1
|
Alecu JE, Sigutova V, Brazdis RM, Lörentz S, Bogiongko ME, Nursaitova A, Regensburger M, Roybon L, Galler KM, Wrasidlo W, Winner B, Prots I. NPT100-18A rescues mitochondrial oxidative stress and neuronal degeneration in human iPSC-based Parkinson's model. BMC Neurosci 2025; 26:8. [PMID: 39875842 PMCID: PMC11773751 DOI: 10.1186/s12868-025-00926-y] [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: 05/10/2024] [Accepted: 01/10/2025] [Indexed: 01/30/2025] Open
Abstract
BACKGROUND Parkinson's disease (PD) is a neurodegenerative disorder characterized by protein aggregates mostly consisting of misfolded alpha-synuclein (αSyn). Progressive degeneration of midbrain dopaminergic neurons (mDANs) and nigrostriatal projections results in severe motor symptoms. While the preferential loss of mDANs has not been fully understood yet, the cell type-specific vulnerability has been linked to a unique intracellular milieu, influenced by dopamine metabolism, high demand for mitochondrial activity, and increased level of oxidative stress (OS). These factors have been shown to adversely impact αSyn aggregation. Reciprocally, αSyn aggregates, in particular oligomers, can impair mitochondrial functions and exacerbate OS. Recent drug-discovery studies have identified a series of small molecules, including NPT100-18A, which reduce αSyn oligomerization by preventing misfolding and dimerization. NPT100-18A and structurally similar compounds (such as NPT200-11/UCB0599, currently being assessed in clinical studies) point towards a promising new approach for disease-modification. METHODS Induced pluripotent stem cell (iPSC)-derived mDANs from PD patients with a monoallelic SNCA locus duplication and unaffected controls were treated with NPT100-18A. αSyn aggregation was evaluated biochemically and reactive oxygen species (ROS) levels were assessed in living mDANs using fluorescent dyes. Adenosine triphosphate (ATP) levels were measured using a luminescence-based assay, and neuronal cell death was evaluated by immunocytochemistry. RESULTS Compared to controls, patient-derived mDANs exhibited higher cytoplasmic and mitochondrial ROS probe levels, reduced ATP-related signals, and increased activation of caspase-3, reflecting early neuronal cell death. NPT100-18A-treatment rescued cleaved caspase-3 levels to control levels and, importantly, attenuated mitochondrial oxidative stress probe levels in a compartment-specific manner and, at higher concentrations, increased ATP signals. CONCLUSIONS Our findings demonstrate that NPT100-18A limits neuronal degeneration in a human in vitro model of PD. In addition, we provide first mechanistic insights into how a compartment-specific antioxidant effect in mitochondria might contribute to the neuroprotective effects of NPT100-18A.
Collapse
Affiliation(s)
- Julian E Alecu
- Department of Stem Cell Biology, University Hospital Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
| | - Veronika Sigutova
- Department of Stem Cell Biology, University Hospital Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
- Department of Operative Dentistry and Periodontology, University Hospital Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
| | - Razvan-Marius Brazdis
- Department of Psychiatry and Psychotherapy, University Hospital Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
| | - Sandra Lörentz
- Department of Stem Cell Biology, University Hospital Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
| | - Marios Evangelos Bogiongko
- Department of Stem Cell Biology, University Hospital Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
| | - Anara Nursaitova
- Department of Stem Cell Biology, University Hospital Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
| | - Martin Regensburger
- Department of Stem Cell Biology, University Hospital Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander University of Erlangen- Nürnberg, Erlangen, Germany
- Center for Rare Diseases Erlangen (ZSEER), University Hospital Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
| | - Laurent Roybon
- Department of Neurodegenerative Science, the MiND program, Van Andel Institute, Grand Rapids, MI, USA
| | - Kerstin M Galler
- Department of Operative Dentistry and Periodontology, University Hospital Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
| | - Wolfgang Wrasidlo
- Neuropore Therapies, Inc, San Diego, CA, USA
- Department of Neuroscience, University of California, San Diego, La Jolla, CA, USA
| | - Beate Winner
- Department of Stem Cell Biology, University Hospital Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
- Center for Rare Diseases Erlangen (ZSEER), University Hospital Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
| | - Iryna Prots
- Department of Operative Dentistry and Periodontology, University Hospital Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany.
| |
Collapse
|
2
|
Nardi FV, Maisumu G, Zhou Y, Liang B, Yakoub AM. Protocol for generation of PD modeling induced neurons and detection of α-synuclein forms. STAR Protoc 2024; 5:103447. [PMID: 39549235 PMCID: PMC11609654 DOI: 10.1016/j.xpro.2024.103447] [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: 07/02/2024] [Revised: 08/19/2024] [Accepted: 10/18/2024] [Indexed: 11/18/2024] Open
Abstract
Alpha-synuclein (α-Syn) is an important molecule in the pathogenesis of Parkinson's disease and Alzheimer's disease-related dementias such as Lewy body dementia, forming multiple pathological species. In vitro disease models, including human neurons and α-Syn-transfected cells, are instrumental to understand synucleinopathies or test new therapies. Here, we provide a detailed protocol to generate human neurons derived from induced pluripotent stem cells (iPSCs), and HEK cells, with α-Syn mutations. We also describe multiple assays to determine the various α-Syn forms.
Collapse
Affiliation(s)
- Francesco V Nardi
- Department of Medicine, Harvard Medical School, and Brigham and Women's Hospital, Boston, MA 02115, USA; Biomedical Engineering Program, College of Engineering and Mines, University of North Dakota, Grand Forks, ND 58202, USA
| | - Gulimiheranmu Maisumu
- Department of Medicine, Harvard Medical School, and Brigham and Women's Hospital, Boston, MA 02115, USA; Biomedical Engineering Program, College of Engineering and Mines, University of North Dakota, Grand Forks, ND 58202, USA
| | - You Zhou
- Department of Medicine, Harvard Medical School, and Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Bo Liang
- Biomedical Engineering Program, College of Engineering and Mines, University of North Dakota, Grand Forks, ND 58202, USA
| | - Abraam M Yakoub
- Department of Medicine, Harvard Medical School, and Brigham and Women's Hospital, Boston, MA 02115, USA.
| |
Collapse
|
3
|
Ioghen OC, Gaina G, Lambrescu I, Manole E, Pop S, Niculescu TM, Mosoia O, Ceafalan LC, Popescu BO. Bacterial products initiation of alpha-synuclein pathology: an in vitro study. Sci Rep 2024; 14:30306. [PMID: 39639092 PMCID: PMC11621565 DOI: 10.1038/s41598-024-81020-x] [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: 03/15/2024] [Accepted: 11/22/2024] [Indexed: 12/07/2024] Open
Abstract
Parkinson's Disease (PD) is a prevalent and escalating neurodegenerative disorder with significant societal implications. Despite being considered a proteinopathy, in which the aggregation of α-synuclein is the main pathological change, the intricacies of PD initiation remain elusive. Recent evidence suggests a potential link between gut microbiota and PD initiation, emphasizing the need to explore the effects of microbiota-derived molecules on neuronal cells. In this study, we exposed dopaminergic-differentiated SH-SY5Y cells to microbial molecules such as lipopolysaccharide (LPS), rhamnolipid, curli CsgA and phenol soluble modulin α-1 (PSMα1). We assessed cellular viability, cytotoxicity, growth curves and α-synuclein levels by performing MTS, LDH, real-time impedance readings, qRT-PCR and Western Blot assays respectively. Statistical analysis revealed that rhamnolipid exhibited concentration-dependent effects, reducing viability and inducing cytotoxicity at higher concentrations, increasing α-synuclein mRNA and protein levels with negative effects on cell morphology and adhesion. Furthermore, LPS exposure also increased α-synuclein levels. Curli CsgA and PSMα-1 showed minimal or no changes. Our findings suggest that microbiota-derived molecules, particularly rhamnolipid and LPS, impact dopaminergic neurons by increasing α-synuclein levels. This study highlights the potential involvement of gut microbiota in initiating the upregulation of α-synuclein that may further initiate PD, indicating the complex interplay between microbiota and neuronal cells.
Collapse
Grants
- 31PFE/30.12.2021 Ministry of Research, Innovation, and Digitalization in Romania
- 31PFE/30.12.2021 Ministry of Research, Innovation, and Digitalization in Romania
- 31PFE/30.12.2021 Ministry of Research, Innovation, and Digitalization in Romania
- 31PFE/30.12.2021 Ministry of Research, Innovation, and Digitalization in Romania
- 31PFE/30.12.2021 Ministry of Research, Innovation, and Digitalization in Romania
- 31PFE/30.12.2021 Ministry of Research, Innovation, and Digitalization in Romania
- 31PFE/30.12.2021 Ministry of Research, Innovation, and Digitalization in Romania
- 31PFE/30.12.2021 Ministry of Research, Innovation, and Digitalization in Romania
- 31PFE/30.12.2021 Ministry of Research, Innovation, and Digitalization in Romania
Collapse
Affiliation(s)
- Octavian Costin Ioghen
- "Victor Babeș" National Institute of Pathology, 050096, Bucharest, Romania
- "Carol Davila" University of Medicine and Pharmacy, 050474, Bucharest, Romania
| | - Gisela Gaina
- "Victor Babeș" National Institute of Pathology, 050096, Bucharest, Romania
- "Carol Davila" University of Medicine and Pharmacy, 050474, Bucharest, Romania
| | - Ioana Lambrescu
- "Victor Babeș" National Institute of Pathology, 050096, Bucharest, Romania
- "Carol Davila" University of Medicine and Pharmacy, 050474, Bucharest, Romania
| | - Emilia Manole
- "Victor Babeș" National Institute of Pathology, 050096, Bucharest, Romania
| | - Sevinci Pop
- "Victor Babeș" National Institute of Pathology, 050096, Bucharest, Romania
| | | | - Oana Mosoia
- "Victor Babeș" National Institute of Pathology, 050096, Bucharest, Romania
| | - Laura Cristina Ceafalan
- "Victor Babeș" National Institute of Pathology, 050096, Bucharest, Romania.
- "Carol Davila" University of Medicine and Pharmacy, 050474, Bucharest, Romania.
| | - Bogdan Ovidiu Popescu
- "Victor Babeș" National Institute of Pathology, 050096, Bucharest, Romania
- "Carol Davila" University of Medicine and Pharmacy, 050474, Bucharest, Romania
| |
Collapse
|
4
|
Nakamura Y, Sawai T, Kakiuchi K, Arawaka S. Neuronal activity promotes secretory autophagy for the extracellular release of α-synuclein. J Biol Chem 2024; 300:107419. [PMID: 38815862 PMCID: PMC11253543 DOI: 10.1016/j.jbc.2024.107419] [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: 11/07/2023] [Revised: 05/15/2024] [Accepted: 05/19/2024] [Indexed: 06/01/2024] Open
Abstract
Extracellular secretion is an essential mechanism for α-synuclein (α-syn) proteostasis. Although it has been reported that neuronal activity affects α-syn secretion, the underlying mechanisms remain unclear. Here, we investigated the autophagic processes that regulate the physiological release of α-syn in mouse primary cortical neurons and SH-SY5Y cells. Stimulating neuronal activity with glutamate or depolarization with high KCl enhanced α-syn secretion. This glutamate-induced α-syn secretion was blocked by a mixture of NMDA receptor antagonist AP5 and AMPA receptor antagonist NBQX, as well as by cytosolic Ca2+ chelator BAPTA-AM. Additionally, mTOR inhibitor rapamycin increased α-syn and p62/SQSTM1 (p62) secretion, and this effect of rapamycin was reduced in primary cortical neurons deficient in the autophagy regulator beclin 1 (derived from BECN1+/- mice). Glutamate-induced α-syn and p62 secretion was suppressed by the knockdown of ATG5, which is required for autophagosome formation. Glutamate increased LC3-II generation and decreased intracellular p62 levels, and the increase in LC3-II levels was blocked by BAPTA-AM. Moreover, glutamate promoted co-localization of α-syn with LC3-positive puncta, but not with LAMP1-positive structures in the neuronal somas. Glutamate-induced α-syn and p62 secretion were also reduced by the knockdown of RAB8A, which is required for autophagosome fusion with the plasma membrane. Collectively, these findings suggest that stimulating neuronal activity mediates autophagic α-syn secretion in a cytosolic Ca2+-dependent manner, and autophagosomes may participate in autophagic secretion by functioning as α-syn carriers.
Collapse
Affiliation(s)
- Yoshitsugu Nakamura
- Division of Neurology, Department of Internal Medicine IV, Osaka Medical and Pharmaceutical University Faculty of Medicine, Takatsuki, Osaka, Japan
| | - Taiki Sawai
- Division of Neurology, Department of Internal Medicine IV, Osaka Medical and Pharmaceutical University Faculty of Medicine, Takatsuki, Osaka, Japan
| | - Kensuke Kakiuchi
- Division of Neurology, Department of Internal Medicine IV, Osaka Medical and Pharmaceutical University Faculty of Medicine, Takatsuki, Osaka, Japan
| | - Shigeki Arawaka
- Division of Neurology, Department of Internal Medicine IV, Osaka Medical and Pharmaceutical University Faculty of Medicine, Takatsuki, Osaka, Japan.
| |
Collapse
|
5
|
Amagai R, Otomo R, Yoshioka S, Nagano H, Hashimoto N, Sakakibara R, Tanaka T, Okado-Matsumoto A. C-terminal truncation is a prominent post-translational modification of human erythrocyte α-synuclein. J Biochem 2024; 175:649-658. [PMID: 38308089 DOI: 10.1093/jb/mvae012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 01/09/2024] [Accepted: 01/25/2024] [Indexed: 02/04/2024] Open
Abstract
α-Synuclein (α-Syn) is a protein related to synucleinopathies with high expression in the central nervous system and erythrocytes which are a major source of peripheral α-Syn. Recent reports have suggested the presence of α-Syn within extracellular vesicles (EVs) derived from erythrocytes, potentially contributing to the pathogenesis of synucleinopathies. While Lewy bodies, intracellular inclusions containing aggregated α-Syn, are prominently observed within the brain, their occurrence in peripheral neurons implies the dissemination of synucleinopathy pathology throughout the body via the propagation of α-Syn. In this study, we found erythrocytes and circulating EVs obtained from plasma contained α-Syn, which was separated into four major forms using high-resolution clear native-PAGE and isoelectric focusing. Notably, erythrocyte α-Syn was classified into full-length and C-terminal truncated forms, with truncation observed between Y133 and Q134 as determined by LC-MS/MS analysis. Our finding revealed that C-terminally truncated α-Syn, which was previously reported to exist solely within the brain, was also present in erythrocytes and circulating EVs obtained from plasma.
Collapse
Affiliation(s)
- Ryosuke Amagai
- Laboratory of Biochemistry, Department of Biology, Faculty of Science, Toho University, 2-2-1 Miyama, Funabashi, Chiba 274-8510, Japan
| | - Riki Otomo
- Laboratory of Biochemistry, Department of Biology, Faculty of Science, Toho University, 2-2-1 Miyama, Funabashi, Chiba 274-8510, Japan
| | - Sakura Yoshioka
- Laboratory of Biochemistry, Department of Biology, Faculty of Science, Toho University, 2-2-1 Miyama, Funabashi, Chiba 274-8510, Japan
| | - Hidekazu Nagano
- Department of Molecular Diagnosis, Chiba University Graduate School of Medicine, Chiba, Chiba 260-8670, Japan
| | - Naoko Hashimoto
- Department of Molecular Diagnosis, Chiba University Graduate School of Medicine, Chiba, Chiba 260-8670, Japan
| | - Ryuji Sakakibara
- Division of Neurology, Department of Internal Medicine, Sakura Medical Center, Toho University, Sakura, Chiba 285-8741, Japan
| | - Tomoaki Tanaka
- Department of Molecular Diagnosis, Chiba University Graduate School of Medicine, Chiba, Chiba 260-8670, Japan
| | - Ayako Okado-Matsumoto
- Laboratory of Biochemistry, Department of Biology, Faculty of Science, Toho University, 2-2-1 Miyama, Funabashi, Chiba 274-8510, Japan
| |
Collapse
|
6
|
Laferrière F, Sabatier L, Claverol S, De Giorgi F, Ichas F. Biases in α-synuclein immuno-quantitation: a core problem for basic and ancillary studies of Parkinson's disease and multiple system atrophy. Transl Neurodegener 2024; 13:15. [PMID: 38528639 PMCID: PMC10962071 DOI: 10.1186/s40035-024-00408-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 03/01/2024] [Indexed: 03/27/2024] Open
Affiliation(s)
| | | | | | | | - François Ichas
- Univ. Bordeaux, CNRS, IMN, UMR 5293, 33000, Bordeaux, France
- DiSTeBA, Univ. Salento, Anatomia Umana, Lecce, Italy
| |
Collapse
|
7
|
Gregorio I, Russo L, Torretta E, Barbacini P, Contarini G, Pacinelli G, Bizzotto D, Moriggi M, Braghetta P, Papaleo F, Gelfi C, Moro E, Cescon M. GBA1 inactivation in oligodendrocytes affects myelination and induces neurodegenerative hallmarks and lipid dyshomeostasis in mice. Mol Neurodegener 2024; 19:22. [PMID: 38454456 PMCID: PMC10921719 DOI: 10.1186/s13024-024-00713-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 02/20/2024] [Indexed: 03/09/2024] Open
Abstract
BACKGROUND Mutations in the β-glucocerebrosidase (GBA1) gene do cause the lysosomal storage Gaucher disease (GD) and are among the most frequent genetic risk factors for Parkinson's disease (PD). So far, studies on both neuronopathic GD and PD primarily focused on neuronal manifestations, besides the evaluation of microglial and astrocyte implication. White matter alterations were described in the central nervous system of paediatric type 1 GD patients and were suggested to sustain or even play a role in the PD process, although the contribution of oligodendrocytes has been so far scarcely investigated. METHODS We exploited a system to study the induction of central myelination in vitro, consisting of Oli-neu cells treated with dibutyryl-cAMP, in order to evaluate the expression levels and function of β-glucocerebrosidase during oligodendrocyte differentiation. Conduritol-B-epoxide, a β-glucocerebrosidase irreversible inhibitor was used to dissect the impact of β-glucocerebrosidase inactivation in the process of myelination, lysosomal degradation and α-synuclein accumulation in vitro. Moreover, to study the role of β-glucocerebrosidase in the white matter in vivo, we developed a novel mouse transgenic line in which β-glucocerebrosidase function is abolished in myelinating glia, by crossing the Cnp1-cre mouse line with a line bearing loxP sequences flanking Gba1 exons 9-11, encoding for β-glucocerebrosidase catalytic domain. Immunofluorescence, western blot and lipidomic analyses were performed in brain samples from wild-type and knockout animals in order to assess the impact of genetic inactivation of β-glucocerebrosidase on myelination and on the onset of early neurodegenerative hallmarks, together with differentiation analysis in primary oligodendrocyte cultures. RESULTS Here we show that β-glucocerebrosidase inactivation in oligodendrocytes induces lysosomal dysfunction and inhibits myelination in vitro. Moreover, oligodendrocyte-specific β-glucocerebrosidase loss-of-function was sufficient to induce in vivo demyelination and early neurodegenerative hallmarks, including axonal degeneration, α-synuclein accumulation and astrogliosis, together with brain lipid dyshomeostasis and functional impairment. CONCLUSIONS Our study sheds light on the contribution of oligodendrocytes in GBA1-related diseases and supports the need for better characterizing oligodendrocytes as actors playing a role in neurodegenerative diseases, also pointing at them as potential novel targets to set a brake to disease progression.
