressing human mutant SNCAA53T under the control of the mouse Prion promoter (MoPrP), or wildtype (WT) mice. The expression of GFP and human aSYN was validated by immunocytochemistry in midbrain neuron cultures upon differentiation of mESC lines using stromal cell-derived inducing activity. These mESC lines can help to study the impact of human aSYN expression in neurons and oligodendrocytes, and also trace GFP-expressing midbrain neurons.
Collapse
Affiliation(s)
- Margarita Chumarina
- Cell Stem Cell Laboratory for CNS Disease Modeling, Department of Experimental Medical Science, Lund University, 22184 Lund, Sweden; MultiPark and Lund Stem Cell Center, Lund University, 22184 Lund, Sweden
| | - Carla Azevedo
- Cell Stem Cell Laboratory for CNS Disease Modeling, Department of Experimental Medical Science, Lund University, 22184 Lund, Sweden; MultiPark and Lund Stem Cell Center, Lund University, 22184 Lund, Sweden
| | - Julie Bigarreau
- Cell Stem Cell Laboratory for CNS Disease Modeling, Department of Experimental Medical Science, Lund University, 22184 Lund, Sweden
| | - Clémentine Vignon
- Cell Stem Cell Laboratory for CNS Disease Modeling, Department of Experimental Medical Science, Lund University, 22184 Lund, Sweden
| | - Kwang-Soo Kim
- Molecular Neurobiology Laboratory, Department of Psychiatry and Program in Neuroscience, McLean Hospital/Harvard Medical School, Belmont, MA 02478, USA; Harvard Stem Cell Institute, McLean Hospital/Harvard Medical School, Belmont, MA 02478, USA
| | - Jia-Yi Li
- Neural Plasticity and Repair Unit, Department of Experimental Medical Science, Wallenberg Neuroscience Center, Lund University, Lund, Sweden; Neuroscience Institute, College of Life and Health Sciences, Northeastern University, Shenyang, 110015, PR China
| | - Laurent Roybon
- Cell Stem Cell Laboratory for CNS Disease Modeling, Department of Experimental Medical Science, Lund University, 22184 Lund, Sweden; MultiPark and Lund Stem Cell Center, Lund University, 22184 Lund, Sweden.
| |
Collapse
|
15
|
Matrone C, Dzamko N, Madsen P, Nyegaard M, Pohlmann R, Søndergaard RV, Lassen LB, Andresen TL, Halliday GM, Jensen PH, Nielsen MS. Mannose 6-Phosphate Receptor Is Reduced in -Synuclein Overexpressing Models of Parkinsons Disease. PLoS One 2016; 11:e0160501. [PMID: 27509067 PMCID: PMC4979956 DOI: 10.1371/journal.pone.0160501] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Accepted: 07/20/2016] [Indexed: 12/03/2022] Open
Abstract
Increasing evidence points to defects in autophagy as a common denominator in most neurodegenerative conditions. Progressive functional decline in the autophagy-lysosomal pathway (ALP) occurs with age, and the consequent impairment in protein processing capacity has been associated with a higher risk of neurodegeneration. Defects in cathepsin D (CD) processing and α-synuclein degradation causing its accumulation in lysosomes are particularly relevant for the development of Parkinson's disease (PD). However, the mechanism by which alterations in CD maturation and α-synuclein degradation leads to autophagy defects in PD neurons is still uncertain. Here we demonstrate that MPR300 shuttling between endosomes and the trans Golgi network is altered in α-synuclein overexpressing neurons. Consequently, CD is not correctly trafficked to lysosomes and cannot be processed to generate its mature active form, leading to a reduced CD-mediated α-synuclein degradation and α-synuclein accumulation in neurons. MPR300 is downregulated in brain from α-synuclein overexpressing animal models and in PD patients with early diagnosis. These data indicate MPR300 as crucial player in the autophagy-lysosomal dysfunctions reported in PD and pinpoint MRP300 as a potential biomarker for PD.
