1
|
Duarte-Zambrano F, Alfonso-Cedeño DF, Barrero JA, Rodríguez-Vanegas LA, Moreno-Cárdenas V, Olarte-Díaz A, Arboleda G, Arboleda H. Genetic variants associated with idiopathic Parkinson's disease in Latin America: A systematic review. Neurogenetics 2025; 26:43. [PMID: 40178685 PMCID: PMC11968493 DOI: 10.1007/s10048-025-00817-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2025] [Accepted: 03/02/2025] [Indexed: 04/05/2025]
Abstract
Idiopathic Parkinson's disease (PD) constitutes a complex trait influenced by genetic, environmental, and lifestyle factors, with an estimated heritability of nearly 30%. However, a large proportion of the heritable variation linked to PD remains uncertain, partly due to ancestral bias. Expanding research into Hispanic populations can contribute to address this gap. To review the evidence of genetic variants associated with idiopathic PD in Latin America. A PRISMA-compliant systematic review was conducted in MEDLINE, EMBASE and LILACS, compiling studies published up to February 7, 2025. Nineteen case-control studies were included. Two hypothesis-free studies identified rs525496 near H2BW1 as a protective factor and rs356182 in SNCA as a risk factor through XWAS and GWAS, respectively. Seventeen hypothesis-driven studies examined over three hundred variants, identifying nineteen genetic markers; risk factors included one INDEL in NR4A2, CNV burdens in PRKN, SNCA, and PLA2G6, along with fourteen variants in six loci including GBA, APOEε4, MTHFR, LRRK2, and SNCA. Three SNPs in the PICALM, ALDH1A1, and APOE-ε3 loci were identified as protective factors. Additionally, six SNCA variant haplotypes appear to increase PD risk, while two NR4A2 INDELs haplotypes showed mixed effects. This review summarized genetic loci associated with idiopathic PD in Latin American populations evidencing an overlap with European findings as well as novel loci, although awaiting replication and validation. These observations contribute to the understanding of genetic configuration of the disease and highlight the need for further genomic research in underrepresented groups that include local ancestry analysis within admixed cohorts to guide development of personalized treatments and population-specific interventions.
Collapse
Affiliation(s)
- Felipe Duarte-Zambrano
- Semillero de Investigación en Neurociencias, Facultad de Medicina, Universidad Nacional de Colombia, Bogotá, Colombia.
- Grupo de Investigación en Neurociencias y Muerte Celular, Facultad de Medicina E Instituto de Genética, Universidad Nacional de Colombia, Bogotá, Colombia.
| | - David Felipe Alfonso-Cedeño
- Semillero de Investigación en Neurociencias, Facultad de Medicina, Universidad Nacional de Colombia, Bogotá, Colombia
| | - Jorge A Barrero
- Semillero de Investigación en Neurociencias, Facultad de Medicina, Universidad Nacional de Colombia, Bogotá, Colombia
| | | | - Valentina Moreno-Cárdenas
- Semillero de Investigación en Neurociencias, Facultad de Medicina, Universidad Nacional de Colombia, Bogotá, Colombia
| | - Anamaría Olarte-Díaz
- Semillero de Investigación en Neurociencias, Facultad de Medicina, Universidad Nacional de Colombia, Bogotá, Colombia
| | - Gonzalo Arboleda
- Grupo de Investigación en Neurociencias y Muerte Celular, Facultad de Medicina E Instituto de Genética, Universidad Nacional de Colombia, Bogotá, Colombia
- Departamento de Patología, Facultad de Medicina, Universidad Nacional de Colombia, Bogotá, Colombia
| | - Humberto Arboleda
- Semillero de Investigación en Neurociencias, Facultad de Medicina, Universidad Nacional de Colombia, Bogotá, Colombia
- Grupo de Investigación en Neurociencias y Muerte Celular, Facultad de Medicina E Instituto de Genética, Universidad Nacional de Colombia, Bogotá, Colombia
- Departamento de Pediatría E Instituto de Genética, Facultad de Medicina, Universidad Nacional de Colombia, Bogotá, Colombia
| |
Collapse
|
2
|
Frattini E, Faustini G, Lopez G, Carsana EV, Tosi M, Trezzi I, Magni M, Soldà G, Straniero L, Facchi D, Samarani M, Martá-Ariza M, De Luca CMG, Vezzoli E, Pittaro A, Stepanyan A, Silipigni R, Rosety I, Schwamborn JC, Sardi SP, Moda F, Corti S, Comi GP, Blandini F, Tritsch NX, Bortolozzi M, Ferrero S, Cribiù FM, Wisniewski T, Asselta R, Aureli M, Bellucci A, Di Fonzo A. Lewy pathology formation in patient-derived GBA1 Parkinson's disease midbrain organoids. Brain 2025; 148:1242-1257. [PMID: 39570889 PMCID: PMC11967528 DOI: 10.1093/brain/awae365] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 09/13/2024] [Accepted: 10/01/2024] [Indexed: 04/05/2025] Open
Abstract
Fibrillary aggregation of α-synuclein in Lewy body inclusions and nigrostriatal dopaminergic neuron degeneration define Parkinson's disease neuropathology. Mutations in GBA1, encoding glucocerebrosidase, are the most frequent genetic risk factor for Parkinson's disease. However, the lack of reliable experimental models able to reproduce key neuropathological signatures has hampered clarification of the link between mutant glucocerebrosidase and Parkinson's disease pathology. Here, we describe an innovative protocol for the generation of human induced pluripotent stem cell-derived midbrain organoids containing dopaminergic neurons with nigral identity that reproduce characteristics of advanced maturation. When applied to patients with GBA1-related Parkinson's disease, this method enabled the differentiation of midbrain organoids recapitulating dopaminergic neuron loss and fundamental features of Lewy pathology observed in human brains, including the generation of α-synuclein fibrillary aggregates with seeding activity that also propagate pathology in healthy control organoids. Concurrently, we found that the retention of mutant glucocerebrosidase in the endoplasmic reticulum and increased levels of its substrate, glucosylceramide, are determinants of α-synuclein aggregation into Lewy body-like inclusions, and the reduction of glucocerebrosidase activity accelerated α-synuclein pathology by promoting fibrillary α-synuclein deposition. Finally, we demonstrated the efficacy of ambroxol and GZ667161 (two modulators of the glucocerebrosidase pathway in clinical development for the treatment of GBA1-related Parkinson's disease) in reducing α-synuclein pathology in this model, supporting the use of midbrain organoids as a relevant preclinical platform for investigational drug screening.
Collapse
Affiliation(s)
- Emanuele Frattini
- Neurology Unit, IRCCS Foundation Ca’ Granda Ospedale Maggiore Policlinico, Milan 20122, Italy
| | - Gaia Faustini
- Department of Molecular and Translational Medicine, University of Brescia, Brescia 25123, Italy
| | - Gianluca Lopez
- Division of Pathology, IRCCS Foundation Ca’ Granda Ospedale Maggiore Policlinico, University of Milan, Milan 20122, Italy
| | - Emma Veronica Carsana
- Department of Medical Biotechnology and Translational Medicine, University of Milano, Milan 20054, Italy
| | - Mattia Tosi
- Neurology Unit, IRCCS Foundation Ca’ Granda Ospedale Maggiore Policlinico, Milan 20122, Italy
| | - Ilaria Trezzi
- Neurology Unit, IRCCS Foundation Ca’ Granda Ospedale Maggiore Policlinico, Milan 20122, Italy
| | - Manuela Magni
- Neurology Unit, IRCCS Foundation Ca’ Granda Ospedale Maggiore Policlinico, Milan 20122, Italy
| | - Giulia Soldà
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan 20072, Italy
- Medical Genetics and RNA Biology Unit, IRCCS Humanitas Research Hospital, Rozzano, Milan 20089, Italy
| | - Letizia Straniero
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan 20072, Italy
- Medical Genetics and RNA Biology Unit, IRCCS Humanitas Research Hospital, Rozzano, Milan 20089, Italy
| | - Daniele Facchi
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan 20072, Italy
- Medical Genetics and RNA Biology Unit, IRCCS Humanitas Research Hospital, Rozzano, Milan 20089, Italy
| | - Maura Samarani
- Unité de Trafic Membranaire et Pathogénèse, Département de Biologie Cellulaire et de l’Infection, Institut Pasteur, Paris 75015, France
| | - Mitchell Martá-Ariza
- Center for Cognitive Neurology, Department of Neurology, New York University Grossman School of Medicine, New York, NY 10016, USA
- Institut de Neurociències, Universitat Autònoma de Barcelona, Barcelona 08193, Spain
| | - Chiara Maria Giulia De Luca
- Division of Neurology 5 and Neuropathology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan 20133, Italy
| | - Elena Vezzoli
- Advanced Light and Electron Microscopy BioImaging Centre (ALEMBIC), IRCCS San Raffaele Scientific Institute, Milan 20132, Italy
| | - Alessandra Pittaro
- Division of Pathology, IRCCS Foundation Ca’ Granda Ospedale Maggiore Policlinico, University of Milan, Milan 20122, Italy
| | - Astghik Stepanyan
- Unità Operativa Complessa, Chirurgia Generale 3, University Hospital of Padua, Padua 35128, Italy
| | - Rosamaria Silipigni
- Laboratory of Medical Genetics, IRCCS Foundation Ca’ Granda Ospedale Maggiore Policlinico, Milan 20122, Italy
| | - Isabel Rosety
- Luxembourg Centre for Systems Biomedicine (LCSB), Developmental and Cellular Biology, University of Luxembourg, Belvaux L-4367, Luxembourg
| | - Jens C Schwamborn
- Luxembourg Centre for Systems Biomedicine (LCSB), Developmental and Cellular Biology, University of Luxembourg, Belvaux L-4367, Luxembourg
| | - Sergio Pablo Sardi
- Rare and Neurological Diseases Therapeutic Area, Sanofi, Framingham, MA 01701, USA
| | - Fabio Moda
- Division of Neurology 5 and Neuropathology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan 20133, Italy
| | - Stefania Corti
- Neurology Unit, IRCCS Foundation Ca’ Granda Ospedale Maggiore Policlinico, Milan 20122, Italy
- Department of Pathophysiology and Transplantation (DEPT), Dino Ferrari Centre, University of Milan, Milan 20122, Italy
| | - Giacomo P Comi
- Neurology Unit, IRCCS Foundation Ca’ Granda Ospedale Maggiore Policlinico, Milan 20122, Italy
- Department of Pathophysiology and Transplantation (DEPT), Dino Ferrari Centre, University of Milan, Milan 20122, Italy
| | - Fabio Blandini
- Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan 20122, Italy
| | - Nicolas X Tritsch
- Neuroscience Institute, New York University Grossman School of Medicine, New York, NY 10016, USA
- Fresco Institute for Parkinson’s and Movement Disorders, New York University Langone Health, New York, NY 10017, USA
| | - Mario Bortolozzi
- Department of Physics and Astronomy ‘G. Galilei’, University of Padua, Padua 35131, Italy
- Veneto Institute of Molecular Medicine (VIMM), Padua 35129, Italy
| | - Stefano Ferrero
- Division of Pathology, IRCCS Foundation Ca’ Granda Ospedale Maggiore Policlinico, University of Milan, Milan 20122, Italy
- Department of Biomedical, Surgical, and Dental Sciences, University of Milan, Milan 20122, Italy
| | - Fulvia Milena Cribiù
- Division of Pathology, IRCCS Foundation Ca’ Granda Ospedale Maggiore Policlinico, University of Milan, Milan 20122, Italy
| | - Thomas Wisniewski
- Center for Cognitive Neurology, Department of Neurology, New York University Grossman School of Medicine, New York, NY 10016, USA
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA
- Department of Psychiatry, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Rosanna Asselta
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan 20072, Italy
- Medical Genetics and RNA Biology Unit, IRCCS Humanitas Research Hospital, Rozzano, Milan 20089, Italy
| | - Massimo Aureli
- Department of Medical Biotechnology and Translational Medicine, University of Milano, Milan 20054, Italy
| | - Arianna Bellucci
- Department of Molecular and Translational Medicine, University of Brescia, Brescia 25123, Italy
| | - Alessio Di Fonzo
- Neurology Unit, IRCCS Foundation Ca’ Granda Ospedale Maggiore Policlinico, Milan 20122, Italy
| |
Collapse
|
3
|
Jia T, Yang F, Qin F, He Y, Han F, Zhang C. Identification of Common Brain Protein and Genetic Loci Between Parkinson's Disease and Lewy Body Dementia. CNS Neurosci Ther 2025; 31:e70370. [PMID: 40202048 PMCID: PMC11979625 DOI: 10.1111/cns.70370] [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: 03/25/2024] [Revised: 02/25/2025] [Accepted: 03/17/2025] [Indexed: 04/10/2025] Open
Abstract
BACKGROUND Parkinson's disease (PD) and Lewy body dementia (LBD) have many common features, including clinical manifestations, neurochemistry, and pathology, but little is known about their shared brain proteins and genetic factors. METHODS To identify susceptibility-related brain proteins that are shared between PD and LBD patients, proteome-wide association studies (PWASs) were conducted by integrating human brain protein quantitative trait loci (pQTLs) with large-scale genome-wide association studies (GWASs) of both diseases. Subsequently, pleiotropy-informed conditional false discovery rate (pleioFDR) analysis was performed to identify common risk genetic loci between PD and LBD. Finally, the downregulation of these risk genes in different disease states was validated by differential gene expression analysis. RESULTS PWASs identified 12 PD risk proteins and nine LBD risk proteins, among which TMEM175 (zPD = -7.25, PPD = 4.12E-13; zLBD = -6.02, PLBD = 1.75E-09) and DOC2A (zPD = -4.13, PPD = 3.71E-05; zLBD = -3.91, PLBD = 9.08E-05) were shared. PleioFDR analysis revealed that five genetic risk loci mapped to eight genes associated with PD and LBD, including the proteome-wide significant risk gene TMEM175 (ConjFDR = 5.74E-03). Differential expression analysis verified that TMEM175 was significantly downregulated in the midbrains of PD patients (p = 1.19E-02), and further exploration revealed that TMEM175 was also dramatically downregulated in the substantia nigra of PD patients (p = 1.16E-02) and incidental Lewy body disease patients (p = 7.52E-03). Moreover, TMEM175 was significantly downregulated in induced pluripotent stem cell-derived dopaminergic neurons from PD patients (p = 4.60E-02). CONCLUSION Dysregulation of TMEM175 may confer PD and LBD risk and may be partly responsible for their comorbidity. Our results revealed the common genetic risk factors between PD and LBD, which elucidated the shared genetic basis of these diseases.
Collapse
Affiliation(s)
- Tingting Jia
- Mental Health Center and Psychiatric Laboratory, the State Key Laboratory of BiotherapyWest China Hospital of Sichuan UniversityChengduSichuanChina
- Department of Gastroenterology and Hepatology and Sichuan University‐University of Oxford Huaxi Joint Centre for Gastrointestinal CancerWest China Hospital of Sichuan UniversityChengduSichuanChina
| | - Fuhua Yang
- Department of NephrologyThe Sixth People's Hospital of ChengduChengduSichuanChina
| | - Fengqin Qin
- Department of NeurologyThe 3rd Affiliated Hospital of Chengdu Medical CollegeChengduSichuanChina
| | - Yongji He
- Clinical Trial Center, National Medical Products Administration key Laboratory for Clinical Research and Evaluation of Innovative DrugsWest China Hospital Sichuan UniversityChengduChina
| | - Feng Han
- Department of Emergency MedicineHainan General Hospital, Hainan Affiliated Hospital of Hainan Medical UniversityHaikouChina
| | - Chengcheng Zhang
- Mental Health Center and Psychiatric Laboratory, the State Key Laboratory of BiotherapyWest China Hospital of Sichuan UniversityChengduSichuanChina
| |
Collapse
|
4
|
Li Y, Torok J, Ding J, Wang N, Lau C, Kulkarni S, Anand C, Tran J, Cheng M, Lo C, Lu B, Sun Y, Yang X, Raj A, Peng C. Distinguish risk genes functioning at presynaptic or postsynaptic regions and key connectomes associated with pathological α-synuclein spreading. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.11.642462. [PMID: 40161679 PMCID: PMC11952395 DOI: 10.1101/2025.03.11.642462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Previous studies have suggested that pathological α-synuclein (α-Syn) mainly transmits along the neuronal network, but several key questions remain unanswered: (1) How many and which connections in the connectome are necessary for predicting the progression of pathological α-Syn? (2) How to identify risk gene that affects pathology spreading functioning at presynaptic or postsynaptic regions, and are these genes enriched in different cell types? Here, we addressed these key questions with novel mathematical models. Strikingly, the spreading of pathological α-Syn is predominantly determined by the key subnetworks composed of only 2% of the strongest connections in the connectome. We further explored the genes that are responsible for the selective vulnerability of different brain regions to transmission to distinguish the genes that play roles in presynaptic from those in postsynaptic regions. Those risk genes were significantly enriched in microglial cells of presynaptic regions and neurons of postsynaptic regions. Gene regulatory network analyses were then conducted to identify 'key drivers' of genes responsible for selective vulnerability and overlapping with Parkinson's disease risk genes. By identifying and discriminating between key gene mediators of transmission operating at presynaptic and postsynaptic regions, our study has demonstrated for the first time that these are functionally distinct processes.
Collapse
|
5
|
Song X, Liu T, Yu L, Ji Q, Guo X, Zong R, Li Y, Huang G, Xue Q, Fu Q, Liu B, Zheng Y, Chen L, Gao C, Liu H. OTUD5 Protects Dopaminergic Neurons by Promoting the Degradation of α-Synuclein in Parkinson's Disease Model. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2406700. [PMID: 39721018 PMCID: PMC11831440 DOI: 10.1002/advs.202406700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 12/04/2024] [Indexed: 12/28/2024]
Abstract
Defective clearance and accumulation of α-synuclein (α-Syn) is the key pathogenic factor in Parkinson's disease (PD). Recent studies emphasize the importance of E3 ligases in regulating the degradation of α-Syn. However, the molecular mechanisms by which deubiquitinases regulate α-Syn degradation are scarcely studied. In this study, it is found that the protein levels of α-Syn are negatively regulated by ovarian tumor protease deubiquitinase 5 (OTUD5) which protects dopaminergic (DA) neurons in the PD model. Mechanistically, OTUD5 promotes K63-linked polyubiquitination of α-Syn independent of its deubiquitinating enzyme activity and mediates its endolysosomal degradation by recruiting the E3 ligase neural precursor cell expressed developmentally downregulated 4 (NEDD4). Furthermore, OTUD5 conditional knockout in DA neurons results in more severe α-Syn related pathology and dyskinesia after injection of α-Syn preformed fibrils (PFF). Overall, the data unveil a novel mechanism to regulate the degradation of α-Syn and provide a new therapeutic strategy to alleviate DA neurodegeneration.