Collapse
Affiliation(s)
- Ilaria Gregorio
- Department of Molecular Medicine, University of Padova, Via Ugo Bassi 58/B, 35131, Padua, Italy
| | - Loris Russo
- Department of Molecular Medicine, University of Padova, Via Ugo Bassi 58/B, 35131, Padua, Italy
| | - Enrica Torretta
- Laboratory of Proteomics and Lipidomics, IRCCS Orthopedic Institute Galeazzi, Milan, 20161, Italy
| | - Pietro Barbacini
- Department of Biomedical Sciences for Health, University of Milan, 20133, Milan, Italy
| | - Gabriella Contarini
- Genetics of Cognition Laboratory, Neuroscience Area, Istituto Italiano Di Tecnologia, 16163, Genova, Italy
- Department of Biomedical and Technological Sciences, University of Catania, 95125, Catania, Italy
| | - Giada Pacinelli
- Genetics of Cognition Laboratory, Neuroscience Area, Istituto Italiano Di Tecnologia, 16163, Genova, Italy
- Padova Neuroscience Center (PNC), University of Padova, 35131, Padua, Italy
| | - Dario Bizzotto
- Department of Molecular Medicine, University of Padova, Via Ugo Bassi 58/B, 35131, Padua, Italy
| | - Manuela Moriggi
- Department of Biomedical Sciences for Health, University of Milan, 20133, Milan, Italy
| | - Paola Braghetta
- Department of Molecular Medicine, University of Padova, Via Ugo Bassi 58/B, 35131, Padua, Italy
| | - Francesco Papaleo
- Genetics of Cognition Laboratory, Neuroscience Area, Istituto Italiano Di Tecnologia, 16163, Genova, Italy
| | - Cecilia Gelfi
- Laboratory of Proteomics and Lipidomics, IRCCS Orthopedic Institute Galeazzi, Milan, 20161, Italy
- Department of Biomedical Sciences for Health, University of Milan, 20133, Milan, Italy
| | - Enrico Moro
- Department of Molecular Medicine, University of Padova, Via Ugo Bassi 58/B, 35131, Padua, Italy
| | - Matilde Cescon
- Department of Molecular Medicine, University of Padova, Via Ugo Bassi 58/B, 35131, Padua, Italy.
| |
Collapse
|
8
|
Santhosh Kumar S, Naseri NN, Pather SR, Hallacli E, Ndayisaba A, Buenaventura C, Acosta K, Roof J, Fazelinia H, Spruce LA, Luk K, Khurana V, Rhoades E, Shalem O. Sequential CRISPR screening reveals partial NatB inhibition as a strategy to mitigate alpha-synuclein levels in human neurons. SCIENCE ADVANCES 2024; 10:eadj4767. [PMID: 38335281 PMCID: PMC10857481 DOI: 10.1126/sciadv.adj4767] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Accepted: 01/11/2024] [Indexed: 02/12/2024]
Abstract
Alpha-synuclein (αSyn) protein levels correlate with the risk and severity of Parkinson's disease and related neurodegenerative diseases. Lowering αSyn is being actively investigated as a therapeutic modality. Here, we systematically map the regulatory network that controls endogenous αSyn using sequential CRISPR-knockout and -interference screens in an αSyn gene (SNCA)-tagged cell line and induced pluripotent stem cell-derived neurons (iNeurons). We uncover αSyn modifiers at multiple regulatory layers, with amino-terminal acetyltransferase B (NatB) enzymes being the most potent endogenous αSyn modifiers in both cell lines. Amino-terminal acetylation protects the cytosolic αSyn from rapid degradation by the proteasome in a Ube2w-dependent manner. Moreover, we show that pharmacological inhibition of methionyl-aminopeptidase 2, a regulator of NatB complex formation, attenuates endogenous αSyn in iNeurons carrying SNCA triplication. Together, our study reveals several gene networks that control endogenous αSyn, identifies mechanisms mediating the degradation of nonacetylated αSyn, and illustrates potential therapeutic pathways for decreasing αSyn levels in synucleinopathies.
Collapse
Affiliation(s)
- Saranya Santhosh Kumar
- Department of Bioengineering, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, PA, USA
- Center for Cellular and Molecular Therapeutics, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Nima N. Naseri
- Center for Cellular and Molecular Therapeutics, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Chemistry, University of Pennsylvania, Philadelphia, PA, USA
| | - Sarshan R. Pather
- Center for Cellular and Molecular Therapeutics, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Erinc Hallacli
- Division of Movement Disorders and Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Alain Ndayisaba
- Division of Movement Disorders and Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Chris Buenaventura
- Center for Cellular and Molecular Therapeutics, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Karen Acosta
- Department of Chemistry, University of Pennsylvania, Philadelphia, PA, USA
| | - Jennifer Roof
- Proteomics Core Facility, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Hossein Fazelinia
- Proteomics Core Facility, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Biomedical and Health Informatics, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Lynn A. Spruce
- Proteomics Core Facility, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Kelvin Luk
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Vikram Khurana
- Division of Movement Disorders and Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Harvard Stem Cell Institute, Cambridge, MA, USA
| | - Elizabeth Rhoades
- Department of Chemistry, University of Pennsylvania, Philadelphia, PA, USA
| | - Ophir Shalem
- Center for Cellular and Molecular Therapeutics, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
9
|
Dorion MF, Yaqubi M, Senkevich K, Kieran NW, MacDonald A, Chen CXQ, Luo W, Wallis A, Shlaifer I, Hall JA, Dudley RWR, Glass IA, Birth Defects Research Laboratory, Stratton JA, Fon EA, Bartels T, Antel JP, Gan-or Z, Durcan TM, Healy LM. MerTK is a mediator of alpha-synuclein fibril uptake by human microglia. Brain 2024; 147:427-443. [PMID: 37671615 PMCID: PMC10834256 DOI: 10.1093/brain/awad298] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 07/26/2023] [Accepted: 08/29/2023] [Indexed: 09/07/2023] Open
Abstract
Mer tyrosine kinase (MerTK) is a receptor tyrosine kinase that mediates non-inflammatory, homeostatic phagocytosis of diverse types of cellular debris. Highly expressed on the surface of microglial cells, MerTK is of importance in brain development, homeostasis, plasticity and disease. Yet, involvement of this receptor in the clearance of protein aggregates that accumulate with ageing and in neurodegenerative diseases has yet to be defined. The current study explored the function of MerTK in the microglial uptake of alpha-synuclein fibrils which play a causative role in the pathobiology of synucleinopathies. Using human primary and induced pluripotent stem cell-derived microglia, the MerTK-dependence of alpha-synuclein fibril internalization was investigated in vitro. Relevance of this pathway in synucleinopathies was assessed through burden analysis of MERTK variants and analysis of MerTK expression in patient-derived cells and tissues. Pharmacological inhibition of MerTK and siRNA-mediated MERTK knockdown both caused a decreased rate of alpha-synuclein fibril internalization by human microglia. Consistent with the non-inflammatory nature of MerTK-mediated phagocytosis, alpha-synuclein fibril internalization was not observed to induce secretion of pro-inflammatory cytokines such as IL-6 or TNF, and downmodulated IL-1β secretion from microglia. Burden analysis in two independent patient cohorts revealed a significant association between rare functionally deleterious MERTK variants and Parkinson's disease in one of the cohorts (P = 0.002). Despite a small upregulation in MERTK mRNA expression in nigral microglia from Parkinson's disease/Lewy body dementia patients compared to those from non-neurological control donors in a single-nuclei RNA-sequencing dataset (P = 5.08 × 10-21), no significant upregulation in MerTK protein expression was observed in human cortex and substantia nigra lysates from Lewy body dementia patients compared to controls. Taken together, our findings define a novel role for MerTK in mediating the uptake of alpha-synuclein fibrils by human microglia, with possible involvement in limiting alpha-synuclein spread in synucleinopathies such as Parkinson's disease. Upregulation of this pathway in synucleinopathies could have therapeutic values in enhancing alpha-synuclein fibril clearance in the brain.
Collapse
Affiliation(s)
- Marie-France Dorion
- Early Drug Discovery Unit, Montreal Neurological Institute-Hospital, McGill University, Montreal H3A 2B4, Canada
- Neuroimmunology Unit, Montreal Neurological Institute-Hospital, McGill University, Montreal H3A 2B4, Canada
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal H3A 2B4, Canada
| | - Moein Yaqubi
- Neuroimmunology Unit, Montreal Neurological Institute-Hospital, McGill University, Montreal H3A 2B4, Canada
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal H3A 2B4, Canada
| | - Konstantin Senkevich
- McGill Parkinson Program and Neurodegenerative Diseases Group, Montreal Neurological Institute-Hospital, McGill University, Montreal H3A 2B4, Canada
- Department of Human Genetics, McGill University, Montreal H3A 0C7, Canada
| | - Nicholas W Kieran
- Neuroimmunology Unit, Montreal Neurological Institute-Hospital, McGill University, Montreal H3A 2B4, Canada
| | - Adam MacDonald
- Neuroimmunology Unit, Montreal Neurological Institute-Hospital, McGill University, Montreal H3A 2B4, Canada
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal H3A 2B4, Canada
| | - Carol X Q Chen
- Early Drug Discovery Unit, Montreal Neurological Institute-Hospital, McGill University, Montreal H3A 2B4, Canada
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal H3A 2B4, Canada
| | - Wen Luo
- Early Drug Discovery Unit, Montreal Neurological Institute-Hospital, McGill University, Montreal H3A 2B4, Canada
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal H3A 2B4, Canada
| | - Amber Wallis
- UK Dementia Research Institute, University College London, London WC1E 6BT, UK
| | - Irina Shlaifer
- Early Drug Discovery Unit, Montreal Neurological Institute-Hospital, McGill University, Montreal H3A 2B4, Canada
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal H3A 2B4, Canada
| | - Jeffery A Hall
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal H3A 2B4, Canada
| | - Roy W R Dudley
- Department of Pediatric Surgery, Division of Neurosurgery, Montreal Children's Hospital, McGill University Health Centers, Montreal H4A 3J1, Canada
| | - Ian A Glass
- Department of Pediatrics, University of Washington, Seattle, WA 98195, USA
| | | | - Jo Anne Stratton
- Neuroimmunology Unit, Montreal Neurological Institute-Hospital, McGill University, Montreal H3A 2B4, Canada
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal H3A 2B4, Canada
| | - Edward A Fon
- Early Drug Discovery Unit, Montreal Neurological Institute-Hospital, McGill University, Montreal H3A 2B4, Canada
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal H3A 2B4, Canada
- McGill Parkinson Program and Neurodegenerative Diseases Group, Montreal Neurological Institute-Hospital, McGill University, Montreal H3A 2B4, Canada
| | - Tim Bartels
- UK Dementia Research Institute, University College London, London WC1E 6BT, UK
| | - Jack P Antel
- Neuroimmunology Unit, Montreal Neurological Institute-Hospital, McGill University, Montreal H3A 2B4, Canada
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal H3A 2B4, Canada
| | - Ziv Gan-or
- McGill Parkinson Program and Neurodegenerative Diseases Group, Montreal Neurological Institute-Hospital, McGill University, Montreal H3A 2B4, Canada
- Department of Human Genetics, McGill University, Montreal H3A 0C7, Canada
| | - Thomas M Durcan
- Early Drug Discovery Unit, Montreal Neurological Institute-Hospital, McGill University, Montreal H3A 2B4, Canada
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal H3A 2B4, Canada
| | - Luke M Healy
- Neuroimmunology Unit, Montreal Neurological Institute-Hospital, McGill University, Montreal H3A 2B4, Canada
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal H3A 2B4, Canada
| |
Collapse
|
10
|
van der Gaag BL, Deshayes NAC, Breve JJP, Bol JGJM, Jonker AJ, Hoozemans JJM, Courade JP, van de Berg WDJ. Distinct tau and alpha-synuclein molecular signatures in Alzheimer's disease with and without Lewy bodies and Parkinson's disease with dementia. Acta Neuropathol 2024; 147:14. [PMID: 38198008 PMCID: PMC10781859 DOI: 10.1007/s00401-023-02657-y] [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: 07/30/2023] [Revised: 11/10/2023] [Accepted: 11/22/2023] [Indexed: 01/11/2024]
Abstract
Alpha-synuclein (aSyn) pathology is present in approximately 50% of Alzheimer's disease (AD) cases at autopsy and might impact the age-of-onset and disease progression in AD. Here, we aimed to determine whether tau and aSyn profiles differ between AD cases with Lewy bodies (AD-LB), pure AD and Parkinson's disease with dementia (PDD) cases using epitope-, post-translational modification- (PTM) and isoform-specific tau and aSyn antibody panels spanning from the N- to C-terminus. We included the middle temporal gyrus (MTG) and amygdala (AMY) of clinically diagnosed and pathologically confirmed cases and performed dot blotting, western blotting and immunohistochemistry combined with quantitative and morphological analyses. All investigated phospho-tau (pTau) species, except pT181, were upregulated in AD-LB and AD cases compared to PDD and control cases, but no significant differences were observed between AD-LB and AD subjects. In addition, tau antibodies targeting the proline-rich regions and C-terminus showed preferential binding to AD-LB and AD brain homogenates. Antibodies targeting C-terminal aSyn epitopes and pS129 aSyn showed stronger binding to AD-LB and PDD cases compared to AD and control cases. Two pTau species (pS198 and pS396) were specifically detected in the soluble protein fractions of AD-LB and AD subjects, indicative of early involvement of these PTMs in the multimerization process of tau. Other phospho-variants for both tau (pT212/S214, pT231 and pS422) and aSyn (pS129) were only detected in the insoluble protein fraction of AD-LB/AD and AD-LB/PDD cases, respectively. aSyn load was higher in the AMY of AD-LB cases compared to PDD cases, suggesting aggravated aSyn pathology under the presence of AD pathology, while tau load was similar between AD-LB and AD cases. Co-localization of pTau and aSyn could be observed within astrocytes of AD-LB cases within the MTG. These findings highlight a unique pathological signature for AD-LB cases compared to pure AD and PDD cases.
Collapse
Affiliation(s)
- Bram L van der Gaag
- Section Clinical Neuroanatomy and Biobanking, Department of Anatomy and Neurosciences, Amsterdam UMC, Vrije Universiteit, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Program Neurodegeneration, Amsterdam, The Netherlands
| | - Natasja A C Deshayes
- Section Clinical Neuroanatomy and Biobanking, Department of Anatomy and Neurosciences, Amsterdam UMC, Vrije Universiteit, Amsterdam, The Netherlands
| | - John J P Breve
- Section Clinical Neuroanatomy and Biobanking, Department of Anatomy and Neurosciences, Amsterdam UMC, Vrije Universiteit, Amsterdam, The Netherlands
| | - John G J M Bol
- Section Clinical Neuroanatomy and Biobanking, Department of Anatomy and Neurosciences, Amsterdam UMC, Vrije Universiteit, Amsterdam, The Netherlands
| | - Allert J Jonker
- Section Clinical Neuroanatomy and Biobanking, Department of Anatomy and Neurosciences, Amsterdam UMC, Vrije Universiteit, Amsterdam, The Netherlands
| | - Jeroen J M Hoozemans
- Amsterdam Neuroscience, Program Neurodegeneration, Amsterdam, The Netherlands
- Department of Pathology, Amsterdam UMC, Vrije Universiteit, Amsterdam, The Netherlands
| | | | - Wilma D J van de Berg
- Section Clinical Neuroanatomy and Biobanking, Department of Anatomy and Neurosciences, Amsterdam UMC, Vrije Universiteit, Amsterdam, The Netherlands.
- Amsterdam Neuroscience, Program Neurodegeneration, Amsterdam, The Netherlands.
| |
Collapse
|
11
|
Jeong JY, Lee HJ, Kim N, Li Y, Rah JC, Oh WJ. Impaired neuronal activity as a potential factor contributing to the underdeveloped cerebrovasculature in a young Parkinson's disease mouse model. Sci Rep 2023; 13:22613. [PMID: 38114623 PMCID: PMC10730707 DOI: 10.1038/s41598-023-49900-w] [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: 05/22/2023] [Accepted: 12/13/2023] [Indexed: 12/21/2023] Open
Abstract
Misfolding of α-synuclein (α-Syn) in the brain causes cellular dysfunction, leading to cell death in a group of neurons, and consequently causes the progression of Parkinson's disease (PD). Although many studies have demonstrated the pathological connections between vascular dysfunction and neurodegenerative diseases, it remains unclear how neuronal accumulation of α-Syn affects the structural and functional aspects of the cerebrovasculature to accelerate early disease progression. Here, we demonstrated the effect of aberrant α-Syn expression on the brain vasculature using a PD mouse model expressing a familial mutant form of human α-Syn selectively in neuronal cells. We showed that young PD mice have an underdeveloped cerebrovasculature without significant α-Syn accumulation in the vasculature. During the early phase of PD, toxic α-Syn was selectively increased in neuronal cells, while endothelial cell proliferation was decreased in the absence of vascular cell death or neuroinflammation. Instead, we observed altered neuronal activation and minor changes in the activity-dependent gene expression in brain endothelial cells (ECs) in young PD mice. These findings demonstrated that neuronal expression of mutant α-Syn in the early stage of PD induces abnormal neuronal activity and contributes to vascular patterning defects, which could be associated with a reduced angiogenic potential of ECs.
Collapse
Affiliation(s)
- Jin-Young Jeong
- Neurovascular Biology Laboratory, Neurovascular Unit Research Group, Korea Brain Research Institute, Daegu, 41062, South Korea
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology, Daegu, 42988, South Korea
| | - Hyun Jung Lee
- Sensory and Motor System Research Group, Korea Brain Research Institute, Daegu, 41062, South Korea
| | - Namsuk Kim
- Neurovascular Biology Laboratory, Neurovascular Unit Research Group, Korea Brain Research Institute, Daegu, 41062, South Korea
| | - Yan Li
- Neurovascular Biology Laboratory, Neurovascular Unit Research Group, Korea Brain Research Institute, Daegu, 41062, South Korea
| | - Jong-Cheol Rah
- Sensory and Motor System Research Group, Korea Brain Research Institute, Daegu, 41062, South Korea
| | - Won-Jong Oh
- Neurovascular Biology Laboratory, Neurovascular Unit Research Group, Korea Brain Research Institute, Daegu, 41062, South Korea.
| |
Collapse
|
12
|
Bétemps D, Arsac JN, Nicot S, Canal D, Tlili H, Belondrade M, Morignat E, Verchère J, Gaillard D, Bruyère-Ostells L, Mayran C, Lakhdar L, Bougard D, Baron T. Protease-Sensitive and -Resistant Forms of Human and Murine Alpha-Synucleins in Distinct Brain Regions of Transgenic Mice (M83) Expressing the Human Mutated A53T Protein. Biomolecules 2023; 13:1788. [PMID: 38136658 PMCID: PMC10741842 DOI: 10.3390/biom13121788] [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: 09/27/2023] [Revised: 12/06/2023] [Accepted: 12/07/2023] [Indexed: 12/24/2023] Open
Abstract
Human neurodegenerative diseases associated with the misfolding of the alpha-synuclein (aS) protein (synucleinopathies) are similar to prion diseases to the extent that lesions are spread by similar molecular mechanisms. In a transgenic mouse model (M83) overexpressing a mutated (A53T) form of human aS, we had previously found that Protein Misfolding Cyclic Amplification (PMCA) triggered the aggregation of aS, which is associated with a high resistance to the proteinase K (PK) digestion of both human and murine aS, a major hallmark of the disease-associated prion protein. In addition, PMCA was also able to trigger the aggregation of murine aS in C57Bl/6 mouse brains after seeding with sick M83 mouse brains. Here, we show that intracerebral inoculations of M83 mice with C57Bl/6-PMCA samples strikingly shortens the incubation period before the typical paralysis that develops in this transgenic model, demonstrating the pathogenicity of PMCA-aggregated murine aS. In the hind brain regions of these sick M83 mice containing lesions with an accumulation of aS phosphorylated at serine 129, aS also showed a high PK resistance in the N-terminal part of the protein. In contrast to M83 mice, old APPxM83 mice co-expressing human mutated amyloid precursor and presenilin 1 proteins were seen to have an aggregation of aS, especially in the cerebral cortex, hippocampus and striatum, which also contained the highest load of aS phosphorylated at serine 129. This was proven by three techniques: a Western blot analysis of PK-resistant aS; an ELISA detection of aS aggregates; or the identification of aggregates of aS using immunohistochemical analyses of cytoplasmic/neuritic aS deposits. The results obtained with the D37A6 antibody suggest a higher involvement of murine aS in APPxM83 mice than in M83 mice. Our study used novel tools for the molecular study of synucleinopathies, which highlight similarities with the molecular mechanisms involved in prion diseases.
