1
|
Azad MG, Russell T, Gu X, Zhao X, Richardson V, Wijesinghe TP, Babu G, Guo X, Kaya B, Dharmasivam M, Deng Z, Richardson DR. NDRG1 and its Family Members: More than Just Metastasis Suppressor Proteins and Targets of Thiosemicarbazones. J Biol Chem 2025:110230. [PMID: 40378957 DOI: 10.1016/j.jbc.2025.110230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2025] [Revised: 04/19/2025] [Accepted: 05/05/2025] [Indexed: 05/19/2025] Open
Abstract
N-Myc downstream regulated gene-1 (NDRG1) and the other three members of this family (NDRG2, 3, and 4) play various functional roles in the cellular stress response, differentiation, migration, and development. These proteins are involved in regulating key signaling proteins and pathways that are often dysregulated in cancer, such as EGFR, PI3K/AKT, c-Met, and the Wnt pathway. NDRG1 is the primary, well-examined member of the NDRG family, and is generally characterized as a metastasis suppressor that inhibits the first step in metastasis, the epithelial-mesenchymal transition. While NDRG1 is well-studied, emerging evidence suggests NDRG2, NDRG3, and NDRG4 also play significant roles in modulating oncogenic signaling and cellular homeostasis. NDRG family members are regulated by multiple mechanisms, including transcriptional control by hypoxia-inducible factors, p53, and Myc, as well as post-translational modifications such as phosphorylation, ubiquitination, and acetylation. Pharmacological targeting of the NDRG family is a therapeutic strategy against cancer. For instance, di-2-pyridylketone 4,4-dimethyl-3-thiosemicarbazone (Dp44mT) and di-2-pyridylketone-4-cyclohexyl-4-methyl-3-thiosemicarbazone (DpC) have been extensively shown to up-regulate NDRG1 expression, leading to metastasis suppression and inhibition of tumor growth in multiple cancer models. Similarly, targeting NDRG2 demonstrates its pro-apoptotic and anti-proliferative effects, particularly in glioblastoma and colorectal cancer. This review provides a comprehensive analysis of the structural features, regulatory mechanisms, and biological functions of the NDRG family and their roles in cancer and neurodegenerative diseases. Additionally, NDRG1-4 are explored as therapeutic targets in oncology, focusing on recent advances in anti-cancer agents that induce the expression of these proteins. Implications for future research and clinical applications are also discussed.
Collapse
Affiliation(s)
- Mahan Gholam Azad
- Centre for Cancer Cell Biology and Drug Discovery, Griffith University, Nathan, Brisbane, 4111, Queensland, Australia
| | - Tiffany Russell
- Centre for Cancer Cell Biology and Drug Discovery, Griffith University, Nathan, Brisbane, 4111, Queensland, Australia
| | - Xuanling Gu
- Centre for Cancer Cell Biology and Drug Discovery, Griffith University, Nathan, Brisbane, 4111, Queensland, Australia
| | - Xiao Zhao
- Centre for Cancer Cell Biology and Drug Discovery, Griffith University, Nathan, Brisbane, 4111, Queensland, Australia
| | - Vera Richardson
- Centre for Cancer Cell Biology and Drug Discovery, Griffith University, Nathan, Brisbane, 4111, Queensland, Australia
| | - Tharushi P Wijesinghe
- Centre for Cancer Cell Biology and Drug Discovery, Griffith University, Nathan, Brisbane, 4111, Queensland, Australia
| | - Golap Babu
- Centre for Cancer Cell Biology and Drug Discovery, Griffith University, Nathan, Brisbane, 4111, Queensland, Australia
| | - Xinnong Guo
- Centre for Cancer Cell Biology and Drug Discovery, Griffith University, Nathan, Brisbane, 4111, Queensland, Australia
| | - Busra Kaya
- Centre for Cancer Cell Biology and Drug Discovery, Griffith University, Nathan, Brisbane, 4111, Queensland, Australia
| | - Mahendiran Dharmasivam
- Centre for Cancer Cell Biology and Drug Discovery, Griffith University, Nathan, Brisbane, 4111, Queensland, Australia
| | - Zhao Deng
- Centre for Cancer Cell Biology and Drug Discovery, Griffith University, Nathan, Brisbane, 4111, Queensland, Australia
| | - Des R Richardson
- Centre for Cancer Cell Biology and Drug Discovery, Griffith University, Nathan, Brisbane, 4111, Queensland, Australia; Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan.