Collapse
Affiliation(s)
- Xiaomeng Song
- Department of PharmacologySchool of Basic Medical SciencesShandong UniversityJinanShandong250012P. R. China
| | - Tengfei Liu
- Department of PharmacologySchool of Basic Medical SciencesShandong UniversityJinanShandong250012P. R. China
| | - Lu Yu
- Department of PharmacologySchool of Basic Medical SciencesShandong UniversityJinanShandong250012P. R. China
| | - Qiuran Ji
- Department of PharmacologySchool of Basic Medical SciencesShandong UniversityJinanShandong250012P. R. China
| | - Xin Guo
- Department of PharmacologySchool of Basic Medical SciencesShandong UniversityJinanShandong250012P. R. China
| | - Runzhe Zong
- Department of PharmacologySchool of Basic Medical SciencesShandong UniversityJinanShandong250012P. R. China
| | - Yiquan Li
- Department of PharmacologySchool of Basic Medical SciencesShandong UniversityJinanShandong250012P. R. China
| | - Gan Huang
- Department of PharmacologySchool of Basic Medical SciencesShandong UniversityJinanShandong250012P. R. China
| | - Qidi Xue
- Department of PharmacologySchool of Basic Medical SciencesShandong UniversityJinanShandong250012P. R. China
| | - Qingyi Fu
- Department of PharmacologySchool of Basic Medical SciencesShandong UniversityJinanShandong250012P. R. China
| | - Bingyu Liu
- Key Laboratory of Infection and Immunity of Shandong ProvinceSchool of Basic Medical SciencesShandong UniversityJinanShandong250012P. R. China
- Department of ImmunologySchool of Basic Medical SciencesShandong UniversityJinanShandong250012P. R. China
| | - Yi Zheng
- Key Laboratory of Infection and Immunity of Shandong ProvinceSchool of Basic Medical SciencesShandong UniversityJinanShandong250012P. R. China
- Department of ImmunologySchool of Basic Medical SciencesShandong UniversityJinanShandong250012P. R. China
| | - Lin Chen
- Department of PharmacologySchool of Basic Medical SciencesShandong UniversityJinanShandong250012P. R. China
| | - Chengjiang Gao
- Key Laboratory of Infection and Immunity of Shandong ProvinceSchool of Basic Medical SciencesShandong UniversityJinanShandong250012P. R. China
- Department of ImmunologySchool of Basic Medical SciencesShandong UniversityJinanShandong250012P. R. China
| | - Huiqing Liu
- Department of PharmacologySchool of Basic Medical SciencesShandong UniversityJinanShandong250012P. R. China
- Department of Rehabilitation MedicineThe Second HospitalShandong UniversityJinanShandong250012P. R. China
| |
Collapse
|
6
|
La Vitola P, Szegö EM, Pinto-Costa R, Rollar A, Harbachova E, Schapira AH, Ulusoy A, Di Monte DA. Mitochondrial oxidant stress promotes α-synuclein aggregation and spreading in mice with mutated glucocerebrosidase. NPJ Parkinsons Dis 2024; 10:233. [PMID: 39663354 PMCID: PMC11634889 DOI: 10.1038/s41531-024-00842-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 11/19/2024] [Indexed: 12/13/2024] Open
Abstract
In this study, heterozygous expression of a common Parkinson-associated GBA1 variant, the L444P mutation, was found to exacerbate α-synuclein aggregation and spreading in a mouse model of Parkinson-like pathology targeting neurons of the medullary vagal system. These neurons were also shown to become more vulnerable to oxidative and nitrative stress after L444P expression. The latter paralleled neuronal formation of reactive oxygen species and led to a pronounced accumulation of nitrated α-synuclein. A causal relationship linked mutation-induced oxidative/nitrative stress to enhanced α-synuclein aggregation and spreading that could indeed be rescued by neuronal overexpression of mitochondrial superoxide dismutase 2. Further evidence supported a key involvement of mitochondria as sources of reactive oxygen species as well as targets of oxidative and nitrative damage within L444P-expressing neurons. These findings support the conclusion that enhanced vulnerability to mitochondrial oxidative stress should be considered an important mechanism predisposing to pathology conversion in carriers of GBA1 mutations.
Collapse
Affiliation(s)
- Pietro La Vitola
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Eva M Szegö
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Rita Pinto-Costa
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Angela Rollar
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Eugenia Harbachova
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Anthony Hv Schapira
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
- Department of Clinical and Movement Neurosciences, University College London Queen Square Institute of Neurology, Royal Free Campus, London, UK
| | - Ayse Ulusoy
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Donato A Di Monte
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany.
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA.
| |
Collapse
|
7
|
Zhang X, Zhao Y, Jiang L, Hu Y, Liu Z, Xu Q, Wang C, Lei L, Li P, Tan Z, Wu H, Shen L, Jiang H, Yan X, Tang B, Guo J. Exposure factors and clinical characteristics associated with Parkinson's disease in GBA1 variant carriers: A Chinese GBA1-PD intrafamilial survey. Parkinsonism Relat Disord 2024; 130:107212. [PMID: 39581168 DOI: 10.1016/j.parkreldis.2024.107212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Revised: 10/10/2024] [Accepted: 11/16/2024] [Indexed: 11/26/2024]
Abstract
INTRODUCTION Glucosylceramidase beta 1 (GBA1) mutations are a genetic risk factor for Parkinson's disease (PD), though most carriers do not develop the disease. This study aimed to identify exposure factors linked to PD in GBA1 carriers and assess clinical features and the probability of prodromal PD in non-manifesting carriers. METHODS Data from the Parkinson's Disease & Movement Disorders Multicenter Database and Collaborative Network in China was used, including 59 GBA1 non-manifesting carriers, 62 controls, and 107 GBA1-associated PD, of whom 81 were in the early stage. Exposure factors included pesticide/solvent exposure, smoking, alcohol, and tea consumption. Logistic regression assessed the association between exposure factors and PD. Clinical characteristics were evaluated using multiple scales, relevant markers were collected based on the Movement Disorders Society criteria. A naive Bayesian classifier method determined the probability of prodromal PD in GBA1 non-manifesting carriers and controls. RESULTS After adjusting for sociodemographic variables, pesticide/solvent exposure was positively associated with PD in GBA1 carriers (OR 8.40; 95 % CI 2.50-28.20), while smoking was inversely associated with PD (OR 0.18; 95 % CI 0.05-0.62). Rapid eye movement sleep behavior disorder, constipation, hyposmia, and cognitive deficits were more severe in early-stage GBA1-associated PD than in carriers and controls. Clinical symptoms and the probability of prodromal PD were similar between carriers and controls. CONCLUSIONS PD in GBA1 carriers is closely linked to exposure factors. Early-stage GBA1-associated PD shows significant prodromal symptoms, which are not evident in carriers. The probability of prodromal PD in carriers is similar to that in controls.
Collapse
Affiliation(s)
- Xuxiang Zhang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Yuwen Zhao
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Li Jiang
- Department of Neurology, & Multi-Omics Research Center for Brain Disorders, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421000, China
| | - Yuxuan Hu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Zhenhua Liu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China; Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, 410008, China; Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, Hunan, 410008, China
| | - Qian Xu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Chunyu Wang
- Department of Neurology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Lifang Lei
- Department of Neurology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Peishan Li
- Department of Neurology, The Second Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, 830054, China
| | - Zhihui Tan
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Heng Wu
- Department of Neurology, & Multi-Omics Research Center for Brain Disorders, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421000, China
| | - Lu Shen
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China; Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, Hunan, 410008, China; Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Changsha, Hunan, 410008, China; Engineering Research Center of Hunan Province in Cognitive Impairment Disorders, Changsha, Hunan, 410008, China
| | - Hong Jiang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China; Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, 410008, China; Department of Neurology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China; Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Changsha, Hunan, 410008, China
| | - Xinxiang Yan
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Beisha Tang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China; Department of Neurology, & Multi-Omics Research Center for Brain Disorders, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421000, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China; Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, 410008, China; Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, Hunan, 410008, China; Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Changsha, Hunan, 410008, China
| | - Jifeng Guo
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China; Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, 410008, China; Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, Hunan, 410008, China; Department of Neurology, The Second Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, 830054, China; Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Changsha, Hunan, 410008, China.
| |
Collapse
|
8
|
Vatsa N, Brynildsen JK, Goralski TM, Kurgat K, Meyerdirk L, Breton L, DeWeerd D, Brasseur L, Turner L, Becker K, Gallik KL, Bassett DS, Henderson MX. Network analysis of α-synuclein pathology progression reveals p21-activated kinases as regulators of vulnerability. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.22.619411. [PMID: 39484617 PMCID: PMC11526907 DOI: 10.1101/2024.10.22.619411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
α-Synuclein misfolding and progressive accumulation drives a pathogenic process in Parkinson's disease. To understand cellular and network vulnerability to α-synuclein pathology, we developed a framework to quantify network-level vulnerability and identify new therapeutic targets at the cellular level. Full brain α-synuclein pathology was mapped in mice over 9 months. Empirical pathology data was compared to theoretical pathology estimates from a diffusion model of pathology progression along anatomical connections. Unexplained variance in the model enabled us to derive regional vulnerability that we compared to regional gene expression. We identified gene expression patterns that relate to regional vulnerability, including 12 kinases that were enriched in vulnerable regions. Among these, an inhibitor of group II PAKs demonstrated protection from neuron death and α-synuclein pathology, even after delayed compound treatment. This study provides a framework for the derivation of cellular vulnerability from network-based studies and identifies a promising therapeutic pathway for Parkinson's disease.
Collapse
Affiliation(s)
- Naman Vatsa
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Julia K. Brynildsen
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Thomas M. Goralski
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Kevin Kurgat
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Lindsay Meyerdirk
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Libby Breton
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Daniella DeWeerd
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Laura Brasseur
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | | | | | | | - Dani S. Bassett
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
- Department of Electrical & Systems Engineering, University of Pennsylvania, Philadelphia, PA, USA
- Department of Physics & Astronomy, University of Pennsylvania, Philadelphia, PA, USA
- Department of Neurology, University of Pennsylvania, Philadelphia, PA, USA
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA, USA
- Santa Fe Institute, Santa Fe, NM, USA
- Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Michael X. Henderson
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
- Lead Contact
| |
Collapse
|
9
|
Simões JLB, de Carvalho Braga G, Eichler SW, da Silva GB, Bagatini MD. Implications of COVID-19 in Parkinson's disease: the purinergic system in a therapeutic-target perspective to diminish neurodegeneration. Purinergic Signal 2024; 20:487-507. [PMID: 38460075 PMCID: PMC11377384 DOI: 10.1007/s11302-024-09998-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 02/21/2024] [Indexed: 03/11/2024] Open
Abstract
The pathophysiology of Parkinson's disease (PD) is marked by degeneration of dopaminergic neurons in the substantia nigra. With advent of COVID-19, which is closely associated with generalized inflammation and multiple organ dysfunctions, the PD patients may develop severe conditions of disease leading to exacerbated degeneration. This condition is caused by the excessive release of pro-inflammatory markers, called cytokine storm, that is capable of triggering neurodegenerative conditions by affecting the blood-brain barrier (BBB). A possible SARS-CoV-2 infection, in serious cases, may compromise the immune system by triggering a hyperstimulation of the neuroimmune response, similar to the pathological processes found in PD. From this perspective, the inflammatory scenario triggers oxidative stress and, consequently, cellular dysfunction in the nervous tissue. The P2X7R seems to be the key mediator of the neuroinflammatory process, as it acts by increasing the concentration of ATP, allowing the influx of Ca2+ and the occurrence of mutations in the α-synuclein protein, causing activation of this receptor. Thus, modulation of the purinergic system may have therapeutic potential on the effects of PD, as well as on the damage caused by inflammation of the BBB, which may be able to mitigate the neurodegeneration caused by diseases. Considering all the processes of neuroinflammation, oxidative stress, and mitochondrial dysfunction that PD propose, we can conclude that the P2X7 antagonist acts in the prevention of viral diseases, and it also controls purinergic receptors formed by multi-target compounds directed to self-amplification circuits and, therefore, may be a viable strategy to obtain the desired disease-modifying effect. Thus, purinergic system receptor modulations have a high therapeutic potential for neurodegenerative diseases such as PD.
Collapse
Affiliation(s)
| | | | | | - Gilnei Bruno da Silva
- Multicentric Postgraduate Program in Biochemistry and Molecular Biology, State University of Santa Catarina, Lages, SC, Brazil
| | - Margarete Dulce Bagatini
- Graduate Program in Biomedical Sciences, Federal University of Fronteira Sul, Chapecó, SC, Brazil.
| |
Collapse
|
10
|
Zhang X, Wu H, Tang B, Guo J. Clinical, mechanistic, biomarker, and therapeutic advances in GBA1-associated Parkinson's disease. Transl Neurodegener 2024; 13:48. [PMID: 39267121 PMCID: PMC11391654 DOI: 10.1186/s40035-024-00437-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 08/17/2024] [Indexed: 09/14/2024] Open
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disease. The development of PD is closely linked to genetic and environmental factors, with GBA1 variants being the most common genetic risk. Mutations in the GBA1 gene lead to reduced activity of the coded enzyme, glucocerebrosidase, which mediates the development of PD by affecting lipid metabolism (especially sphingolipids), lysosomal autophagy, endoplasmic reticulum, as well as mitochondrial and other cellular functions. Clinically, PD with GBA1 mutations (GBA1-PD) is characterized by particular features regarding the progression of symptom severity. On the therapeutic side, the discovery of the relationship between GBA1 variants and PD offers an opportunity for targeted therapeutic interventions. In this review, we explore the genotypic and phenotypic correlations, etiologic mechanisms, biomarkers, and therapeutic approaches of GBA1-PD and summarize the current state of research and its challenges.
Collapse
Affiliation(s)
- Xuxiang Zhang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Heng Wu
- Department of Neurology, Multi-Omics Research Center for Brain Disorders, The First Affiliated Hospital, University of South China, Hengyang, 421001, China
- Clinical Research Center for Immune-Related Encephalopathy of Hunan Province, Hengyang, 421001, China
| | - Beisha Tang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, 410008, China
- Department of Neurology, Multi-Omics Research Center for Brain Disorders, The First Affiliated Hospital, University of South China, Hengyang, 421001, China
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, 410008, China
- Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Changsha, 410008, China
- Center for Medical Genetics and Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, 410008, China
- Engineering Research Center of Hunan Province in Cognitive Impairment Disorders, Central South University, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Jifeng Guo
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, 410008, China.
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, 410008, China.
- Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Changsha, 410008, China.
- Center for Medical Genetics and Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, 410008, China.
- Engineering Research Center of Hunan Province in Cognitive Impairment Disorders, Central South University, Changsha, 410008, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China.
| |
Collapse
|
11
|
Hertz E, Chen Y, Sidransky E. Gaucher disease provides a unique window into Parkinson disease pathogenesis. Nat Rev Neurol 2024; 20:526-540. [PMID: 39107435 DOI: 10.1038/s41582-024-00999-z] [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] [Accepted: 07/08/2024] [Indexed: 09/04/2024]
Abstract
An exciting development in the field of neurodegeneration is the association between the rare monogenic disorder Gaucher disease and the common complex disorder Parkinson disease (PD). Gaucher disease is a lysosomal storage disorder resulting from an inherited deficiency of the enzyme glucocerebrosidase, encoded by GBA1, which hydrolyses the glycosphingolipids glucosylceramide and glucosylsphingosine. The observation of parkinsonism in a rare subgroup of individuals with Gaucher disease first directed attention to the role of glucocerebrosidase deficiency in the pathogenesis of PD. PD occurs more frequently in people heterozygous for Gaucher GBA1 mutations, and 3-25% of people with Parkinson disease carry a GBA1 variant. However, only a small percentage of individuals with GBA1 variants develop parkinsonism, suggesting that the penetrance is low. Despite over a decade of intense research in this field, including clinical and radiological evaluations, genetic studies and investigations using model systems, the mechanism underlying GBA1-PD is still being pursued. Insights from this association have emphasized the role of lysosomal pathways in parkinsonism. Furthermore, different therapeutic strategies considered or developed for Gaucher disease can now inform drug development for PD.
Collapse
Affiliation(s)
- Ellen Hertz
- Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Yu Chen
- Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Ellen Sidransky
- Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
12
|
Chen Y, Xie WY, Xia D, Zhang MT, Sun YR, Duan WX, Shen Y, Wang F, Qu WM, Huang ZL, Liu CF. GBA-AAV mitigates sleep disruptions and motor deficits in mice with REM sleep behavior disorder. NPJ Parkinsons Dis 2024; 10:142. [PMID: 39095359 PMCID: PMC11297138 DOI: 10.1038/s41531-024-00756-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 07/22/2024] [Indexed: 08/04/2024] Open
Abstract
Sleep disturbances, including rapid eye movement sleep behavior disorder (RBD), excessive daytime sleepiness, and insomnia, are common non-motor manifestations of Parkinson's disease (PD). Little is known about the underlying mechanisms, partly due to the inability of current rodent models to adequately mimic the human PD sleep phenotype. Clinically, increasing studies have reported that variants of the glucocerebrosidase gene (GBA) increase the risk of PD. Here, we developed a mouse model characterized by sleep-wakefulness by injecting α-synuclein preformed fibronectin (PFF) into the sublaterodorsal tegmental nucleus (SLD) of GBA L444P mutant mice and investigated the role of the GBA L444P variant in the transition from rapid eye movement sleep behavior disorder to PD. Initially, we analyzed spectral correlates of REM and NREM sleep in GBA L444P mutant mice. Importantly, EEG power spectral analysis revealed that GBA L444P mutation mice exhibited reduced delta power during non-rapid eye movement (NREM) sleep and increased theta power (8.2-10 Hz) in active rapid eye movement (REM) sleep phases. Our study revealed that GBA L444P-mutant mice, after receiving PFF injections, exhibited increased sleep fragmentation, significant motor and cognitive dysfunctions, and loss of dopaminergic neurons in the substantia nigra. Furthermore, the over-expression of GBA-AAV partially improved these sleep disturbances and motor and cognitive impairments. In conclusion, we present the initial evidence that the GBA L444P mutant mouse serves as an essential tool in understanding the complex sleep disturbances associated with PD. This model further provides insights into potential therapeutic approaches, particularly concerning α-synuclein accumulation and its subsequent pathological consequences.