Collapse
Affiliation(s)
- Dominique Bétemps
- ANSES (French Agency for Food, Environmental and Occupational Health & Safety), University of Lyon, 69364 Lyon, France; (D.B.); (J.-N.A.); (D.C.); (H.T.); (E.M.); (J.V.); (D.G.); (L.L.)
| | - Jean-Noël Arsac
- ANSES (French Agency for Food, Environmental and Occupational Health & Safety), University of Lyon, 69364 Lyon, France; (D.B.); (J.-N.A.); (D.C.); (H.T.); (E.M.); (J.V.); (D.G.); (L.L.)
| | - Simon Nicot
- Pathogenesis and Control of Chronic and Emerging Infections, University of Montpellier, Inserm, Etablissement Français Du Sang, 34493 Montpellier, France; (S.N.); (M.B.); (L.B.-O.); (C.M.); (D.B.)
| | - Dominique Canal
- ANSES (French Agency for Food, Environmental and Occupational Health & Safety), University of Lyon, 69364 Lyon, France; (D.B.); (J.-N.A.); (D.C.); (H.T.); (E.M.); (J.V.); (D.G.); (L.L.)
| | - Habiba Tlili
- ANSES (French Agency for Food, Environmental and Occupational Health & Safety), University of Lyon, 69364 Lyon, France; (D.B.); (J.-N.A.); (D.C.); (H.T.); (E.M.); (J.V.); (D.G.); (L.L.)
| | - Maxime Belondrade
- Pathogenesis and Control of Chronic and Emerging Infections, University of Montpellier, Inserm, Etablissement Français Du Sang, 34493 Montpellier, France; (S.N.); (M.B.); (L.B.-O.); (C.M.); (D.B.)
| | - Eric Morignat
- ANSES (French Agency for Food, Environmental and Occupational Health & Safety), University of Lyon, 69364 Lyon, France; (D.B.); (J.-N.A.); (D.C.); (H.T.); (E.M.); (J.V.); (D.G.); (L.L.)
| | - Jérémy Verchère
- ANSES (French Agency for Food, Environmental and Occupational Health & Safety), University of Lyon, 69364 Lyon, France; (D.B.); (J.-N.A.); (D.C.); (H.T.); (E.M.); (J.V.); (D.G.); (L.L.)
| | - Damien Gaillard
- ANSES (French Agency for Food, Environmental and Occupational Health & Safety), University of Lyon, 69364 Lyon, France; (D.B.); (J.-N.A.); (D.C.); (H.T.); (E.M.); (J.V.); (D.G.); (L.L.)
| | - Lilian Bruyère-Ostells
- Pathogenesis and Control of Chronic and Emerging Infections, University of Montpellier, Inserm, Etablissement Français Du Sang, 34493 Montpellier, France; (S.N.); (M.B.); (L.B.-O.); (C.M.); (D.B.)
| | - Charly Mayran
- Pathogenesis and Control of Chronic and Emerging Infections, University of Montpellier, Inserm, Etablissement Français Du Sang, 34493 Montpellier, France; (S.N.); (M.B.); (L.B.-O.); (C.M.); (D.B.)
| | - Latifa Lakhdar
- ANSES (French Agency for Food, Environmental and Occupational Health & Safety), University of Lyon, 69364 Lyon, France; (D.B.); (J.-N.A.); (D.C.); (H.T.); (E.M.); (J.V.); (D.G.); (L.L.)
| | - Daisy Bougard
- Pathogenesis and Control of Chronic and Emerging Infections, University of Montpellier, Inserm, Etablissement Français Du Sang, 34493 Montpellier, France; (S.N.); (M.B.); (L.B.-O.); (C.M.); (D.B.)
| | - Thierry Baron
- ANSES (French Agency for Food, Environmental and Occupational Health & Safety), University of Lyon, 69364 Lyon, France; (D.B.); (J.-N.A.); (D.C.); (H.T.); (E.M.); (J.V.); (D.G.); (L.L.)
| |
Collapse
|
13
|
Alibekova Long M, Benman WKJ, Petrikas N, Bugaj LJ, Hughes AJ. Enhancing Single-Cell Western Blotting Sensitivity Using Diffusive Analyte Blotting and Antibody Conjugate Amplification. Anal Chem 2023; 95:17894-17902. [PMID: 37974303 DOI: 10.1021/acs.analchem.3c04130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2023]
Abstract
While there are many techniques to achieve highly sensitive, multiplex detection of RNA and DNA from single cells, detecting protein content often suffers from low limits of detection and throughput. Miniaturized, high-sensitivity Western blots on single cells (scWesterns) are attractive because they do not require advanced instrumentation. By physically separating analytes, scWesterns also uniquely mitigate limitations to target protein multiplexing posed by the affinity reagent performance. However, a fundamental limitation of scWesterns is their limited sensitivity for detecting low-abundance proteins, which arises from transport barriers posed by the separation gel against detection species. Here we address the sensitivity by decoupling the electrophoretic separation medium from the detection medium. We transfer scWestern separations to a nitrocellulose blotting medium with distinct mass transfer advantages over traditional in-gel probing, yielding a 5.9-fold improvement in the limit of detection. We next amplify probing of blotted proteins with enzyme-antibody conjugates, which are incompatible with traditional in-gel probing to achieve further improvement in the limit of detection to 1000 molecules, a 120-fold improvement. This enables us to detect 100% of cells in an EGFP-expressing population using fluorescently tagged and enzyme-conjugated antibodies compared to 84.5% of cells using in-gel detection. These results suggest the compatibility of nitrocellulose-immobilized scWesterns with a variety of affinity reagents─not previously accessible for in-gel use─for further signal amplification and detection of low-abundance targets.
Collapse
Affiliation(s)
- Mariia Alibekova Long
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Bioengineering Graduate Group, University of Pennsylvania, Philadelphia, Pennsylvania, 19104, United States
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - William K J Benman
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Bioengineering Graduate Group, University of Pennsylvania, Philadelphia, Pennsylvania, 19104, United States
| | - Nathan Petrikas
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Lukasz J Bugaj
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Bioengineering Graduate Group, University of Pennsylvania, Philadelphia, Pennsylvania, 19104, United States
- Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Alex J Hughes
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Bioengineering Graduate Group, University of Pennsylvania, Philadelphia, Pennsylvania, 19104, United States
- Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Center for Soft and Living Matter, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| |
Collapse
|
14
|
Schorova L, Bedard N, Khayachi A, Ho HH, Bolivar-Pedroso J, Huynh J, Piccirelli M, Wang Y, Plourde M, Luo W, Del Cid-Pellitero E, Shlaifer I, Castellanos-Montiel MJ, Yu Z, Valenzuela DVC, Lacalle-Aurioles M, Kriz A, Ye Y, Durcan TM, Wing SS. USP19 deubiquitinase inactivation regulates α-synuclein ubiquitination and inhibits accumulation of Lewy body-like aggregates in mice. NPJ Parkinsons Dis 2023; 9:157. [PMID: 38017009 PMCID: PMC10684503 DOI: 10.1038/s41531-023-00601-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 11/14/2023] [Indexed: 11/30/2023] Open
Abstract
The USP19 deubiquitinase is found in a locus associated with Parkinson's Disease (PD), interacts with chaperonins, and promotes secretion of α-synuclein (α-syn) through the misfolding-associated protein secretion (MAPS) pathway. Since these processes might modulate the processing of α-syn aggregates in PD, we inactivated USP19 (KO) in mice expressing the A53T mutation of α-syn and in whom α-syn preformed fibrils (PFF) had been injected in the striatum. Compared to WT, KO brains showed decreased accumulation of phospho-synuclein (pSyn) positive aggregates. This improvement was associated with less activation of microglia and improved performance in a tail-suspension test. Exposure of primary neurons from WT and KO mice to PFF in vitro also led to decreased accumulation of pSyn aggregates. KO did not affect uptake of PFF nor propagation of aggregates in the cultured neurons. We conclude that USP19 instead modulates intracellular dynamics of aggregates. At an early time following PFF injection when the number of pSyn-positive neurons were similar in WT and KO brains, the KO neurons contained less aggregates. KO brain aggregates stained more intensely with anti-ubiquitin antibodies. Immunoprecipitation of soluble proteins from WT and KO brains with antibodies to pSyn showed higher levels of ubiquitinated oligomeric species in the KO samples. We propose that the improved pathology in USP19 KO brains may arise from decreased formation or enhanced clearance of the more ubiquitinated aggregates and/or enhanced disassembly towards more soluble oligomeric species. USP19 inhibition may represent a novel therapeutic approach that targets the intracellular dynamics of α-syn complexes.
Collapse
Affiliation(s)
- Lenka Schorova
- Department of Medicine, McGill University and Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Nathalie Bedard
- Department of Medicine, McGill University and Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Anouar Khayachi
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| | - Hung-Hsiang Ho
- Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
| | - Joao Bolivar-Pedroso
- Department of Medicine, McGill University and Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Julie Huynh
- Department of Biochemistry, McGill University, Montreal, QC, Canada
| | - Mikaela Piccirelli
- Department of Medicine, McGill University and Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Yifei Wang
- Department of Medicine, McGill University and Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Marie Plourde
- Department of Medicine, McGill University and Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Wen Luo
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| | | | - Irina Shlaifer
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| | - María José Castellanos-Montiel
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
| | - Ziqi Yu
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| | | | | | - Anita Kriz
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| | - Yihong Ye
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Thomas M Durcan
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada.
- Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada.
| | - Simon S Wing
- Department of Medicine, McGill University and Research Institute of the McGill University Health Centre, Montreal, QC, Canada.
- Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada.
- Department of Biochemistry, McGill University, Montreal, QC, Canada.
| |
Collapse
|
15
|
Paslawski W, Svenningsson P. Elevated ApoE, ApoJ and lipoprotein-bound α-synuclein levels in cerebrospinal fluid from Parkinson's disease patients - Validation in the BioFIND cohort. Parkinsonism Relat Disord 2023; 116:105765. [PMID: 37479568 PMCID: PMC11140586 DOI: 10.1016/j.parkreldis.2023.105765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 06/12/2023] [Accepted: 07/11/2023] [Indexed: 07/23/2023]
Abstract
BACKGROUND The progressive accumulation, aggregation, and spread of α-synuclein (aSN) are common hallmarks of Parkinson's disease (PD) pathology. The genotype of apolipoprotein E (ApoE) influences PD progression. Recently we found that aSN co-localize with apolipoproteins on lipoprotein vesicles. We reported an increased level of ApoE, ApoJ and lipoprotein-bound aSN in CSF from early PD patients compared to matched controls. We also found reduced plasma ApoAI in PD patients. OBJECTIVE In this study, we used the same approach on the BioFIND cohort to validate our previous results and extended the studies to examine correlations with ApoE genotype, demographic variables, clinical symptoms and other biochemical findings reported in the BioFIND cohort. METHODS For the assessment, we used Western-Blot (WB) technique for apolipoproteins measurements in CSF and plasma from PD patients and healthy controls. Further, for measurement of aSN bound to lipoproteins, we combined immunodepletion with the enzyme-linked immunosorbent assay (ELISA). RESULTS Levels of ApoE, ApoJ and lipoprotein bound aSN were significantly increased in CSF from PD patients compared to controls. We also observed decreased levels of ApoAI and ApoJ in plasma from PD patients compared to controls. CONCLUSIONS Concluding, the present data validated our previous findings. Altered lipoproteins appear to be important in early PD pathology and may be involved in mechanisms underlying aSN cell-to-cell transfer in the nervous system and be developed in algorithms for early diagnosis of PD.
Collapse
Affiliation(s)
- Wojciech Paslawski
- Laboratory of Translational Neuropharmacology, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden.
| | - Per Svenningsson
- Laboratory of Translational Neuropharmacology, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden; Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom.
| |
Collapse
|
16
|
Hou X, Chen TH, Koga S, Bredenberg JM, Faroqi AH, Delenclos M, Bu G, Wszolek ZK, Carr JA, Ross OA, McLean PJ, Murray ME, Dickson DW, Fiesel FC, Springer W. Alpha-synuclein-associated changes in PINK1-PRKN-mediated mitophagy are disease context dependent. Brain Pathol 2023; 33:e13175. [PMID: 37259617 PMCID: PMC10467041 DOI: 10.1111/bpa.13175] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 05/05/2023] [Indexed: 06/02/2023] Open
Abstract
Alpha-synuclein (αsyn) aggregates are pathological features of several neurodegenerative conditions including Parkinson disease (PD), dementia with Lewy bodies, and multiple system atrophy (MSA). Accumulating evidence suggests that mitochondrial dysfunction and impairments of the autophagic-lysosomal system can contribute to the deposition of αsyn, which in turn may interfere with health and function of these organelles in a potentially vicious cycle. Here we investigated a potential convergence of αsyn with the PINK1-PRKN-mediated mitochondrial autophagy pathway in cell models, αsyn transgenic mice, and human autopsy brain. PINK1 and PRKN identify and selectively label damaged mitochondria with phosphorylated ubiquitin (pS65-Ub) to mark them for degradation (mitophagy). We found that disease-causing multiplications of αsyn resulted in accumulation of the ubiquitin ligase PRKN in cells. This effect could be normalized by starvation-induced autophagy activation and by CRISPR/Cas9-mediated αsyn knockout. Upon acute mitochondrial damage, the increased levels of PRKN protein contributed to an enhanced pS65-Ub response. We further confirmed increased pS65-Ub-immunopositive signals in mouse brain with αsyn overexpression and in postmortem human disease brain. Of note, increased pS65-Ub was associated with neuronal Lewy body-type αsyn pathology, but not glial cytoplasmic inclusions of αsyn as seen in MSA. While our results add another layer of complexity to the crosstalk between αsyn and the PINK1-PRKN pathway, distinct mechanisms may underlie in cells and brain tissue despite similar outcomes. Notwithstanding, our finding suggests that pS65-Ub may be useful as a biomarker to discriminate different synucleinopathies and may serve as a potential therapeutic target for Lewy body disease.
Collapse
Affiliation(s)
- Xu Hou
- Department of NeuroscienceMayo ClinicJacksonvilleFloridaUSA
| | | | - Shunsuke Koga
- Department of NeuroscienceMayo ClinicJacksonvilleFloridaUSA
| | | | - Ayman H. Faroqi
- Department of NeuroscienceMayo ClinicJacksonvilleFloridaUSA
- Neuroscience PhD ProgramMayo Clinic Graduate School of Biomedical SciencesJacksonvilleFloridaUSA
| | | | - Guojun Bu
- Department of NeuroscienceMayo ClinicJacksonvilleFloridaUSA
- Neuroscience PhD ProgramMayo Clinic Graduate School of Biomedical SciencesJacksonvilleFloridaUSA
| | | | - Jonathan A. Carr
- Division of Neurology, Department of Medicine, Faculty of Medicine and Health SciencesStellenbosch UniversityCape TownSouth Africa
| | - Owen A. Ross
- Department of NeuroscienceMayo ClinicJacksonvilleFloridaUSA
- Neuroscience PhD ProgramMayo Clinic Graduate School of Biomedical SciencesJacksonvilleFloridaUSA
| | - Pamela J. McLean
- Department of NeuroscienceMayo ClinicJacksonvilleFloridaUSA
- Neuroscience PhD ProgramMayo Clinic Graduate School of Biomedical SciencesJacksonvilleFloridaUSA
| | - Melissa E. Murray
- Department of NeuroscienceMayo ClinicJacksonvilleFloridaUSA
- Neuroscience PhD ProgramMayo Clinic Graduate School of Biomedical SciencesJacksonvilleFloridaUSA
| | - Dennis W. Dickson
- Department of NeuroscienceMayo ClinicJacksonvilleFloridaUSA
- Neuroscience PhD ProgramMayo Clinic Graduate School of Biomedical SciencesJacksonvilleFloridaUSA
| | - Fabienne C. Fiesel
- Department of NeuroscienceMayo ClinicJacksonvilleFloridaUSA
- Neuroscience PhD ProgramMayo Clinic Graduate School of Biomedical SciencesJacksonvilleFloridaUSA
| | - Wolfdieter Springer
- Department of NeuroscienceMayo ClinicJacksonvilleFloridaUSA
- Neuroscience PhD ProgramMayo Clinic Graduate School of Biomedical SciencesJacksonvilleFloridaUSA
| |
Collapse
|
17
|
Calogero AM, Basellini MJ, Isilgan HB, Longhena F, Bellucci A, Mazzetti S, Rolando C, Pezzoli G, Cappelletti G. Acetylated α-Tubulin and α-Synuclein: Physiological Interplay and Contribution to α-Synuclein Oligomerization. Int J Mol Sci 2023; 24:12287. [PMID: 37569662 PMCID: PMC10418364 DOI: 10.3390/ijms241512287] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 07/26/2023] [Accepted: 07/28/2023] [Indexed: 08/13/2023] Open
Abstract
Emerging evidence supports that altered α-tubulin acetylation occurs in Parkinson's disease (PD), a neurodegenerative disorder characterized by the deposition of α-synuclein fibrillary aggregates within Lewy bodies and nigrostriatal neuron degeneration. Nevertheless, studies addressing the interplay between α-tubulin acetylation and α-synuclein are lacking. Here, we investigated the relationship between α-synuclein and microtubules in primary midbrain murine neurons and the substantia nigra of post-mortem human brains. Taking advantage of immunofluorescence and Proximity Ligation Assay (PLA), a method allowing us to visualize protein-protein interactions in situ, combined with confocal and super-resolution microscopy, we found that α-synuclein and acetylated α-tubulin colocalized and were in close proximity. Next, we employed an α-synuclein overexpressing cellular model and tested the role of α-tubulin acetylation in α-synuclein oligomer formation. We used the α-tubulin deacetylase HDAC6 inhibitor Tubacin to modulate α-tubulin acetylation, and we evaluated the presence of α-synuclein oligomers by PLA. We found that the increase in acetylated α-tubulin significantly induced α-synuclein oligomerization. In conclusion, we unraveled the link between acetylated α-tubulin and α-synuclein and demonstrated that α-tubulin acetylation could trigger the early step of α-synuclein aggregation. These data suggest that the proper regulation of α-tubulin acetylation might be considered a therapeutic strategy to take on PD.
Collapse
Affiliation(s)
- Alessandra Maria Calogero
- Department of Biosciences, Università degli Studi di Milano, 20133 Milan, Italy; (M.J.B.); (H.B.I.); (S.M.); (C.R.)
- Fondazione Grigioni per il Morbo di Parkinson, 20125 Milan, Italy;
| | - Milo Jarno Basellini
- Department of Biosciences, Università degli Studi di Milano, 20133 Milan, Italy; (M.J.B.); (H.B.I.); (S.M.); (C.R.)
| | - Huseyin Berkcan Isilgan
- Department of Biosciences, Università degli Studi di Milano, 20133 Milan, Italy; (M.J.B.); (H.B.I.); (S.M.); (C.R.)
| | - Francesca Longhena
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy; (F.L.); (A.B.)
| | - Arianna Bellucci
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy; (F.L.); (A.B.)
| | - Samanta Mazzetti
- Department of Biosciences, Università degli Studi di Milano, 20133 Milan, Italy; (M.J.B.); (H.B.I.); (S.M.); (C.R.)
- Fondazione Grigioni per il Morbo di Parkinson, 20125 Milan, Italy;
| | - Chiara Rolando
- Department of Biosciences, Università degli Studi di Milano, 20133 Milan, Italy; (M.J.B.); (H.B.I.); (S.M.); (C.R.)
| | - Gianni Pezzoli
- Fondazione Grigioni per il Morbo di Parkinson, 20125 Milan, Italy;
- Parkinson Institute, ASST-Pini-CTO, 20126 Milan, Italy
| | - Graziella Cappelletti
- Department of Biosciences, Università degli Studi di Milano, 20133 Milan, Italy; (M.J.B.); (H.B.I.); (S.M.); (C.R.)
- Center of Excellence on Neurodegenerative Diseases, Università degli Studi di Milano, 20133 Milan, Italy
| |
Collapse
|
18
|
Ladouce R, Combes GF, Trajković K, Drmić Hofman I, Merćep M. Oxime blot: A novel method for reliable and sensitive detection of carbonylated proteins in diverse biological systems. Redox Biol 2023; 63:102743. [PMID: 37207613 DOI: 10.1016/j.redox.2023.102743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 05/11/2023] [Indexed: 05/21/2023] Open
Abstract
Oxidative stress and oxidative protein damage occur in various biological processes and diseases. The carbonyl group on amino acid side chains is the most widely used protein oxidation biomarker. Carbonyl groups are commonly detected indirectly through their reaction with 2,4-dinitrophenylhydrazine (DNPH) and subsequent labeling with an anti-DNP antibody. However, the DNPH immunoblotting method lacks protocol standardization, exhibits technical bias, and has low reliability. To overcome these shortcomings, we have developed a new blotting method in which the carbonyl group reacts with the biotin-aminooxy probe to form a chemically stable oxime bond. The reaction speed and the extent of the carbonyl group derivatization are increased by adding a p-phenylenediamine (pPDA) catalyst under neutral pH conditions. These improvements are crucial since they ensure that the carbonyl derivatization reaction reaches a plateau within hours and increases the sensitivity and robustness of protein carbonyl detection. Furthermore, derivatization under pH-neutral conditions facilitates a good SDS-PAGE protein migration pattern, avoids protein loss by acidic precipitation, and is directly compatible with protein immunoprecipitation. This work describes the new Oxime blot method and demonstrates its use in detecting protein carbonylation in complex matrices from diverse biological samples.