| |
Collapse
|
2
|
Szunyogh S, Carroll E, Wade-Martins R. Recent developments in gene therapy for Parkinson's disease. Mol Ther 2025; 33:2052-2064. [PMID: 40121531 DOI: 10.1016/j.ymthe.2025.03.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 03/07/2025] [Accepted: 03/18/2025] [Indexed: 03/25/2025] Open
Abstract
Parkinson's disease (PD) is a progressive, neurodegenerative disorder for which there is currently no cure. Gene therapy has emerged as a novel approach offering renewed hope for the development of treatments that meaningfully alter the course of the disease. In this review, we explore various gene therapy strategies currently being developed targeting key aspects of PD pathogenesis: the restoration of the dopamine system by delivering genes involved in dopamine biosynthesis, reinforcing the inhibitory signaling pathways through glutamic acid decarboxylase (GAD) delivery to increase GABA production, enhancing neuronal survival and development by introducing various neurotrophic factors, delivery of genes to complement recessive familial PD mutations to correct mitochondrial dysfunction, restoring lysosomal function through delivery of GBA1 to increase glucocerebrosidase (GCase) activity, and reducing α-synuclein levels by reducing or silencing SNCA expression. Despite promising early work, challenges remain in developing safe, effective, and long-lasting gene therapies. Key considerations include optimizing viral vectors for targeted delivery, achieving controlled and sustained gene expression using different promoters, minimizing immune responses, and increasing transgene delivery capacity. Future prospects may involve combinatory strategies targeting multiple pathways, such as multi-gene constructs delivered via high-capacity viral systems.
Collapse
Affiliation(s)
- Sandor Szunyogh
- Oxford Parkinson's Disease Centre, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3QX, UK; Kavli Institute for Nanoscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, South Parks Road, Oxford OX1 3QU, UK
| | - Emily Carroll
- Oxford Parkinson's Disease Centre, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3QX, UK; Kavli Institute for Nanoscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, South Parks Road, Oxford OX1 3QU, UK
| | - Richard Wade-Martins
- Oxford Parkinson's Disease Centre, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3QX, UK; Kavli Institute for Nanoscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, South Parks Road, Oxford OX1 3QU, UK.
| |
Collapse
|
3
|
Luo Y, Funayama M, Hatano T, Li Y, Yoshino H, Yamashita S, Mori A, Nakamura R, Hashizume Y, Yoshida M, Riku Y, Kanzato N, Hattori N. Identification of DAGLB variants in Japanese early-onset Parkinson's disease. J Neural Transm (Vienna) 2025:10.1007/s00702-025-02926-y. [PMID: 40244389 DOI: 10.1007/s00702-025-02926-y] [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: 02/20/2025] [Accepted: 04/04/2025] [Indexed: 04/18/2025]
Abstract
Hereditary factors play a significant role in the development of Parkinson's disease and the identification of causative genes is ongoing. Biallelic variants in Diacylglycerol lipase β (DAGLB) are related to early-onset Parkinson's disease (EOPD) in the Chinese population, and have also been identified in an Algerian case. To date, no EOPD cases with DAGLB variants have been reported among Japanese patients. This study was conducted to clarify the occurrence of DAGLB variants among Japanese EOPD patients. We screened 270 patients with sporadic EOPD (male: female ratio, 1.37:1; mean age at onset ± standard deviation, 37.32 ± 7.91 years), and 276 patients with suspected autosomal recessive Parkinson's disease (ARPD, male: female ratio, 0.75:1; mean age at onset ± standard deviation, 58.86 ± 14.67 years). Genetic screening of all coding exons and flanking splicing regions was performed by Sanger sequencing. We identified two rare biallelic variants in two patients, both from consanguineous families. One variant was a homozygous frameshift variant (c.1770_1771del, p.Tyr591ProfsTer26), which was predicted to be pathogenic. The other was a missense variant (c.1444T > C, p.Tyr482His) and was predicted to be benign, with co-segregation ruled out for this variant. We identified a pathogenic variant in the DAGLB gene. Together with previous reports, these findings provide further evidence that loss-of-function variants in DAGLB are involved in EOPD in the Japanese population.
Collapse
Affiliation(s)
- Yue Luo
- Department of Neurology, Faculty of Medicine, Juntendo University, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Manabu Funayama
- Department of Neurology, Faculty of Medicine, Juntendo University, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan.
- Research Institute for Diseases of Old Age, Graduate School of Medicine, Juntendo University, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan.