Collapse
Affiliation(s)
- Ying Chen
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, 215004, Suzhou, China
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, 215123, Suzhou, Jiangsu, China
| | - Wei-Ye Xie
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, 215004, Suzhou, China
| | - Dong Xia
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, 215123, Suzhou, Jiangsu, China
| | - Mu-Tian Zhang
- Department of Pharmacology, School of Basic Medical Sciences; State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 200032, Shanghai, China
| | - Yan-Rui Sun
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, 215004, Suzhou, China
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, 215123, Suzhou, Jiangsu, China
| | - Wen-Xiang Duan
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, 215123, Suzhou, Jiangsu, China
| | - Yun Shen
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, 215004, Suzhou, China
| | - Fen Wang
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, 215004, Suzhou, China
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, 215123, Suzhou, Jiangsu, China
| | - Wei-Min Qu
- Department of Pharmacology, School of Basic Medical Sciences; State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 200032, Shanghai, China
| | - Zhi-Li Huang
- Department of Pharmacology, School of Basic Medical Sciences; State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 200032, Shanghai, China.
| | - Chun-Feng Liu
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, 215004, Suzhou, China.
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, 215123, Suzhou, Jiangsu, China.
- Department of Neurology, Xiongan Xuanwu Hospital, 071700, Xiongan, China.
| |
Collapse
|
13
|
Dadgar-Kiani E, Bieri G, Melki R, Hossain A, Gitler AD, Lee JH. Neuromodulation modifies α-synuclein spreading dynamics in vivo and the pattern is predicted by changes in whole-brain function. Brain Stimul 2024; 17:938-946. [PMID: 39096960 PMCID: PMC11416857 DOI: 10.1016/j.brs.2024.07.021] [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: 04/16/2024] [Revised: 07/08/2024] [Accepted: 07/31/2024] [Indexed: 08/05/2024] Open
Abstract
BACKGROUND Many neurodegenerative disease treatments, such as deep brain stimulation for Parkinson's Disease, can alleviate symptoms by primarily compensating for circuit dysfunctions. However, the stimulation's effect on the underlying disease progression remains relatively unknown. Here, we report that neuromodulation can not only modulate circuit function but also modulate the in vivo spreading dynamics of α-synuclein pathology, the primary pathological hallmark observed in Parkinson's Disease. METHODS In a mouse model, pre-formed fibrils were injected into the striatum to induce widespread α-synuclein pathology. Two days after fibril injection, mice were treated for two weeks with daily optogenetic stimulation of the Secondary Motor Area, Layer V. Whole brains were then extracted, immunolabeled, cleared, and imaged with light-sheet fluorescent microscopy. RESULTS Repeated optogenetic stimulation led to a decrease in pathology at the site of stimulation and at various cortical and subcortical regions, while the contralateral cortex saw a consistent increase. Aligning the pathology changes with optogenetic-fMRI measured brain activity, we found that the changes in pathology and brain function had similar spatial locations but opposite polarity. CONCLUSION These results demonstrate the ability to modulate and predict whole brain pathology changes using neuromodulation, opening a new horizon for investigating optimized neuromodulation therapies.
Collapse
Affiliation(s)
- Ehsan Dadgar-Kiani
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, 94305, USA; Department of Bioengineering, Stanford University, Stanford, CA, 94305, USA
| | - Gregor Bieri
- Department of Genetics, Stanford University, CA, 94305, USA
| | - Ronald Melki
- Institut François Jacob, MIRCen, CEA and Laboratory of Neurodegenerative Diseases, CNRS, Fontenay-aux-Roses, France
| | - Aronee Hossain
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, 94305, USA
| | - Aaron D Gitler
- Department of Genetics, Stanford University, CA, 94305, USA; Chan Zuckerberg Biohub, San Francisco, CA, 94158, USA
| | - Jin Hyung Lee
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, 94305, USA; Department of Bioengineering, Stanford University, Stanford, CA, 94305, USA; Department of Electrical Engineering, Stanford University, CA, 94305, USA; Department of Neurosurgery, Stanford University, Stanford, CA, 94305, USA.
| |
Collapse
|
14
|
Skrahin A, Horowitz M, Istaiti M, Skrahina V, Lukas J, Yahalom G, Cohen ME, Revel-Vilk S, Goker-Alpan O, Becker-Cohen M, Hassin-Baer S, Svenningsson P, Rolfs A, Zimran A. GBA1-Associated Parkinson's Disease Is a Distinct Entity. Int J Mol Sci 2024; 25:7102. [PMID: 39000225 PMCID: PMC11241486 DOI: 10.3390/ijms25137102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 06/24/2024] [Accepted: 06/25/2024] [Indexed: 07/16/2024] Open
Abstract
GBA1-associated Parkinson's disease (GBA1-PD) is increasingly recognized as a distinct entity within the spectrum of parkinsonian disorders. This review explores the unique pathophysiological features, clinical progression, and genetic underpinnings that differentiate GBA1-PD from idiopathic Parkinson's disease (iPD). GBA1-PD typically presents with earlier onset and more rapid progression, with a poor response to standard PD medications. It is marked by pronounced cognitive impairment and a higher burden of non-motor symptoms compared to iPD. Additionally, patients with GBA1-PD often exhibit a broader distribution of Lewy bodies within the brain, accentuating neurodegenerative processes. The pathogenesis of GBA1-PD is closely associated with mutations in the GBA1 gene, which encodes the lysosomal enzyme beta-glucocerebrosidase (GCase). In this review, we discuss two mechanisms by which GBA1 mutations contribute to disease development: 'haploinsufficiency,' where a single functional gene copy fails to produce a sufficient amount of GCase, and 'gain of function,' where the mutated GCase acquires harmful properties that directly impact cellular mechanisms for alpha-synuclein degradation, leading to alpha-synuclein aggregation and neuronal cell damage. Continued research is advancing our understanding of how these mechanisms contribute to the development and progression of GBA1-PD, with the 'gain of function' mechanism appearing to be the most plausible. This review also explores the implications of GBA1 mutations for therapeutic strategies, highlighting the need for early diagnosis and targeted interventions. Currently, small molecular chaperones have shown the most promising clinical results compared to other agents. This synthesis of clinical, pathological, and molecular aspects underscores the assertion that GBA1-PD is a distinct clinical and pathobiological PD phenotype, necessitating specific management and research approaches to better understand and treat this debilitating condition.
Collapse
Affiliation(s)
- Aliaksandr Skrahin
- Rare Disease Consulting RCV GmbH, Leibnizstrasse 58, 10629 Berlin, Germany
| | - Mia Horowitz
- Shmunis School of Biomedicine and Cancer Research, Faculty of Life Sciences, Tel Aviv University, 6997801 Ramat Aviv, Israel
| | - Majdolen Istaiti
- Gaucher Unit, Shaare Zedek Medical Center, 9103102 Jerusalem, Israel
- Agyany Pharma Ltd., 9695614 Jerusalem, Israel
| | | | - Jan Lukas
- Translational Neurodegeneration Section Albrecht Kossel, Department of Neurology, University Medical Center Rostock, 18147 Rostock, Germany
- Center for Transdisciplinary Neurosciences Rostock (CTNR), University Medical Center Rostock, University of Rostock, 18147 Rostock, Germany
| | - Gilad Yahalom
- Department of Neurology and Movement Disorders Unit, Shaare Zedek Medical Center, 9103102 Jerusalem, Israel
- Faculty of Medicine, Hebrew University of Jerusalem, 9112102 Jerusalem, Israel
| | - Mikhal E. Cohen
- Department of Neurology and Movement Disorders Unit, Shaare Zedek Medical Center, 9103102 Jerusalem, Israel
- Faculty of Medicine, Hebrew University of Jerusalem, 9112102 Jerusalem, Israel
| | - Shoshana Revel-Vilk
- Gaucher Unit, Shaare Zedek Medical Center, 9103102 Jerusalem, Israel
- Faculty of Medicine, Hebrew University of Jerusalem, 9112102 Jerusalem, Israel
| | - Ozlem Goker-Alpan
- Lysosomal and Rare Disorders Research and Treatment Center, Fairfax, VA 22030, USA
| | | | - Sharon Hassin-Baer
- Movement Disorders Institute, Department of Neurology, Chaim Sheba Medical Center, 5262101 Tel-Hashomer, Israel
- Department of Neurology and Neurosurgery, Faculty of Medical and Health Sciences, Tel Aviv University, 6997801 Tel-Aviv, Israel
| | - Per Svenningsson
- Department of Clinical Neuroscience, Karolinska Institute, 17177 Stockholm, Sweden
- Department of Basal and Clinical Neuroscience, King’s College London, London SE5 9RT, UK
| | - Arndt Rolfs
- Rare Disease Consulting RCV GmbH, Leibnizstrasse 58, 10629 Berlin, Germany
- Agyany Pharma Ltd., 9695614 Jerusalem, Israel
- Medical Faculty, University of Rostock, 18055 Rostock, Germany
| | - Ari Zimran
- Gaucher Unit, Shaare Zedek Medical Center, 9103102 Jerusalem, Israel
- Agyany Pharma Ltd., 9695614 Jerusalem, Israel
- Faculty of Medicine, Hebrew University of Jerusalem, 9112102 Jerusalem, Israel
| |
Collapse
|
15
|
Huang X, Zhu Z, Du M, Wu C, Fu J, Zhang J, Tan W, Wu B, Liu L, Liao ZB. FMOD Alleviates Depression-Like Behaviors by Targeting the PI3K/AKT/mTOR Signaling After Traumatic Brain Injury. Neuromolecular Med 2024; 26:24. [PMID: 38864941 PMCID: PMC11169026 DOI: 10.1007/s12017-024-08793-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2024] [Accepted: 05/26/2024] [Indexed: 06/13/2024]
Abstract
Depression frequently occurs following traumatic brain injury (TBI). However, the role of Fibromodulin (FMOD) in TBI-related depression is not yet clear. Previous studies have suggested FMOD as a potential key factor in TBI, yet its association with depression post-TBI and underlying mechanisms are not well understood. Serum levels of FMOD were measured in patients with traumatic brain injury using qPCR. The severity of depression was assessed using the self-depression scale (SDS). Neurological function, depressive state, and cognitive function in mice were assessed using the modified Neurological Severity Score (mNSS), forced swimming test (FST), tail suspension test (TST), Sucrose Preference Test (SPT), and morris water maze (MWM). The morphological features of mouse hippocampal synapses and neuronal dendritic spines were revealed through immunofluorescence, transmission electron microscopy, and Golgi-Cox staining. The protein expression levels of FMOD, MAP2, SYP, and PSD95, as well as the phosphorylation levels of the PI3K/AKT/mTOR signaling pathway, were detected through Western blotting. FMOD levels were decreased in TBI patients' serum. Overexpression of FMOD preserved neuronal function and alleviated depression-like behaviour, increased synaptic protein expression, and induced ultrastructural changes in hippocampal neurons. The increased phosphorylation of PI3K, AKT, and mTOR suggested the involvement of the PI3K/AKT/mTOR signaling pathway in FMOD's protective effects. FMOD exhibits potential as a therapeutic target for depression related to TBI, with its protective effects potentially mediated through the PI3K/AKT/mTOR signaling pathway.
Collapse
Affiliation(s)
- Xuekang Huang
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuanjiagang, Yuzhong District, Chongqing, 400016, China
| | - Ziyu Zhu
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuanjiagang, Yuzhong District, Chongqing, 400016, China
| | - Mengran Du
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuanjiagang, Yuzhong District, Chongqing, 400016, China
| | - Chenrui Wu
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuanjiagang, Yuzhong District, Chongqing, 400016, China
| | - Jiayuanyuan Fu
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuanjiagang, Yuzhong District, Chongqing, 400016, China
| | - Jie Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuanjiagang, Yuzhong District, Chongqing, 400016, China
| | - Weilin Tan
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuanjiagang, Yuzhong District, Chongqing, 400016, China
| | - Biying Wu
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuanjiagang, Yuzhong District, Chongqing, 400016, China
| | - Lian Liu
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuanjiagang, Yuzhong District, Chongqing, 400016, China
| | - Z B Liao
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuanjiagang, Yuzhong District, Chongqing, 400016, China.
| |
Collapse
|
16
|
Walton S, Fenyi A, Tittle T, Sidransky E, Pal G, Choi S, Melki R, Killinger BA, Kordower JH. Neither alpha-synuclein fibril strain nor host murine genotype influences seeding efficacy. NPJ Parkinsons Dis 2024; 10:105. [PMID: 38773124 PMCID: PMC11109094 DOI: 10.1038/s41531-024-00679-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 03/07/2024] [Indexed: 05/23/2024] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative disease characterized by progressive motor symptoms and alpha-synuclein (αsyn) aggregation in the nervous system. For unclear reasons, PD patients with certain GBA1 mutations (GBA-PD) have a more aggressive clinical progression. Two testable hypotheses that can potentially account for this phenomenon are that GBA1 mutations promote αsyn spread or drive the generation of highly pathogenic αsyn polymorphs (i.e., strains). We tested these hypotheses by treating homozygous GBA1 D409V knockin (KI) mice with human α-syn-preformed fibrils (PFFs) and treating wild-type mice (WT) with several αsyn-PFF polymorphs amplified from brain autopsy samples collected from patients with idiopathic PD and GBA-PD patients with either homozygous or heterozygous GBA1 mutations. Robust phosphorylated-αsyn (PSER129) positive pathology was observed at the injection site (i.e., the olfactory bulb granule cell layer) and throughout the brain six months following PFF injection. The PFF seeding efficiency and degree of spread were similar regardless of the mouse genotype or PFF polymorphs. We found that PFFs amplified from the human brain, regardless of patient genotype, were generally more effective seeders than wholly synthetic PFFs (i.e., non-amplified); however, PFF concentration differed between these two studies, which might also account for the observed differences. To investigate whether the molecular composition of pathology differed between different seeding conditions, we performed Biotinylation by Antibody Recognition on PSER129 (BAR-PSER129). We found that for BAR-PSER129, the endogenous PSER129 pool dominated identified interactions, and thus, very few potential interactions were explicitly identified for seeded pathology. However, we found Dynactin Subunit 2 (Dctn2) interaction was shared across all PFF conditions, and NCK Associated Protein 1 (Nckap1) and Adaptor Related Protein Complex 3 Subunit Beta 2 (Ap3b2) were unique to PFFs amplified from GBA-PD brains of heterozygous mutation carriers. In conclusion, both the genotype and αsyn strain had little effect on overall seeding efficacy and global PSER129-interactions.
Collapse
Affiliation(s)
- Sara Walton
- ASU-Banner Neurodegenerative Disease Research Center and School of Life Sciences, Arizona State University, Tempe, AZ, USA
| | - Alexis Fenyi
- Institut Francois Jacob (MIRCen), CEA and Laboratory of Neurodegenerative Diseases, CNRS, Fontenay-Aux-Roses Cedex, France
| | - Tyler Tittle
- Graduate College, Rush University Medical Center, Chicago, IL, USA
| | - Ellen Sidransky
- Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Gian Pal
- Department of Neurology, Division of Movement Disorders, Rutgers - Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Solji Choi
- Graduate College, Rush University Medical Center, Chicago, IL, USA
| | - Ronald Melki
- Institut Francois Jacob (MIRCen), CEA and Laboratory of Neurodegenerative Diseases, CNRS, Fontenay-Aux-Roses Cedex, France
| | | | - Jeffrey H Kordower
- ASU-Banner Neurodegenerative Disease Research Center and School of Life Sciences, Arizona State University, Tempe, AZ, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| |
Collapse
|
17
|
Liu J, Huang Y, Qian T, Chen J, Ding Y, Lai Z, Zhong X, Lai M, Zhang H, Wang Y, Wang H, Peng Y. Exploring the neuroprotective role of artesunate in mouse models of anti-NMDAR encephalitis: insights from molecular mechanisms and transmission electron microscopy. Cell Commun Signal 2024; 22:269. [PMID: 38745240 PMCID: PMC11094908 DOI: 10.1186/s12964-024-01652-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 05/06/2024] [Indexed: 05/16/2024] Open
Abstract
BACKGROUND The pathway involving PTEN-induced putative kinase 1 (PINK1) and PARKIN plays a crucial role in mitophagy, a process activated by artesunate (ART). We propose that patients with anti-N-methyl-D-aspartate receptor (NMDAR) encephalitis exhibit insufficient mitophagy, and ART enhances mitophagy via the PINK1/PARKIN pathway, thereby providing neuroprotection. METHODS Adult female mice aged 8-10 weeks were selected to create a passive transfer model of anti-NMDAR encephalitis. We conducted behavioral tests on these mice within a set timeframe. Techniques such as immunohistochemistry, immunofluorescence, and western blotting were employed to assess markers including PINK1, PARKIN, LC3B, p62, caspase3, and cleaved caspase3. The TUNEL assay was utilized to detect neuronal apoptosis, while transmission electron microscopy (TEM) was used to examine mitochondrial autophagosomes. Primary hippocampal neurons were cultured, treated, and then analyzed through immunofluorescence for mtDNA, mtROS, TMRM. RESULTS In comparison to the control group, mitophagy levels in the experimental group were not significantly altered, yet there was a notable increase in apoptotic neurons. Furthermore, markers indicative of mitochondrial leakage and damage were found to be elevated in the experimental group compared to the control group, but these markers showed improvement following ART treatment. ART was effective in activating the PINK1/PARKIN pathway, enhancing mitophagy, and diminishing neuronal apoptosis. Behavioral assessments revealed that ART ameliorated symptoms in mice with anti-NMDAR encephalitis in the passive transfer model (PTM). The knockdown of PINK1 led to a reduction in mitophagy levels, and subsequent ART intervention did not alleviate symptoms in the anti-NMDAR encephalitis PTM mice, indicating that ART's therapeutic efficacy is mediated through the activation of the PINK1/PARKIN pathway. CONCLUSIONS At the onset of anti-NMDAR encephalitis, mitochondrial damage is observed; however, this damage is mitigated by the activation of mitophagy via the PINK1/PARKIN pathway. This regulatory feedback mechanism facilitates the removal of damaged mitochondria, prevents neuronal apoptosis, and consequently safeguards neural tissue. ART activates the PINK1/PARKIN pathway to enhance mitophagy, thereby exerting neuroprotective effects and may achieve therapeutic goals in treating anti-NMDAR encephalitis.