Collapse
Affiliation(s)
- Romain Ladouce
- Mediterranean Institute for Life Sciences (MedILS), Meštrovićevo šetalište 45, 21000, Split, Croatia
| | - Guillaume Fabien Combes
- Mediterranean Institute for Life Sciences (MedILS), Meštrovićevo šetalište 45, 21000, Split, Croatia; Center of Excellence for Science and Technology-Integration of Mediterranean Region (STIM), Faculty of Science, University of Split, 21000, Split, Croatia.
| | - Katarina Trajković
- Mediterranean Institute for Life Sciences (MedILS), Meštrovićevo šetalište 45, 21000, Split, Croatia; Center of Excellence for Science and Technology-Integration of Mediterranean Region (STIM), Faculty of Science, University of Split, 21000, Split, Croatia
| | - Irena Drmić Hofman
- University Department of Health Studies, University of Split, 21000, Split, Croatia; School of Medicine, University of Split, 21000, Split, Croatia
| | - Mladen Merćep
- Mediterranean Institute for Life Sciences (MedILS), Meštrovićevo šetalište 45, 21000, Split, Croatia; Department of Biotechnology, University of Rijeka, Radmile Matejčić 2, 51000, Rijeka, Croatia; Zora Foundation, Ruđera Boškovića 21, 21000, Split, Croatia.
| |
Collapse
|
19
|
Sánchez-Vidaña DI, Li J, Abokyi S, Chan JNM, Ngai SPC, Lau BWM. In vitro methods in autophagy research: Applications in neurodegenerative diseases and mood disorders. Front Mol Neurosci 2023; 16:1168948. [PMID: 37122628 PMCID: PMC10130388 DOI: 10.3389/fnmol.2023.1168948] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Accepted: 03/14/2023] [Indexed: 05/02/2023] Open
Abstract
Background Autophagy is a conserved physiological intracellular mechanism responsible for the degradation and recycling of cytoplasmic constituents (e.g., damaged organelles, and protein aggregates) to maintain cell homeostasis. Aberrant autophagy has been observed in neurodegenerative diseases, including Alzheimer's Disease (AD), Parkinson's Disease (PD), Amyotrophic Lateral Sclerosis (ALS), and Huntington's Disease (HD), and recently aberrant autophagy has been associated with mood disorders, such as depression. Several in vitro methods have been developed to study the complex and tightly regulated mechanisms of autophagy. In vitro methods applied to autophagy research are used to identify molecular key players involved in dysfunctional autophagy and to screen autophagy regulators with therapeutic applications in neurological diseases and mood disorders. Therefore, the aims of this narrative review are (1) to compile information on the cell-based methods used in autophagy research, (2) to discuss their application, and (3) to create a catalog of traditional and novel in vitro methods applied in neurodegenerative diseases and depression. Methods Pubmed and Google Scholar were used to retrieve relevant in vitro studies on autophagy mechanisms in neurological diseases and depression using a combination of search terms per mechanism and disease (e.g., "macroautophagy" and "Alzheimer's disease"). A total of 37 studies were included (14 in PD, 8 in AD, 5 in ALS, 5 in %, and 5 in depression). Results A repertoire of traditional and novel approaches and techniques was compiled and discussed. The methods used in autophagy research focused on the mechanisms of macroautophagy, microautophagy, and chaperone-mediated autophagy. The in vitro tools presented in this review can be applied to explore pathophysiological mechanisms at a molecular level and to screen for potential therapeutic agents and their mechanism of action, which can be of great importance to understanding disease biology and potential therapeutic options in the context of neurodegenerative disorders and depression. Conclusion This is the first review to compile, discuss, and provide a catalog of traditional and novel in vitro models applied to neurodegenerative disorders and depression.
Collapse
Affiliation(s)
- Dalinda Isabel Sánchez-Vidaña
- Department of Rehabilitation Sciences, Faculty of Health and Social Sciences, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China
- Mental Health Research Centre, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China
| | - Jingjing Li
- Department of Rehabilitation Sciences, Faculty of Health and Social Sciences, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China
| | - Samuel Abokyi
- School of Optometry, Faculty of Health and Social Sciences, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China
| | - Jackie Ngai-Man Chan
- Department of Rehabilitation Sciences, Faculty of Health and Social Sciences, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China
| | - Shirley Pui-Ching Ngai
- Department of Rehabilitation Sciences, Faculty of Health and Social Sciences, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China
| | - Benson Wui-Man Lau
- Department of Rehabilitation Sciences, Faculty of Health and Social Sciences, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China
- Mental Health Research Centre, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China
| |
Collapse
|
20
|
Song S, Tu D, Meng C, Liu J, Wilson B, Wang Q, Shih YYI, Gao HM, Hong JS. Dysfunction of the noradrenergic system drives inflammation, α-synucleinopathy, and neuronal loss in mouse colon. Front Immunol 2023; 14:1083513. [PMID: 36845109 PMCID: PMC9950510 DOI: 10.3389/fimmu.2023.1083513] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Accepted: 02/01/2023] [Indexed: 02/12/2023] Open
Abstract
Clinical and pathological evidence revealed that α-synuclein (α-syn) pathology seen in PD patients starts in the gut and spreads via anatomically connected structures from the gut to the brain. Our previous study demonstrated that depletion of central norepinephrine (NE) disrupted brain immune homeostasis, producing a spatiotemporal order of neurodegeneration in the mouse brain. The purpose of this study was 1) to determine the role of peripheral noradrenergic system in the maintenance of gut immune homeostasis and in the pathogenesis of PD and 2) to investigate whether NE-depletion induced PD-like α-syn pathological changes starts from the gut. For these purposes, we investigated time-dependent changes of α-synucleinopathy and neuronal loss in the gut following a single injection of DSP-4 (a selective noradrenergic neurotoxin) to A53T-SNCA (human mutant α-syn) over-expression mice. We found DPS-4 significantly reduced the tissue level of NE and increased immune activities in gut, characterized by increased number of phagocytes and proinflammatory gene expression. Furthermore, a rapid-onset of α-syn pathology was observed in enteric neurons after 2 weeks and delayed dopaminergic neurodegeneration in the substantia nigra was detected after 3-5 months, associated with the appearance of constipation and impaired motor function, respectively. The increased α-syn pathology was only observed in large, but not in the small, intestine, which is similar to what was observed in PD patients. Mechanistic studies reveal that DSP-4-elicited upregulation of NADPH oxidase (NOX2) initially occurred only in immune cells during the acute intestinal inflammation stage, and then spread to enteric neurons and mucosal epithelial cells during the chronic inflammation stage. The upregulation of neuronal NOX2 correlated well with the extent of α-syn aggregation and subsequent enteric neuronal loss, suggesting that NOX2-generated reactive oxygen species play a key role in α-synucleinopathy. Moreover, inhibiting NOX2 by diphenyleneiodonium or restoring NE function by salmeterol (a β2-receptor agonist) significantly attenuated colon inflammation, α-syn aggregation/propagation, and enteric neurodegeneration in the colon and ameliorated subsequent behavioral deficits. Taken together, our model of PD shows a progressive pattern of pathological changes from the gut to the brain and suggests a potential role of the noradrenergic dysfunction in the pathogenesis of PD.
Collapse
Affiliation(s)
- Sheng Song
- Neuropharmacology Section, Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, United States
- Biomedical Research Imaging Center, University of North Caroline at Chapel Hill, Chapel Hill, NC, United States
| | - Dezhen Tu
- Neuropharmacology Section, Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, United States
- Ministry of Education (MOE) Key Laboratory of Model Animal for Disease Study, Institute for Brain Sciences, Jiangsu Key Laboratory of Molecular Medicine, Model Animal Research Center, School of medicine, Nanjing University, Nanjing, China
| | - Chengbo Meng
- In Vivo Neurobiology Group, Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, United States
| | - Jie Liu
- Neuropharmacology Section, Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, United States
| | - Belinda Wilson
- Neuropharmacology Section, Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, United States
| | - Qingshan Wang
- National-Local Joint Engineering Research Center for Drug-Research and Development (R & D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, China
| | - Yen-Yu Ian Shih
- Biomedical Research Imaging Center, University of North Caroline at Chapel Hill, Chapel Hill, NC, United States
| | - Hui-Ming Gao
- Ministry of Education (MOE) Key Laboratory of Model Animal for Disease Study, Institute for Brain Sciences, Jiangsu Key Laboratory of Molecular Medicine, Model Animal Research Center, School of medicine, Nanjing University, Nanjing, China
| | - Jau-Shyong Hong
- Neuropharmacology Section, Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, United States
| |
Collapse
|
21
|
Herman S, Djaldetti R, Mollenhauer B, Offen D. CSF-derived extracellular vesicles from patients with Parkinson's disease induce symptoms and pathology. Brain 2023; 146:209-224. [PMID: 35881523 DOI: 10.1093/brain/awac261] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 06/23/2022] [Accepted: 07/01/2022] [Indexed: 01/11/2023] Open
Abstract
Parkinson's disease is characterized by the gradual appearance of intraneuronal inclusions that are primarily composed of misfolded α-synuclein protein, leading to cytotoxicity and neural death. Recent in vitro and in vivo studies suggest that misfolded α-synuclein may spread transcellularly in a prion-like manner, inducing pathological aggregates in healthy neurons, and is disseminated via secretion of extracellular vesicles. Accordingly, extracellular vesicles derived from brain lysates and CSF of patients with Parkinson's disease were shown to facilitate α-synuclein aggregation in healthy cells. Prompted by the hypothesis of Braak and colleagues that the olfactory bulb is one of the primary propagation sites for the initiation of Parkinson's disease, we sought to investigate the role of extracellular vesicles in the spread of α-synuclein and progression of Parkinson's disease through the olfactory bulb. Extracellular vesicles derived from the CSF of patients diagnosed with Parkinson's disease or with a non-synucleinopathy neurodegenerative disorder were administered intranasally to healthy mice, once daily over 4 days. Three months later, mice were subjected to motor and non-motor tests. Functional impairments were elucidated by histochemical analysis of midbrain structures relevant to Parkinson's disease pathology, 8 months after EVs treatment. Mice treated with extracellular vesicles from the patients with Parkinson's disease displayed multiple symptoms consistent with prodromal and clinical-phase Parkinson's disease such as hyposmia, motor behaviour impairments and high anxiety levels. Furthermore, their midbrains showed widespread α-synuclein aggregations, dopaminergic neurodegeneration, neuroinflammation and altered autophagy activity. Several unconventional pathologies were also observed, such as α-synuclein aggregations in the red nucleus, growth of premature grey hair and astrogliosis. Collectively, these data indicate that intranasally administered extracellular vesicles derived from the CSF of patients with Parkinson's disease can propagate α-synuclein aggregation in vivo and trigger Parkinson's disease-like symptoms and pathology in healthy mice.
Collapse
Affiliation(s)
- Shay Herman
- Department of Human Genetics and Biochemistry, Sackler School of Medicine, and Felsenstein Medical Research Center, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Ruth Djaldetti
- Department of Neurology, Rabin Medical Center-Beilinson Hospital, Petach Tikva 4941492, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Brit Mollenhauer
- Department of Neurology, University Medical Center Göttingen, Göttingen, Germany.,Paracelsus-Elena-Klinik, Kassel, Germany
| | - Daniel Offen
- Department of Human Genetics and Biochemistry, Sackler School of Medicine, and Felsenstein Medical Research Center, Tel Aviv University, Tel Aviv 6997801, Israel.,Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 6997801, Israel
| |
Collapse
|
22
|
Lassozé S, de Guilhem de Lataillade A, Oullier T, Neunslist M, Leclair-Visonneau L, Derkinderen P, Paillusson S. Comparison of commercially available antibodies for the detection of phosphorylated alpha-synuclein in primary culture of ENS. Neurogastroenterol Motil 2022; 34:e14354. [PMID: 35279896 DOI: 10.1111/nmo.14354] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 02/04/2022] [Accepted: 02/08/2022] [Indexed: 12/24/2022]
Abstract
BACKGROUND It is now well established that phosphorylated alpha-synuclein histopathology, the pathologic hallmark of Parkinson's disease (PD) is not limited to the brain but also extends to the enteric nervous system (ENS). This observation led to the hypothesis that the ENS could play a pivotal role in the development of PD. Research on the enteric synucleinopathy has, however, been hampered by difficulties in detecting phosphorylated alpha-synuclein in the ENS by Western blotting, even when the transferred membrane is fixed with an optimized protocol. This suggests that the available antibodies used in previous studies lacked of sensitivity for the detection of phosphorylated alpha-synuclein at Ser129 in enteric neurons. Here, we evaluated three recent commercially available phospho-alpha-synuclein antibodies and compared them to two antibodies used in previous research. METHODS The specificity and sensitivity of the 5 antibodies were evaluated by Western blot performed with recombinant alpha-synuclein and with protein lysates from rat primary cultures of ENS. In primary culture of ENS, additional experiments were performed with the most specific antibody in order to modulate alpha-synuclein phosphorylation and to validate its utilization in immunofluorescence experiments. RESULTS The rabbit monoclonal antibody D1R1R uniquely and robustly detected endogenous phosphorylated alpha-synuclein at Ser129 in rat primary culture of ENS without any non-specific bands, allowing for a reliable analysis of phosphorylated alpha-synuclein regulation by pharmacologic means. CONCLUSIONS AND INFERENCES Using D1R1R antibody together with the optimized protocol for membrane fixation may help deciphering the signaling pathways involved in enteric alpha-synuclein post-translational regulation in PD.
Collapse
Affiliation(s)
- Simon Lassozé
- Université de Nantes, Inserm, TENS, The Enteric Nervous System in Gut and Brain Diseases, IMAD, Nantes, France.,Department of Neurology, CHU Nantes, Nantes, France
| | - Adrien de Guilhem de Lataillade
- Université de Nantes, Inserm, TENS, The Enteric Nervous System in Gut and Brain Diseases, IMAD, Nantes, France.,Department of Neurology, CHU Nantes, Nantes, France
| | - Thibauld Oullier
- Université de Nantes, Inserm, TENS, The Enteric Nervous System in Gut and Brain Diseases, IMAD, Nantes, France
| | - Michel Neunslist
- Université de Nantes, Inserm, TENS, The Enteric Nervous System in Gut and Brain Diseases, IMAD, Nantes, France
| | - Laurène Leclair-Visonneau
- Université de Nantes, Inserm, TENS, The Enteric Nervous System in Gut and Brain Diseases, IMAD, Nantes, France.,Department of Neurology, CHU Nantes, Nantes, France
| | - Pascal Derkinderen
- Université de Nantes, Inserm, TENS, The Enteric Nervous System in Gut and Brain Diseases, IMAD, Nantes, France.,Department of Neurology, CHU Nantes, Nantes, France
| | - Sébastien Paillusson
- Université de Nantes, Inserm, TENS, The Enteric Nervous System in Gut and Brain Diseases, IMAD, Nantes, France
| |
Collapse
|
23
|
Leite K, Garg P, Spitzner FP, Guerin Darvas S, Bähr M, Priesemann V, Kügler S. α-Synuclein Impacts on Intrinsic Neuronal Network Activity Through Reduced Levels of Cyclic AMP and Diminished Numbers of Active Presynaptic Terminals. Front Mol Neurosci 2022; 15:868790. [PMID: 35721317 PMCID: PMC9199018 DOI: 10.3389/fnmol.2022.868790] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 04/08/2022] [Indexed: 11/23/2022] Open
Abstract
α-synuclein (α-Syn) is intimately linked to synucleinopathies like Parkinson’s disease and dementia with Lewy bodies. However, the pathophysiological mechanisms that are triggered by this protein are still largely enigmatic. α-Syn overabundance may cause neurodegeneration through protein accumulation and mitochondrial deterioration but may also result in pathomechanisms independent from neuronal cell death. One such proposed pathological mechanism is the influence of α-Syn on non-stimulated, intrinsic brain activity. This activity is responsible for more than 90% of the brain’s energyconsumption, and is thus thought to play an eminent role in basic brain functionality. Here we report that α-Syn substantially disrupts intrinsic neuronal network burst activity in a long-term neuronal cell culture model. Mechanistically, the impairment of network activity originates from reduced levels of cyclic AMP and cyclic AMP-mediated signaling as well as from diminished numbers of active presynaptic terminals. The profound reduction of network activity due to α-Syn was mediated only by intracellularly expressed α-Syn, but not by α-Syn that is naturally released by neurons. Conversely, extracellular pre-formed fibrils of α-Syn mimicked the effect of intracellular α-Syn, suggesting that they trigger an off-target mechanism that is not activated by naturally released α-Syn. A simulation-based model of the network activity in our cultures demonstrated that even subtle effect sizes in reducing outbound connectivity, i.e., loss of active synapses, can cause substantial global reductions in non-stimulated network activity. These results suggest that even low-level loss of synaptic output capabilities caused by α-Syn may result in significant functional impairments in terms of intrinsic neuronal network activity. Provided that our model holds true for the human brain, then α-Syn may cause significant functional lesions independent from neurodegeneration.
Collapse
Affiliation(s)
- Kristian Leite
- Department of Neurology, University Medical Center Göttingen, Göttingen, Germany
| | - Pretty Garg
- Department of Neurology, University Medical Center Göttingen, Göttingen, Germany
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Göttingen, Germany
| | - F. Paul Spitzner
- Neural Systems Theory group, Max-Planck-Institute for Dynamics and Self-Organization, Göttingen, Germany
| | - Sofia Guerin Darvas
- Department of Neurology, University Medical Center Göttingen, Göttingen, Germany
| | - Mathias Bähr
- Department of Neurology, University Medical Center Göttingen, Göttingen, Germany
| | - Viola Priesemann
- Neural Systems Theory group, Max-Planck-Institute for Dynamics and Self-Organization, Göttingen, Germany
- Institute for the Dynamics of Complex Systems, University of Göttingen, Göttingen, Germany
| | - Sebastian Kügler
- Department of Neurology, University Medical Center Göttingen, Göttingen, Germany
- *Correspondence: Sebastian Kügler
| |
Collapse
|
24
|
Anti-Aggregative Effect of the Antioxidant DJ-1 on the TPPP/p25-Derived Pathological Associations of Alpha-Synuclein. Cells 2021; 10:cells10112909. [PMID: 34831132 PMCID: PMC8616041 DOI: 10.3390/cells10112909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 10/22/2021] [Accepted: 10/25/2021] [Indexed: 11/16/2022] Open
Abstract
DJ-1, a multi-functional protein with antioxidant properties, protects dopaminergic neurons against Parkinson’s disease (PD). The oligomerization/assembly of alpha-synuclein (SYN), promoted by Tubulin Polymerization Promoting Protein (TPPP/p25), is fatal in the early stage of PD. The pathological assembly of SYN with TPPP/p25 inhibits their proteolytic degradation. In this work, we identified DJ-1 as a new interactive partner of TPPP/p25, and revealed its influence on the association of TPPP/p25 with SYN. DJ-1 did not affect the TPPP/p25-derived tubulin polymerization; however, it did impede the toxic assembly of TPPP/p25 with SYN. The interaction of DJ-1 with TPPP/p25 was visualized in living human cells by fluorescence confocal microscopy coupled with Bifunctional Fluorescence Complementation (BiFC). While the transfected DJ-1 displayed homogeneous intracellular distribution, the TPPP/p25-DJ-1 complex was aligned along the microtubule network. The anti-aggregative effect of DJ-1 on the pathological TPPP/p25-SYN assemblies was established by the decrease in the intensity of their intracellular fluorescence (BiFC signal) and the increase in the proteolytic degradation of SYN complexed with TPPP/p25 due to the DJ-1-derived disassembly of SYN with TPPP/p25. These data obtained with HeLa and SH-SY5Y cells revealed the protective effect of DJ-1 against toxic SYN assemblies, which assigns a new function to the antioxidant sensor DJ-1.
Collapse
|
25
|
Lai TT, Kim YJ, Nguyen PT, Koh YH, Nguyen TT, Ma HI, Kim YE. Temporal Evolution of Inflammation and Neurodegeneration With Alpha-Synuclein Propagation in Parkinson's Disease Mouse Model. Front Integr Neurosci 2021; 15:715190. [PMID: 34675786 PMCID: PMC8523784 DOI: 10.3389/fnint.2021.715190] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 09/01/2021] [Indexed: 12/30/2022] Open
Abstract
According to a few studies, α-synuclein (αSyn) propagation has been suggested to play a key role in the pathomechanism of Parkinson's disease (PD), but neurodegeneration and the involvement of inflammation in its pathologic progression are not well understood with regard to temporal relationship. In this study, with the help of the PD mouse model injected with intrastriatal αSyn preformed fibril (PFF), the temporal evolution of αSyn propagation, inflammation, and neurodegeneration was explored in the perspective of the striatum and the whole brain. In the PFF-injected striatum, inflammatory response cells, including microglia and astrocytes, were activated at the earliest stage and reduced with time, and the phosphorylated form of αSyn accumulation increased behind it. Afterward, the degeneration of striatal dopaminergic neurons became significant with the conspicuity of behavioral phenotype. Similar patterns of forefront eruption of inflammation and then followed by αSyn propagation were noted in the opposite striatum, which were not injured by PFF injection. In analyzing the whole brain, inflammatory responses were activated at the earliest stage, and the soluble αSyn expression increased concurrently. The inflammatory response decreased afterward, and the accumulation of the insoluble form of αSyn increased behind it. Our results suggested that the inflammatory response may precede the accumulation of the pathologic form of αSyn; thereafter, the neurodegeneration and motor dysfunction followed αSyn proliferation in the PD mouse model. From this model, recognizing the temporal relationship between inflammation, αSyn propagation, and neurodegeneration may be helpful in establishing the PD animal model and monitoring the effect of interventional therapy.