Collapse
Affiliation(s)
- Carmela Matrone
- Department of Biomedicine, Faculty of Health, Aarhus University, 8000 Aarhus C, Denmark
- * E-mail: ;
| | - Nicolas Dzamko
- Neuroscience Research Australia, Sydney, NSW 2031, and School of Medical Sciences, Faculty of Medicine, The University of New South Wales, Sydney, NSW 2052, Australia
| | - Peder Madsen
- Department of Biomedicine, Faculty of Health, Aarhus University, 8000 Aarhus C, Denmark
- Research Initiative on Blood Brain and Drug Delivery, The Lundbeck Foundation, 8000 Aarhus C, Denmark
| | - Mette Nyegaard
- Department of Biomedicine, Faculty of Health, Aarhus University, 8000 Aarhus C, Denmark
| | - Regina Pohlmann
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, 48149 Münster, Germany
| | - Rikke V. Søndergaard
- Research Initiative on Blood Brain and Drug Delivery, The Lundbeck Foundation, 8000 Aarhus C, Denmark
- Department of Micro- and Nanotechnology, Technical University of Denmark (DTU) Nanotech, DTU, 2800 Lyngby, Denmark
| | - Louise B. Lassen
- Department of Biomedicine, Faculty of Health, Aarhus University, 8000 Aarhus C, Denmark
| | - Thomas L. Andresen
- Research Initiative on Blood Brain and Drug Delivery, The Lundbeck Foundation, 8000 Aarhus C, Denmark
- Department of Micro- and Nanotechnology, Technical University of Denmark (DTU) Nanotech, DTU, 2800 Lyngby, Denmark
| | - Glenda M. Halliday
- Neuroscience Research Australia, Sydney, NSW 2031, and School of Medical Sciences, Faculty of Medicine, The University of New South Wales, Sydney, NSW 2052, Australia
| | - Poul Henning Jensen
- Department of Biomedicine, Faculty of Health, Aarhus University, 8000 Aarhus C, Denmark
| | - Morten S. Nielsen
- Department of Biomedicine, Faculty of Health, Aarhus University, 8000 Aarhus C, Denmark
- Research Initiative on Blood Brain and Drug Delivery, The Lundbeck Foundation, 8000 Aarhus C, Denmark
- * E-mail: ;
| |
Collapse
|
16
|
5-HT2A Receptor Binding in the Frontal Cortex of Parkinson's Disease Patients and Alpha-Synuclein Overexpressing Mice: A Postmortem Study. PARKINSONS DISEASE 2016; 2016:3682936. [PMID: 27579212 PMCID: PMC4989080 DOI: 10.1155/2016/3682936] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Revised: 06/24/2016] [Accepted: 07/05/2016] [Indexed: 01/01/2023]
Abstract
The 5-HT2A receptor is highly involved in aspects of cognition and executive function and seen to be affected in neurodegenerative diseases like Alzheimer's disease and related to the disease pathology. Even though Parkinson's disease (PD) is primarily a motor disorder, reports of impaired executive function are also steadily being associated with this disease. Not much is known about the pathophysiology behind this. The aim of this study was thereby twofold: (1) to investigate 5-HT2A receptor binding levels in Parkinson's brains and (2) to investigate whether PD associated pathology, alpha-synuclein (AS) overexpression, could be associated with 5-HT2A alterations. Binding density for the 5-HT2A-specific radioligand [3H]-MDL 100.907 was measured in membrane suspensions of frontal cortex tissue from PD patients. Protein levels of AS were further measured using western blotting. Results showed higher AS levels accompanied by increased 5-HT2A receptor binding in PD brains. In a separate study, we looked for changes in 5-HT2A receptors in the prefrontal cortex in 52-week-old transgenic mice overexpressing human AS. We performed region-specific 5-HT2A receptor binding measurements followed by gene expression analysis. The transgenic mice showed lower 5-HT2A binding in the frontal association cortex that was not accompanied by changes in gene expression levels. This study is one of the first to look at differences in serotonin receptor levels in PD and in relation to AS overexpression.
Collapse
|
17
|
Dehay B, Decressac M, Bourdenx M, Guadagnino I, Fernagut PO, Tamburrino A, Bassil F, Meissner WG, Bezard E. Targeting α-synuclein: Therapeutic options. Mov Disord 2016; 31:882-8. [PMID: 26926119 DOI: 10.1002/mds.26568] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Revised: 01/09/2016] [Accepted: 01/14/2016] [Indexed: 12/21/2022] Open
Abstract
The discovery of the central role of α-synuclein (αSyn) in the pathogenesis of Parkinson's disease (PD) has powered, in the last decade, the emergence of novel relevant models of this condition based on viral vector-mediated expression of the disease-causing protein or inoculation of toxic species of αSyn. Although the development of these powerful tools and models has provided considerable insights into the mechanisms underlying neurodegeneration in PD, it has also been translated into the expansion of the landscape of preclinical therapeutic strategies. Much attention is now brought to the proteotoxic mechanisms induced by αSyn and how to block them using strategies inspired by intrinsic cellular pathways such as the enhancement of cellular clearance by the lysosomal-autophagic system, through proteasome-mediated degradation or through immunization. The important effort undertaken by several laboratories and consortia to tackle these issues and identify novel targets warrants great promise for the discovery not only of neuroprotective approaches but also of restorative strategies for PD and other synucleinopathies. In this viewpoint, we summarize the latest advances in this new area of PD research and will discuss promising approaches and ongoing challenges. © 2016 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Benjamin Dehay
- Université de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, Bordeaux, France.,CNRS, Institut des Maladies Neurodégénératives, UMR 5293, Bordeaux, France
| | | | - Mathieu Bourdenx
- Université de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, Bordeaux, France.,CNRS, Institut des Maladies Neurodégénératives, UMR 5293, Bordeaux, France
| | | | - Pierre-Olivier Fernagut
- Université de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, Bordeaux, France.,CNRS, Institut des Maladies Neurodégénératives, UMR 5293, Bordeaux, France
| | - Anna Tamburrino
- Telethon Institute of Genetics and Medicine, Pozzuoli, Italy
| | - Fares Bassil
- Université de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, Bordeaux, France.,CNRS, Institut des Maladies Neurodégénératives, UMR 5293, Bordeaux, France
| | - Wassilios G Meissner
- Université de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, Bordeaux, France.,CNRS, Institut des Maladies Neurodégénératives, UMR 5293, Bordeaux, France.,Department of Neurology, University Hospital Bordeaux, Bordeaux, France
| | - Erwan Bezard