Collapse
Affiliation(s)
- Jingsi Liu
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Yingyi Huang
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
- Department of Neurology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, 510641, China
| | - Tinglin Qian
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Jinyu Chen
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Yuewen Ding
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Zhaohui Lai
- Department of Neurology, Ganzhou People's Hospital, Ganzhou, 341000, China
| | - Xinghua Zhong
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Mingjun Lai
- Department of Neurology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, 510641, China
| | - Huili Zhang
- Department of Neurology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, 510641, China
| | - Yuanyuan Wang
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| | - Honghao Wang
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| | - Yu Peng
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
18
|
Hull A, Atilano ML, Gergi L, Kinghorn KJ. Lysosomal storage, impaired autophagy and innate immunity in Gaucher and Parkinson's diseases: insights for drug discovery. Philos Trans R Soc Lond B Biol Sci 2024; 379:20220381. [PMID: 38368939 PMCID: PMC10874704 DOI: 10.1098/rstb.2022.0381] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 11/08/2023] [Indexed: 02/20/2024] Open
Abstract
Impairment of autophagic-lysosomal pathways is increasingly being implicated in Parkinson's disease (PD). GBA1 mutations cause the lysosomal storage disorder Gaucher disease (GD) and are the commonest known genetic risk factor for PD. GBA1 mutations have been shown to cause autophagic-lysosomal impairment. Defective autophagic degradation of unwanted cellular constituents is associated with several pathologies, including loss of normal protein homeostasis, particularly of α-synuclein, and innate immune dysfunction. The latter is observed both peripherally and centrally in PD and GD. Here, we will discuss the mechanistic links between autophagy and immune dysregulation, and the possible role of these pathologies in communication between the gut and brain in these disorders. Recent work in a fly model of neuronopathic GD (nGD) revealed intestinal autophagic defects leading to gastrointestinal dysfunction and immune activation. Rapamycin treatment partially reversed the autophagic block and reduced immune activity, in association with increased survival and improved locomotor performance. Alterations in the gut microbiome are a critical driver of neuroinflammation, and studies have revealed that eradication of the microbiome in nGD fly and mouse models of PD ameliorate brain inflammation. Following these observations, lysosomal-autophagic pathways, innate immune signalling and microbiome dysbiosis are discussed as potential therapeutic targets in PD and GD. This article is part of a discussion meeting issue 'Understanding the endo-lysosomal network in neurodegeneration'.
Collapse
Affiliation(s)
- Alexander Hull
- Department of Genetics, Evolution & Environment, Institute of Healthy Ageing, Darwin Building, Gower Street, London WC1E 6BT, UK
| | - Magda L Atilano
- Department of Genetics, Evolution & Environment, Institute of Healthy Ageing, Darwin Building, Gower Street, London WC1E 6BT, UK
| | - Laith Gergi
- Department of Genetics, Evolution & Environment, Institute of Healthy Ageing, Darwin Building, Gower Street, London WC1E 6BT, UK
| | - Kerri J Kinghorn
- Department of Genetics, Evolution & Environment, Institute of Healthy Ageing, Darwin Building, Gower Street, London WC1E 6BT, UK
| |
Collapse
|
19
|
Liang H, Ma Z, Zhong W, Liu J, Sugimoto K, Chen H. Regulation of mitophagy and mitochondrial function: Natural compounds as potential therapeutic strategies for Parkinson's disease. Phytother Res 2024; 38:1838-1862. [PMID: 38356178 DOI: 10.1002/ptr.8156] [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: 09/20/2023] [Revised: 01/27/2024] [Accepted: 01/29/2024] [Indexed: 02/16/2024]
Abstract
Mitochondrial damage is associated with the development of Parkinson's disease (PD), indicating that mitochondrial-targeted treatments could hold promise as disease-modifying approaches for PD. Notably, natural compounds have demonstrated the ability to modulate mitochondrial-related processes. In this review article, we discussed the possible neuroprotective mechanisms of natural compounds against PD in modulating mitophagy and mitochondrial function. A comprehensive literature search on natural compounds related to the treatment of PD by regulating mitophagy and mitochondrial function was conducted from PubMed, Web of Science and Chinese National Knowledge Infrastructure databases from their inception until April 2023. We summarize recent advancements in mitophagy's molecular mechanisms, including upstream and downstream processes, and its relationship with PD-related genes or proteins. Importantly, we highlight how natural compounds can therapeutically regulate various mitochondrial processes through multiple targets and pathways to alleviate oxidative stress, neuroinflammation, Lewy's body aggregation and apoptosis, which are key contributors to PD pathogenesis. Unlike the single-target strategy of modern medicine, natural compounds provide neuroprotection against PD by modulating various mitochondrial-related processes, including ameliorating mitophagy by targeting the PINK1/parkin pathway, the NIX/BNIP3 pathway, and autophagosome formation (i.e., LC3 and p62). Given the prevalence of mitochondrial damage in various neurodegenerative diseases, exploring the exact mechanism of natural compounds on mitophagy and mitochondrial dysfunction could shed light on the development of highly effective disease-modifying or adjuvant therapies targeting PD and other neurodegenerative disorders.
Collapse
Affiliation(s)
- Hao Liang
- Department of Acupuncture, Heilongjiang Academy of Traditional Chinese Medicine, Harbin, China
| | - Zhenwang Ma
- Department of Acupuncture, Heilongjiang Academy of Traditional Chinese Medicine, Harbin, China
| | - Wei Zhong
- Department of Rheumatology and Immunology, Affiliated Qiqihar Hospital, Southern Medical University, Qiqihar, China
| | - Jia Liu
- Department of Neurology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
- Institute for Brain Disorders, Beijing University of Chinese Medicine, Beijing, China
| | - Kazuo Sugimoto
- Department of Neurology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
- Institute for Brain Disorders, Beijing University of Chinese Medicine, Beijing, China
| | - Hong Chen
- Department of Acupuncture, Heilongjiang Academy of Traditional Chinese Medicine, Harbin, China
- Department of TCM Geriatric, Southern Medical University, Guangzhou, China
| |
Collapse
|
20
|
Kulesskaya N, Bhattacharjee A, Holmström KM, Vuorio P, Henriques A, Callizot N, Huttunen HJ. HER-096 is a CDNF-derived brain-penetrating peptidomimetic that protects dopaminergic neurons in a mouse synucleinopathy model of Parkinson's disease. Cell Chem Biol 2024; 31:593-606.e9. [PMID: 38039968 DOI: 10.1016/j.chembiol.2023.11.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 09/28/2023] [Accepted: 11/07/2023] [Indexed: 12/03/2023]
Abstract
Cerebral dopamine neurotrophic factor (CDNF) is an unconventional neurotropic factor that modulates unfolded protein response (UPR) pathway signaling and alleviates endoplasmic reticulum (ER) stress providing cytoprotective effects in different models of neurodegenerative disorders. Here, we developed a brain-penetrating peptidomimetic compound based on human CDNF. This compound called HER-096 shows similar potency and mechanism of action as CDNF, and promotes dopamine neuron survival, reduces α-synuclein aggregation and modulates UPR signaling in in vitro models. HER-096 is metabolically stable and able to penetrate to cerebrospinal (CSF) and brain interstitial fluids (ISF) after subcutaneous administration, with an extended CSF and brain ISF half-life compared to plasma. Subcutaneously administered HER-096 modulated UPR pathway activity, protected dopamine neurons, and reduced α-synuclein aggregates and neuroinflammation in substantia nigra of aged mice with synucleinopathy. Peptidomimetic HER-096 is a candidate for development of a disease-modifying therapy for Parkinson's disease with a patient-friendly route of administration.
Collapse
|
21
|
Zhang Z, Wang R, Zhou H, Wu D, Cao Y, Zhang C, Sun H, Mu C, Hao Z, Ren H, Wang N, Yu S, Zhang J, Tao M, Wang C, Liu Y, Liu L, Liu Y, Zang J, Wang G. Inhibition of EHMT1/2 rescues synaptic damage and motor impairment in a PD mouse model. Cell Mol Life Sci 2024; 81:128. [PMID: 38472451 DOI: 10.1007/s00018-024-05176-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 01/16/2024] [Accepted: 02/14/2024] [Indexed: 03/14/2024]
Abstract
Epigenetic dysregulation that leads to alterations in gene expression and is suggested to be one of the key pathophysiological factors of Parkinson's disease (PD). Here, we found that α-synuclein preformed fibrils (PFFs) induced histone H3 dimethylation at lysine 9 (H3K9me2) and increased the euchromatic histone methyltransferases EHMT1 and EHMT2, which were accompanied by neuronal synaptic damage, including loss of synapses and diminished expression levels of synaptic-related proteins. Furthermore, the levels of H3K9me2 at promoters in genes that encode the synaptic-related proteins SNAP25, PSD95, Synapsin 1 and vGLUT1 were increased in primary neurons after PFF treatment, which suggests a linkage between H3K9 dimethylation and synaptic dysfunction. Inhibition of EHMT1/2 with the specific inhibitor A-366 or shRNA suppressed histone methylation and alleviated synaptic damage in primary neurons that were treated with PFFs. In addition, the synaptic damage and motor impairment in mice that were injected with PFFs were repressed by treatment with the EHMT1/2 inhibitor A-366. Thus, our findings reveal the role of histone H3 modification by EHMT1/2 in synaptic damage and motor impairment in a PFF animal model, suggesting the involvement of epigenetic dysregulation in PD pathogenesis.
Collapse
Affiliation(s)
- Zhixiong Zhang
- Laboratory of Molecular Neuropathology, Department of Pharmacology, Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, Jiangsu, China
| | - Rui Wang
- Laboratory of Molecular Neuropathology, Department of Pharmacology, Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, Jiangsu, China
| | - Hui Zhou
- Laboratory of Molecular Neuropathology, Department of Pharmacology, Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, Jiangsu, China
| | - Dan Wu
- Laboratory of Molecular Neuropathology, Department of Pharmacology, Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, Jiangsu, China
| | - Yifan Cao
- Laboratory of Molecular Neuropathology, Department of Pharmacology, Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, Jiangsu, China
| | - Chuang Zhang
- Laboratory of Molecular Neuropathology, Department of Pharmacology, Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, Jiangsu, China
| | - Hongyang Sun
- Laboratory of Molecular Neuropathology, Department of Pharmacology, Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, Jiangsu, China
| | - Chenchen Mu
- Laboratory of Molecular Neuropathology, Department of Pharmacology, Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, Jiangsu, China
| | - Zongbing Hao
- Laboratory of Molecular Neuropathology, Department of Pharmacology, Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, Jiangsu, China
| | - Haigang Ren
- Laboratory of Molecular Neuropathology, Department of Pharmacology, Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, Jiangsu, China
- Jiangsu Provincial Medical Innovation Center of Trauma Medicine, Institute of Trauma Medicine, Soochow University, Suzhou, 215123, Jiangsu, China
- MOE Key Laboratory of Geriatric Diseases and Immunology, Soochow University, Suzhou, 215123, Jiangsu, China
| | - Nana Wang
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, 215163, China
| | - Shuang Yu
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, 215163, China
| | - Jingzhong Zhang
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, 215163, China
| | - Mengdan Tao
- School of Pharmacy, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, 211166, China
| | - Can Wang
- School of Pharmacy, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, 211166, China
| | - Yan Liu
- School of Pharmacy, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, 211166, China
| | - Liu Liu
- Department of Pharmacy, The First Affiliated Hospital of Hunan Normal University, Hunan Provincial People's Hospital, Changsha, 410005, China
| | - Yanli Liu
- College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, Jiangsu, China
| | - Jianye Zang
- Hefei National Laboratory for Physical Sciences at Microscale CAS Center for Excellence in Biomacromolecules, Collaborative Innovation Center of Chemistry for Life Sciences, and School of Life Sciences, University of Science and Technology of China, 96 Jinzhai Road, Hefei, 230026, Anhui, China
| | - Guanghui Wang
- Laboratory of Molecular Neuropathology, Department of Pharmacology, Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, Jiangsu, China.
- MOE Key Laboratory of Geriatric Diseases and Immunology, Soochow University, Suzhou, 215123, Jiangsu, China.
- Center of Translational Medicine, First People's Hospital of Taicang, Taicang Affiliated Hospital of Soochow University, Suzhou, 215400, China.
| |
Collapse
|
22
|
Lubben N, Brynildsen JK, Webb CM, Li HL, Leyns CEG, Changolkar L, Zhang B, Meymand ES, O'Reilly M, Madaj Z, DeWeerd D, Fell MJ, Lee VMY, Bassett DS, Henderson MX. LRRK2 kinase inhibition reverses G2019S mutation-dependent effects on tau pathology progression. Transl Neurodegener 2024; 13:13. [PMID: 38438877 PMCID: PMC10910783 DOI: 10.1186/s40035-024-00403-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 02/07/2024] [Indexed: 03/06/2024] Open
Abstract
BACKGROUND Mutations in leucine-rich repeat kinase 2 (LRRK2) are the most common cause of familial Parkinson's disease (PD). These mutations elevate the LRRK2 kinase activity, making LRRK2 kinase inhibitors an attractive therapeutic. LRRK2 kinase activity has been consistently linked to specific cell signaling pathways, mostly related to organelle trafficking and homeostasis, but its relationship to PD pathogenesis has been more difficult to define. LRRK2-PD patients consistently present with loss of dopaminergic neurons in the substantia nigra but show variable development of Lewy body or tau tangle pathology. Animal models carrying LRRK2 mutations do not develop robust PD-related phenotypes spontaneously, hampering the assessment of the efficacy of LRRK2 inhibitors against disease processes. We hypothesized that mutations in LRRK2 may not be directly related to a single disease pathway, but instead may elevate the susceptibility to multiple disease processes, depending on the disease trigger. To test this hypothesis, we have previously evaluated progression of α-synuclein and tau pathologies following injection of proteopathic seeds. We demonstrated that transgenic mice overexpressing mutant LRRK2 show alterations in the brain-wide progression of pathology, especially at older ages. METHODS Here, we assess tau pathology progression in relation to long-term LRRK2 kinase inhibition. Wild-type or LRRK2G2019S knock-in mice were injected with tau fibrils and treated with control diet or diet containing LRRK2 kinase inhibitor MLi-2 targeting the IC50 or IC90 of LRRK2 for 3-6 months. Mice were evaluated for tau pathology by brain-wide quantitative pathology in 844 brain regions and subsequent linear diffusion modeling of progression. RESULTS Consistent with our previous work, we found systemic alterations in the progression of tau pathology in LRRK2G2019S mice, which were most pronounced at 6 months. Importantly, LRRK2 kinase inhibition reversed these effects in LRRK2G2019S mice, but had minimal effect in wild-type mice, suggesting that LRRK2 kinase inhibition is likely to reverse specific disease processes in G2019S mutation carriers. Additional work may be necessary to determine the potential effect in non-carriers. CONCLUSIONS This work supports a protective role of LRRK2 kinase inhibition in G2019S carriers and provides a rational workflow for systematic evaluation of brain-wide phenotypes in therapeutic development.
Collapse
Affiliation(s)
- Noah Lubben
- Department of Neurodegenerative Science, Van Andel Institute, 333 Bostwick Ave NE, Grand Rapids, MI, 49503, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
| | - Julia K Brynildsen
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Connor M Webb
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, Institute On Aging and Center for Neurodegenerative Disease Research, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Howard L Li
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, Institute On Aging and Center for Neurodegenerative Disease Research, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Cheryl E G Leyns
- Neuroscience Discovery, Merck & Co., Inc., Boston, MA, 02115, USA
| | - Lakshmi Changolkar
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, Institute On Aging and Center for Neurodegenerative Disease Research, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Bin Zhang
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, Institute On Aging and Center for Neurodegenerative Disease Research, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Emily S Meymand
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, Institute On Aging and Center for Neurodegenerative Disease Research, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Mia O'Reilly
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Zach Madaj
- Bioinformatics and Biostatistics Core, Van Andel Institute, 333 Bostwick Ave., NE, Grand Rapids, MI, 49503, USA
| | - Daniella DeWeerd
- Department of Neurodegenerative Science, Van Andel Institute, 333 Bostwick Ave NE, Grand Rapids, MI, 49503, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
| | - Matthew J Fell
- Neuroscience Discovery, Merck & Co., Inc., Boston, MA, 02115, USA
| | - Virginia M Y Lee
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, Institute On Aging and Center for Neurodegenerative Disease Research, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Dani S Bassett
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Department of Electrical and Systems Engineering, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Department of Physics and Astronomy, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Department of Neurology, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Santa Fe Institute, Santa Fe, NM, 87501, USA
| | - Michael X Henderson
- Department of Neurodegenerative Science, Van Andel Institute, 333 Bostwick Ave NE, Grand Rapids, MI, 49503, USA.
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA.
| |
Collapse
|
23
|
Mercado G, Kaeufer C, Richter F, Peelaerts W. Infections in the Etiology of Parkinson's Disease and Synucleinopathies: A Renewed Perspective, Mechanistic Insights, and Therapeutic Implications. JOURNAL OF PARKINSON'S DISEASE 2024; 14:1301-1329. [PMID: 39331109 PMCID: PMC11492057 DOI: 10.3233/jpd-240195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 09/06/2024] [Indexed: 09/28/2024]
Abstract
Increasing evidence suggests a potential role for infectious pathogens in the etiology of synucleinopathies, a group of age-related neurodegenerative disorders including Parkinson's disease (PD), multiple system atrophy and dementia with Lewy bodies. In this review, we discuss the link between infections and synucleinopathies from a historical perspective, present emerging evidence that supports this link, and address current research challenges with a focus on neuroinflammation. Infectious pathogens can elicit a neuroinflammatory response and modulate genetic risk in PD and related synucleinopathies. The mechanisms of how infections might be linked with synucleinopathies as well as the overlap between the immune cellular pathways affected by virulent pathogens and disease-related genetic risk factors are discussed. Here, an important role for α-synuclein in the immune response against infections is emerging. Critical methodological and knowledge gaps are addressed, and we provide new future perspectives on how to address these gaps. Understanding how infections and neuroinflammation influence synucleinopathies will be essential for the development of early diagnostic tools and novel therapies.