Collapse
Affiliation(s)
- Thuy Thi Lai
- Department of Biomedical Gerontology, Graduate School of Hallym University, Chuncheon, South Korea.,Department of Neurology, Hallym University Sacred Heart Hospital, Hallym University, Anyang, South Korea.,Hallym Neurological Institute, Hallym University, Anyang, South Korea
| | - Yun Joong Kim
- Department of Neurology, Yongin Severance Hospital, Yonsei University College of Medicine, Yongin, South Korea
| | - Phuong Thi Nguyen
- Department of Biomedical Gerontology, Graduate School of Hallym University, Chuncheon, South Korea.,Ilsong Institute of Life Science, Hallym University, Anyang, South Korea
| | - Young Ho Koh
- Department of Biomedical Gerontology, Graduate School of Hallym University, Chuncheon, South Korea.,Ilsong Institute of Life Science, Hallym University, Anyang, South Korea
| | - Tinh Thi Nguyen
- Department of Biomedical Gerontology, Graduate School of Hallym University, Chuncheon, South Korea.,Ilsong Institute of Life Science, Hallym University, Anyang, South Korea
| | - Hyeo-Il Ma
- Department of Neurology, Hallym University Sacred Heart Hospital, Hallym University, Anyang, South Korea.,Hallym Neurological Institute, Hallym University, Anyang, South Korea
| | - Young Eun Kim
- Department of Neurology, Hallym University Sacred Heart Hospital, Hallym University, Anyang, South Korea.,Hallym Neurological Institute, Hallym University, Anyang, South Korea
| |
Collapse
|
26
|
Mohamed NV, Sirois J, Ramamurthy J, Mathur M, Lépine P, Deneault E, Maussion G, Nicouleau M, Chen CXQ, Abdian N, Soubannier V, Cai E, Nami H, Thomas RA, Wen D, Tabatabaei M, Beitel LK, Singh Dolt K, Karamchandani J, Stratton JA, Kunath T, Fon EA, Durcan TM. Midbrain organoids with an SNCA gene triplication model key features of synucleinopathy. Brain Commun 2021; 3:fcab223. [PMID: 34632384 PMCID: PMC8495137 DOI: 10.1093/braincomms/fcab223] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 08/06/2021] [Accepted: 08/16/2021] [Indexed: 02/05/2023] Open
Abstract
SNCA, the first gene associated with Parkinson's disease, encodes the α-synuclein protein, the predominant component within pathological inclusions termed Lewy bodies. The presence of Lewy bodies is one of the classical hallmarks found in the brain of patients with Parkinson's disease, and Lewy bodies have also been observed in patients with other synucleinopathies. However, the study of α-synuclein pathology in cells has relied largely on two-dimensional culture models, which typically lack the cellular diversity and complex spatial environment found in the brain. Here, to address this gap, we use three-dimensional midbrain organoids, differentiated from human-induced pluripotent stem cells derived from patients carrying a triplication of the SNCA gene and from CRISPR/Cas9 corrected isogenic control iPSCs. These human midbrain organoids recapitulate key features of α-synuclein pathology observed in the brains of patients with synucleinopathies. In particular, we find that SNCA triplication human midbrain organoids express elevated levels of α-synuclein and exhibit an age-dependent increase in α-synuclein aggregation, manifested by the presence of both oligomeric and phosphorylated forms of α-synuclein. These phosphorylated α-synuclein aggregates were found in both neurons and glial cells and their time-dependent accumulation correlated with a selective reduction in dopaminergic neuron numbers. Thus, human midbrain organoids from patients carrying SNCA gene multiplication can reliably model key pathological features of Parkinson's disease and provide a powerful system to study the pathogenesis of synucleinopathies.
Collapse
Affiliation(s)
- Nguyen-Vi Mohamed
- Early Drug Discovery Unit (EDDU), Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal, Quebec H3A 2B4, Canada
| | - Julien Sirois
- Early Drug Discovery Unit (EDDU), Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal, Quebec H3A 2B4, Canada
| | - Janani Ramamurthy
- Early Drug Discovery Unit (EDDU), Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal, Quebec H3A 2B4, Canada
| | - Meghna Mathur
- Early Drug Discovery Unit (EDDU), Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal, Quebec H3A 2B4, Canada
| | - Paula Lépine
- Early Drug Discovery Unit (EDDU), Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal, Quebec H3A 2B4, Canada
| | - Eric Deneault
- Early Drug Discovery Unit (EDDU), Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal, Quebec H3A 2B4, Canada
| | - Gilles Maussion
- Early Drug Discovery Unit (EDDU), Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal, Quebec H3A 2B4, Canada
| | - Michael Nicouleau
- Early Drug Discovery Unit (EDDU), Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal, Quebec H3A 2B4, Canada
| | - Carol X-Q Chen
- Early Drug Discovery Unit (EDDU), Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal, Quebec H3A 2B4, Canada
| | - Narges Abdian
- Early Drug Discovery Unit (EDDU), Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal, Quebec H3A 2B4, Canada
| | - Vincent Soubannier
- Early Drug Discovery Unit (EDDU), Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal, Quebec H3A 2B4, Canada
| | - Eddie Cai
- Early Drug Discovery Unit (EDDU), Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal, Quebec H3A 2B4, Canada
| | - Harris Nami
- Early Drug Discovery Unit (EDDU), Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal, Quebec H3A 2B4, Canada
| | - Rhalena A Thomas
- Early Drug Discovery Unit (EDDU), Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal, Quebec H3A 2B4, Canada
| | - Dingke Wen
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu 610063, China
| | - Mahdieh Tabatabaei
- Early Drug Discovery Unit (EDDU), Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal, Quebec H3A 2B4, Canada.,C-BIG Biorepository (C-BIG), Montreal Neurological Institute-Hospital, McGill University, Montreal, Quebec H3A 2B4, Canada
| | - Lenore K Beitel
- Early Drug Discovery Unit (EDDU), Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal, Quebec H3A 2B4, Canada
| | - Karamjit Singh Dolt
- Centre for Regenerative Medicine, School of Biological Sciences, University of Edinburgh, Edinburgh EH16 4UU, UK
| | - Jason Karamchandani
- Early Drug Discovery Unit (EDDU), Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal, Quebec H3A 2B4, Canada.,C-BIG Biorepository (C-BIG), Montreal Neurological Institute-Hospital, McGill University, Montreal, Quebec H3A 2B4, Canada
| | - Jo Anne Stratton
- Montreal Neurological Institute-Hospital, McGill University, Montreal, Quebec H3A 2B4, Canada
| | - Tilo Kunath
- Centre for Regenerative Medicine, School of Biological Sciences, University of Edinburgh, Edinburgh EH16 4UU, UK
| | - Edward A Fon
- Early Drug Discovery Unit (EDDU), Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal, Quebec H3A 2B4, Canada
| | - Thomas M Durcan
- Early Drug Discovery Unit (EDDU), Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal, Quebec H3A 2B4, Canada
| |
Collapse
|
27
|
Albert K, Raymundo DP, Panhelainen A, Eesmaa A, Shvachiy L, Araújo GR, Chmielarz P, Yan X, Singh A, Cordeiro Y, Palhano FL, Foguel D, Luk KC, Domanskyi A, Voutilainen MH, Huttunen HJ, Outeiro TF, Saarma M, Almeida MS, Airavaara M. Cerebral dopamine neurotrophic factor reduces α-synuclein aggregation and propagation and alleviates behavioral alterations in vivo. Mol Ther 2021; 29:2821-2840. [PMID: 33940158 PMCID: PMC8417450 DOI: 10.1016/j.ymthe.2021.04.035] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 02/11/2021] [Accepted: 04/27/2021] [Indexed: 01/16/2023] Open
Abstract
A molecular hallmark in Parkinson's disease (PD) pathogenesis are α-synuclein aggregates. Cerebral dopamine neurotrophic factor (CDNF) is an atypical growth factor that is mostly resident in the endoplasmic reticulum but exerts its effects both intracellularly and extracellularly. One of the beneficial effects of CDNF can be protecting neurons from the toxic effects of α-synuclein. Here, we investigated the effects of CDNF on α-synuclein aggregation in vitro and in vivo. We found that CDNF directly interacts with α-synuclein with a KD = 23 ± 6 nM and reduces its auto-association. Using nuclear magnetic resonance (NMR) spectroscopy, we identified interaction sites on the CDNF protein. Remarkably, CDNF reduces the neuronal internalization of α-synuclein fibrils and induces the formation of insoluble phosphorylated α-synuclein inclusions. Intra-striatal CDNF administration alleviates motor deficits in rodents challenged with α-synuclein fibrils, though it did not reduce the number of phosphorylated α-synuclein inclusions in the substantia nigra. CDNF's beneficial effects on rodent behavior appear not to be related to the number of inclusions formed in the current context, and further study of its effects on the aggregation mechanism in vivo are needed. Nonetheless, the interaction of CDNF with α-synuclein, modifying its aggregation, spreading, and associated behavioral alterations, provides novel insights into the potential of CDNF as a therapeutic strategy in PD and other synucleinopathies.
Collapse
Affiliation(s)
- Katrina Albert
- Institute of Biotechnology, HiLIFE, University of Helsinki, 00014 Helsinki, Finland
| | - Diana P Raymundo
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21.941-902, Brazil; Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, 37073 Göttingen, Germany; Protein Advanced Biochemistry, CENABIO, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21.941-902, Brazil
| | - Anne Panhelainen
- Institute of Biotechnology, HiLIFE, University of Helsinki, 00014 Helsinki, Finland; Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Ave Eesmaa
- Institute of Biotechnology, HiLIFE, University of Helsinki, 00014 Helsinki, Finland
| | - Liana Shvachiy
- Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, 37073 Göttingen, Germany; Centro Cardiovascular da Universidade de Lisboa, Faculdade de Medicina, Universidade de Lisboa, Av Prof Egas Moniz, 1649-028 Lisbon, Portugal
| | - Gabriela R Araújo
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21.941-902, Brazil; Protein Advanced Biochemistry, CENABIO, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21.941-902, Brazil
| | - Piotr Chmielarz
- Institute of Biotechnology, HiLIFE, University of Helsinki, 00014 Helsinki, Finland; Department of Brain Biochemistry, Maj Institute of Pharmacology, Polish Academy of Sciences, Krakow 31-343, Poland
| | - Xu Yan
- Neuroscience Center, HiLIFE, University of Helsinki, 00014 Helsinki, Finland
| | - Aastha Singh
- Institute of Biotechnology, HiLIFE, University of Helsinki, 00014 Helsinki, Finland
| | - Yraima Cordeiro
- Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21.941-902, Brazil
| | - Fernando L Palhano
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21.941-902, Brazil
| | - Debora Foguel
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21.941-902, Brazil
| | - Kelvin C Luk
- Center for Neurodegenerative Disease Research, Department of Pathology and Laboratory Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Andrii Domanskyi
- Institute of Biotechnology, HiLIFE, University of Helsinki, 00014 Helsinki, Finland
| | - Merja H Voutilainen
- Institute of Biotechnology, HiLIFE, University of Helsinki, 00014 Helsinki, Finland; Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Henri J Huttunen
- Neuroscience Center, HiLIFE, University of Helsinki, 00014 Helsinki, Finland; Herantis Pharma Plc, 20520 Espoo, Finland
| | - Tiago F Outeiro
- Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, 37073 Göttingen, Germany; Max Planck Institute for Experimental Medicine, 37075 Göttingen, Germany; Institute of Neuroscience, The Medical School, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Mart Saarma
- Institute of Biotechnology, HiLIFE, University of Helsinki, 00014 Helsinki, Finland
| | - Marcius S Almeida
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21.941-902, Brazil; Protein Advanced Biochemistry, CENABIO, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21.941-902, Brazil.
| | - Mikko Airavaara
- Institute of Biotechnology, HiLIFE, University of Helsinki, 00014 Helsinki, Finland; Neuroscience Center, HiLIFE, University of Helsinki, 00014 Helsinki, Finland; Faculty of Pharmacy, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
28
|
Gegg ME, Verona G, Schapira AHV. Glucocerebrosidase deficiency promotes release of α-synuclein fibrils from cultured neurons. Hum Mol Genet 2021; 29:1716-1728. [PMID: 32391886 PMCID: PMC7322566 DOI: 10.1093/hmg/ddaa085] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 04/23/2020] [Accepted: 04/24/2020] [Indexed: 01/17/2023] Open
Abstract
Mutations in the GBA gene, which encodes the lysosomal enzyme glucocerebrosidase (GCase), are the most important genetic risk factor for Parkinson disease (PD). GCase activity is also decreased in sporadic PD brains and with normal ageing. Loss of GCase activity impairs the autophagy lysosomal pathway resulting in increased α-synuclein (α-syn) levels. Furthermore, elevated α-syn results in decreased GCase activity. Although the role of α-syn in PD remains unclear, evidence indicates that aggregated α-syn fibrils are a pathogenic species in PD, passing between neurons and inducing endogenous native α-syn to aggregate; spreading pathology through the brain. We have investigated if preformed α-syn fibrils (PFFs) impair GCase activity in mouse cortical neurons and differentiated dopaminergic cells, and whether GCase deficiency in these models increased the transfer of α-syn pathology to naïve cells. Neurons treated with PFFs induced endogenous α-syn to become insoluble and phosphorylated at Ser129 to a greater extent than monomeric α-syn-treatment. PFFs, but not monomeric α-syn, inhibited lysosomal GCase activity in these cells and induced the unfolded protein response. Neurons in which GCase was inhibited by conduritol β-epoxide did not increase the amount of insoluble monomeric α-syn or its phosphorylation status. Instead the release of α-syn fibrils from GCase deficient cells was significantly increased. Co-culture studies showed that the transfer of α-syn pathology to naïve cells was greater from GCase deficient cells. This study suggests that GCase deficiency increases the spread of α-syn pathology and likely contributes to the earlier age of onset and increased cognitive decline associated with GBA-PD.
Collapse
Affiliation(s)
- Matthew E Gegg
- Department of Clinical and Movement Neuroscience, UCL Queen Square Institute of Neurology, Rowland Hill Street, London NW3 2PF, UK
| | - Guglielmo Verona
- Wolfson Drug Discovery Unit, Centre for Amyloidosis and Acute Phase Proteins, Division of Medicine, University College London, London NW3 2PF, UK
| | - Anthony H V Schapira
- Department of Clinical and Movement Neuroscience, UCL Queen Square Institute of Neurology, Rowland Hill Street, London NW3 2PF, UK
| |
Collapse
|
29
|
Brazdis RM, Alecu JE, Marsch D, Dahms A, Simmnacher K, Lörentz S, Brendler A, Schneider Y, Marxreiter F, Roybon L, Winner B, Xiang W, Prots I. Demonstration of brain region-specific neuronal vulnerability in human iPSC-based model of familial Parkinson's disease. Hum Mol Genet 2021; 29:1180-1191. [PMID: 32160287 PMCID: PMC7206857 DOI: 10.1093/hmg/ddaa039] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 02/29/2020] [Accepted: 03/03/2020] [Indexed: 12/12/2022] Open
Abstract
Parkinson’s disease (PD) is a neurodegenerative disorder characterized by protein inclusions mostly composed of aggregated forms of α-synuclein (α-Syn) and by the progressive degeneration of midbrain dopaminergic neurons (mDANs), resulting in motor symptoms. While other brain regions also undergo pathologic changes in PD, the relevance of α-Syn aggregation for the preferential loss of mDANs in PD pathology is not completely understood yet. To elucidate the mechanisms of the brain region-specific neuronal vulnerability in PD, we modeled human PD using human-induced pluripotent stem cells (iPSCs) from familial PD cases with a duplication (Dupl) of the α-Syn gene (SNCA) locus. Human iPSCs from PD Dupl patients and a control individual were differentiated into mDANs and cortical projection neurons (CPNs). SNCA dosage increase did not influence the differentiation efficiency of mDANs and CPNs. However, elevated α-Syn pathology, as revealed by enhanced α-Syn insolubility and phosphorylation, was determined in PD-derived mDANs compared with PD CPNs. PD-derived mDANs exhibited higher levels of reactive oxygen species and protein nitration levels compared with CPNs, which might underlie elevated α-Syn pathology observed in mDANs. Finally, increased neuronal death was observed in PD-derived mDANs compared to PD CPNs and to control mDANs and CPNs. Our results reveal, for the first time, a higher α-Syn pathology, oxidative stress level, and neuronal death rate in human PD mDANs compared with PD CPNs from the same patient. The finding implies the contribution of pathogenic α-Syn, probably induced by oxidative stress, to selective vulnerability of substantia nigra dopaminergic neurons in human PD.
Collapse
Affiliation(s)
- Razvan-Marius Brazdis
- Department of Stem Cell Biology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen 91054, Germany.,Department of Psychiatry and Psychotherapy, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen 91054, Germany
| | - Julian E Alecu
- Department of Stem Cell Biology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen 91054, Germany
| | - Daniel Marsch
- Institute of Biochemistry (Emil-Fischer-Center), Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen 91054, Germany
| | - Annika Dahms
- Institute of Biochemistry (Emil-Fischer-Center), Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen 91054, Germany
| | - Katrin Simmnacher
- Department of Stem Cell Biology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen 91054, Germany
| | - Sandra Lörentz
- Department of Stem Cell Biology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen 91054, Germany
| | - Anna Brendler
- Department of Stem Cell Biology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen 91054, Germany
| | - Yanni Schneider
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen 91054, Germany
| | - Franz Marxreiter
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen 91054, Germany
| | - Laurent Roybon
- Stem Cell Laboratory for CNS Disease Modeling, Department of Experimental Medical Science, Lund University, Lund 22184, Sweden
| | - Beate Winner
- Department of Stem Cell Biology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen 91054, Germany
| | - Wei Xiang
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen 91054, Germany
| | - Iryna Prots
- Department of Stem Cell Biology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen 91054, Germany
| |
Collapse
|
30
|
Boutros SW, Raber J, Unni VK. Effects of Alpha-Synuclein Targeted Antisense Oligonucleotides on Lewy Body-Like Pathology and Behavioral Disturbances Induced by Injections of Pre-Formed Fibrils in the Mouse Motor Cortex. JOURNAL OF PARKINSONS DISEASE 2021; 11:1091-1115. [PMID: 34057097 PMCID: PMC8461707 DOI: 10.3233/jpd-212566] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Background: Alpha-synuclein (αsyn) characterizes neurodegenerative diseases known as synucleinopathies. The phosphorylated form (psyn) is the primary component of protein aggregates known as Lewy bodies (LBs), which are the hallmark of diseases such as Parkinson’s disease (PD). Synucleinopathies might spread in a prion-like fashion, leading to a progressive emergence of symptoms over time. αsyn pre-formed fibrils (PFFs) induce LB-like pathology in wild-type (WT) mice, but questions remain about their progressive spread and their associated effects on behavioral performance. Objective: To characterize the behavioral, cognitive, and pathological long-term effects of LB-like pathology induced after bilateral motor cortex PFF injection in WT mice and to assess the ability of mouse αsyn-targeted antisense oligonucleotides (ASOs) to ameliorate those effects. Methods: We induced LB-like pathology in the motor cortex and connected brain regions of male WT mice using PFFs. Three months post-PFF injection (mpi), we assessed behavioral and cognitive performance. We then delivered a targeted ASO via the ventricle and assessed behavioral and cognitive performance 5 weeks later, followed by pathological analysis. Results: At 3 and 6 mpi, PFF-injected mice showed mild, progressive behavioral deficits. The ASO reduced total αsyn and psyn protein levels, and LB-like pathology, but was also associated with some deleterious off-target effects not involving lowering of αsyn, such as a decline in body weight and impairments in motor function. Conclusions: These results increase understanding of the progressive nature of the PFF model and support the therapeutic potential of ASOs, though more investigation into effects of ASO-mediated reduction in αsyn on brain function is needed.