- International Collaborative Research Administration, Juntendo University, Tokyo, Japan.
| | - Taku Hatano
- Department of Neurology, Faculty of Medicine, Juntendo University, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Yuanzhe Li
- Department of Neurology, Faculty of Medicine, Juntendo University, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
- Department of Diagnosis, Prevention and Treatment of Dementia, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Hiroyo Yoshino
- Research Institute for Diseases of Old Age, Graduate School of Medicine, Juntendo University, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Satoshi Yamashita
- Department of Neurology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
- Department of Neurology, International University of Health and Welfare Narita Hospital, Narita, Chiba, Japan
| | - Akira Mori
- Department of Neurology, Kumamoto Southern Regional Hospital, Kumamoto, Japan
| | - Ryoichi Nakamura
- Department of Neurology, Japanese Red Cross Aichi Medical Center Nagoya Daiichi Hospital, Nagoya, Aichi, Japan
- Department of Neurology, Aichi Medical University School of Medicine, Nagakute, Aichi, Japan
| | - Yoshio Hashizume
- Choju Medical Institute, Fukushimura Hospital, Toyohashi, Aichi, Japan
| | - Mari Yoshida
- Institute for Medical Science of Aging, Aichi Medical University, Nagakute, Aichi, Japan
| | - Yuichi Riku
- Institute for Medical Science of Aging, Aichi Medical University, Nagakute, Aichi, Japan
| | - Naomi Kanzato
- Department of Neurology, Okinawa Prefectural Southern Medical Center & Children's Medical Center, Haebaru-cho, Okinawa, Japan
| | - Nobutaka Hattori
- Department of Neurology, Faculty of Medicine, Juntendo University, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan.
- Research Institute for Diseases of Old Age, Graduate School of Medicine, Juntendo University, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan.
- International Collaborative Research Administration, Juntendo University, Tokyo, Japan.
- Department of Diagnosis, Prevention and Treatment of Dementia, Graduate School of Medicine, Juntendo University, Tokyo, Japan.
- Neurodegenerative Disorders Collaborative Laboratory, RIKEN Center for Brain Science, Saitama, Japan.
| |
Collapse
|
4
|
Daida K, Yoshino H, Malik L, Baker B, Ishiguro M, Genner R, Paquette K, Li Y, Nishioka K, Masuzugawa S, Hirano M, Takahashi K, Kolmogorov M, Billingsley KJ, Funayama M, Blauwendraat C, Hattori N. The Utility of Long-Read Sequencing in Diagnosing Early Onset Parkinson's Disease. Ann Neurol 2025; 97:753-765. [PMID: 39699073 PMCID: PMC11889530 DOI: 10.1002/ana.27155] [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] [Received: 07/30/2024] [Revised: 11/27/2024] [Accepted: 11/27/2024] [Indexed: 12/20/2024]
Abstract
OBJECTIVE Variants in PRKN and PINK1 are the leading cause of early-onset autosomal recessive Parkinson's disease, yet many cases remain genetically unresolved. We previously identified a 7 megabases complex structural variant in a pair of monozygotic twins using Oxford Nanopore Technologies (ONT) long-read sequencing. This study aims to determine if ONT long-read sequencing can detect a second variant in other unresolved early-onset Parkinson's disease (EOPD) cases with 1 heterozygous PRKN or PINK1 variant. METHODS ONT long-read sequencing was performed on EOPD patients with 1 reported PRKN/PINK1 pathogenic variant, with onset age under 50. Positive controls included EOPD patients with 2 known PRKN pathogenic variants. Initial testing involved short-read targeted panel sequencing for single nucleotide variants and multiplex ligation-dependent probe amplification for copy number variants. RESULTS A total of 47 patients were studied (PRKN "one-variant," n = 23; PINK1 "one-variant," n = 12; PRKN "two-variants," n = 12). ONT long-read sequencing identified a second pathogenic variant in 26% of PRKN "one-variant" patients (6/23), but none in PINK1 "one-variant" patients (0/12). Detected variants included 1 complex inversion, 2 structural variant overlaps, and 3 duplications. In the PRKN "two-variants" group, both variants were identified in all patients (100%, 12/12). INTERPRETATION ONT long-read sequencing effectively identifies pathogenic structural variants in the PRKN locus missed by conventional methods. It should be considered for unresolved EOPD cases when a second variant is not detected through conventional approaches. ANN NEUROL 2025;97:753-765.