Collapse
Affiliation(s)
- Gabriela Mercado
- Division of Neurobiology, Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Christopher Kaeufer
- Center for Systems Neuroscience, Hannover, Germany
- Department of Pharmacology, Toxicology and Pharmacy, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Franziska Richter
- Department of Pharmacology, Toxicology and Pharmacy, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Wouter Peelaerts
- Laboratory for Virology and Gene Therapy, Department of Pharmacy and Pharmaceutical Sciences, KU Leuven, Leuven, Belgium
| |
Collapse
|
24
|
Xie H, Yang Y, Sun Q, Li ZY, Ni MH, Chen ZH, Li SN, Dai P, Cui YY, Cao XY, Jiang N, Du LJ, Yu Y, Yan LF, Cui GB. Abnormalities of cerebral blood flow and the regional brain function in Parkinson's disease: a systematic review and multimodal neuroimaging meta-analysis. Front Neurol 2023; 14:1289934. [PMID: 38162449 PMCID: PMC10755479 DOI: 10.3389/fneur.2023.1289934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 11/15/2023] [Indexed: 01/03/2024] Open
Abstract
Background Parkinson's disease (PD) is a neurodegenerative disease with high incidence rate. Resting state functional magnetic resonance imaging (rs-fMRI), as a widely used method for studying neurodegenerative diseases, has not yet been combined with two important indicators, amplitude low-frequency fluctuation (ALFF) and cerebral blood flow (CBF), for standardized analysis of PD. Methods In this study, we used seed-based d-mapping and permutation of subject images (SDM-PSI) software to investigate the changes in ALFF and CBF of PD patients. After obtaining the regions of PD with changes in ALFF or CBF, we conducted a multimodal analysis to identify brain regions where ALFF and CBF changed together or could not synchronize. Results The final study included 31 eligible trials with 37 data sets. The main analysis results showed that the ALFF of the left striatum and left anterior thalamic projection decreased in PD patients, while the CBF of the right superior frontal gyrus decreased. However, the results of multimodal analysis suggested that there were no statistically significant brain regions. In addition, the decrease of ALFF in the left striatum and the decrease of CBF in the right superior frontal gyrus was correlated with the decrease in clinical cognitive scores. Conclusion PD patients had a series of spontaneous brain activity abnormalities, mainly involving brain regions related to the striatum-thalamic-cortex circuit, and related to the clinical manifestations of PD. Among them, the left striatum and right superior frontal gyrus are more closely related to cognition. Systematic review registration https://www.crd.york.ac.uk/ PROSPERO (CRD42023390914).
Collapse
Affiliation(s)
- Hao Xie
- Department of Radiology and Functional and Molecular Imaging Key Lab of Shaanxi Province, Tangdu Hospital, Fourth Military Medical University (Air Force Medical University), Xi’an, Shaanxi, China
| | - Yang Yang
- Department of Radiology and Functional and Molecular Imaging Key Lab of Shaanxi Province, Tangdu Hospital, Fourth Military Medical University (Air Force Medical University), Xi’an, Shaanxi, China
| | - Qian Sun
- Department of Radiology and Functional and Molecular Imaging Key Lab of Shaanxi Province, Tangdu Hospital, Fourth Military Medical University (Air Force Medical University), Xi’an, Shaanxi, China
| | - Ze-Yang Li
- Department of Radiology and Functional and Molecular Imaging Key Lab of Shaanxi Province, Tangdu Hospital, Fourth Military Medical University (Air Force Medical University), Xi’an, Shaanxi, China
| | - Min-Hua Ni
- Department of Radiology and Functional and Molecular Imaging Key Lab of Shaanxi Province, Tangdu Hospital, Fourth Military Medical University (Air Force Medical University), Xi’an, Shaanxi, China
| | - Zhu-Hong Chen
- Department of Radiology and Functional and Molecular Imaging Key Lab of Shaanxi Province, Tangdu Hospital, Fourth Military Medical University (Air Force Medical University), Xi’an, Shaanxi, China
| | - Si-Ning Li
- Department of Radiology and Functional and Molecular Imaging Key Lab of Shaanxi Province, Tangdu Hospital, Fourth Military Medical University (Air Force Medical University), Xi’an, Shaanxi, China
- Faculty of Medical Technology, Xi’an Medical University, Xi’an, Shaanxi, China
| | - Pan Dai
- Department of Radiology and Functional and Molecular Imaging Key Lab of Shaanxi Province, Tangdu Hospital, Fourth Military Medical University (Air Force Medical University), Xi’an, Shaanxi, China
- Faculty of Medical Technology, Xi’an Medical University, Xi’an, Shaanxi, China
| | - Yan-Yan Cui
- Department of Radiology and Functional and Molecular Imaging Key Lab of Shaanxi Province, Tangdu Hospital, Fourth Military Medical University (Air Force Medical University), Xi’an, Shaanxi, China
- Faculty of Medical Technology, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, China
| | - Xin-Yu Cao
- Department of Radiology and Functional and Molecular Imaging Key Lab of Shaanxi Province, Tangdu Hospital, Fourth Military Medical University (Air Force Medical University), Xi’an, Shaanxi, China
- Faculty of Medical Technology, Medical School of Yan’an University, Yan’an, Shaanxi, China
| | - Nan Jiang
- Department of Radiology and Functional and Molecular Imaging Key Lab of Shaanxi Province, Tangdu Hospital, Fourth Military Medical University (Air Force Medical University), Xi’an, Shaanxi, China
| | - Li-Juan Du
- Department of Radiology and Functional and Molecular Imaging Key Lab of Shaanxi Province, Tangdu Hospital, Fourth Military Medical University (Air Force Medical University), Xi’an, Shaanxi, China
| | - Ying Yu
- Department of Radiology and Functional and Molecular Imaging Key Lab of Shaanxi Province, Tangdu Hospital, Fourth Military Medical University (Air Force Medical University), Xi’an, Shaanxi, China
| | - Lin-Feng Yan
- Department of Radiology and Functional and Molecular Imaging Key Lab of Shaanxi Province, Tangdu Hospital, Fourth Military Medical University (Air Force Medical University), Xi’an, Shaanxi, China
| | - Guang-Bin Cui
- Department of Radiology and Functional and Molecular Imaging Key Lab of Shaanxi Province, Tangdu Hospital, Fourth Military Medical University (Air Force Medical University), Xi’an, Shaanxi, China
| |
Collapse
|
25
|
Bigi A, Cascella R, Cecchi C. α-Synuclein oligomers and fibrils: partners in crime in synucleinopathies. Neural Regen Res 2023; 18:2332-2342. [PMID: 37282450 PMCID: PMC10360081 DOI: 10.4103/1673-5374.371345] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023] Open
Abstract
The misfolding and aggregation of α-synuclein is the general hallmark of a group of devastating neurodegenerative pathologies referred to as synucleinopathies, such as Parkinson's disease, dementia with Lewy bodies, and multiple system atrophy. In such conditions, a range of different misfolded aggregates, including oligomers, protofibrils, and fibrils, are present both in neurons and glial cells. Growing experimental evidence supports the proposition that soluble oligomeric assemblies, formed during the early phases of the aggregation process, are the major culprits of neuronal toxicity; at the same time, fibrillar conformers appear to be the most efficient at propagating among interconnected neurons, thus contributing to the spreading of α-synuclein pathology. Moreover, α-synuclein fibrils have been recently reported to release soluble and highly toxic oligomeric species, responsible for an immediate dysfunction in the recipient neurons. In this review, we discuss the current knowledge about the plethora of mechanisms of cellular dysfunction caused by α-synuclein oligomers and fibrils, both contributing to neurodegeneration in synucleinopathies.
Collapse
Affiliation(s)
- Alessandra Bigi
- Department of Experimental and Clinical Biomedical Sciences, Section of Biochemistry, University of Florence, Florence, Italy
| | - Roberta Cascella
- Department of Experimental and Clinical Biomedical Sciences, Section of Biochemistry, University of Florence, Florence, Italy
| | - Cristina Cecchi
- Department of Experimental and Clinical Biomedical Sciences, Section of Biochemistry, University of Florence, Florence, Italy
| |
Collapse
|
26
|
Labrador-Garrido A, Zhong S, Hughes L, Keshiya S, Kim WS, Halliday GM, Dzamko N. Live cell in situ lysosomal GCase activity correlates to alpha-synuclein levels in human differentiated neurons with LRRK2 and GBA1 mutations. Front Cell Neurosci 2023; 17:1229213. [PMID: 37908374 PMCID: PMC10613732 DOI: 10.3389/fncel.2023.1229213] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 09/20/2023] [Indexed: 11/02/2023] Open
Abstract
Introduction Heterozygous mutations in GBA1, which encodes the lysosomal hydrolase glucocerebrosidase (GCase), are a common risk factor for the neurodegenerative movement disorder Parkinson's disease (PD). Consequently, therapeutic options targeting the GCase enzyme are in development. An important aspect of this development is determining the effect of potential modifying compounds on GCase activity, which can be complicated by the different methods and substrate probes that are commonly employed for this purpose. Methods In this study, we employed the GCase substrate probe 5-(pentafluorobenzoylamino)fluorescein di-D-glucopyranoside (PFB-FDGlu) in combination with live cell imaging to measure GCase activity in situ in the lysosome. Results The live cell assay was validated using the GCase inhibitor conduritol-B-epoxide and with GBA1 knockout neural cells and was then used to assess GCase activity in iPSC differentiated into neural stem cells and neurons that were obtained from idiopathic PD patients and PD patients with the LRRK2 G2019S and GBA N370S mutations, as well as controls (n = 4 per group). Heterogeneity in GCase activity was observed across all groups. However, a significant inverse correlation between GCase activity and levels of alpha-synuclein protein was observed. Discussion The live cell imaging assay for GCase activity could be useful for further understanding the role of GCase in PD and screening potential modifying compounds in differentiated human cell models.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Nicolas Dzamko
- School of Medical Sciences, Faculty of Medicine and Health and the Charles Perkins Centre, University of Sydney, Camperdown, NSW, Australia
| |
Collapse
|
27
|
Keefe AJ, Gabrych DR, Zhu Y, Vocadlo DJ, Silverman MA. Axonal Transport of Lysosomes Is Unaffected in Glucocerebrosidase-Inhibited iPSC-Derived Forebrain Neurons. eNeuro 2023; 10:ENEURO.0079-23.2023. [PMID: 37816595 PMCID: PMC10576257 DOI: 10.1523/eneuro.0079-23.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 09/18/2023] [Accepted: 09/20/2023] [Indexed: 10/12/2023] Open
Abstract
Lysosomes are acidic organelles that traffic throughout neurons delivering catabolic enzymes to distal regions of the cell and maintaining degradative demands. Loss of function mutations in the gene GBA encoding the lysosomal enzyme glucocerebrosidase (GCase) cause the lysosomal storage disorder Gaucher's disease (GD) and are the most common genetic risk factor for synucleinopathies like Parkinson's disease (PD) and dementia with Lewy bodies (DLB). GCase degrades the membrane lipid glucosylceramide (GlcCer) and mutations in GBA, or inhibiting its activity, results in the accumulation of GlcCer and disturbs the composition of the lysosomal membrane. The lysosomal membrane serves as the platform to which intracellular trafficking complexes are recruited and activated. Here, we investigated whether lysosomal trafficking in axons was altered by inhibition of GCase with the pharmacological agent Conduritol B Epoxide (CBE). Using live cell imaging in human male induced pluripotent human stem cell (iPSC)-derived forebrain neurons, we demonstrated that lysosomal transport was similar in both control and CBE-treated neurons. Furthermore, we tested whether lysosomal rupture, a process implicated in various neurodegenerative disorders, was affected by inhibition of GCase. Using L-leucyl-L-leucine methyl ester (LLoME) to induce lysosomal membrane damage and immunocytochemical staining for markers of lysosomal rupture, we found no difference in susceptibility to rupture between control and CBE-treated neurons. These results suggest the loss of GCase activity does not contribute to neurodegenerative disease by disrupting either lysosomal transport or rupture.
Collapse
Affiliation(s)
- A J Keefe
- Department of Biological Sciences, Simon Fraser University, Burnaby, British Columbia V5A 1S6, Canada
| | - D R Gabrych
- Department of Biological Sciences, Simon Fraser University, Burnaby, British Columbia V5A 1S6, Canada
| | - Y Zhu
- Department of Chemistry, Simon Fraser University, Burnaby, British Columbia V5A 1S6, Canada
| | - D J Vocadlo
- Centre for Cell Biology, Development, and Disease, Simon Fraser University, Burnaby, British Columbia V5A 1S6, Canada
- Department of Chemistry, Simon Fraser University, Burnaby, British Columbia V5A 1S6, Canada
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia V5A 1S6, Canada
| | - M A Silverman
- Department of Biological Sciences, Simon Fraser University, Burnaby, British Columbia V5A 1S6, Canada
- Centre for Cell Biology, Development, and Disease, Simon Fraser University, Burnaby, British Columbia V5A 1S6, Canada
| |
Collapse
|
28
|
Brynildsen JK, Rajan K, Henderson MX, Bassett DS. Network models to enhance the translational impact of cross-species studies. Nat Rev Neurosci 2023; 24:575-588. [PMID: 37524935 PMCID: PMC10634203 DOI: 10.1038/s41583-023-00720-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/17/2023] [Indexed: 08/02/2023]
Abstract
Neuroscience studies are often carried out in animal models for the purpose of understanding specific aspects of the human condition. However, the translation of findings across species remains a substantial challenge. Network science approaches can enhance the translational impact of cross-species studies by providing a means of mapping small-scale cellular processes identified in animal model studies to larger-scale inter-regional circuits observed in humans. In this Review, we highlight the contributions of network science approaches to the development of cross-species translational research in neuroscience. We lay the foundation for our discussion by exploring the objectives of cross-species translational models. We then discuss how the development of new tools that enable the acquisition of whole-brain data in animal models with cellular resolution provides unprecedented opportunity for cross-species applications of network science approaches for understanding large-scale brain networks. We describe how these tools may support the translation of findings across species and imaging modalities and highlight future opportunities. Our overarching goal is to illustrate how the application of network science tools across human and animal model studies could deepen insight into the neurobiology that underlies phenomena observed with non-invasive neuroimaging methods and could simultaneously further our ability to translate findings across species.
Collapse
Affiliation(s)
- Julia K Brynildsen
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Kanaka Rajan
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Michael X Henderson
- Parkinson's Disease Center, Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA
| | - Dani S Bassett
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA.
- Department of Electrical & Systems Engineering, University of Pennsylvania, Philadelphia, PA, USA.
- Department of Physics & Astronomy, University of Pennsylvania, Philadelphia, PA, USA.
- Department of Neurology, University of Pennsylvania, Philadelphia, PA, USA.
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA, USA.
- Santa Fe Institute, Santa Fe, NM, USA.
| |
Collapse
|
29
|
Dues DJ, Nguyen APT, Becker K, Ma J, Moore DJ. Hippocampal subfield vulnerability to α-synuclein pathology precedes neurodegeneration and cognitive dysfunction. NPJ Parkinsons Dis 2023; 9:125. [PMID: 37640722 PMCID: PMC10462636 DOI: 10.1038/s41531-023-00574-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Accepted: 08/22/2023] [Indexed: 08/31/2023] Open
Abstract
Cognitive dysfunction is a salient feature of Parkinson's disease (PD) and Dementia with Lewy bodies (DLB). The onset of dementia reflects the spread of Lewy pathology throughout forebrain structures. The mere presence of Lewy pathology, however, provides limited indication of cognitive status. Thus, it remains unclear whether Lewy pathology is the de facto substrate driving cognitive dysfunction in PD and DLB. Through application of α-synuclein fibrils in vivo, we sought to examine the influence of pathologic inclusions on cognition. Following stereotactic injection of α-synuclein fibrils within the mouse forebrain, we measured the burden of α-synuclein pathology at 1-, 3-, and 6-months post-injection within subregions of the hippocampus and cortex. Under this paradigm, the hippocampal CA2/3 subfield was especially susceptible to α-synuclein pathology. Strikingly, we observed a drastic reduction of pathology in the CA2/3 subfield across time-points, consistent with the consolidation of α-synuclein pathology into dense somatic inclusions followed by neurodegeneration. Silver-positive degenerating neurites were observed prior to neuronal loss, suggesting that this might be an early feature of fibril-induced neurotoxicity and a precursor to neurodegeneration. Critically, mice injected with α-synuclein fibrils developed progressive deficits in spatial learning and memory. These findings support that the formation of α-synuclein inclusions in the mouse forebrain precipitate neurodegenerative changes that recapitulate features of Lewy-related cognitive dysfunction.