Collapse
Affiliation(s)
- Sydney Weber Boutros
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, USA
| | - Jacob Raber
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, USA.,Department of Neurology, Oregon Health & Science University, Portland, OR, USA.,Departments of Psychiatry and Radiation Medicine, Division of Neuroscience, ONPRC, Oregon Health & Science University, Portland, OR, USA
| | - Vivek K Unni
- Department of Neurology, Oregon Health & Science University, Portland, OR, USA.,Jungers Center for Neurosciences Research and OHSU Parkinson Center, Oregon Health & Science University, Portland, OR, USA
| |
Collapse
|
31
|
Monoamine Oxidase-B Inhibition Facilitates α-Synuclein Secretion In Vitro and Delays Its Aggregation in rAAV-Based Rat Models of Parkinson's Disease. J Neurosci 2021; 41:7479-7491. [PMID: 34290084 DOI: 10.1523/jneurosci.0476-21.2021] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Revised: 07/12/2021] [Accepted: 07/15/2021] [Indexed: 12/26/2022] Open
Abstract
Cell-to-cell transmission of α-synuclein (α-syn) pathology is considered to underlie the spread of neurodegeneration in Parkinson's disease (PD). Previous studies have demonstrated that α-syn is secreted under physiological conditions in neuronal cell lines and primary neurons. However, the molecular mechanisms that regulate extracellular α-syn secretion remain unclear. In this study, we found that inhibition of monoamine oxidase-B (MAO-B) enzymatic activity facilitated α-syn secretion in human neuroblastoma SH-SY5Y cells. Both inhibition of MAO-B by selegiline or rasagiline and siRNA-mediated knock-down of MAO-B facilitated α-syn secretion. However, TVP-1022, the S-isomer of rasagiline that is 1000 times less active, failed to facilitate α-syn secretion. Additionally, the MAO-B inhibition-induced increase in α-syn secretion was unaffected by brefeldin A, which inhibits endoplasmic reticulum (ER)/Golgi transport, but was blocked by probenecid and glyburide, which inhibit ATP-binding cassette (ABC) transporter function. MAO-B inhibition preferentially facilitated the secretion of detergent-insoluble α-syn protein and decreased its intracellular accumulation under chloroquine-induced lysosomal dysfunction. Moreover, in a rat model (male Sprague Dawley rats) generated by injecting recombinant adeno-associated virus (rAAV)-A53T α-syn, subcutaneous administration of selegiline delayed the striatal formation of Ser129-phosphorylated α-syn aggregates, and mitigated loss of nigrostriatal dopaminergic neurons. Selegiline also delayed α-syn aggregation and dopaminergic neuronal loss in a cell-to-cell transmission rat model (male Sprague Dawley rats) generated by injecting rAAV-wild-type α-syn and externally inoculating α-syn fibrils into the striatum. These findings suggest that MAO-B inhibition modulates the intracellular clearance of detergent-insoluble α-syn via the ABC transporter-mediated non-classical secretion pathway, and temporarily suppresses the formation and transmission of α-syn aggregates.SIGNIFICANCE STATEMENT The identification of a neuroprotective agent that slows or stops the progression of motor impairments is required to treat Parkinson's disease (PD). The process of α-synuclein (α-syn) aggregation is thought to underlie neurodegeneration in PD. Here, we demonstrated that pharmacological inhibition or knock-down of monoamine oxidase-B (MAO-B) in SH-SY5Y cells facilitated α-syn secretion via a non-classical pathway involving an ATP-binding cassette (ABC) transporter. MAO-B inhibition preferentially facilitated secretion of detergent-insoluble α-syn protein and reduced its intracellular accumulation under chloroquine-induced lysosomal dysfunction. Additionally, MAO-B inhibition by selegiline protected A53T α-syn-induced nigrostriatal dopaminergic neuronal loss and suppressed the formation and cell-to-cell transmission of α-syn aggregates in rat models. We therefore propose a new function of MAO-B inhibition that modulates α-syn secretion and aggregation.
Collapse
|
32
|
Ghosh S, Won SJ, Wang J, Fong R, Butler NJM, Moss A, Wong C, Pan J, Sanchez J, Huynh A, Wu L, Manfredsson FP, Swanson RA. α-synuclein aggregates induce c-Abl activation and dopaminergic neuronal loss by a feed-forward redox stress mechanism. Prog Neurobiol 2021; 202:102070. [PMID: 33951536 PMCID: PMC8210833 DOI: 10.1016/j.pneurobio.2021.102070] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 01/21/2021] [Accepted: 04/27/2021] [Indexed: 11/21/2022]
Abstract
Oxidative stress and α-synuclein aggregation both drive neurodegeneration in Parkinson's disease, and the protein kinase c-Abl provides a potential amplifying link between these pathogenic factors. Suppressing interactions between these factors may thus be a viable therapeutic approach for this disorder. To evaluate this possibility, pre-formed α-synuclein fibrils (PFFs) were used to induce α-synuclein aggregation in neuronal cultures. Exposure to PFFs induced oxidative stress and c-Abl activation in wild-type neurons. By contrast, α-synuclein - deficient neurons, which cannot form α-synuclein aggregates, failed to exhibit either oxidative stress or c-Abl activation. N-acetyl cysteine, a thiol repletion agent that supports neuronal glutathione metabolism, suppressed the PFF - induced redox stress and c-Abl activation in the wild-type neurons, and likewise suppressed α-synuclein aggregation. Parallel findings were observed in mouse brain: PFF-induced α-synuclein aggregation in the substantia nigra was associated with redox stress, c-Abl activation, and dopaminergic neuronal loss, along with microglial activation and motor impairment, all of which were attenuated with oral N-acetyl cysteine. Similar results were obtained using AAV-mediated α-synuclein overexpression as an alternative means of driving α-synuclein aggregation in vivo. These findings show that α-synuclein aggregates induce c-Abl activation by a redox stress mechanism. c-Abl activation in turn promotes α-synuclein aggregation, in a feed-forward interaction. The capacity of N-acetyl cysteine to interrupt this interaction adds mechanistic support its consideration as a therapeutic in Parkinson's disease.
Collapse
Affiliation(s)
- Soumitra Ghosh
- Department of Neurology, University of California San Francisco, United States; Neurology Service, San Francisco Veterans Affairs Health Care System, United States
| | - Seok Joon Won
- Department of Neurology, University of California San Francisco, United States; Neurology Service, San Francisco Veterans Affairs Health Care System, United States
| | - Jiejie Wang
- Department of Neurology, University of California San Francisco, United States; Neurology Service, San Francisco Veterans Affairs Health Care System, United States
| | - Rebecca Fong
- Department of Neurology, University of California San Francisco, United States; Neurology Service, San Francisco Veterans Affairs Health Care System, United States
| | - Nicholas J M Butler
- Department of Neurology, University of California San Francisco, United States; Neurology Service, San Francisco Veterans Affairs Health Care System, United States
| | - Arianna Moss
- Department of Neurology, University of California San Francisco, United States; Neurology Service, San Francisco Veterans Affairs Health Care System, United States
| | - Candance Wong
- Department of Neurology, University of California San Francisco, United States; Neurology Service, San Francisco Veterans Affairs Health Care System, United States
| | - June Pan
- Department of Neurology, University of California San Francisco, United States; Neurology Service, San Francisco Veterans Affairs Health Care System, United States
| | - Jennifer Sanchez
- Department of Neurology, University of California San Francisco, United States; Neurology Service, San Francisco Veterans Affairs Health Care System, United States
| | - Annie Huynh
- Department of Neurology, University of California San Francisco, United States; Neurology Service, San Francisco Veterans Affairs Health Care System, United States
| | - Long Wu
- Department of Neurology, University of California San Francisco, United States; Neurology Service, San Francisco Veterans Affairs Health Care System, United States
| | - Fredric P Manfredsson
- Parkinson's Disease Research Unit, Department of Neurobiology, Barrow Neurological Institute, Phoenix, AZ, 85013, United States
| | - Raymond A Swanson
- Department of Neurology, University of California San Francisco, United States; Neurology Service, San Francisco Veterans Affairs Health Care System, United States.
| |
Collapse
|
33
|
Bu LL, Liu YQ, Shen Y, Fan Y, Yu WB, Jiang DL, Tang YL, Yang YJ, Wu P, Zuo CT, Koprich JB, Liu FT, Wu JJ, Wang J. Neuroprotection of Exendin-4 by Enhanced Autophagy in a Parkinsonian Rat Model of α-Synucleinopathy. Neurotherapeutics 2021; 18:962-978. [PMID: 33723752 PMCID: PMC8423983 DOI: 10.1007/s13311-021-01018-5] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/26/2021] [Indexed: 12/14/2022] Open
Abstract
Glucagon-like peptide-1 (GLP-1) receptor stimulation ameliorates parkinsonian motor and non-motor deficits in both experimental animals and patients; however, the disease-modifying mechanisms of GLP-1 receptor activation have remained unknown. The present study investigated whether exendin-4 (a GLP-1 analogue) can rescue motor deficits and exert disease-modifying effects in a parkinsonian rat model of α-synucleinopathy. This model was established by unilaterally injecting AAV-9-A53T-α-synuclein into the right substantia nigra pars compacta, followed by 4 or 8 weeks of twice-daily intraperitoneal injections of exendin-4 (5 μg/kg/day) starting at 2 weeks after AAV-9-A53T-α-synuclein injections. Positron emission tomography/computed tomography (PET/CT) scanning and immunostaining established that treatment with exendin-4 attenuated tyrosine-hydroxylase-positive neuronal loss and terminal denervation and mitigated the decrease in expression of vesicular monoamine transporter 2 within the nigrostriatal dopaminergic systems of rats injected with AAV-9-A53T-α-synuclein. It also mitigated the parkinsonian motor deficits assessed in behavioral tests. Furthermore, through both in vivo and in vitro models of Parkinson's disease, we showed that exendin-4 promoted autophagy and mediated degradation of pathological α-synuclein, the effects of which were counteracted by 3-methyladenine or chloroquine, the autophagic inhibitors. Additionally, exendin-4 attenuated dysregulation of the PI3K/Akt/mTOR pathway in rats injected with AAV-9-A53T-α-synuclein. Taken together, our results demonstrate that exendin-4 treatment relieved behavioral deficits, dopaminergic degeneration, and pathological α-synuclein aggregation in a parkinsonian rat model of α-synucleinopathy and that these effects were mediated by enhanced autophagy via inhibiting the PI3K/Akt/mTOR pathway. In light of the safety and tolerance of exendin-4 administration, our results suggest that exendin-4 may represent a promising disease-modifying treatment for Parkinson's disease.
Collapse
Affiliation(s)
- Lu-Lu Bu
- Department of Neurology & National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, 200040 China
| | - Yi-Qi Liu
- Department of Neurology & National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, 200040 China
| | - Yan Shen
- Department of Neurology & National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, 200040 China
| | - Yun Fan
- Department of Neurology & National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, 200040 China
| | - Wen-Bo Yu
- Department of Neurology & National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, 200040 China
| | - Dong-Lang Jiang
- PET Center, Huashan Hospital, Fudan University, Shanghai, 200235 China
| | - Yi-Lin Tang
- Department of Neurology & National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, 200040 China
| | - Yu-Jie Yang
- Department of Neurology & National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, 200040 China
| | - Ping Wu
- PET Center, Huashan Hospital, Fudan University, Shanghai, 200235 China
| | - Chuan-Tao Zuo
- PET Center, Huashan Hospital, Fudan University, Shanghai, 200235 China
| | - James B. Koprich
- Department of Neurology & National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, 200040 China
- Krembil Research Institute, Toronto Western Hospital, University Health Network, Toronto, ON M5T 2S8 Canada
| | - Feng-Tao Liu
- Department of Neurology & National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, 200040 China
| | - Jian-Jun Wu
- Department of Neurology & National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, 200040 China
| | - Jian Wang
- Department of Neurology & National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, 200040 China
| |
Collapse
|
34
|
Psol M, Darvas SG, Leite K, Mahajani SU, Bähr M, Kügler S. Dementia with Lewy bodies-associated ß-synuclein mutations V70M and P123H cause mutation-specific neuropathological lesions. Hum Mol Genet 2021; 30:247-264. [PMID: 33760043 DOI: 10.1093/hmg/ddab036] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 01/20/2021] [Accepted: 01/20/2021] [Indexed: 12/26/2022] Open
Abstract
Beta (ß)-synuclein (ß-Syn) has long been considered to be an attenuator for the neuropathological effects caused by the Parkinson's disease-related alpha (α)-synuclein (α-Syn) protein. However, recent studies demonstrated that overabundant ß-Syn can form aggregates and induce neurodegeneration in central nervous system (CNS) neurons in vitro and in vivo, albeit at a slower pace as compared with α-Syn. Here, we demonstrate that ß-Syn mutants V70M, detected in a sporadic case of dementia with Lewy bodies (DLB), and P123H, detected in a familial case of DLB, robustly aggravate the neurotoxic potential of ß-Syn. Intriguingly, the two mutations trigger mutually exclusive pathways. ß-Syn V70M enhances morphological mitochondrial deterioration and degeneration of dopaminergic and non-dopaminergic neurons, but it has no influence on neuronal network activity. Conversely, ß-Syn P123H silences neuronal network activity, but it does not aggravate neurodegeneration. ß-Syn wild type (WT), V70M and P123H formed proteinase K-resistant intracellular fibrils within neurons, albeit with less stable C-termini as compared with α-Syn. Under cell-free conditions, ß-Syn V70M demonstrated a much slower pace of fibril formation as compared with WT ß-Syn, and P123H fibrils present with a unique phenotype characterized by large numbers of short, truncated fibrils. Thus, it is possible that V70M and P123H cause structural alterations in ß-Syn, which are linked to their distinct neuropathological profiles. The extent of the lesions caused by these neuropathological profiles is almost identical to that of overabundant α-Syn and is thus likely to be directly involved into the etiology of DLB. Overall, this study provides insights into distinct disease mechanisms caused by mutations of ß-Syn.
Collapse
Affiliation(s)
- Maryna Psol
- Department of Neurology, University Medicine Göttingen, Göttingen 37073, Germany
| | - Sofia Guerin Darvas
- Department of Neurology, University Medicine Göttingen, Göttingen 37073, Germany
| | - Kristian Leite
- Department of Neurology, University Medicine Göttingen, Göttingen 37073, Germany
| | - Sameehan U Mahajani
- Department of Neurology, University Medicine Göttingen, Göttingen 37073, Germany
| | - Mathias Bähr
- Department of Neurology, University Medicine Göttingen, Göttingen 37075, Germany
| | - Sebastian Kügler
- Department of Neurology, University Medicine Göttingen, Göttingen 37073, Germany
| |
Collapse
|
35
|
Systemic α-synuclein injection triggers selective neuronal pathology as seen in patients with Parkinson's disease. Mol Psychiatry 2021; 26:556-567. [PMID: 31758091 PMCID: PMC7850975 DOI: 10.1038/s41380-019-0608-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 11/07/2019] [Accepted: 11/13/2019] [Indexed: 12/25/2022]
Abstract
Parkinson's disease (PD) is an α-synucleinopathy characterized by the progressive loss of specific neuronal populations. Here, we develop a novel approach to transvascularly deliver proteins of complex quaternary structures, including α-synuclein preformed fibrils (pff). We show that a single systemic administration of α-synuclein pff triggers pathological transformation of endogenous α-synuclein in non-transgenic rats, which leads to neurodegeneration in discrete brain regions. Specifically, pff-exposed animals displayed a progressive deterioration in gastrointestinal and olfactory functions, which corresponded with the presence of cellular pathology in the central and enteric nervous systems. The α-synuclein pathology generated was both time dependent and region specific. Interestingly, the most significant neuropathological changes were observed in those brain regions affected in the early stages of PD. Our data therefore demonstrate for the first time that a single, transvascular administration of α-synuclein pff can lead to selective regional neuropathology resembling the premotor stage of idiopathic PD. Furthermore, this novel delivery approach could also be used to deliver a range of other pathogenic, as well as therapeutic, protein cargos transvascularly to the brain.
Collapse
|
36
|
Matsuo K, Yabuki Y, Melki R, Bousset L, Owada Y, Fukunaga K. Crucial Role of FABP3 in αSyn-Induced Reduction of Septal GABAergic Neurons and Cognitive Decline in Mice. Int J Mol Sci 2021; 22:E400. [PMID: 33401521 PMCID: PMC7795765 DOI: 10.3390/ijms22010400] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Revised: 12/27/2020] [Accepted: 12/29/2020] [Indexed: 12/13/2022] Open
Abstract
In synucleinopathies, while motor symptoms are thought to be attributed to the accumulation of misfolded α-synuclein (αSyn) in nigral dopaminergic neurons, it remains to be elucidated how cognitive decline arises. Here, we investigated the effects of distinct αSyn strains on cognition and the related neuropathology in the medial septum/diagonal band (MS/DB), a key region for cognitive processing. Bilateral injection of αSyn fibrils into the dorsal striatum potently impaired cognition in mice. The cognitive decline was accompanied by accumulation of phosphorylated αSyn at Ser129 and reduction of gamma-aminobutyric acid (GABA)-ergic but not cholinergic neurons in the MS/DB. Since we have demonstrated that fatty acid-binding protein 3 (FABP3) is critical for αSyn neurotoxicity in nigral dopaminergic neurons, we investigated whether FABP3 also participates in αSyn pathology in the MS/DB and cognitive decline. FABP3 was highly expressed in GABAergic but rarely in cholinergic neurons in the MS/DB. Notably, Fabp3 deletion antagonized the accumulation of phosphorylated αSyn, decrease in GABAergic neurons, and cognitive impairment caused by αSyn fibrils. Overall, the present study indicates that FABP3 mediates αSyn neurotoxicity in septal GABAergic neurons and the resultant cognitive impairment, and that FABP3 in this subpopulation could be a therapeutic target for dementia in synucleinopathies.
Collapse
Affiliation(s)
- Kazuya Matsuo
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan; (K.M.); (Y.Y.)
| | - Yasushi Yabuki
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan; (K.M.); (Y.Y.)
- Department of Genomic Neurology, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto 860-0811, Japan
| | - Ronald Melki
- Laboratory of Neurodegenerative Diseases, CEA, Institut François Jacob (MIRcen) and CNRS, 18 Route du Panorama, 92265 Fontenay-aux-Roses, France; (R.M.); (L.B.)
| | - Luc Bousset
- Laboratory of Neurodegenerative Diseases, CEA, Institut François Jacob (MIRcen) and CNRS, 18 Route du Panorama, 92265 Fontenay-aux-Roses, France; (R.M.); (L.B.)
| | - Yuji Owada
- Department of Organ Anatomy, Graduate School of Medicine, Tohoku University, Sendai 980-0872, Japan;
| | - Kohji Fukunaga
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan; (K.M.); (Y.Y.)
| |
Collapse
|
37
|
Simmnacher K, Krach F, Schneider Y, Alecu JE, Mautner L, Klein P, Roybon L, Prots I, Xiang W, Winner B. Unique signatures of stress-induced senescent human astrocytes. Exp Neurol 2020; 334:113466. [PMID: 32949572 DOI: 10.1016/j.expneurol.2020.113466] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 09/11/2020] [Accepted: 09/14/2020] [Indexed: 12/15/2022]
Abstract
Senescence was recently linked to neurodegeneration and astrocytes are one of the major cell types to turn senescent under neurodegenerative conditions. Senescent astrocytes were detected in Parkinson's disease (PD) patients' brains besides reactive astrocytes, yet the difference between senescent and reactive astrocytes is unclear. We aimed to characterize senescent astrocytes in comparison to reactive astrocytes and investigate differences and similarities. In a cell culture model of human fetal astrocytes, we determined a unique senescent transcriptome distinct from reactive astrocytes, which comprises dysregulated pathways. Both, senescent and reactive human astrocytes activated a proinflammatory pattern. Astrocyte senescence was at least partially depending on active mechanistic-target-of-rapamycin (mTOR) and DNA-damage response signaling, both drivers of senescence. To further investigate how PD and senescence connect to each other, we asked if a PD-linked environmental factor induces senescence and if senescence impairs midbrain neurons. We could show that the PD-linked pesticide rotenone causes astrocyte senescence. We further delineate, that the senescent secretome exaggerates rotenone-induced neurodegeneration in midbrain neurons differentiated from human induced pluripotent stem cells (hiPSC) of PD patients with alpha-synuclein gene (SNCA) locus duplication.
Collapse
Affiliation(s)
- Katrin Simmnacher
- Department of Stem Cell Biology, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Florian Krach
- Department of Stem Cell Biology, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany; Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Yanni Schneider
- Department of Molecular Neurology, FAU Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Julian E Alecu
- Department of Stem Cell Biology, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Lena Mautner
- Department of Stem Cell Biology, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Paulina Klein
- Department of Stem Cell Biology, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Laurent Roybon
- Stem Cell Laboratory for CNS Disease Modeling, MultiPark and Lund Stem Cell Center, Department of Experimental Medical Science, Faculty of Medicine, Lund University, 22184 Lund, Sweden
| | - Iryna Prots
- Department of Stem Cell Biology, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Wei Xiang
- Department of Molecular Neurology, FAU Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Beate Winner
- Department of Stem Cell Biology, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany.
| |
Collapse
|
38
|
Gezer AO, Kochmanski J, VanOeveren SE, Cole-Strauss A, Kemp CJ, Patterson JR, Miller KM, Kuhn NC, Herman DE, McIntire A, Lipton JW, Luk KC, Fleming SM, Sortwell CE, Bernstein AI. Developmental exposure to the organochlorine pesticide dieldrin causes male-specific exacerbation of α-synuclein-preformed fibril-induced toxicity and motor deficits. Neurobiol Dis 2020; 141:104947. [PMID: 32422283 PMCID: PMC7343230 DOI: 10.1016/j.nbd.2020.104947] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 04/22/2020] [Accepted: 05/13/2020] [Indexed: 12/21/2022] Open
Abstract
Human and animal studies have shown that exposure to the organochlorine pesticide dieldrin is associated with increased risk of Parkinson's disease (PD). Previous work showed that developmental dieldrin exposure increased neuronal susceptibility to MPTP toxicity in male C57BL/6 mice, possibly via changes in dopamine (DA) packaging and turnover. However, the relevance of the MPTP model to PD pathophysiology has been questioned. We therefore studied dieldrin-induced neurotoxicity in the α-synuclein (α-syn)-preformed fibril (PFF) model, which better reflects the α-syn pathology and toxicity observed in PD pathogenesis. Specifically, we used a "two-hit" model to determine whether developmental dieldrin exposure increases susceptibility to α-syn PFF-induced synucleinopathy. Dams were fed either dieldrin (0.3 mg/kg, every 3-4 days) or vehicle corn oil starting 1 month prior to breeding and continuing through weaning of pups at postnatal day 22. At 12 weeks of age, male and female offspring received intrastriatal α-syn PFF or control saline injections. Consistent with the male-specific increased susceptibility to MPTP, our results demonstrate that developmental dieldrin exposure exacerbates PFF-induced toxicity in male mice only. Specifically, in male offspring, dieldrin exacerbated PFF-induced motor deficits on the challenging beam and increased DA turnover in the striatum 6 months after PFF injection. However, male offspring showed neither exacerbation of phosphorylated α-syn aggregation (pSyn) in the substantia nigra (SN) at 1 or 2 months post-PFF injection, nor exacerbation of PFF-induced TH and NeuN loss in the SN 6 months post-PFF injection. Collectively, these data indicate that developmental dieldrin exposure produces a male-specific exacerbation of synucleinopathy-induced behavioral and biochemical deficits. This sex-specific result is consistent with both previous work in the MPTP model, our previously reported sex-specific effects of this exposure paradigm on the male and female epigenome, and the higher prevalence and more severe course of PD in males. The novel two-hit environmental toxicant/PFF exposure paradigm established in this project can be used to explore the mechanisms by which other PD-related exposures alter neuronal vulnerability to synucleinopathy in sporadic PD.