- Université de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, Bordeaux, France.,CNRS, Institut des Maladies Neurodégénératives, UMR 5293, Bordeaux, France
| |
Collapse
|
18
|
Gellhaar S, Marcellino D, Abrams MB, Galter D. Chronic L-DOPA induces hyperactivity, normalization of gait and dyskinetic behavior in MitoPark mice. GENES BRAIN AND BEHAVIOR 2016; 14:260-70. [PMID: 25752644 PMCID: PMC4405092 DOI: 10.1111/gbb.12210] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/14/2014] [Revised: 10/08/2014] [Accepted: 11/04/2014] [Indexed: 12/12/2022]
Abstract
Dopamine (DA) replacement therapy continues to be the gold standard treatment for Parkinson's disease (PD), as it improves key motor symptoms including bradykinesia and gait disturbances. With time, treatment induces side effects in the majority of patients, known as L-DOPA-induced dyskinesia (LID), which are often studied in animals by the use of unilateral, toxin-induced rodent models. In this study, we used the progressive, genetic PD model MitoPark to specifically evaluate bilateral changes in motor behavior following long-term L-DOPA treatment at three different stages of striatal DA depletion. Besides locomotor activity, we assessed changes in gait with two automated gait analysis systems and the development of dyskinetic behavior. Long-term treatment with a moderate, clinically relevant dose of L-DOPA (8 mg/kg) gradually produced age-dependent hyperactivity in MitoPark mice. In voluntary and forced gait analyses, we show that MitoPark mice with severe DA depletion have distinct gait characteristics, which are normalized to control levels following long-term L-DOPA treatment. The cylinder test showed an age-dependent and gradual development of bilateral LID. Significant increase in striatal FosB and prodynorphin expression was found to accompany the behavior changes. Taken together, we report that MitoPark mice model both behavioral and biochemical characteristics of long-term L-DOPA treatment in PD patients and provide a novel, consistent and progressive animal model of dyskinesia to aid in the discovery and evaluation of better treatment options to counteract LID.
Collapse
Affiliation(s)
- S Gellhaar
- Department of Neuroscience, Karolinska Institute, Stockholm, Sweden
| | | | | | | |
Collapse
|
19
|
Dehay B, Vila M, Bezard E, Brundin P, Kordower JH. Alpha-synuclein propagation: New insights from animal models. Mov Disord 2015; 31:161-8. [PMID: 26347034 DOI: 10.1002/mds.26370] [Citation(s) in RCA: 89] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Revised: 07/03/2015] [Accepted: 07/15/2015] [Indexed: 12/21/2022] Open
Abstract
Aggregation of alpha-synuclein is implicated in several neurodegenerative diseases collectively termed synucleinopathies. Emerging evidence strongly implicates cell-to-cell transmission of misfolded alpha-synuclein as a common pathogenetic mechanism in synucleinopathies. The impact of alpha-synuclein pathology on neuronal dysfunction and behavioral impairments is being explored in animal models. This review provides an update on how research in animal models supports the concept that misfolded alpha-synuclein spreads from cell to cell and describes how findings in animal models might relate to the disease process in humans. Finally, we discuss the current underlying molecular and cellular mechanisms and future therapeutic strategies targeting alpha-synuclein propagation.
Collapse
Affiliation(s)
- Benjamin Dehay
- Université de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, Bordeaux, France.,CNRS, Institut des Maladies Neurodégénératives, UMR 5293, Bordeaux, France
| | - Miquel Vila
- Neurodegenerative Diseases Research Group, Vall d'Hebron Research Institute, Centro Investigación Biomédica en Red Enfermedades Neurodegenerativas, Barcelona, Spain.,Department of Biochemistry and Molecular Biology, Autonomous University of Barcelona, Bellaterra, Barcelona, Spain.,Catalan Institution for Research and Advanced Studies, Barcelona, Spain
| | - Erwan Bezard
- Université de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, Bordeaux, France.,CNRS, Institut des Maladies Neurodégénératives, UMR 5293, Bordeaux, France
| | - Patrik Brundin
- Laboratory for Translational Parkinson's Disease Research, Center for Neurodegenerative Science, Van Andel Research Institute, Grand Rapids, Michigan, USA
| | - Jeffrey H Kordower
- Department of Neurological Sciences, The Graduate College, Rush University, Chicago, Illinois, USA
| |
Collapse
|
20
|
Zhang SN, Li XZ, Lu F, Liu SM. Cerebral potential biomarkers discovery and metabolic pathways analysis of α-synucleinopathies and the dual effects of Acanthopanax senticosus Harms on central nervous system through metabolomics analysis. JOURNAL OF ETHNOPHARMACOLOGY 2015; 163:264-272. [PMID: 25660332 DOI: 10.1016/j.jep.2015.01.046] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Revised: 01/12/2015] [Accepted: 01/15/2015] [Indexed: 06/04/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Acanthopanax senticosus Harms (AS), also called "Ciwujia" in Chinese and "Siberian ginseng" in the Siberian Taiga region, is the herb used in traditional medicinal systems of China, Russia, Japan and Korea for the treatment of various nervous and cerebrovascular diseases. AIM OF THE STUDY Our pre-study has showed that AS can significantly suppress α-synuclein overexpression and toxicity. Neuronal protein α-synuclein is a key player in the development of neurodegenerative diseases called α-synucleinopathies. Identifying the potential biomarkers related to α-synucleinopathies may facilitate understanding the pathogenesis of the diseases and the safe application of AS in the clinic. METHODS AND RESULTS Ultra-performance liquid chromatography-quadrupole time-of-flight-mass spectrometry (UPLC-QTOF-MS) coupled with pattern recognition methods was integrated to examine the cerebral metabolic signature of human α-synuclein transgenic mice and the effects of AS on central nervous system (CNS) in pathology and physiology. Totally, 17 differentially expressed metabolites in wild type (WT) group and 26 in A30P mutant (A30P) group were identified and considered as potential biomarkers. Among them, 11 endogenous metabolites in WT+AS group and 18 in A30P+AS group were involved in the anti-α-synucleinopathies mechanism of AS. However, western blot and metabolomics analysis showed the effects of AS on CNS in physiology were opposite to those in pathology, which may cause potential neurotoxicity. CONCLUSIONS This study demonstrated that endogenous metabolites perturbation was involved in the pathogenesis of α-synucleinopathies and AS produced the dual effects on pathological and physiological CNS.