Collapse
Affiliation(s)
- Dylan J Dues
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA
| | - An Phu Tran Nguyen
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA
| | - Katelyn Becker
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA
| | - Jiyan Ma
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA
- Chinese Institute for Brain Research, Beijing, China
| | - Darren J Moore
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA.
| |
Collapse
|
30
|
Oftedal L, Lange J, Pedersen KF, Erga AH, Dalen I, Tysnes OB, Alves G, Maple-Grødem J. Early GCase activity is a predictor of long-term cognitive decline in Parkinson's disease. Transl Neurodegener 2023; 12:41. [PMID: 37635244 PMCID: PMC10463992 DOI: 10.1186/s40035-023-00373-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: 03/09/2023] [Accepted: 07/27/2023] [Indexed: 08/29/2023] Open
Affiliation(s)
- Linn Oftedal
- Centre for Movement Disorders, Centre for Brain Health, Stavanger University Hospital, 4011, Stavanger, Norway
| | - Johannes Lange
- Centre for Movement Disorders, Centre for Brain Health, Stavanger University Hospital, 4011, Stavanger, Norway
- Department of Chemistry, Bioscience and Environmental Engineering, University of Stavanger, 4021, Stavanger, Norway
| | - Kenn Freddy Pedersen
- Centre for Movement Disorders, Centre for Brain Health, Stavanger University Hospital, 4011, Stavanger, Norway
- Department of Neurology, Stavanger University Hospital, 4011, Stavanger, Norway
| | - Aleksander Hagen Erga
- Centre for Movement Disorders, Centre for Brain Health, Stavanger University Hospital, 4011, Stavanger, Norway
- Department of Social Studies, University of Stavanger, 4021, Stavanger, Norway
| | - Ingvild Dalen
- Section of Biostatistics, Department of Research, Stavanger University Hospital, 4011, Stavanger, Norway
| | - Ole-Bjørn Tysnes
- Department of Clinical Medicine, University of Bergen, 5021, Bergen, Norway
- Department of Neurology, Haukeland University Hospital, 5053, Bergen, Norway
| | - Guido Alves
- Centre for Movement Disorders, Centre for Brain Health, Stavanger University Hospital, 4011, Stavanger, Norway
- Department of Chemistry, Bioscience and Environmental Engineering, University of Stavanger, 4021, Stavanger, Norway
- Department of Neurology, Stavanger University Hospital, 4011, Stavanger, Norway
| | - Jodi Maple-Grødem
- Centre for Movement Disorders, Centre for Brain Health, Stavanger University Hospital, 4011, Stavanger, Norway.
- Department of Chemistry, Bioscience and Environmental Engineering, University of Stavanger, 4021, Stavanger, Norway.
| |
Collapse
|
31
|
Walton S, Fenyi A, Tittle T, Sidransky E, Pal G, Choi S, Melki R, Killinger BA, Kordower JH. Neither alpha-synuclein-preformed fibrils derived from patients with GBA1 mutations nor the host murine genotype significantly influence seeding efficacy in the mouse olfactory bulb. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.24.554646. [PMID: 37662402 PMCID: PMC10473741 DOI: 10.1101/2023.08.24.554646] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
Parkinson's disease (PD) is a neurodegenerative disease characterized by progressive motor symptoms and alpha-synuclein (αsyn) aggregation in the nervous system. For unclear reasons, PD patients with certain GBA mutations (GBA-PD) have a more aggressive clinical progression. Two testable hypotheses that can potentially account for this phenomenon are that GBA1 mutations promote αsyn spread or drive the generation of highly pathogenic αsyn polymorphs (i.e., strains). We tested these hypotheses by treating homozygous GBA1 D409V knockin (KI) mice with human α-syn-preformed fibrils (PFFs) and treating wild-type mice (WT) with several αsyn-PFF polymorphs amplified from brain autopsy samples collected from patients with idiopathic PD and GBA-PD patients with either homozygous or heterozygous GBA1 mutations. Robust phosphorylated-αsyn (PSER129) positive pathology was observed at the injection site (i.e., the olfactory bulb granular layer) and throughout the brain six months following PFF injection. The PFF seeding efficiency and degree of spread were similar regardless of the mouse genotype or PFF polymorphs. We found that PFFs amplified from the human brain, regardless of patient genotype, were generally more effective seeders than wholly synthetic PFFs (i.e., non-amplified); however, PFF concentration differed between these two studies, and this might also account for the observed differences. To investigate whether the molecular composition of pathology differed between different seeding conditions, we permed Biotinylation by Antibody Recognition on PSER129 (BAR-PSER129). We found that for BAR-PSER129, the endogenous PSER129 pool dominated identified interactions, and thus, very few potential interactions were explicitly identified for seeded pathology. However, we found Dctn2 interaction was shared across all PFF conditions, and Nckap1 and Ap3b2 were unique to PFFs amplified from GBA-PD brains of heterozygous mutation carriers. In conclusion, both the genotype and αsyn strain had little effect on overall seeding efficacy and global PSER129-interactions.
Collapse
Affiliation(s)
- Sara Walton
- ASU-Banner Neurodegenerative Disease Research Center and School of Life Sciences, Arizona State University, Tempe, AZ, USA
| | - Alexis Fenyi
- Institut Francois Jacob (MIRCen), CEA and Laboratory of Neurodegenerative Diseases, CNRS, Fontenay-Aux-Roses Cedex, France
| | - Tyler Tittle
- Graduate College, Rush University Medical Center, Chicago, Illinois 60612
| | - Ellen Sidransky
- Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Gian Pal
- Department of Neurology, Division of Movement Disorders, Rutgers - Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Solji Choi
- Graduate College, Rush University Medical Center, Chicago, Illinois 60612
| | - Ronald Melki
- Institut Francois Jacob (MIRCen), CEA and Laboratory of Neurodegenerative Diseases, CNRS, Fontenay-Aux-Roses Cedex, France
| | - Bryan A Killinger
- Graduate College, Rush University Medical Center, Chicago, Illinois 60612
| | - Jeffrey H Kordower
- ASU-Banner Neurodegenerative Disease Research Center and School of Life Sciences, Arizona State University, Tempe, AZ, USA
| |
Collapse
|
32
|
Mi X, Chen L, Xie J, Song N. Linking Genetic Risks to Pathological α-Synuclein Transmission in Parkinson's Disease. Neurosci Bull 2023; 39:1186-1188. [PMID: 36737591 PMCID: PMC10313604 DOI: 10.1007/s12264-023-01031-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Accepted: 11/19/2022] [Indexed: 02/05/2023] Open
Affiliation(s)
- Xiaoqing Mi
- Department of Physiology, School of Basic Medicine, Institute of Brain Science and Disease, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, 266071, China
| | - Lei Chen
- Department of Physiology, School of Basic Medicine, Institute of Brain Science and Disease, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, 266071, China
| | - Junxia Xie
- Department of Physiology, School of Basic Medicine, Institute of Brain Science and Disease, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, 266071, China.
| | - Ning Song
- Department of Physiology, School of Basic Medicine, Institute of Brain Science and Disease, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, 266071, China.
| |
Collapse
|
33
|
Iemolo A, De Risi M, Giordano N, Torromino G, Somma C, Cavezza D, Colucci M, Mancini M, de Iure A, Granata R, Picconi B, Calabresi P, De Leonibus E. Synaptic mechanisms underlying onset and progression of memory deficits caused by hippocampal and midbrain synucleinopathy. NPJ Parkinsons Dis 2023; 9:92. [PMID: 37328503 DOI: 10.1038/s41531-023-00520-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 05/15/2023] [Indexed: 06/18/2023] Open
Abstract
Cognitive deficits, including working memory, and visuospatial deficits are common and debilitating in Parkinson's disease. α-synucleinopathy in the hippocampus and cortex is considered as the major risk factor. However, little is known about the progression and specific synaptic mechanisms underlying the memory deficits induced by α-synucleinopathy. Here, we tested the hypothesis that pathologic α-Synuclein (α-Syn), initiated in different brain regions, leads to distinct onset and progression of the pathology. We report that overexpression of human α-Syn in the murine mesencephalon leads to late onset memory impairment and sensorimotor deficits accompanied by reduced dopamine D1 expression in the hippocampus. In contrast, human α-Syn overexpression in the hippocampus leads to early memory impairment, altered synaptic transmission and plasticity, and decreased expression of GluA1 AMPA-type glutamate receptors. These findings identify the synaptic mechanisms leading to memory impairment induced by hippocampal α-synucleinopathy and provide functional evidence of the major neuronal networks involved in disease progression.
Collapse
Affiliation(s)
- Attilio Iemolo
- Telethon Institute of Genetics and Medicine, Via dei Campi Flegrei 34, Pozzuoli, Naples, Italy
- Institute of Genetics and Biophysics (IGB), Consiglio Nazionale delle Ricerche (CNR), via Pietro Castellino 111, Naples, Italy
| | - Maria De Risi
- Telethon Institute of Genetics and Medicine, Via dei Campi Flegrei 34, Pozzuoli, Naples, Italy
- Institute of Biochemistry and Cell Biology, Consiglio Nazionale delle Ricerche (CNR), Via Ramarini 33, Monterotondo Scalo, Rome, Italy
| | - Nadia Giordano
- Telethon Institute of Genetics and Medicine, Via dei Campi Flegrei 34, Pozzuoli, Naples, Italy
| | - Giulia Torromino
- Institute of Biochemistry and Cell Biology, Consiglio Nazionale delle Ricerche (CNR), Via Ramarini 33, Monterotondo Scalo, Rome, Italy
- University of Naples Federico II, Department of Humanistic Studies, Naples, Italy
| | - Cristina Somma
- Telethon Institute of Genetics and Medicine, Via dei Campi Flegrei 34, Pozzuoli, Naples, Italy
| | - Diletta Cavezza
- Institute of Biochemistry and Cell Biology, Consiglio Nazionale delle Ricerche (CNR), Via Ramarini 33, Monterotondo Scalo, Rome, Italy
| | - Martina Colucci
- Telethon Institute of Genetics and Medicine, Via dei Campi Flegrei 34, Pozzuoli, Naples, Italy
- Institute of Genetics and Biophysics (IGB), Consiglio Nazionale delle Ricerche (CNR), via Pietro Castellino 111, Naples, Italy
| | - Maria Mancini
- Institute of Neuroscience (IN), Consiglio Nazionale delle Ricerche (CNR), via Raoul Follereau 3, Vedano al Lambro, Monza e Brianza, Italy
| | - Antonio de Iure
- Lab. Experimental Neurophysiology, IRCCS San Raffaele, Rome, 00166, Italy
| | - Rocco Granata
- Institute of Biochemistry and Cell Biology, Consiglio Nazionale delle Ricerche (CNR), Via Ramarini 33, Monterotondo Scalo, Rome, Italy
| | - Barbara Picconi
- Lab. Experimental Neurophysiology, IRCCS San Raffaele, Rome, 00166, Italy
- Telematic University San Raffaele, Rome, 00166, Italy
| | - Paolo Calabresi
- Neurological Clinic, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168, Rome, Italy
- Neurology, Department of Neuroscience, Faculty of Medicine, Università Cattolica del "Sacro Cuore", 00168, Rome, Italy
| | - Elvira De Leonibus
- Telethon Institute of Genetics and Medicine, Via dei Campi Flegrei 34, Pozzuoli, Naples, Italy.
- Institute of Genetics and Biophysics (IGB), Consiglio Nazionale delle Ricerche (CNR), via Pietro Castellino 111, Naples, Italy.
- Institute of Biochemistry and Cell Biology, Consiglio Nazionale delle Ricerche (CNR), Via Ramarini 33, Monterotondo Scalo, Rome, Italy.
| |
Collapse
|
34
|
Chatterjee D, Krainc D. Mechanisms of Glucocerebrosidase Dysfunction in Parkinson's Disease. J Mol Biol 2023; 435:168023. [PMID: 36828270 PMCID: PMC10247409 DOI: 10.1016/j.jmb.2023.168023] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 02/15/2023] [Accepted: 02/17/2023] [Indexed: 02/24/2023]
Abstract
Beta-glucocerebrosidase is a lysosomal hydrolase, encoded by GBA1 that represents the most common risk gene associated with Parkinson's disease (PD) and Lewy Body Dementia. Glucocerebrosidase dysfunction has been also observed in the absence of GBA1 mutations across different genetic and sporadic forms of PD and related disorders, suggesting a broader role of glucocerebrosidase in neurodegeneration. In this review, we highlight recent advances in mechanistic characterization of glucocerebrosidase function as the foundation for development of novel therapeutics targeting glucocerebrosidase in PD and related disorders.
Collapse
Affiliation(s)
- Diptaman Chatterjee
- Ken and Ruth Davee Department of Neurology, Northwestern University, Feinberg School of Medicine, Chicago, IL, USA. https://twitter.com/NeilChatterBox
| | - Dimitri Krainc
- Ken and Ruth Davee Department of Neurology, Northwestern University, Feinberg School of Medicine, Chicago, IL, USA; Simpson Querrey Center for Neurogenetics, Northwestern University, Feinberg School of Medicine, Chicago, IL, USA.
| |
Collapse
|
35
|
Leyns CEG, Prigent A, Beezhold B, Yao L, Hatcher NG, Tao P, Kang J, Suh E, Van Deerlin VM, Trojanowski JQ, Lee VMY, Kennedy ME, Fell MJ, Henderson MX. Glucocerebrosidase activity and lipid levels are related to protein pathologies in Parkinson's disease. NPJ Parkinsons Dis 2023; 9:74. [PMID: 37169750 PMCID: PMC10175254 DOI: 10.1038/s41531-023-00517-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Accepted: 04/28/2023] [Indexed: 05/13/2023] Open
Abstract
Parkinson's disease (PD) and dementia with Lewy bodies (DLB) are progressive neurodegenerative diseases characterized by the accumulation of misfolded α-synuclein in the form of Lewy pathology. While most cases are sporadic, there are rare genetic mutations that cause disease and more common variants that increase incidence of disease. The most prominent genetic mutations for PD and DLB are in the GBA1 and LRRK2 genes. GBA1 mutations are associated with decreased glucocerebrosidase activity and lysosomal accumulation of its lipid substrates, glucosylceramide and glucosylsphingosine. Previous studies have shown a link between this enzyme and lipids even in sporadic PD. However, it is unclear how the protein pathologies of disease are related to enzyme activity and glycosphingolipid levels. To address this gap in knowledge, we examined quantitative protein pathology, glucocerebrosidase activity and lipid substrates in parallel from 4 regions of 91 brains with no neurological disease, idiopathic, GBA1-linked, or LRRK2-linked PD and DLB. We find that several biomarkers are altered with respect to mutation and progression to dementia. We found mild association of glucocerebrosidase activity with disease, but a strong association of glucosylsphingosine with α-synuclein pathology, irrespective of genetic mutation. This association suggests that Lewy pathology precipitates changes in lipid levels related to progression to dementia.
Collapse
Affiliation(s)
- Cheryl E G Leyns
- Merck & Co., Inc., 33 Avenue Louis Pasteur, Boston, MA, 02115, USA
| | - Alice Prigent
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, 49503, USA
| | - Brenna Beezhold
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, 49503, USA
| | - Lihang Yao
- Merck & Co., Inc., 770 Sumneytown Pk, West Point, PA, 19486, USA
| | - Nathan G Hatcher
- Merck & Co., Inc., 770 Sumneytown Pk, West Point, PA, 19486, USA
| | - Peining Tao
- Merck & Co., Inc., 33 Avenue Louis Pasteur, Boston, MA, 02115, USA
| | - John Kang
- Merck & Co., Inc., 33 Avenue Louis Pasteur, Boston, MA, 02115, USA
| | - EunRan Suh
- Institute on Aging and Center for Neurodegenerative Disease Research, Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Vivianna M Van Deerlin
- Institute on Aging and Center for Neurodegenerative Disease Research, Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - John Q Trojanowski
- Institute on Aging and Center for Neurodegenerative Disease Research, Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Virginia M Y Lee
- Institute on Aging and Center for Neurodegenerative Disease Research, Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | | | - Matthew J Fell
- Merck & Co., Inc., 33 Avenue Louis Pasteur, Boston, MA, 02115, USA
| | - Michael X Henderson
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, 49503, USA.
| |
Collapse
|
36
|
Dues DJ, Nguyen APT, Becker K, Ma J, Moore DJ. Hippocampal subfield vulnerability to α-synuclein pathology precedes neurodegeneration and cognitive dysfunction. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.12.536572. [PMID: 37090590 PMCID: PMC10120695 DOI: 10.1101/2023.04.12.536572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/25/2023]
Abstract
Cognitive dysfunction is a salient feature of Parkinson's disease (PD) and Dementia with Lewy bodies (DLB). The onset of dementia reflects the spread of Lewy pathology throughout forebrain structures. The mere presence of Lewy pathology, however, provides limited indication of cognitive status. Thus, it remains unclear whether Lewy pathology is the de facto substrate driving cognitive dysfunction in PD and DLB. Through application of α-synuclein fibrils in vivo , we sought to examine the influence of pathologic inclusions on cognition. Following stereotactic injection of α-synuclein fibrils within the mouse forebrain, we measured the burden of α-synuclein pathology at 1-, 3-, and 6-months post-injection within subregions of the hippocampus and cortex. Under this paradigm, the hippocampal CA2/3 subfield was especially susceptible to α- synuclein pathology. Strikingly, we observed a drastic reduction of pathology in the CA2/3 subfield across time-points, consistent with the consolidation of α-synuclein pathology into dense somatic inclusions followed by neurodegeneration. Silver-positive degenerating neurites were observed prior to neuronal loss, suggesting that this might be an early feature of fibril-induced neurotoxicity and a precursor to neurodegeneration. Critically, mice injected with α-synuclein fibrils developed progressive deficits in spatial learning and memory. These findings support that the formation of α-synuclein inclusions in the mouse forebrain precipitate neurodegenerative changes that recapitulate features of Lewy-related cognitive dysfunction. Highlights Mice injected with α-synuclein fibrils develop hippocampal and cortical α- synuclein pathology with a dynamic regional burden at 1-, 3-, and 6-months post-injection.Silver-positive neuronal processes are an early and enduring degenerative feature of the fibril model, while extensive neurodegeneration of the hippocampal CA2/3 subfield is detected at 6-months post-injection.Mice exhibit progressive hippocampal-dependent spatial learning and memory deficits.Forebrain injection of α-synuclein fibrils may be used to model aspects of Lewy-related cognitive dysfunction.