Collapse
Affiliation(s)
- Aysegul O Gezer
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI, United States of America; Cell and Molecular Biology Graduate Program, College of Natural Sciences, Michigan State University, East Lansing, MI, United States of America; College of Osteopathic Medicine, Michigan State University, East Lansing, MI, United States of America
| | - Joseph Kochmanski
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI, United States of America
| | - Sarah E VanOeveren
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI, United States of America
| | - Allyson Cole-Strauss
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI, United States of America
| | - Christopher J Kemp
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI, United States of America
| | - Joseph R Patterson
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI, United States of America
| | - Kathryn M Miller
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI, United States of America
| | - Nathan C Kuhn
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI, United States of America
| | - Danielle E Herman
- Department of Pharmaceutical Sciences, College of Pharmacy, Northeast Ohio Medical University, Rootstown, OH, United States of America
| | - Alyssa McIntire
- Department of Pharmaceutical Sciences, College of Pharmacy, Northeast Ohio Medical University, Rootstown, OH, United States of America
| | - Jack W Lipton
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI, United States of America; Mercy Health St. Mary's, Grand Rapids, MI, United States of America
| | - Kelvin C Luk
- Department of Pathology and Laboratory Medicine, Center for Neurodegenerative Disease Research, University of Pennsylvania, Philadelphia, PA, United States of America
| | - Sheila M Fleming
- Department of Pharmaceutical Sciences, College of Pharmacy, Northeast Ohio Medical University, Rootstown, OH, United States of America
| | - Caryl E Sortwell
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI, United States of America; Mercy Health St. Mary's, Grand Rapids, MI, United States of America
| | - Alison I Bernstein
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI, United States of America; Mercy Health St. Mary's, Grand Rapids, MI, United States of America.
| |
Collapse
|
39
|
Kasongo DW, de Leo G, Vicario N, Leanza G, Legname G. Chronic α-Synuclein Accumulation in Rat Hippocampus Induces Lewy Bodies Formation and Specific Cognitive Impairments. eNeuro 2020; 7:ENEURO.0009-20.2020. [PMID: 32393581 PMCID: PMC7307628 DOI: 10.1523/eneuro.0009-20.2020] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 04/06/2020] [Accepted: 04/07/2020] [Indexed: 12/21/2022] Open
Abstract
Occurrence of Lewy bodies (LBs)/Lewy neurites (LNs) containing misfolded fibrillar α-synuclein (α-syn) is one of the pathologic hallmarks of memory impairment-linked synucleinopathies, such as Parkinson's disease (PD) and dementia with LBs (DLB). While it has been shown that brainstem LBs may contribute to motor symptoms, the neuropathological substrates for cognitive symptoms are still elusive. Here, recombinant mouse α-syn fibrils were bilaterally injected in the hippocampus of female Sprague Dawley rats, which underwent behavioral testing for sensorimotor and spatial learning and memory abilities. No sensorimotor deficits affecting Morris water maze task performance were observed, nor was any reference memory disturbances detectable in injected animals. By contrast, significant impairments in working memory performance became evident at 12 months postinjection. These deficits were associated to a time-dependent increase in the levels of phosphorylated α-syn at Ser129 and in the stereologically estimated numbers of proteinase K (PK)-resistant α-syn aggregates within the hippocampus. Interestingly, pathologic α-syn aggregates were found in the entorhinal cortex and, by 12 months postinjection, also in the vertical limb of the diagonal band and the piriform cortices. No pathologic α-syn deposits were found within the substantia nigra (SN), the ventral tegmental area (VTA), or the striatum, nor was any loss of dopaminergic, noradrenergic, or cholinergic neurons detected in α-syn-injected animals, compared with controls. This would suggest that the behavioral impairments seen in the α-syn-injected animals might be determined by the long-term α-syn neuropathology, rather than by neurodegeneration per se, thus leading to the onset of working memory deficits.
Collapse
Affiliation(s)
- Danielle Walu Kasongo
- B.R.A.I.N. Laboratory for Neurogenesis and Repair, Department of Life Sciences, University of Trieste, Trieste 34127, Italy
- Department of Neuroscience, Scuola Internazionale Superiore di Studi Avanzati (SISSA), Laboratory of Prion Biology, Trieste 34127, Italy
| | - Gioacchino de Leo
- B.R.A.I.N. Laboratory for Neurogenesis and Repair, Department of Life Sciences, University of Trieste, Trieste 34127, Italy
| | - Nunzio Vicario
- Section of Physiology, Department of Biomedical and Biotechnological Sciences, University of Catania, Catania 95123, Italy
| | - Giampiero Leanza
- B.R.A.I.N. Laboratory for Neurogenesis and Repair, Department of Life Sciences, University of Trieste, Trieste 34127, Italy
- Molecular Preclinical and Translational Imaging Research Centre - IMPRonTE, University of Catania, Catania 95125, Italy
- Department of Drug Sciences, University of Catania, Catania 95125, Italy
| | - Giuseppe Legname
- Department of Neuroscience, Scuola Internazionale Superiore di Studi Avanzati (SISSA), Laboratory of Prion Biology, Trieste 34127, Italy
| |
Collapse
|
40
|
Yoshinaga S, Yamanaka T, Miyazaki H, Okuzumi A, Hiyama A, Murayama S, Nukina N. Preserved proteinase K-resistant core after amplification of alpha-synuclein aggregates: Implication to disease-related structural study. Biochem Biophys Res Commun 2020; 522:655-661. [PMID: 31785806 DOI: 10.1016/j.bbrc.2019.11.142] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Accepted: 11/20/2019] [Indexed: 11/17/2022]
Abstract
Many pathological proteins related to neurodegenerative diseases are misfolded, aggregating to form amyloid fibrils during pathogenesis. One of the pathological proteins, alpha-synuclein (α-syn), accumulates in the brains of Parkinson disease (PD), dementia with Lewy bodies (DLB) and multiple system atrophy (MSA), which are designated as synucleinopathies. Recently, structural properties of abnormal accumulated proteins are suggested to determine the disease phenotype. However, the biochemical and structural characteristics of those accumulated proteins are still poorly understood. We previously reported the sequence and seed-structure-dependent polymorphic fibrils of α-syn and the polymorphism was identified by proteinase K-resistant cores determined by mass spectrometry (MS) analysis. In this study, we applied this method to analyze α-syn aggregates of MSA and DLB. To perform MS analysis on proteinase K-resistant cores, we first performed amplification of α-syn aggregates by seeding reaction and protein misfolding cyclic amplification (PMCA) to obtain a sufficient amount of aggregates. Using SDS insoluble fraction of the disease brain, we successfully amplified enough α-syn aggregates for MS analysis. We differentiated between mouse and human α-syn aggregates by MS analysis on proteinase K-resistant cores of the aggregates before and after amplification. The results suggest that structural properties of amplified α-syn fibrils are preserved after PMCA and these methods can be applicable in the study of pathological proteins of the neurodegenerative disorders.
Collapse
Affiliation(s)
- Saki Yoshinaga
- Laboratory of Structural Neuropathology, Doshisha University Graduate School of Brain Science, 1-3 Miyakodanitatara, Kyotanabe-shi, Kyoto, 610-0394, Japan
| | - Tomoyuki Yamanaka
- Laboratory of Structural Neuropathology, Doshisha University Graduate School of Brain Science, 1-3 Miyakodanitatara, Kyotanabe-shi, Kyoto, 610-0394, Japan.
| | - Haruko Miyazaki
- Laboratory of Structural Neuropathology, Doshisha University Graduate School of Brain Science, 1-3 Miyakodanitatara, Kyotanabe-shi, Kyoto, 610-0394, Japan
| | - Ayami Okuzumi
- Laboratory of Structural Neuropathology, Doshisha University Graduate School of Brain Science, 1-3 Miyakodanitatara, Kyotanabe-shi, Kyoto, 610-0394, Japan; Department of Neurology, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Akiko Hiyama
- Laboratory of Structural Neuropathology, Doshisha University Graduate School of Brain Science, 1-3 Miyakodanitatara, Kyotanabe-shi, Kyoto, 610-0394, Japan
| | - Shigeo Murayama
- Department of Neurology and Neuropathology (the Brain Bank for Aging Research), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology, 35-2, Sakae-chou, Itabashi-ku, Tokyo, 173-0015, Japan
| | - Nobuyuki Nukina
- Laboratory of Structural Neuropathology, Doshisha University Graduate School of Brain Science, 1-3 Miyakodanitatara, Kyotanabe-shi, Kyoto, 610-0394, Japan.
| |
Collapse
|
41
|
Mendsaikhan A, Tooyama I, Bellier JP, Serrano GE, Sue LI, Lue LF, Beach TG, Walker DG. Characterization of lysosomal proteins Progranulin and Prosaposin and their interactions in Alzheimer's disease and aged brains: increased levels correlate with neuropathology. Acta Neuropathol Commun 2019; 7:215. [PMID: 31864418 PMCID: PMC6925443 DOI: 10.1186/s40478-019-0862-8] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2019] [Accepted: 12/02/2019] [Indexed: 12/15/2022] Open
Abstract
Progranulin (PGRN) is a protein encoded by the GRN gene with multiple identified functions including as a neurotrophic factor, tumorigenic growth factor, anti-inflammatory cytokine and regulator of lysosomal function. A single mutation in the human GRN gene resulting in reduced PGRN expression causes types of frontotemporal lobar degeneration resulting in frontotemporal dementia. Prosaposin (PSAP) is also a multifunctional neuroprotective secreted protein and regulator of lysosomal function. Interactions of PGRN and PSAP affect their functional properties. Their roles in Alzheimer's disease (AD), the leading cause of dementia, have not been defined. In this report, we examined in detail the cellular expression of PGRN in middle temporal gyrus samples of a series of human brain cases (n = 45) staged for increasing plaque pathology. Immunohistochemistry showed PGRN expression in cortical neurons, microglia, cerebral vessels and amyloid beta (Aβ) plaques, while PSAP expression was mainly detected in neurons and Aβ plaques, and to a limited extent in astrocytes. We showed that there were increased levels of PGRN protein in AD cases and corresponding increased levels of PSAP. Levels of PGRN and PSAP protein positively correlated with amyloid beta (Aβ), with PGRN levels correlating with phosphorylated tau (serine 205) levels in these samples. Although PGRN colocalized with lysosomal-associated membrane protein-1 in neurons, most PGRN associated with Aβ plaques did not. Aβ plaques with PGRN and PSAP deposits were identified in the low plaque non-demented cases suggesting this was an early event in plaque formation. We did not observe PGRN-positive neurofibrillary tangles. Co-immunoprecipitation studies of PGRN from brain samples identified only PSAP associated with PGRN, not sortilin or other known PGRN-binding proteins, under conditions used. Most PGRN associated with Aβ plaques were immunoreactive for PSAP showing a high degree of colocalization of these proteins that did not change between disease groups. As PGRN supplementation has been considered as a therapeutic approach for AD, the possible involvement of PGRN and PSAP interactions in AD pathology needs to be further considered.
Collapse
|
42
|
Uemura N, Yagi H, Uemura MT, Yamakado H, Takahashi R. Limited spread of pathology within the brainstem of α-synuclein BAC transgenic mice inoculated with preformed fibrils into the gastrointestinal tract. Neurosci Lett 2019; 716:134651. [PMID: 31783082 DOI: 10.1016/j.neulet.2019.134651] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2019] [Revised: 11/15/2019] [Accepted: 11/23/2019] [Indexed: 12/01/2022]
Abstract
Parkinson's disease (PD) is pathologically characterized by intraneuronal α-synuclein (α-Syn) aggregates called Lewy bodies (LBs) as well as the loss of dopaminergic neurons in the substantia nigra pars compacta (SNpc). On the basis of autopsy studies, Braak et al. hypothesized that Lewy pathology initially occurs in the enteric nervous system, subsequently spreading to the dorsal motor nucleus of the vagus nerve (dmX) and then ascending in the brainstem to the SNpc. However, this hypothetical progression lacks adequate experimental evidence. We previously reported that inoculation of α-Syn preformed fibrils (PFFs) into the gastric wall of wild-type (WT) mice induced LB-like α-Syn aggregates in the dmX via the vagus nerve. However, α-Syn pathology did not spread beyond the dmX up to 12 months postinoculation. In the present study, we inoculated α-Syn PFFs into the gastric wall of bacterial artificial chromosome (BAC) transgenic mice harboring the human α-Syn gene with an A53 T mutation and analyzed the pathology. The transgenic mice had ∼1.5-fold overexpression of α-Syn in the brains and ∼6-fold overexpression of α-Syn in the stomach compared with WT mice. After inoculation of α-Syn PFFs, the transgenic mice developed a higher number of phosphorylated α-Syn (p-α-Syn)-positive neurons in the dmX compared with similarly inoculated WT mice. However, the number of p-α-Syn-positive neurons in the dmX decreased over time, and α-Syn pathology was not observed in other brain regions except in the ambiguous nucleus up to 8 months postinoculation. Taken together, BAC transgenic expression of α-Syn facilitated induction of α-Syn pathology in the brainstem, but not subsequent caudo-rostral spread in accordance with Braak's hypothesis.
Collapse
Affiliation(s)
- Norihito Uemura
- Department of Neurology, Kyoto University Graduate School of Medicine, Kyoto, Japan.
| | - Hisashi Yagi
- Department of Chemistry and Biotechnology, Graduate School of Sustainability Science, Tottori University, Tottori, Japan; Center for Research on Green Sustainable Chemistry, Tottori University, Tottori, Japan
| | - Maiko T Uemura
- Department of Neurology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Hodaka Yamakado
- Department of Neurology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Ryosuke Takahashi
- Department of Neurology, Kyoto University Graduate School of Medicine, Kyoto, Japan.
| |
Collapse
|
43
|
Kuo YM, Nwankwo EI, Nussbaum RL, Rogers J, Maccecchini ML. Translational inhibition of α-synuclein by Posiphen normalizes distal colon motility in transgenic Parkinson mice. AMERICAN JOURNAL OF NEURODEGENERATIVE DISEASE 2019; 8:1-15. [PMID: 30906671 PMCID: PMC6420700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Accepted: 01/01/2019] [Indexed: 06/09/2023]
Abstract
Parkinson disease (PD) is a neurodegenerative disease with motor as well as non-motor symptoms, including gastrointestinal dysfunction. In humans, these precede the motor symptoms by decades. Previously developed and characterized transgenic mice expressing the mutant human α-synuclein gene (SNCA) (either A53T or A30P), but not the endogenous mouse Snca, serve as models for familial PD. These animals demonstrate both robust abnormalities in enteric nervous system (ENS) function as well as synuclein-immunoreactive aggregates in ENS ganglia by 3 months of age, recapitulating early gastrointestinal abnormalities seen before the gait impairment characteristics of human and murine PD. Posiphen is a translational inhibitor of α-synuclein that targets the 5' untranslated region (UTR) of SNCA mRNA and could be a potential drug for the treatment of PD. However, its efficacy in ameliorating symptoms of PD has not yet been evaluated. Here, we used these transgenic mouse models to investigate the efficacy of Posiphen in reversing the gastrointestinal dysfunction. We show that Posiphen normalizes the colonic motility of both transgenic mouse models, although it did not affect the Whole Gut Transit Time (WGTT). Pharmacokinetics studies revealed that Posiphen is more abundant in the brain than in blood, in agreement with its lipophilicity, and the main metabolite is N8-NorPosiphen, a molecule with similar properties as Posiphen. The brain Posiphen levels necessary to effect optimal function were calculated and compared with efficacious brain levels from previous studies, showing that a 2-3 mM concentration of Posiphen and metabolites is sufficient for functional efficacy. Finally, 10 mg/kg Posiphen reduced α-synuclein levels in the gut of hSNCAA53T mice treated for twenty-one weeks, while 50 and 65 mg/kg Posiphen reduced α-synuclein levels in the brain of hSNCAA53T mice treated for twenty-one days. In conclusion, this is the first study showing the preclinical efficacy of Posiphen in normalizing the colonic motility in mouse models of gastrointestinal dysfunction in early PD. This result is in agreement with the ability of Posiphen to reach the nervous system, and its mechanism of action, the translational inhibition of α-synuclein expression. These significant findings support further development of Posiphen as a drug for the treatment of PD.
Collapse
Affiliation(s)
- Yien-Ming Kuo
- Institute for Human Genetics, University of California San FranciscoSan Francisco, CA 94143, USA
| | - Ejike Innocent Nwankwo
- Neurochemistry Laboratory, Department of Psychiatry-Neuroscience, Massachusetts General Hospital (East), Harvard Medical SchoolCNY2, Building 149, Charlestown, MA 02129, USA
| | - Robert L Nussbaum
- Invitae Corporation1400 16th Street, San Francisco, CA 94103
- Department of Medicine, University of California San FranciscoSan Francisco, CA 94143, USA
- Institute for Human Genetics, University of California San FranciscoSan Francisco, CA 94143, USA
| | - Jack Rogers
- Neurochemistry Laboratory, Department of Psychiatry-Neuroscience, Massachusetts General Hospital (East), Harvard Medical SchoolCNY2, Building 149, Charlestown, MA 02129, USA
| | | |
Collapse
|
44
|
Prigent A, Gonzales J, Durand T, Le Berre-Scoul C, Rolli-Derkinderen M, Neunlist M, Derkinderen P. Acute inflammation down-regulates alpha-synuclein expression in enteric neurons. J Neurochem 2019; 148:746-760. [PMID: 30589944 DOI: 10.1111/jnc.14656] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Revised: 11/22/2018] [Accepted: 12/21/2018] [Indexed: 12/15/2022]
Abstract
The protein alpha-synuclein whose expression is strongly implicated in Parkinson's disease (PD) is not only expressed in the CNS but also in the enteric nervous system (ENS). The growing body of evidence suggesting that gastrointestinal inflammation is involved in the development of PD led us to investigate the effects of inflammation on alpha-synuclein expression in primary culture of rat ENS and in mice with dextran sulfate sodium-induced colitis. Using western blot and qPCR, we found that both lipopolysaccharide and a combination of tumor necrosis factor-α and interleukin 1-β decreased the expression levels of alpha-synuclein in primary culture of rat ENS, an effect that was prevented in the presence of the p38 inhibitors SB203580 and BIRB 796. Lipopolysaccharide and tumor necrosis factor-α/interleukin 1-β had no effect on alpha-synuclein expression in primary culture of rat CNS and in human erythroid leukemia cells. In mice, acute but not chronic dextran sulfate sodium-induced colitis was associated with a decreased expression of colonic alpha-synuclein. As a whole, our findings indicate that acute inflammatory insults down-regulate alpha-synuclein expression in the ENS via a p38 pathway. They provide new insights into the widely discussed concepts of alpha-synuclein expression and aggregation in the ENS in PD and raise issues about the possible role of gastrointestinal inflammation in the development of PD. OPEN SCIENCE BADGES: This article has received a badge for *Open Materials* because it provided all relevant information to reproduce the study in the manuscript. The complete Open Science Disclosure form for this article can be found at the end of the article. More information about the Open Practices badges can be found at https://cos.io/our-services/open-science-badges/.