Collapse
Affiliation(s)
- Shuai-Nan Zhang
- Chinese Medicine Toxicological Laboratory, Heilongjiang University of Chinese Medicine, Harbin 150040, PR China
| | - Xu-Zhao Li
- Chinese Medicine Toxicological Laboratory, Heilongjiang University of Chinese Medicine, Harbin 150040, PR China
| | - Fang Lu
- Chinese Medicine Toxicological Laboratory, Heilongjiang University of Chinese Medicine, Harbin 150040, PR China.
| | - Shu-Min Liu
- Chinese Medicine Toxicological Laboratory, Heilongjiang University of Chinese Medicine, Harbin 150040, PR China; Drug Safety Evaluation Center, Heilongjiang University of Chinese Medicine, Harbin 150040, PR China.
| |
Collapse
|
21
|
Progressive dopaminergic alterations and mitochondrial abnormalities in LRRK2 G2019S knock-in mice. Neurobiol Dis 2015; 78:172-95. [PMID: 25836420 DOI: 10.1016/j.nbd.2015.02.031] [Citation(s) in RCA: 166] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Revised: 02/19/2015] [Accepted: 02/21/2015] [Indexed: 01/19/2023] Open
Abstract
Mutations in the LRRK2 gene represent the most common genetic cause of late onset Parkinson's disease. The physiological and pathological roles of LRRK2 are yet to be fully determined but evidence points towards LRRK2 mutations causing a gain in kinase function, impacting on neuronal maintenance, vesicular dynamics and neurotransmitter release. To explore the role of physiological levels of mutant LRRK2, we created knock-in (KI) mice harboring the most common LRRK2 mutation G2019S in their own genome. We have performed comprehensive dopaminergic, behavioral and neuropathological analyses in this model up to 24months of age. We find elevated kinase activity in the brain of both heterozygous and homozygous mice. Although normal at 6months, by 12months of age, basal and pharmacologically induced extracellular release of dopamine is impaired in both heterozygous and homozygous mice, corroborating previous findings in transgenic models over-expressing mutant LRRK2. Via in vivo microdialysis measurement of basal and drug-evoked extracellular release of dopamine and its metabolites, our findings indicate that exocytotic release from the vesicular pool is impaired. Furthermore, profound mitochondrial abnormalities are evident in the striatum of older homozygous G2019S KI mice, which are consistent with mitochondrial fission arrest. We anticipate that this G2019S mouse line will be a useful pre-clinical model for further evaluation of early mechanistic events in LRRK2 pathogenesis and for second-hit approaches to model disease progression.
Collapse
|
22
|
Esteves AR, Swerdlow RH, Cardoso SM. LRRK2, a puzzling protein: insights into Parkinson's disease pathogenesis. Exp Neurol 2014; 261:206-16. [PMID: 24907399 DOI: 10.1016/j.expneurol.2014.05.025] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2014] [Accepted: 05/26/2014] [Indexed: 01/10/2023]
Abstract
Leucine-rich repeat kinase 2 (LRRK2) is a large, ubiquitous protein of unknown function. Mutations in the gene encoding LRRK2 have been linked to familial and sporadic Parkinson's disease (PD) cases. The LRRK2 protein is a single polypeptide that displays GTPase and kinase activity. Kinase and GTPase domains are involved in different cellular signaling pathways. Despite several experimental studies associating LRRK2 protein with various intracellular membranes and vesicular structures such as endosomal/lysosomal compartments, the mitochondrial outer membrane, lipid rafts, microtubule-associated vesicles, the golgi complex, and the endoplasmic reticulum its broader physiologic function(s) remain unidentified. Additionally, the cellular distribution of LRRK2 may indicate its role in several different pathways, such as the ubiquitin-proteasome system, the autophagic-lysosomal pathway, intracellular trafficking, and mitochondrial dysfunction. This review discusses potential mechanisms through which LRRK2 may mediate neurodegeneration and cause PD.