Collapse
Affiliation(s)
- Dylan J. Dues
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, Michigan, USA
| | - An Phu Tran Nguyen
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, Michigan, USA
| | - Katelyn Becker
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, Michigan, USA
| | - Jiyan Ma
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, Michigan, USA
| | - Darren J. Moore
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, Michigan, USA
| |
Collapse
|
37
|
Skiteva O, Yao N, Mantas I, Zhang X, Perlmann T, Svenningsson P, Chergui K. Aberrant somatic calcium channel function in cNurr1 and LRRK2-G2019S mice. NPJ Parkinsons Dis 2023; 9:56. [PMID: 37029193 PMCID: PMC10082048 DOI: 10.1038/s41531-023-00500-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 03/23/2023] [Indexed: 04/09/2023] Open
Abstract
In Parkinson's disease (PD), axons of dopaminergic (DA) neurons in the substantia nigra pars compacta (SNc) degenerate before their cell bodies. Calcium influx during pacemaker firing might contribute to neuronal loss, but it is not known if dysfunctions of voltage-gated calcium channels (VGCCs) occur in DA neurons somata and axon terminals. We investigated T-type and L-type VGCCs in SNc-DA neurons of two mouse models of PD: mice with a deletion of the Nurr1 gene in DA neurons from an adult age (cNurr1 mice), and mice bearing the G2019S mutation in the gene coding for LRRK2 (G2019S mice). Adult cNurr1 mice displayed motor and DA deficits, while middle-aged G2019S mice did not. The number and morphology of SNc-DA neurons, most of their intrinsic membrane properties and pacemaker firing were unaltered in cNurr1 and G2019S mice compared to their control and wild-type littermates. L-type VGCCs contributed to the pacemaker firing of SNc-DA neurons in G2019S mice, but not in control, wild-type, and cNurr1 mice. In cNurr1 mice, but not G2019S mice, the contribution of T-type VGCCs to the pacemaker firing of SNc-DA neurons was reduced, and somatic dopamine-D2 autoreceptors desensitized more. Altered contribution of L-type and T-type VGCCs to the pacemaker firing was not observed in the presence of a LRRK2 kinase inhibitor in G2019S mice, and in the presence of a flavonoid with antioxidant activity in G2019S and cNurr1 mice. The role of L-type and T-type VGCCs in controlling dopamine release from axon terminals in the striatum was unaltered in cNurr1 and G2019S mice. Our findings uncover opposite changes, linked to oxidative stress, in the function of two VGCCs in DA neurons somata, but not axon terminals, in two different experimental PD models.
Collapse
Affiliation(s)
- Olga Skiteva
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Ning Yao
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Ioannis Mantas
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Xiaoqun Zhang
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Thomas Perlmann
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Per Svenningsson
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Karima Chergui
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
38
|
Simons E, Fleming SM. Role of rodent models in advancing precision medicine for Parkinson's disease. HANDBOOK OF CLINICAL NEUROLOGY 2023; 193:3-16. [PMID: 36803818 DOI: 10.1016/b978-0-323-85555-6.00002-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/18/2023]
Abstract
With a current lack of disease-modifying treatments, an initiative toward implementing a precision medicine approach for treating Parkinson's disease (PD) has emerged. However, challenges remain in how to define and apply precision medicine in PD. To accomplish the goal of optimally targeted and timed treatment for each patient, preclinical research in a diverse population of rodent models will continue to be an essential part of the translational path to identify novel biomarkers for patient diagnosis and subgrouping, understand PD disease mechanisms, identify new therapeutic targets, and screen therapeutics prior to clinical testing. This review highlights the most common rodent models of PD and discusses how these models can contribute to defining and implementing precision medicine for the treatment of PD.
Collapse
Affiliation(s)
- Emily Simons
- Department of Pharmaceutical Sciences, Northeast Ohio Medical University, Rootstown, OH, United States
| | - Sheila M Fleming
- Department of Pharmaceutical Sciences, Northeast Ohio Medical University, Rootstown, OH, United States.
| |
Collapse
|
39
|
Abstract
Parkinson's disease (PD) is clinically, pathologically, and genetically heterogeneous, resisting distillation to a single, cohesive disorder. Instead, each affected individual develops a virtually unique form of Parkinson's syndrome. Clinical manifestations consist of variable motor and nonmotor features, and myriad overlaps are recognized with other neurodegenerative conditions. Although most commonly characterized by alpha-synuclein protein pathology throughout the central and peripheral nervous systems, the distribution varies and other pathologies commonly modify PD or trigger similar manifestations. Nearly all PD is genetically influenced. More than 100 genes or genetic loci have been identified, and most cases likely arise from interactions among many common and rare genetic variants. Despite its complex architecture, insights from experimental genetic dissection coalesce to reveal unifying biological themes, including synaptic, lysosomal, mitochondrial, andimmune-mediated mechanisms of pathogenesis. This emerging understanding of Parkinson's syndrome, coupled with advances in biomarkers and targeted therapies, presages successful precision medicine strategies.
Collapse
Affiliation(s)
- Hui Ye
- Department of Neurology, Baylor College of Medicine, Houston, Texas, USA; ,
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, Texas, USA
| | - Laurie A Robak
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, Texas, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA;
| | - Meigen Yu
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, Texas, USA
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas, USA;
| | - Matthew Cykowski
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Houston, Texas, USA;
- Department of Neurology, Houston Methodist Hospital, Houston, Texas, USA
| | - Joshua M Shulman
- Department of Neurology, Baylor College of Medicine, Houston, Texas, USA; ,
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, Texas, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA;
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas, USA;
- Center for Alzheimer's and Neurodegenerative Diseases, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
40
|
GBA1 Gene Mutations in α-Synucleinopathies-Molecular Mechanisms Underlying Pathology and Their Clinical Significance. Int J Mol Sci 2023; 24:ijms24032044. [PMID: 36768367 PMCID: PMC9917178 DOI: 10.3390/ijms24032044] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 01/17/2023] [Accepted: 01/18/2023] [Indexed: 01/22/2023] Open
Abstract
α-Synucleinopathies comprise a group of neurodegenerative diseases characterized by altered accumulation of a protein called α-synuclein inside neurons and glial cells. This aggregation leads to the formation of intraneuronal inclusions, Lewy bodies, that constitute the hallmark of α-synuclein pathology. The most prevalent α-synucleinopathies are Parkinson's disease (PD), dementia with Lewy bodies (DLB), and multiple system atrophy (MSA). To date, only symptomatic treatment is available for these disorders, hence new approaches to their therapy are needed. It has been observed that GBA1 mutations are one of the most impactful risk factors for developing α-synucleinopathies such as PD and DLB. Mutations in the GBA1 gene, which encodes a lysosomal hydrolase β-glucocerebrosidase (GCase), cause a reduction in GCase activity and impaired α-synuclein metabolism. The most abundant GBA1 gene mutations are N370S or N409S, L444P/L483P and E326K/E365K. The mechanisms by which GCase impacts α-synuclein aggregation are poorly understood and need to be further investigated. Here, we discuss some of the potential interactions between α-synuclein and GCase and show how GBA1 mutations may impact the course of the most prevalent α-synucleinopathies.
Collapse
|
41
|
Mahoney-Crane CL, Viswanathan M, Russell D, Curtiss RAC, Freire J, Bobba SS, Coyle SD, Kandebo M, Yao L, Wan BL, Hatcher NG, Smith SM, Marcus JN, Volpicelli-Daley LA. Neuronopathic GBA1L444P Mutation Accelerates Glucosylsphingosine Levels and Formation of Hippocampal Alpha-Synuclein Inclusions. J Neurosci 2023; 43:501-521. [PMID: 36639889 PMCID: PMC9864632 DOI: 10.1523/jneurosci.0680-22.2022] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 09/09/2022] [Accepted: 11/10/2022] [Indexed: 12/12/2022] Open
Abstract
The most common genetic risk factor for Parkinson's disease (PD) is heterozygous mutations GBA1, which encodes for the lysosomal enzyme, glucocerebrosidase. Reduced glucocerebrosidase activity associates with an accumulation of abnormal α-synuclein (α-syn) called Lewy pathology, which characterizes PD. PD patients heterozygous for the neuronotypic GBA1L444P mutation (GBA1+/L444P) have a 5.6-fold increased risk of cognitive impairments. In this study, we used GBA1+/L444P mice of either sex to determine its effects on lipid metabolism, expression of synaptic proteins, behavior, and α-syn inclusion formation. At 3 months of age, GBA1+/L444P mice demonstrated impaired contextual fear conditioning, and increased motor activity. Hippocampal levels of vGLUT1 were selectively reduced in GBA1+/L444P mice. We show, using mass spectrometry, that GBA1L444P expression increased levels of glucosylsphingosine, but not glucosylceramide, in the brains and serum of GBA1+/L444P mice. Templated induction of α-syn pathology in mice showed an increase in α-syn inclusion formation in the hippocampus of GBA1+/L444P mice compared with GBA1+/+ mice, but not in the cortex, or substantia nigra pars compacta. Pathologic α-syn reduced SNc dopamine neurons by 50% in both GBA1+/+ and GBA1+/L444P mice. Treatment with a GlcCer synthase inhibitor did not affect abundance of α-syn inclusions in the hippocampus or rescue dopamine neuron loss. Overall, these data suggest the importance of evaluating the contribution of elevated glucosylsphingosine to PD phenotypes. Further, our data suggest that expression of neuronotypic GBA1L444P may cause defects in the hippocampus, which may be a mechanism by which cognitive decline is more prevalent in individuals with GBA1-PD.SIGNIFICANCE STATEMENT Parkinson's disease (PD) and dementia with Lewy bodies (DLB) are both pathologically characterized by abnormal α-synuclein (α-syn). Mutant GBA1 is a risk factor for both PD and DLB. Our data show the expression of neuronotypic GBA1L444P impairs behaviors related to hippocampal function, reduces expression of a hippocampal excitatory synaptic protein, and that the hippocampus is more susceptible to α-syn inclusion formation. Further, our data strengthen support for the importance of evaluating the contribution of glucosylsphingosine to PD phenotypes. These outcomes suggest potential mechanisms by which GBA1L444P contributes to the cognitive symptoms clinically observed in PD and DLB. Our findings also highlight the importance of glucosylsphingosine as a relevant biomarker for future therapeutics.
Collapse
Affiliation(s)
- Casey L Mahoney-Crane
- Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, Alabama 35294
| | - Megha Viswanathan
- Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, Alabama 35294
| | - Dreson Russell
- Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, Alabama 35294
| | - Rachel A C Curtiss
- Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, Alabama 35294
| | - Jennifer Freire
- Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, Alabama 35294
| | - Sai Sumedha Bobba
- Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, Alabama 35294
| | - Sean D Coyle
- Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, Alabama 35294
| | - Monika Kandebo
- Neuroscience Discovery, Merck & Company, Inc, West Point, Pennsylvania 19486
| | - Lihang Yao
- Neuroscience Discovery, Merck & Company, Inc, West Point, Pennsylvania 19486
| | - Bang-Lin Wan
- Neuroscience Discovery, Merck & Company, Inc, West Point, Pennsylvania 19486
| | - Nathan G Hatcher
- Neuroscience Discovery, Merck & Company, Inc, West Point, Pennsylvania 19486
| | - Sean M Smith
- Neuroscience Discovery, Merck & Company, Inc, West Point, Pennsylvania 19486
| | - Jacob N Marcus
- Neuroscience Discovery, Merck & Company, Inc, West Point, Pennsylvania 19486
| | - Laura A Volpicelli-Daley
- Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, Alabama 35294
| |
Collapse
|
42
|
Han Y, He Z. Concomitant protein pathogenesis in Parkinson's disease and perspective mechanisms. Front Aging Neurosci 2023; 15:1189809. [PMID: 37181621 PMCID: PMC10174460 DOI: 10.3389/fnagi.2023.1189809] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 04/11/2023] [Indexed: 05/16/2023] Open
Abstract
Comorbidity is a common phenotype in Parkinson's disease (PD). Patients with PD not only have motor deficit symptoms, but also have heterogeneous non-motor symptoms, including cognitive impairment and emotional changes, which are the featured symptoms observed in patients with Alzheimer's disease (AD), frontotemporal dementia (FTD) and cerebrovascular disease. Moreover, autopsy studies have also confirmed the concomitant protein pathogenesis, such as the co-existences of α-synuclein, amyloid-β and tau pathologies in PD and AD patients' brains. Here, we briefly summarize the recent reports regarding the comorbidity issues in PD from both clinical observations and neuropathological evidences. Furthermore, we provide some discussion about the perspective potential mechanisms underlying such comorbidity phenomenon, with a focus on PD and related neurodegenerative diseases.
Collapse
Affiliation(s)
- Yuliang Han
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
- University of the Chinese Academy of Sciences, Beijing, China
| | - Zhuohao He
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
- University of the Chinese Academy of Sciences, Beijing, China
- Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
- *Correspondence: Zhuohao He,
| |
Collapse
|
43
|
Peelaerts W, Baekelandt V. ⍺-Synuclein Structural Diversity and the Cellular Environment in ⍺-Synuclein Transmission Models and Humans. Neurotherapeutics 2023; 20:67-82. [PMID: 37052776 PMCID: PMC10119367 DOI: 10.1007/s13311-023-01365-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/04/2023] [Indexed: 04/14/2023] Open
Abstract
Parkinson's disease (PD), dementia with Lewy bodies (DLB), and multiple system atrophy (MSA) are termed synucleinopathies, disorders that are characterized by the intracellular aggregation of the protein ɑ-synuclein. The cellular tropism of synuclein pathology in these syndromes is notably distinct since in the Lewy disorders, PD and DLB, ɑSyn forms aggregates in neurons whereas in MSA ɑSyn forms aggregates in oligodendrocytes. Studies examining ɑSyn pathology in experimental models and in human brain have now identified fibrillar ɑSyn with unique but distinct molecular signatures, suggesting that the structure of these ɑSyn fibrils might be closely tied to their cellular ontogeny. In contrast to the native structural heterogeneity of ɑSyn in vitro, the conformational landscape of fibrillar ɑSyn in human brain and in vivo transmission models appears to be remarkably uniform. Here, we review the studies by which we propose a hypothesis that the cellular host environment might be in part responsible for how ɑSyn filaments assemble into phenotype-specific strains. We postulate that the maturation of ɑSyn strains develops as a function of their in vivo transmission routes and cell-specific risk factors. The impact of the cellular environment on the structural diversity of ɑSyn might have important implications for the design of preclinical studies and their use for the development of ɑSyn-based biomarkers and therapeutic strategies. By combining phenotype-specific fibrils and relevant synucleinopathy transmission models, preclinical models might more closely reflect unique disease phenotypes.
Collapse
Affiliation(s)
- Wouter Peelaerts
- Laboratory for Neurobiology and Gene Therapy, Department of Neurosciences, Leuven Brain Institute, KU Leuven, Leuven, Belgium
- Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
| | - Veerle Baekelandt
- Laboratory for Neurobiology and Gene Therapy, Department of Neurosciences, Leuven Brain Institute, KU Leuven, Leuven, Belgium.
| |
Collapse
|
44
|
Yao N, Skiteva O, Chergui K. Age- and Sex-Dependent Behavioral and Neurochemical Alterations in hLRRK2-G2019S BAC Mice. Biomolecules 2022; 13:biom13010051. [PMID: 36671436 PMCID: PMC9856037 DOI: 10.3390/biom13010051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 12/22/2022] [Accepted: 12/23/2022] [Indexed: 12/29/2022] Open
Abstract
The G2019S mutation in the leucine-rich repeat kinase 2 (LRRK2) gene is associated with late-onset Parkinson's disease (PD). Although PD affects men and women differently, longitudinal studies examining sex- and age-dependent alterations in mice carrying the G2019S mutation are limited. We examined if behavioral and neurochemical dysfunctions, as well as neurodegeneration, occur in male and female BAC LRRK2-hG2019S (G2019S) mice, compared to their age-matched wild type littermates, at four age ranges. In the open field test, hyperlocomotion was observed in 10-12 month old male and 2-4.5 months old female G2019S mice. In the pole test, motor coordination was impaired in male G2019S mice from 15 months of age and in 20-21 months old female G2019S mice. In the striatum of G2019S male and female mice, the amounts of tyrosine hydroxylase (TH), measured with Western blotting, were unaltered. However, we found a decreased expression of the dopamine transporter in 20-21 month old male G2019S mice. The number of TH-positive neurons in the substantia nigra compacta was unaltered in 20-21 month old male and female G2019S mice. These results identify sex- and age-dependent differences in the occurrence of motor and neurochemical deficits in BAC LRRK2-hG2019S mice, and no degeneration of DA neurons.
Collapse
|
45
|
Dadgar-Kiani E, Bieri G, Melki R, Gitler AD, Lee JH. Mesoscale connections and gene expression empower whole-brain modeling of α-synuclein spread, aggregation, and decay dynamics. Cell Rep 2022; 41:111631. [PMID: 36351406 PMCID: PMC10840492 DOI: 10.1016/j.celrep.2022.111631] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 09/19/2022] [Accepted: 10/18/2022] [Indexed: 11/11/2022] Open
Abstract
An emerging view regarding neurodegenerative diseases is that discreet seeding of misfolded proteins leads to widespread pathology. However, the mechanisms by which misfolded proteins seed distinct brain regions and cause differential whole-brain pathology remain elusive. We used whole-brain tissue clearing and high-resolution imaging to longitudinally map pathology in an α-synuclein pre-formed fibril injection model of Parkinson's disease. Cleared brains at different time points of disease progression were quantitatively segmented and registered to a standardized atlas, revealing distinct phases of spreading and decline. We then fit a computational model with parameters that represent α-synuclein pathology spreading, aggregation, decay, and gene expression pattern to this longitudinal dataset. Remarkably, our model can generalize to predicting α-synuclein spreading patterns from several distinct brain regions and can even estimate their origins. This model empowers mechanistic understanding and accurate prediction of disease progression, paving the way for the development and testing of therapeutic interventions.