Collapse
Affiliation(s)
- Alice Prigent
- Inserm, U1235, Nantes, France.,University Nantes, Nantes, France
| | - Jacques Gonzales
- Inserm, U1235, Nantes, France.,University Nantes, Nantes, France
| | | | | | | | - Michel Neunlist
- Inserm, U1235, Nantes, France.,University Nantes, Nantes, France
| | - Pascal Derkinderen
- Inserm, U1235, Nantes, France.,University Nantes, Nantes, France.,Department of Neurology, CHU Nantes, Nantes, France
| |
Collapse
|
45
|
Extracellular aggregated alpha synuclein primarily triggers lysosomal dysfunction in neural cells prevented by trehalose. Sci Rep 2019; 9:544. [PMID: 30679445 PMCID: PMC6345801 DOI: 10.1038/s41598-018-35811-8] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Accepted: 10/29/2018] [Indexed: 01/30/2023] Open
Abstract
Cell-to-cell propagation of aggregated alpha synuclein (aSyn) has been suggested to play an important role in the progression of alpha synucleinopathies. A critical step for the propagation process is the accumulation of extracellular aSyn within recipient cells. Here, we investigated the trafficking of distinct exogenous aSyn forms and addressed the mechanisms influencing their accumulation in recipient cells. The aggregated aSyn species (oligomers and fibrils) exhibited more pronounced accumulation within recipient cells than aSyn monomers. In particular, internalized extracellular aSyn in the aggregated forms was able to seed the aggregation of endogenous aSyn. Following uptake, aSyn was detected along endosome-to-lysosome and autophagosome-to-lysosome routes. Intriguingly, aggregated aSyn resulted in lysosomal activity impairment, accompanied by the accumulation of dilated lysosomes. Moreover, analysis of autophagy-related protein markers suggested decreased autophagosome clearance. In contrast, the endocytic pathway, proteasome activity, and mitochondrial homeostasis were not substantially affected in recipient cells. Our data suggests that extracellularly added aggregated aSyn primarily impairs lysosomal activity, consequently leading to aSyn accumulation within recipient cells. Importantly, the autophagy inducer trehalose prevented lysosomal alterations and attenuated aSyn accumulation within aSyn-exposed cells. Our study underscores the importance of lysosomes for the propagation of aSyn pathology, thereby proposing these organelles as interventional targets.
Collapse
|
46
|
Issa AR, Sun J, Petitgas C, Mesquita A, Dulac A, Robin M, Mollereau B, Jenny A, Chérif-Zahar B, Birman S. The lysosomal membrane protein LAMP2A promotes autophagic flux and prevents SNCA-induced Parkinson disease-like symptoms in the Drosophila brain. Autophagy 2018; 14:1898-1910. [PMID: 29989488 PMCID: PMC6152503 DOI: 10.1080/15548627.2018.1491489] [Citation(s) in RCA: 97] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Revised: 06/05/2018] [Accepted: 06/13/2018] [Indexed: 02/03/2023] Open
Abstract
The autophagy-lysosome pathway plays a fundamental role in the clearance of aggregated proteins and protection against cellular stress and neurodegenerative conditions. Alterations in autophagy processes, including macroautophagy and chaperone-mediated autophagy (CMA), have been described in Parkinson disease (PD). CMA is a selective autophagic process that depends on LAMP2A (lysosomal-associated membrane protein 2A), a mammal and bird-specific membrane glycoprotein that translocates cytosolic proteins containing a KFERQ-like peptide motif across the lysosomal membrane. Drosophila reportedly lack CMA and use endosomal microautophagy (eMI) as an alternative selective autophagic process. Here we report that neuronal expression of human LAMP2A protected Drosophila against starvation and oxidative stress, and delayed locomotor decline in aging flies without extending their lifespan. LAMP2A also prevented the progressive locomotor and oxidative defects induced by neuronal expression of PD-associated human SNCA (synuclein alpha) with alanine-to-proline mutation at position 30 (SNCAA30P). Using KFERQ-tagged fluorescent biosensors, we observed that LAMP2A expression stimulated selective autophagy in the adult brain and not in the larval fat body, but did not increase this process under starvation conditions. Noteworthy, we found that neurally expressed LAMP2A markedly upregulated levels of Drosophila Atg5, a key macroautophagy initiation protein, and that it increased the density of Atg8a/LC3-positive puncta, which reflects the formation of autophagosomes. Furthermore, LAMP2A efficiently prevented accumulation of the autophagy defect marker Ref(2)P/p62 in the adult brain under acute oxidative stress. These results indicate that LAMP2A can potentiate autophagic flux in the Drosophila brain, leading to enhanced stress resistance and neuroprotection. ABBREVIATIONS Act5C: actin 5C; a.E.: after eclosion; Atg5: autophagy-related 5; Atg8a/LC3: autophagy-related 8a; CMA: chaperone-mediated autophagy; DHE: dihydroethidium; elav: embryonic lethal abnormal vision; eMI: endosomal microautophagy; ESCRT: endosomal sorting complexes required for transport; GABARAP: GABA typeA receptor-associated protein; Hsc70-4: heat shock protein cognate 4; HSPA8/Hsc70: heat shock protein family A (Hsp70) member 8; LAMP2: lysosomal associated membrane protein 2; MDA: malondialdehyde; PA-mCherry: photoactivable mCherry; PBS: phosphate-buffered saline; PCR: polymerase chain reaction; PD: Parkinson disease; Ref(2)P/p62: refractory to sigma P; ROS: reactive oxygen species; RpL32/rp49: ribosomal protein L32; RT-PCR: reverse transcription polymerase chain reaction; SING: startle-induced negative geotaxis; SNCA/α-synuclein: synuclein alpha; SQSTM1/p62: sequestosome 1; TBS: Tris-buffered saline; UAS: upstream activating sequence.
Collapse
Affiliation(s)
- Abdul-Raouf Issa
- Genes Circuits Rhythms and Neuropathology, Brain Plasticity Unit, CNRS, ESPCI Paris, PSL Research University, Paris, France
| | - Jun Sun
- Genes Circuits Rhythms and Neuropathology, Brain Plasticity Unit, CNRS, ESPCI Paris, PSL Research University, Paris, France
| | - Céline Petitgas
- Genes Circuits Rhythms and Neuropathology, Brain Plasticity Unit, CNRS, ESPCI Paris, PSL Research University, Paris, France
| | - Ana Mesquita
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, New York, NY, USA
| | - Amina Dulac
- Genes Circuits Rhythms and Neuropathology, Brain Plasticity Unit, CNRS, ESPCI Paris, PSL Research University, Paris, France
| | - Marion Robin
- ENSL, UCBL, CNRS, LBMC, UMS 3444 Biosciences Lyon Gerland, Université de Lyon, Lyon, France
| | - Bertrand Mollereau
- ENSL, UCBL, CNRS, LBMC, UMS 3444 Biosciences Lyon Gerland, Université de Lyon, Lyon, France
| | - Andreas Jenny
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, New York, NY, USA
| | - Baya Chérif-Zahar
- Genes Circuits Rhythms and Neuropathology, Brain Plasticity Unit, CNRS, ESPCI Paris, PSL Research University, Paris, France
| | - Serge Birman
- Genes Circuits Rhythms and Neuropathology, Brain Plasticity Unit, CNRS, ESPCI Paris, PSL Research University, Paris, France
| |
Collapse
|
47
|
Xie P, Horio F, Fujii I, Zhao J, Shinohara M, Matsukura M. A novel polysaccharide derived from algae extract inhibits cancer progression via JNK, not via the p38 MAPK signaling pathway. Int J Oncol 2018; 52:1380-1390. [PMID: 29512724 PMCID: PMC5873927 DOI: 10.3892/ijo.2018.4297] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Accepted: 02/06/2018] [Indexed: 02/05/2023] Open
Abstract
Cancer has long been one of the most malignant diseases worldwide. Processes in cancer cells are often mediated by Jun N-terminal kinase (JNK), p38 mitogen-activated protein kinase (p38 MAPK) and other signaling pathways. Traditional therapies are often problematic. Recently, a novel polysaccharide derived from algae extract was investigated due to the increasing interest in biological activities of compounds from marine organisms. The effect of this novel polysaccharide on human MKN45 gastric carcinoma cells was determined previously. The current aimed to determine whether the polysaccharide affects other types of cancer, and the deeper mechanisms involved in the process. Human MCF-7 breast cancer cells were used to investigate the novel polysaccharide for its role in the cell growth and migration, and determine the mechanisms affected. MTT assay, nuclear staining and fluorescence activated cell sorting analysis demonstrated that the novel polysaccharide reduced the viability of MCF-7 cells by inducing cell apoptosis and arresting the cells at G2/M phase. Results of western blot analysis demonstrated that phosphorylation of JNK and expression of p53, caspase-9 and caspase-3 were upregulated in the polysaccharide-treated MCF-7 cells. SP600125, an inhibitor of JNK, maintained MCF-7 cell viability, prevented cell apoptosis and cycle arrest, and downregulated the polysaccharide-induced protein phosphorylation/expression. However, a migration assay demonstrated that the novel polysaccharide did not change the migration of MCF-7 cells, as well as the expression of p38 MAPK, and matrix metalloproteinase-9 and -2. Taken together, the current study demonstrated that the novel polysaccharide suppressed cancer cell growth, induced cancer cell apoptosis and cell cycle arrest via JNK signaling, but had no effect on cancer cell migration and p38 MAPK signaling.
Collapse
Affiliation(s)
- Peiyu Xie
- Laboratory of Clinical Pharmacology and Therapeutics, Faculty of Pharmaceutical Sciences, Sojo University, Kumamoto, Kumamoto 860-0822, Japan
| | - Fukuko Horio
- Laboratory of Clinical Pharmacology and Therapeutics, Faculty of Pharmaceutical Sciences, Sojo University, Kumamoto, Kumamoto 860-0822, Japan
| | - Isao Fujii
- Laboratory of Clinical Pharmacology and Therapeutics, Faculty of Pharmaceutical Sciences, Sojo University, Kumamoto, Kumamoto 860-0822, Japan
| | - Jien Zhao
- Ashikita Institution for Developmental Disabilities, Ashikita, Kumamoto 869-541, Japan
| | - Makoto Shinohara
- Ashikita Institution for Developmental Disabilities, Ashikita, Kumamoto 869-541, Japan
| | - Makoto Matsukura
- Laboratory of Clinical Pharmacology and Therapeutics, Faculty of Pharmaceutical Sciences, Sojo University, Kumamoto, Kumamoto 860-0822, Japan
| |
Collapse
|
48
|
Zhao Y, Perera G, Takahashi-Fujigasaki J, Mash DC, Vonsattel JPG, Uchino A, Hasegawa K, Jeremy Nichols R, Holton JL, Murayama S, Dzamko N, Halliday GM. Reduced LRRK2 in association with retromer dysfunction in post-mortem brain tissue from LRRK2 mutation carriers. Brain 2018; 141:486-495. [PMID: 29253086 PMCID: PMC5837795 DOI: 10.1093/brain/awx344] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Revised: 10/03/2017] [Accepted: 10/25/2017] [Indexed: 12/25/2022] Open
Abstract
Missense mutations in leucine-rich repeat kinase 2 (LRRK2) are pathogenic for familial Parkinson's disease. However, it is unknown whether levels of LRRK2 protein in the brain are altered in patients with LRRK2-associated Parkinson's disease. Because LRRK2 mutations are relatively rare, accounting for approximately 1% of all Parkinson's disease, we accessioned cases from five international brain banks to investigate levels of the LRRK2 protein, and other genetically associated Parkinson's disease proteins. Brain tissue was obtained from 17 LRRK2 mutation carriers (12 with the G2019S mutation and five with the I2020T mutation) and assayed by immunoblot. Compared to matched controls and idiopathic Parkinson's disease cases, we found levels of LRRK2 protein were reduced in the LRRK2 mutation cases. We also measured a decrease in two other proteins genetically implicated in Parkinson's disease, the core retromer component, vacuolar protein sorting associated protein 35 (VPS35), and the lysosomal hydrolase, glucocerebrosidase (GBA). Moreover, the classical retromer cargo protein, cation-independent mannose-6-phosphate receptor (MPR300, encoded by IGF2R), was also reduced in the LRRK2 mutation cohort and protein levels of the receptor were correlated to levels of LRRK2. These results provide new data on LRRK2 protein expression in brain tissue from LRRK2 mutation carriers and support a relationship between LRRK2 and retromer dysfunction in LRRK2-associated Parkinson's disease brain.
Collapse
Affiliation(s)
- Ye Zhao
- Brain and Mind Centre, Sydney Medical School, University of Sydney, Camperdown, 2050, Australia
- School of Medical Sciences, University of NSW, Kensington, 2033, Australia
- Neuroscience Research Australia, Randwick, 2031, Australia
| | - Gayathri Perera
- Brain and Mind Centre, Sydney Medical School, University of Sydney, Camperdown, 2050, Australia
- Neuroscience Research Australia, Randwick, 2031, Australia
| | - Junko Takahashi-Fujigasaki
- Department of Neuropathology, Brain Bank for Aging Research, Tokyo Metropolitan Geriatric0 Hospital and Institute of Gerontology, Tokyo, 173-0015, Japan
| | - Deborah C Mash
- University of Miami Brain Endowment Bank™, University of Miami Miller School of Medicine, Miami, Florida, 33136, USA
| | - Jean Paul G Vonsattel
- New York Brain Bank, Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York, 10032, USA
| | - Akiko Uchino
- Department of Neuropathology, Brain Bank for Aging Research, Tokyo Metropolitan Geriatric0 Hospital and Institute of Gerontology, Tokyo, 173-0015, Japan
| | - Kazuko Hasegawa
- Department of Neurology, Sagamihara National Hospital, Kangawa, 252-0315, Japan
| | - R Jeremy Nichols
- Parkinson’s Institute and Clinical Center, Sunnyvale, California, 94085, USA
| | - Janice L Holton
- Queen Square Brain Bank, UCL Institute of Neurology, University College London, London, WC1N 1PJ, UK
| | - Shigeo Murayama
- Department of Neuropathology, Brain Bank for Aging Research, Tokyo Metropolitan Geriatric0 Hospital and Institute of Gerontology, Tokyo, 173-0015, Japan
| | - Nicolas Dzamko
- Brain and Mind Centre, Sydney Medical School, University of Sydney, Camperdown, 2050, Australia
- School of Medical Sciences, University of NSW, Kensington, 2033, Australia
- Neuroscience Research Australia, Randwick, 2031, Australia
| | - Glenda M Halliday
- Brain and Mind Centre, Sydney Medical School, University of Sydney, Camperdown, 2050, Australia
- School of Medical Sciences, University of NSW, Kensington, 2033, Australia
- Neuroscience Research Australia, Randwick, 2031, Australia
| |
Collapse
|
49
|
Moritz CP. Tubulin or Not Tubulin: Heading Toward Total Protein Staining as Loading Control in Western Blots. Proteomics 2018; 17. [PMID: 28941183 DOI: 10.1002/pmic.201600189] [Citation(s) in RCA: 122] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Revised: 09/13/2017] [Indexed: 12/30/2022]
Abstract
Western blotting is an analytical method widely used for detecting and (semi-)quantifying specific proteins in given samples. Western blots are continuously applied and developed by the protein community. This review article focuses on a significant, but not yet well-established, improvement concerning the internal loading control as a prerequisite to accurately quantifying Western blots. Currently, housekeeping proteins (HKPs) like actin, tubulin, or GAPDH are often used to check for equal loading or to compensate potential loading differences. However, this loading control has multiple drawbacks. Staining of the total protein on the blotting membrane has emerged as a better loading control. Total protein staining (TPS) represents the actual loading amount more accurately than HKPs due to minor technical and biological variation. Further, the broad dynamic range of TPS solves the issue of HKPs that commonly fail to show loading differences above small loading amounts of 0.5-10 μg. Although these and further significant advantages have been demonstrated over the past 10 years, only a small percentage of laboratories take advantage of it. The objective of this review article is to collect and compare information about TPS options and to invite users to reconsider their applied loading control. Nine benefits of TPS are discussed and seven different variants are critically evaluated by comparing technical details. Consequently, this review article offers an orientation in selecting the appropriate staining type. I conclude that TPS should be the preferred loading control in future Western blot approaches.
Collapse
Affiliation(s)
- Christian P Moritz
- Synaptopathies and Autoantibodies, Faculty of Medicine, University Jean Monnet, Saint-Étienne, France.,Institut NeuroMyoGène, Team Synaptopathies and Autoantibodies, Lyon/Saint-Étienne, France
| |
Collapse
|
50
|
Minakaki G, Menges S, Kittel A, Emmanouilidou E, Schaeffner I, Barkovits K, Bergmann A, Rockenstein E, Adame A, Marxreiter F, Mollenhauer B, Galasko D, Buzás EI, Schlötzer-Schrehardt U, Marcus K, Xiang W, Lie DC, Vekrellis K, Masliah E, Winkler J, Klucken J. Autophagy inhibition promotes SNCA/alpha-synuclein release and transfer via extracellular vesicles with a hybrid autophagosome-exosome-like phenotype. Autophagy 2018; 14:98-119. [PMID: 29198173 DOI: 10.1080/15548627.2017.1395992] [Citation(s) in RCA: 209] [Impact Index Per Article: 29.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The autophagy-lysosome pathway (ALP) regulates intracellular homeostasis of the cytosolic protein SNCA/alpha-synuclein and is impaired in synucleinopathies, including Parkinson disease and dementia with Lewy bodies (DLB). Emerging evidence suggests that ALP influences SNCA release, but the underlying cellular mechanisms are not well understood. Several studies identified SNCA in exosome/extracellular vesicle (EV) fractions. EVs are generated in the multivesicular body compartment and either released upon its fusion with the plasma membrane, or cleared via the ALP. We therefore hypothesized that inhibiting ALP clearance 1) enhances SNCA release via EVs by increasing extracellular shuttling of multivesicular body contents, 2) alters EV biochemical profile, and 3) promotes SNCA cell-to-cell transfer. Indeed, ALP inhibition increased the ratio of extra- to intracellular SNCA and upregulated SNCA association with EVs in neuronal cells. Ultrastructural analysis revealed a widespread, fused multivesicular body-autophagosome compartment. Biochemical characterization revealed the presence of autophagosome-related proteins, such as LC3-II and SQSTM1. This distinct "autophagosome-exosome-like" profile was also identified in human cerebrospinal fluid (CSF) EVs. After a single intracortical injection of SNCA-containing EVs derived from CSF into mice, human SNCA colocalized with endosome and neuronal markers. Prominent SNCA immunoreactivity and a higher number of neuronal SNCA inclusions were observed after DLB patient CSF EV injections. In summary, this study provides compelling evidence that a) ALP inhibition increases SNCA in neuronal EVs, b) distinct ALP components are present in EVs, and c) CSF EVs transfer SNCA from cell to cell in vivo. Thus, macroautophagy/autophagy may regulate EV protein composition and consequently progression in synucleinopathies.
Collapse
Affiliation(s)
- Georgia Minakaki
- a Department of Molecular Neurology , University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) , Erlangen , Germany
| | - Stefanie Menges
- a Department of Molecular Neurology , University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) , Erlangen , Germany
| | - Agnes Kittel
- b Department of Pharmacology, Institute of Experimental Medicine, Hungarian Academy of Sciences , Semmelweis University , Budapest , Hungary
| | - Evangelia Emmanouilidou
- c Department of Neuroscience, Center for Basic Research , Biomedical Research Foundation of the Academy of Athens , Athens , Greece
| | | | - Katalin Barkovits
- e Medizinisches Proteom-Center, Medical Faculty , Ruhr University Bochum , Bochum , Germany
| | - Anna Bergmann
- a Department of Molecular Neurology , University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) , Erlangen , Germany
| | - Edward Rockenstein
- f Department of Neurosciences , University of California , San Diego , CA USA
| | - Anthony Adame
- f Department of Neurosciences , University of California , San Diego , CA USA
| | - Franz Marxreiter
- a Department of Molecular Neurology , University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) , Erlangen , Germany
| | - Brit Mollenhauer
- g Paracelsus-Elena-Klinik, Kassel and Departments of Neuropathology & Neurosurgery , & University Medical Center , Göttingen
| | - Douglas Galasko
- f Department of Neurosciences , University of California , San Diego , CA USA
| | - Edit Irén Buzás
- h Department of Genetics, Cell and Immunobiology , Semmelweis University , Budapest , Hungary
| | | | - Katrin Marcus
- e Medizinisches Proteom-Center, Medical Faculty , Ruhr University Bochum , Bochum , Germany
| | - Wei Xiang
- d Institute of Biochemistry , FAU , Erlangen , Germany
| | | | - Kostas Vekrellis
- c Department of Neuroscience, Center for Basic Research , Biomedical Research Foundation of the Academy of Athens , Athens , Greece
| | - Eliezer Masliah
- f Department of Neurosciences , University of California , San Diego , CA USA.,j Department of Pathology , University of California , San Diego , CA USA
| | - Jürgen Winkler
- a Department of Molecular Neurology , University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) , Erlangen , Germany
| | - Jochen Klucken
- a Department of Molecular Neurology , University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) , Erlangen , Germany
| |
Collapse
|