Collapse
Affiliation(s)
- A Raquel Esteves
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Portugal
| | - Russell H Swerdlow
- University of Kansas Alzheimer's Disease Center, University of Kansas Medical Center, Kansas City, KS, USA
| | - Sandra M Cardoso
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Portugal; Faculty of Medicine, University of Coimbra, Coimbra, Portugal.
| |
Collapse
|
23
|
Divergent α-synuclein solubility and aggregation properties in G2019S LRRK2 Parkinson's disease brains with Lewy Body pathology compared to idiopathic cases. Neurobiol Dis 2013; 58:183-90. [PMID: 23747310 PMCID: PMC3752970 DOI: 10.1016/j.nbd.2013.05.017] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2012] [Revised: 04/18/2013] [Accepted: 05/22/2013] [Indexed: 11/23/2022] Open
Abstract
Mutations in LRRK2 are the most common genetic cause of Parkinson's disease (PD). The most prevalent LRRK2 mutation is the G2019S coding change, located in the kinase domain of this complex multi-domain protein. The majority of G2019S autopsy cases feature typical Lewy Body pathology with a clinical phenotype almost indistinguishable from idiopathic PD (iPD). Here we have investigated the biochemical characteristics of α-synuclein in G2019S LRRK2 PD post-mortem material, in comparison to pathology-matched iPD. Immunohistochemistry with pS129 α-synuclein antibody showed that the medulla is heavily affected with pathology in G2019S PD whilst the basal ganglia (BG), limbic and frontal cortical regions demonstrated comparable pathology scores between G2019S PD and iPD. Significantly lower levels of the highly aggregated α-synuclein species in urea–SDS fractions were observed in G2019S cases compared to iPD in the BG and limbic cortex. Our data, albeit from a small number of cases, highlight a difference in the biochemical properties of aggregated α-synuclein in G2019S linked PD compared to iPD, despite a similar histopathological presentation. This divergence in solubility is most notable in the basal ganglia, a region that is affected preclinically and is damaged before overt dopaminergic cell death. We compared α-synuclein biochemistry from LRRK2 G2019S and idiopathic PD brains. We used four G2019S PD post-mortem brains and pathology matched idiopathic PD cases. G2019S PD and idiopathic PD cases show comparable Limbic stage Lewy Body pathology. Minimal SDS-insoluble α-synuclein seen in G2019S PD in contrast to idiopathic PD We propose a divergent nature of α-synuclein pathogenic species in G2019S PD.
Collapse
|
24
|
Petit GH, Berkovich E, Hickery M, Kallunki P, Fog K, Fitzer-Attas C, Brundin P. Rasagiline ameliorates olfactory deficits in an alpha-synuclein mouse model of Parkinson's disease. PLoS One 2013; 8:e60691. [PMID: 23573275 PMCID: PMC3616111 DOI: 10.1371/journal.pone.0060691] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2012] [Accepted: 03/03/2013] [Indexed: 12/23/2022] Open
Abstract
Impaired olfaction is an early pre-motor symptom of Parkinson's disease. The neuropathology underlying olfactory dysfunction in Parkinson's disease is unknown, however α-synuclein accumulation/aggregation and altered neurogenesis might play a role. We characterized olfactory deficits in a transgenic mouse model of Parkinson's disease expressing human wild-type α-synuclein under the control of the mouse α-synuclein promoter. Preliminary clinical observations suggest that rasagiline, a monoamine oxidase-B inhibitor, improves olfaction in Parkinson's disease. We therefore examined whether rasagiline ameliorates olfactory deficits in this Parkinson's disease model and investigated the role of olfactory bulb neurogenesis. α-Synuclein mice were progressively impaired in their ability to detect odors, to discriminate between odors, and exhibited alterations in short-term olfactory memory. Rasagiline treatment rescued odor detection and odor discrimination abilities. However, rasagiline did not affect short-term olfactory memory. Finally, olfactory changes were not coupled to alterations in olfactory bulb neurogenesis. We conclude that rasagiline reverses select olfactory deficits in a transgenic mouse model of Parkinson's disease. The findings correlate with preliminary clinical observations suggesting that rasagiline ameliorates olfactory deficits in Parkinson's disease.
Collapse
Affiliation(s)
- Géraldine H Petit
- Neuronal Survival Unit, Wallenberg Neuroscience Center, Department of Experimental Medical Science, BMC B11, Lund University, Lund, Sweden.
| | | | | | | | | | | | | |
Collapse
|
25
|
Guerreiro PS, Huang Y, Gysbers A, Cheng D, Gai WP, Outeiro TF, Halliday GM. LRRK2 interactions with α-synuclein in Parkinson's disease brains and in cell models. J Mol Med (Berl) 2013; 91:513-22. [PMID: 23183827 PMCID: PMC3611031 DOI: 10.1007/s00109-012-0984-y] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2012] [Revised: 11/12/2012] [Accepted: 11/15/2012] [Indexed: 02/07/2023]
Abstract
Mutations in the genes encoding leucine-rich repeat kinase 2 (LRRK2) and α-synuclein are associated with both autosomal dominant and idiopathic forms of Parkinson's disease (PD). α-Synuclein is the main protein in Lewy bodies, hallmark inclusions present in both sporadic and familial PD. We show that in PD brain tissue, the levels of LRRK2 are positively related to the increase in α-synuclein phosphorylation and aggregation in affected brain regions (amygdala and anterior cingulate cortex), but not in the unaffected visual cortex. In disease-affected regions, we show co-localization of these two proteins in neurons and Lewy body inclusions. Further, in vitro experiments show a molecular interaction between α-synuclein and LRRK2 under endogenous and over-expression conditions. In a cell culture model of α-synuclein inclusion formation, LRRK2 co-localizes with the α-synuclein inclusions, and knocking down LRRK2 increases the number of smaller inclusions. In addition to providing strong evidence for an interaction between LRRK2 and α-synuclein, our results shed light on the complex relationship between these two proteins in the brains of patients with PD and the underlying molecular mechanisms of the disease.