Collapse
Affiliation(s)
- Ehsan Dadgar-Kiani
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA 94305, USA; Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Gregor Bieri
- Department of Genetics, Stanford University, Stanford, CA 94305, USA
| | - Ronald Melki
- Institut François Jacob, MIRCen, CEA and Laboratory of Neurodegenerative Diseases, CNRS, 92265 Fontenay-Aux-Roses, France
| | - Aaron D Gitler
- Department of Genetics, Stanford University, Stanford, CA 94305, USA; Chan Zuckerberg Biohub, San Francisco, CA 94158, USA.
| | - Jin Hyung Lee
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA 94305, USA; Department of Bioengineering, Stanford University, Stanford, CA 94305, USA; Department of Electrical Engineering, Stanford University, Stanford, CA 94305, USA; Department of Neurosurgery, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
46
|
Lashuel HA, Mahul-Mellier AL, Novello S, Hegde RN, Jasiqi Y, Altay MF, Donzelli S, DeGuire SM, Burai R, Magalhães P, Chiki A, Ricci J, Boussouf M, Sadek A, Stoops E, Iseli C, Guex N. Revisiting the specificity and ability of phospho-S129 antibodies to capture alpha-synuclein biochemical and pathological diversity. NPJ Parkinsons Dis 2022; 8:136. [PMID: 36266318 PMCID: PMC9584898 DOI: 10.1038/s41531-022-00388-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 08/30/2022] [Indexed: 11/08/2022] Open
Abstract
Antibodies against phosphorylated alpha-synuclein (aSyn) at S129 have emerged as the primary tools to investigate, monitor, and quantify aSyn pathology in the brain and peripheral tissues of patients with Parkinson's disease and other neurodegenerative diseases. Herein, we demonstrate that the co-occurrence of multiple pathology-associated C-terminal post-translational modifications (PTMs) (e.g., phosphorylation at Tyrosine 125 or truncation at residue 133 or 135) differentially influences the detection of pS129-aSyn species by pS129-aSyn antibodies. These observations prompted us to systematically reassess the specificity of the most commonly used pS129 antibodies against monomeric and aggregated forms of pS129-aSyn in mouse brain slices, primary neurons, mammalian cells and seeding models of aSyn pathology formation. We identified two antibodies that are insensitive to pS129 neighboring PTMs. Although most pS129 antibodies showed good performance in detecting aSyn aggregates in cells, neurons and mouse brain tissue containing abundant aSyn pathology, they also showed cross-reactivity towards other proteins and often detected non-specific low and high molecular weight bands in aSyn knock-out samples that could be easily mistaken for monomeric or high molecular weight aSyn species. Our observations suggest that not all pS129 antibodies capture the biochemical and morphological diversity of aSyn pathology, and all should be used with the appropriate protein standards and controls when investigating aSyn under physiological conditions. Finally, our work underscores the need for more pS129 antibodies that are not sensitive to neighboring PTMs and more thorough characterization and validation of existing and new antibodies.
Collapse
Affiliation(s)
- Hilal A Lashuel
- Laboratory of Molecular and Chemical Biology of Neurodegeneration, School of Life Sciences, Brain Mind Institute, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015, Lausanne, Switzerland.
| | - Anne-Laure Mahul-Mellier
- Laboratory of Molecular and Chemical Biology of Neurodegeneration, School of Life Sciences, Brain Mind Institute, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015, Lausanne, Switzerland
| | - Salvatore Novello
- Laboratory of Molecular and Chemical Biology of Neurodegeneration, School of Life Sciences, Brain Mind Institute, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015, Lausanne, Switzerland
| | - Ramanath Narayana Hegde
- Laboratory of Molecular and Chemical Biology of Neurodegeneration, School of Life Sciences, Brain Mind Institute, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015, Lausanne, Switzerland
| | - Yllza Jasiqi
- Laboratory of Molecular and Chemical Biology of Neurodegeneration, School of Life Sciences, Brain Mind Institute, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015, Lausanne, Switzerland
| | - Melek Firat Altay
- Laboratory of Molecular and Chemical Biology of Neurodegeneration, School of Life Sciences, Brain Mind Institute, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015, Lausanne, Switzerland
| | - Sonia Donzelli
- Laboratory of Molecular and Chemical Biology of Neurodegeneration, School of Life Sciences, Brain Mind Institute, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015, Lausanne, Switzerland
| | - Sean M DeGuire
- Laboratory of Molecular and Chemical Biology of Neurodegeneration, School of Life Sciences, Brain Mind Institute, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015, Lausanne, Switzerland
| | - Ritwik Burai
- Laboratory of Molecular and Chemical Biology of Neurodegeneration, School of Life Sciences, Brain Mind Institute, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015, Lausanne, Switzerland
| | - Pedro Magalhães
- Laboratory of Molecular and Chemical Biology of Neurodegeneration, School of Life Sciences, Brain Mind Institute, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015, Lausanne, Switzerland
| | - Anass Chiki
- Laboratory of Molecular and Chemical Biology of Neurodegeneration, School of Life Sciences, Brain Mind Institute, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015, Lausanne, Switzerland
| | - Jonathan Ricci
- Laboratory of Molecular and Chemical Biology of Neurodegeneration, School of Life Sciences, Brain Mind Institute, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015, Lausanne, Switzerland
| | - Manel Boussouf
- Laboratory of Molecular and Chemical Biology of Neurodegeneration, School of Life Sciences, Brain Mind Institute, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015, Lausanne, Switzerland
| | - Ahmed Sadek
- Laboratory of Molecular and Chemical Biology of Neurodegeneration, School of Life Sciences, Brain Mind Institute, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015, Lausanne, Switzerland
| | - Erik Stoops
- ADx NeuroSciences, Technologiepark 94, Ghent, Belgium
| | - Christian Iseli
- Bioinformatics Competence Center, Ecole Polytechnique Fédérale de Lausanne, 1015, Lausanne, Switzerland
- Bioinformatics Competence Center, University of Lausanne, 1015, Lausanne, Switzerland
| | - Nicolas Guex
- Bioinformatics Competence Center, Ecole Polytechnique Fédérale de Lausanne, 1015, Lausanne, Switzerland
- Bioinformatics Competence Center, University of Lausanne, 1015, Lausanne, Switzerland
| |
Collapse
|
47
|
Alpha-Synuclein: The Spark That Flames Dopaminergic Neurons, In Vitro and In Vivo Evidence. Int J Mol Sci 2022; 23:ijms23179864. [PMID: 36077253 PMCID: PMC9456396 DOI: 10.3390/ijms23179864] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 08/15/2022] [Accepted: 08/21/2022] [Indexed: 11/16/2022] Open
Abstract
Mitochondria, α-syn fibrils and the endo-lysosomal system are key players in the pathophysiology of Parkinson’s disease. The toxicity of α-syn is amplified by cell-to-cell transmission and aggregation of endogenous species in newly invaded neurons. Toxicity of α-syn PFF was investigated using primary cultures of dopaminergic neurons or on aged mice after infusion in the SNpc and combined with mild inhibition of GBA. In primary dopaminergic neurons, application of α-syn PFF induced a progressive cytotoxicity associated with mitochondrial dysfunction, oxidative stress, and accumulation of lysosomes suggesting that exogenous α-syn reached the lysosome (from the endosome). Counteracting the α-syn endocytosis with a clathrin inhibitor, dopaminergic neuron degeneration was prevented. In vivo, α-syn PFF induced progressive neurodegeneration of dopaminergic neurons associated with motor deficits. Histology revealed progressive aggregation of α-syn and microglial activation and accounted for the seeding role of α-syn, injection of which acted as a spark suggesting a triggering of cell-to-cell toxicity. We showed for the first time that a localized SNpc α-syn administration combined with a slight lysosomal deficiency and aging triggered a progressive lesion. The cellular and animal models described could help in the understanding of the human disease and might contribute to the development of new therapies.
Collapse
|
48
|
Riboldi GM, Vialle RA, Navarro E, Udine E, de Paiva Lopes K, Humphrey J, Allan A, Parks M, Henderson B, Astudillo K, Argyrou C, Zhuang M, Sikder T, Oriol Narcis J, Kumar SD, Janssen W, Sowa A, Comi GP, Di Fonzo A, Crary JF, Frucht SJ, Raj T. Transcriptome deregulation of peripheral monocytes and whole blood in GBA-related Parkinson's disease. Mol Neurodegener 2022; 17:52. [PMID: 35978378 PMCID: PMC9386994 DOI: 10.1186/s13024-022-00554-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 06/29/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Genetic mutations in beta-glucocerebrosidase (GBA) represent the major genetic risk factor for Parkinson's disease (PD). GBA participates in both the endo-lysosomal pathway and the immune response, two important mechanisms involved in the pathogenesis of PD. However, modifiers of GBA penetrance have not yet been fully elucidated. METHODS We characterized the transcriptomic profiles of circulating monocytes in a population of patients with PD and healthy controls (CTRL) with and without GBA variants (n = 23 PD/GBA, 13 CTRL/GBA, 56 PD, 66 CTRL) and whole blood (n = 616 PD, 362 CTRL, 127 PD/GBA, 165 CTRL/GBA). Differential expression analysis, pathway enrichment analysis, and outlier detection were performed. Ultrastructural characterization of isolated CD14+ monocytes in the four groups was also performed through electron microscopy. RESULTS We observed hundreds of differentially expressed genes and dysregulated pathways when comparing manifesting and non-manifesting GBA mutation carriers. Specifically, when compared to idiopathic PD, PD/GBA showed dysregulation in genes involved in alpha-synuclein degradation, aging and amyloid processing. Gene-based outlier analysis confirmed the involvement of lysosomal, membrane trafficking, and mitochondrial processing in manifesting compared to non-manifesting GBA-carriers, as also observed at the ultrastructural levels. Transcriptomic results were only partially replicated in an independent cohort of whole blood samples, suggesting cell-type specific changes. CONCLUSIONS Overall, our transcriptomic analysis of primary monocytes identified gene targets and biological processes that can help in understanding the pathogenic mechanisms associated with GBA mutations in the context of PD.
Collapse
Affiliation(s)
- Giulietta Maria Riboldi
- The Marlene and Paolo Fresco Institute for Parkinson’s Disease and Movement Disorders, New York University Langone Health, 222 East 41st street, New York, NY 10017 USA
| | - Ricardo A. Vialle
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029 USA
- Ronald M. Loeb Center for Alzheimer’s disease, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029 USA
- Department of Genetics and Genomic Sciences & Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1498, New York, NY 10029 USA
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1137, New York, NY 10029 USA
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL USA
| | - Elisa Navarro
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029 USA
- Ronald M. Loeb Center for Alzheimer’s disease, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029 USA
- Department of Genetics and Genomic Sciences & Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1498, New York, NY 10029 USA
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1137, New York, NY 10029 USA
- Department of Biochemistry and Molecular Biology (Universidad Complutense de Madrid) & Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Evan Udine
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029 USA
- Ronald M. Loeb Center for Alzheimer’s disease, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029 USA
- Department of Genetics and Genomic Sciences & Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1498, New York, NY 10029 USA
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1137, New York, NY 10029 USA
| | - Katia de Paiva Lopes
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029 USA
- Ronald M. Loeb Center for Alzheimer’s disease, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029 USA
- Department of Genetics and Genomic Sciences & Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1498, New York, NY 10029 USA
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1137, New York, NY 10029 USA
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL USA
| | - Jack Humphrey
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029 USA
- Ronald M. Loeb Center for Alzheimer’s disease, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029 USA
- Department of Genetics and Genomic Sciences & Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1498, New York, NY 10029 USA
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1137, New York, NY 10029 USA
| | - Amanda Allan
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029 USA
- Ronald M. Loeb Center for Alzheimer’s disease, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029 USA
- Department of Genetics and Genomic Sciences & Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1498, New York, NY 10029 USA
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1137, New York, NY 10029 USA
| | - Madison Parks
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029 USA
- Ronald M. Loeb Center for Alzheimer’s disease, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029 USA
- Department of Genetics and Genomic Sciences & Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1498, New York, NY 10029 USA
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1137, New York, NY 10029 USA
| | - Brooklyn Henderson
- The Marlene and Paolo Fresco Institute for Parkinson’s Disease and Movement Disorders, New York University Langone Health, 222 East 41st street, New York, NY 10017 USA
| | - Kelly Astudillo
- The Marlene and Paolo Fresco Institute for Parkinson’s Disease and Movement Disorders, New York University Langone Health, 222 East 41st street, New York, NY 10017 USA
| | - Charalambos Argyrou
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029 USA
- Ronald M. Loeb Center for Alzheimer’s disease, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029 USA
- Department of Genetics and Genomic Sciences & Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1498, New York, NY 10029 USA
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1137, New York, NY 10029 USA
| | - Maojuan Zhuang
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029 USA
- Ronald M. Loeb Center for Alzheimer’s disease, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029 USA
- Department of Genetics and Genomic Sciences & Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1498, New York, NY 10029 USA
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1137, New York, NY 10029 USA
| | - Tamjeed Sikder
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029 USA
- Ronald M. Loeb Center for Alzheimer’s disease, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029 USA
- Department of Pathology, Icahn School of Medicine at Mount Sinai, 1468 Madison Avenue, Annenberg Building, 15th Floor, New York, NY 10029 USA
- Neuropathology Brain Bank & Research CoRE, Icahn School of Medicine at Mount Sinai, 1425 Madison Avenue, Room 9-22, New York, NY 10029 USA
| | - J. Oriol Narcis
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029 USA
- Ronald M. Loeb Center for Alzheimer’s disease, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029 USA
- Department of Genetics and Genomic Sciences & Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1498, New York, NY 10029 USA
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1137, New York, NY 10029 USA
| | - Shilpa Dilip Kumar
- Microscopy Core and Advanced Bioimaging Center at the Icahn School of Medicine at Mount Sinai Center, 1468 Madison Avenue, Room 18-250, New York, NY 10029 USA
| | - William Janssen
- Microscopy Core and Advanced Bioimaging Center at the Icahn School of Medicine at Mount Sinai Center, 1468 Madison Avenue, Room 18-250, New York, NY 10029 USA
| | - Allison Sowa
- Department of Pathology, Icahn School of Medicine at Mount Sinai, 1468 Madison Avenue, Annenberg Building, 15th Floor, New York, NY 10029 USA
| | - Giacomo P. Comi
- Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Neurology Unit, Milan, Italy
- Dino Ferrari Center, Neuroscience Section, Department of Pathophysiology and Transplantation, University of Milan, Via Francesco Sforza, 35, 20122 Milano, MI Italy
| | - Alessio Di Fonzo
- Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Neurology Unit, Milan, Italy
- Dino Ferrari Center, Neuroscience Section, Department of Pathophysiology and Transplantation, University of Milan, Via Francesco Sforza, 35, 20122 Milano, MI Italy
| | - John F. Crary
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029 USA
- Ronald M. Loeb Center for Alzheimer’s disease, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029 USA
- Department of Pathology, Icahn School of Medicine at Mount Sinai, 1468 Madison Avenue, Annenberg Building, 15th Floor, New York, NY 10029 USA
- Neuropathology Brain Bank & Research CoRE, Icahn School of Medicine at Mount Sinai, 1425 Madison Avenue, Room 9-22, New York, NY 10029 USA
| | - Steven J. Frucht
- The Marlene and Paolo Fresco Institute for Parkinson’s Disease and Movement Disorders, New York University Langone Health, 222 East 41st street, New York, NY 10017 USA
| | - Towfique Raj
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029 USA
- Ronald M. Loeb Center for Alzheimer’s disease, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029 USA
- Department of Genetics and Genomic Sciences & Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1498, New York, NY 10029 USA
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1137, New York, NY 10029 USA
- Icahn School of Medicine at Mount Sinai, 1425 Madison Avenue, ICAHN 10-70E, New York, NY 10029–6574 USA
| |
Collapse
|
49
|
Bellucci A, Longhena F, Spillantini MG. The Role of Rab Proteins in Parkinson's Disease Synaptopathy. Biomedicines 2022; 10:biomedicines10081941. [PMID: 36009486 PMCID: PMC9406004 DOI: 10.3390/biomedicines10081941] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 07/31/2022] [Accepted: 08/08/2022] [Indexed: 12/29/2022] Open
Abstract
In patients affected by Parkinson's disease (PD), the most common neurodegenerative movement disorder, the brain is characterized by the loss of dopaminergic neurons in the nigrostriatal system, leading to dyshomeostasis of the basal ganglia network activity that is linked to motility dysfunction. PD mostly arises as an age-associated sporadic disease, but several genetic forms also exist. Compelling evidence supports that synaptic damage and dysfunction characterize the very early phases of either sporadic or genetic forms of PD and that this early PD synaptopathy drives retrograde terminal-to-cell body degeneration, culminating in neuronal loss. The Ras-associated binding protein (Rab) family of small GTPases, which is involved in the maintenance of neuronal vesicular trafficking, synaptic architecture and function in the central nervous system, has recently emerged among the major players in PD synaptopathy. In this manuscript, we provide an overview of the main findings supporting the involvement of Rabs in either sporadic or genetic PD pathophysiology, and we highlight how Rab alterations participate in the onset of early synaptic damage and dysfunction.
Collapse
Affiliation(s)
- Arianna Bellucci
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
- Correspondence: ; Tel.: +39-0303-717-380
| | - Francesca Longhena
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
- Department of Clinical Neurosciences, University of Cambridge, Clifford Albutt Building, Cambridge CB2 0AH, UK
| | - Maria Grazia Spillantini
- Department of Clinical Neurosciences, University of Cambridge, Clifford Albutt Building, Cambridge CB2 0AH, UK
| |
Collapse
|
50
|
Wang L, Lin G, Zuo Z, Li Y, Byeon SK, Pandey A, Bellen HJ. Neuronal activity induces glucosylceramide that is secreted via exosomes for lysosomal degradation in glia. SCIENCE ADVANCES 2022; 8:eabn3326. [PMID: 35857503 PMCID: PMC9278864 DOI: 10.1126/sciadv.abn3326] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 05/27/2022] [Indexed: 05/06/2023]
Abstract
Recessive variants in GBA1 cause Gaucher disease, a prevalent form of lysosome storage disease. GBA1 encodes a lysosomal enzyme that hydrolyzes glucosylceramide (GlcCer) into glucose and ceramide. Its loss causes lysosomal dysfunction and increased levels of GlcCer. We generated a null allele of the Drosophila ortholog Gba1b by inserting the Gal4 using CRISPR-Cas9. Here, we show that Gba1b is expressed in glia but not in neurons. Glial-specific knockdown recapitulates the defects found in Gba1b mutants, and these can be rescued by glial expression of human GBA1. We show that GlcCer is synthesized upon neuronal activity, and it is transported from neurons to glia through exosomes. Furthermore, we found that glial TGF-β/BMP induces the transfer of GlcCer from neurons to glia and that the White protein, an ABCG transporter, promotes GlcCer trafficking to glial lysosomes for degradation.
Collapse
Affiliation(s)
- Liping Wang
- Program in Developmental Biology, Baylor College of Medicine, Houston, TX 77030, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, TX 77030, USA
| | - Guang Lin
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, TX 77030, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Zhongyuan Zuo
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, TX 77030, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Yarong Li
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, TX 77030, USA
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA
| | - Seul Kee Byeon
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905, USA
| | - Akhilesh Pandey
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905, USA
- Center for Individualized Medicine, Mayo Clinic, Rochester, MN 55905, USA
- Manipal Academy of Higher Education, Manipal, Karnataka 576 104, India
| | - Hugo J. Bellen
- Program in Developmental Biology, Baylor College of Medicine, Houston, TX 77030, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, TX 77030, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|