Collapse
Affiliation(s)
- Patrícia Silva Guerreiro
- />Cell and Molecular Neuroscience Unit, Instituto de Medicina Molecular, Lisbon, 04250 Portugal
- />Instituto de Fisiologia, Faculdade de Medicina, Universidade de Lisboa, Av. Professor Egas Moniz, 1649-028, Lisbon, 04250 Portugal
- />Department of Neurodegeneration and Restorative Research, University Medizin Goettingen, Waldweg 33, Goettingen, 37073 Germany
| | - Yue Huang
- />Neuroscience Research Australia and The University of New South Wales (UNSW), Sydney, 2031 Australia
| | - Amanda Gysbers
- />Neuroscience Research Australia and The University of New South Wales (UNSW), Sydney, 2031 Australia
| | - Danni Cheng
- />Neuroscience Research Australia and The University of New South Wales (UNSW), Sydney, 2031 Australia
| | - Wei Ping Gai
- />Department of Human Physiology, The Centre for Neuroscience, Flinders University School of South Australia, Flinders, 5042 Australia
| | - Tiago Fleming Outeiro
- />Cell and Molecular Neuroscience Unit, Instituto de Medicina Molecular, Lisbon, 04250 Portugal
- />Instituto de Fisiologia, Faculdade de Medicina, Universidade de Lisboa, Av. Professor Egas Moniz, 1649-028, Lisbon, 04250 Portugal
- />Department of Neurodegeneration and Restorative Research, University Medizin Goettingen, Waldweg 33, Goettingen, 37073 Germany
| | - Glenda Margaret Halliday
- />Neuroscience Research Australia and The University of New South Wales (UNSW), Sydney, 2031 Australia
- />Neuroscience Research Australia, Barker Street, Randwick, NSW 2031 Australia
| |
Collapse
|
26
|
Mandemakers W, Snellinx A, O'Neill MJ, de Strooper B. LRRK2 expression is enriched in the striosomal compartment of mouse striatum. Neurobiol Dis 2012; 48:582-93. [PMID: 22850484 DOI: 10.1016/j.nbd.2012.07.017] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2012] [Revised: 07/13/2012] [Accepted: 07/20/2012] [Indexed: 01/24/2023] Open
Abstract
In spite of a clear genetic link between Parkinson's disease (PD) and mutations in LRRK2, cellular localization and physiological function of LRRK2 remain debated. Here we demonstrate the immunohistochemical localization of LRRK2 in adult mouse and early postnatal mouse brain development. Antibody specificity is verified by absence of specific staining in LRRK2 knockout mouse brain. Although LRRK2 is expressed in various mouse brain regions (i.e. cortex, thalamus, hippocampus, cerebellum), strongest expression is detected in striatum, whereas LRRK2 protein expression in substantia nigra pars compacta in contrast is low. LRRK2 is highly expressed in striatal medium spiny neurons (MSN) and few cholinergic interneurons. LRRK2 expression is undetectable in other interneurons, oligodendrocytes or astrocytes of the striatum. Interestingly, LRRK2 expression is associated with striosome specific markers (i.e. MOR1, RASGRP1). Analysis of LRRK2 expression during early postnatal development and in LRRK2 knockout mice, demonstrates that LRRK2 is not required for generation or maintenance of the striosome compartment. Comparing LRRK2-WT, LRRK2-R1441G transgenic and non-transgenic mice, changes of LRRK2 expression in striosome/matrix compartments can be detected. The findings rule out a specific requirement of LRRK2 in striosome genesis but suggest a functional role for LRRK2 in striosomes.
Collapse
|
27
|
Daher JPL, Pletnikova O, Biskup S, Musso A, Gellhaar S, Galter D, Troncoso JC, Lee MK, Dawson TM, Dawson VL, Moore DJ. Neurodegenerative phenotypes in an A53T α-synuclein transgenic mouse model are independent of LRRK2. Hum Mol Genet 2012; 21:2420-31. [PMID: 22357653 DOI: 10.1093/hmg/dds057] [Citation(s) in RCA: 77] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Mutations in the genes encoding LRRK2 and α-synuclein cause autosomal dominant forms of familial Parkinson's disease (PD). Fibrillar forms of α-synuclein are a major component of Lewy bodies, the intracytoplasmic proteinaceous inclusions that are a pathological hallmark of idiopathic and certain familial forms of PD. LRRK2 mutations cause late-onset familial PD with a clinical, neurochemical and, for the most part, neuropathological phenotype that is indistinguishable from idiopathic PD. Importantly, α-synuclein-positive Lewy bodies are the most common pathology identified in the brains of PD subjects harboring LRRK2 mutations. These observations may suggest that LRRK2 functions in a common pathway with α-synuclein to regulate its aggregation. To explore the potential pathophysiological interaction between LRRK2 and α-synuclein in vivo, we modulated LRRK2 expression in a well-established human A53T α-synuclein transgenic mouse model with transgene expression driven by the hindbrain-selective prion protein promoter. Deletion of LRRK2 or overexpression of human G2019S-LRRK2 has minimal impact on the lethal neurodegenerative phenotype that develops in A53T α-synuclein transgenic mice, including premature lethality, pre-symptomatic behavioral deficits and human α-synuclein or glial neuropathology. We also find that endogenous or human LRRK2 and A53T α-synuclein do not interact together to influence the number of nigrostriatal dopaminergic neurons. Taken together, our data suggest that α-synuclein-related pathology, which occurs predominantly in the hindbrain of this A53T α-synuclein mouse model, occurs largely independently from LRRK2 expression. These observations fail to provide support for a pathophysiological interaction of LRRK2 and α-synuclein in vivo, at least within neurons of the mouse hindbrain.
Collapse
Affiliation(s)
- João Paulo L Daher
- NeuroRegeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Hansen C, Angot E, Bergström AL, Steiner JA, Pieri L, Paul G, Outeiro TF, Melki R, Kallunki P, Fog K, Li JY, Brundin P. α-Synuclein propagates from mouse brain to grafted dopaminergic neurons and seeds aggregation in cultured human cells. J Clin Invest 2011; 121:715-25. [PMID: 21245577 DOI: 10.1172/jci43366] [Citation(s) in RCA: 645] [Impact Index Per Article: 49.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2010] [Accepted: 11/17/2010] [Indexed: 12/12/2022] Open
Abstract
Post-mortem analyses of brains from patients with Parkinson disease who received fetal mesencephalic transplants show that α-synuclein-containing (α-syn-containing) Lewy bodies gradually appear in grafted neurons. Here, we explored whether intercellular transfer of α-syn from host to graft, followed by seeding of α-syn aggregation in recipient neurons, can contribute to this phenomenon. We assessed α-syn cell-to-cell transfer using microscopy, flow cytometry, and high-content screening in several coculture model systems. Coculturing cells engineered to express either GFP- or DsRed-tagged α-syn resulted in a gradual increase in double-labeled cells. Importantly, α-syn-GFP derived from 1 neuroblastoma cell line localized to red fluorescent aggregates in other cells expressing DsRed-α-syn, suggesting a seeding effect of transmitted α-syn. Extracellular α-syn was taken up by cells through endocytosis and interacted with intracellular α-syn. Next, following intracortical injection of recombinant α-syn in rats, we found neuronal uptake was attenuated by coinjection of an endocytosis inhibitor. Finally, we demonstrated in vivo transfer of α-syn between host cells and grafted dopaminergic neurons in mice overexpressing human α-syn. In summary, intercellularly transferred α-syn interacts with cytoplasmic α-syn and can propagate α-syn pathology. These results suggest that α-syn propagation is a key element in the progression of Parkinson disease pathology.
Collapse
Affiliation(s)
- Christian Hansen
- Neuronal Survival Unit, Wallenberg Neuroscience Center, Lund University, Lund, Sweden
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Carballo-Carbajal I, Weber-Endress S, Rovelli G, Chan D, Wolozin B, Klein CL, Patenge N, Gasser T, Kahle PJ. Leucine-rich repeat kinase 2 induces alpha-synuclein expression via the extracellular signal-regulated kinase pathway. Cell Signal 2010; 22:821-7. [PMID: 20074637 PMCID: PMC3163153 DOI: 10.1016/j.cellsig.2010.01.006] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2009] [Accepted: 01/05/2010] [Indexed: 10/20/2022]
Abstract
Mutations in leucine-rich repeat kinase 2 (LRRK2) are the most frequent cause of autosomal-dominant Parkinson's disease (PD). The second known autosomal-dominant PD gene (SNCA) encodes alpha-synuclein, which is deposited in Lewy bodies, the neuropathological hallmark of PD. LRRK2 contains a kinase domain with homology to mitogen-activated protein kinase kinase kinases (MAPKKKs) and its activity has been suggested to be a key factor in LRRK2-associated PD. Here we investigated the role of LRRK2 in signal transduction pathways to identify putative PD-relevant downstream targets. Over-expression of wild-type [wt]LRRK2 in human embryonic kidney HEK293 cells selectively activated the extracellular signal-regulated kinase (ERK) module. PD-associated mutants G2019S and R1441C, but not kinase-dead LRRK2, induced ERK phosphorylation to the same extent as [wt]LRRK2, indicating that this effect is kinase-dependent. However, ERK activation by mutant R1441C and G2019S was significantly slower than that for [wt]LRRK2, despite similar levels of expression. Furthermore, induction of the ERK module by LRRK2 was associated to a small but significant induction of SNCA, which was suppressed by treatment with the selective MAPK/ERK kinase inhibitor U0126. This pathway linking the two dominant PD genes LRRK2 and SNCA may offer an interesting target for drug therapy in both familial and sporadic disease.
Collapse
Affiliation(s)
- Iria Carballo-Carbajal
- Department of Neurodegeneration, Hertie Institute for Clinical Brain Research, University Clinics Tübingen, Otfried-Müller-Strasse 27, 72076 Tübingen, Germany
| | - Susanne Weber-Endress
- Department of Neurodegeneration, Hertie Institute for Clinical Brain Research, University Clinics Tübingen, Otfried-Müller-Strasse 27, 72076 Tübingen, Germany
| | - Giorgio Rovelli
- Nervous System Research, Novartis Pharma Ltd, Klybeckstrasse 141, 4057 Basel, Switzerland
| | - Diane Chan
- Department of Pharmacology, Boston University School of Medicine, 80 E Concord Street, Boston, MA 02118, USA
| | - Benjamin Wolozin
- Department of Pharmacology, Boston University School of Medicine, 80 E Concord Street, Boston, MA 02118, USA
| | - Christian L. Klein
- Department of Neurodegeneration, Hertie Institute for Clinical Brain Research, University Clinics Tübingen, Otfried-Müller-Strasse 27, 72076 Tübingen, Germany
| | - Nadja Patenge
- Department of Neurodegeneration, Hertie Institute for Clinical Brain Research, University Clinics Tübingen, Otfried-Müller-Strasse 27, 72076 Tübingen, Germany
| | - Thomas Gasser
- Department of Neurodegeneration, Hertie Institute for Clinical Brain Research, University Clinics Tübingen, Otfried-Müller-Strasse 27, 72076 Tübingen, Germany
| | - Philipp J. Kahle
- Department of Neurodegeneration, Hertie Institute for Clinical Brain Research, University Clinics Tübingen, Otfried-Müller-Strasse 27, 72076 Tübingen, Germany
| |
Collapse
|
30
|
Leucine-rich repeat kinase 2 regulates the progression of neuropathology induced by Parkinson's-disease-related mutant alpha-synuclein. Neuron 2010; 64:807-27. [PMID: 20064389 DOI: 10.1016/j.neuron.2009.11.006] [Citation(s) in RCA: 395] [Impact Index Per Article: 28.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/03/2009] [Indexed: 10/20/2022]
Abstract
Mutations in alpha-synuclein and Leucine-rich repeat kinase 2 (LRRK2) are linked to autosomal dominant forms of Parkinson's disease (PD). However, little is known about any potential pathophysiological interplay between these two PD-related genes. Here we show in transgenic mice that although overexpression of LRRK2 alone did not cause neurodegeneration, the presence of excess LRRK2 greatly accelerated the progression of neuropathological abnormalities developed in PD-related A53T alpha-synuclein transgenic mice. Moreover, we found that LRRK2 promoted the abnormal aggregation and somatic accumulation of alpha-synuclein in A53T mice, which likely resulted from the impairment of microtubule dynamics, Golgi organization, and the ubiquitin-proteasome pathway. Conversely, genetic ablation of LRRK2 preserved the Golgi structure and suppressed the aggregation and somatic accumulation of alpha-synuclein, and thereby delayed the progression of neuropathology in A53T mice. These findings demonstrate that overexpression of LRRK2 enhances alpha-synuclein-mediated cytotoxicity and suggest inhibition of LRRK2 expression as a potential therapeutic option for ameliorating alpha-synuclein-induced neurodegeneration.
Collapse
|
31
|
Curcumin exposure induces expression of the Parkinson's disease-associated leucine-rich repeat kinase 2 (LRRK2) in rat mesencephalic cells. Neurosci Lett 2010; 468:120-4. [DOI: 10.1016/j.neulet.2009.10.081] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2009] [Revised: 10/21/2009] [Accepted: 10/24/2009] [Indexed: 11/19/2022]
|
32
|
Yue Z. LRRK2 in Parkinson's disease: in vivo models and approaches for understanding pathogenic roles. FEBS J 2009; 276:6445-54. [PMID: 19804414 DOI: 10.1111/j.1742-4658.2009.07343.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
The recent discovery of the genetic causes for Parkinson's disease (PD) is fruitful; however, the continuing revelation of PD-related genes is rapidly outpacing the functional characterization of the gene products. Although the discovery of multiple PD-related genes places PD as one of the most complex multigenetic diseases of the brain, it will undoubtedly facilitate the unfolding of a central pathogenic pathway and an understanding of the etiology of PD. Recent findings of pathogenic mutations in leucine-rich repeat kinase 2 (LRRK2) (PARK8) that are linked to the most common familial forms and some sporadic forms of PD provide a unique opportunity to gain insight into the pathogenesis of PD. Despite rapid growth in biochemical, structural and in vitro cell culture studies of LRRK2, the in vivo characterizations of LRRK2 function generally fall short and are largely limited to invertebrates. The investigation of LRRK2 or homologs of LRRK2 in nonmammalian models provides important clues with respect to the cellular functions of LRRK2, but an elucidation of the physiology and pathophysiology of LRRK2 relevant to PD would still depend on mammalian models established by multiple genetic approaches, followed by rigorous examination of the models for pathological process. This minireview summarizes previous studies of genes for ROCO and LRRK2 homologs in slime mold, nematode worms and fruit flies. It also discusses the results obtained from available mouse models of LRRK2 that begin to provide information for understanding LRRK2-mediated pathogenesis in PD.
Collapse
Affiliation(s)
- Zhenyu Yue
- Department of Neurology and Neuroscience, Mount Sinai School of Medicine, New York, NY 10029, USA.
| |
Collapse
|