Collapse
Affiliation(s)
- Kensuke Daida
- Integrative Neurogenomics Unit, Laboratory of NeurogeneticsNational Institute on Aging, National Institutes of HealthBethesdaMDUSA
- Center for Alzheimer's and Related Dementias (CARD)National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of HealthBethesdaMDUSA
- Department of Neurology, Faculty of MedicineJuntendo UniversityTokyoJapan
| | - Hiroyo Yoshino
- Research Institute for Diseases of Old Age, Graduate School of MedicineJuntendo UniversityTokyoJapan
| | - Laksh Malik
- Center for Alzheimer's and Related Dementias (CARD)National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of HealthBethesdaMDUSA
| | - Breeana Baker
- Center for Alzheimer's and Related Dementias (CARD)National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of HealthBethesdaMDUSA
| | - Mayu Ishiguro
- Department of Neurology, Faculty of MedicineJuntendo UniversityTokyoJapan
| | - Rylee Genner
- Center for Alzheimer's and Related Dementias (CARD)National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of HealthBethesdaMDUSA
| | - Kimberly Paquette
- Center for Alzheimer's and Related Dementias (CARD)National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of HealthBethesdaMDUSA
| | - Yuanzhe Li
- Department of Neurology, Faculty of MedicineJuntendo UniversityTokyoJapan
- Department of Diagnosis, Prevention and Treatment of Dementia, Graduate School of MedicineJuntendo UniversityTokyoJapan
| | - Kenya Nishioka
- Department of NeurologyJuntendo Tokyo Koto Geriatric Medical CenterTokyoJapan
| | | | - Makito Hirano
- Department of NeurologyKindai University Faculty of MedicineOsakaJapan
| | - Kenta Takahashi
- Division of Neurology and Gerontology, Department of Internal Medicine, School of MedicineIwate Medical UniversityMoriokaJapan
| | - Mikhail Kolmogorov
- Center for Cancer ResearchNational Cancer Institute, National Institutes of HealthBethesdaMDUSA
| | - Kimberley J. Billingsley
- Molecular Genetics Section, Laboratory of NeurogeneticsNational Institute on Aging, National Institutes of HealthBethesdaMDUSA
| | - Manabu Funayama
- Department of Neurology, Faculty of MedicineJuntendo UniversityTokyoJapan
- Research Institute for Diseases of Old Age, Graduate School of MedicineJuntendo UniversityTokyoJapan
| | - Cornelis Blauwendraat
- Integrative Neurogenomics Unit, Laboratory of NeurogeneticsNational Institute on Aging, National Institutes of HealthBethesdaMDUSA
- Center for Alzheimer's and Related Dementias (CARD)National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of HealthBethesdaMDUSA
| | - Nobutaka Hattori
- Department of Neurology, Faculty of MedicineJuntendo UniversityTokyoJapan
- Research Institute for Diseases of Old Age, Graduate School of MedicineJuntendo UniversityTokyoJapan
- Neurodegenerative Disorders Collaborative Laboratory, RIKEN Center for Brain ScienceWakoJapan
| |
Collapse
|
5
|
Braccia A, Elia AE, Devigili G, Lombardi R, Luppino A, Mazzetti S, Panteghini C, Colangelo I, Suerz M, Portaleone SM, Perilli AM, De Luca CMG, Ciullini A, Dellarole IL, Telese R, Garavaglia B, Moda F, Eleopra R. α-Synuclein distribution in olfactory mucosa and skin nerves in Parkinson disease associated with an EIF4G1 gene mutation. J Neuropathol Exp Neurol 2025; 84:286-292. [PMID: 39864405 DOI: 10.1093/jnen/nlaf001] [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] [Indexed: 01/28/2025] Open
Abstract
The EIF4G1 gene has been considered an autosomal dominant cause of Parkinson disease (PD), even if its role is still debated. The objective of this study was to describe the phenotype and α-synuclein distribution in peripheral tissues in 2 related PD patients (mother and daughter), who are carriers of the same variant in exon 10 of EIF4G1 (c.1216G>A, p.Gly406Arg). We used the Burghart Sniffin Sticks test for olfactory function. α-Synuclein distribution in the olfactory mucosa and skin samples was analyzed using RT-QuIC, double immunofluorescence, and immunohistochemical staining. Both patients presented with a mild motor syndrome associated with hyposmia as prominent traits; pathological α-synuclein deposits were found in the olfactory mucosa but not in the skin. The phenotype and the findings in peripheral tissues suggest that PARK18 could manifest as a "benign" form of PD associated with hyposmia, with a slow progression and sparse α-synuclein accumulation in the peripheral nervous system.
Collapse
Affiliation(s)
- Arianna Braccia
- Department of Clinical Neurosciences, Parkinson and Movement Disorders Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Antonio Emanuele Elia
- Department of Clinical Neurosciences, Parkinson and Movement Disorders Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Grazia Devigili
- Department of Clinical Neurosciences, Parkinson and Movement Disorders Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Raffaella Lombardi
- Neuroalgology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Alessia Luppino
- Department of Clinical Neurosciences, Parkinson and Movement Disorders Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Samanta Mazzetti
- Department of Clinical Neurosciences, Parkinson and Movement Disorders Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Celeste Panteghini
- Medical Genetics and Neurogenetics Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Isabel Colangelo
- Medical Genetics and Neurogenetics Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Marta Suerz
- Medical Genetics and Neurogenetics Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Sara Maria Portaleone
- Department of Health Sciences, Otolaryngology Unit, San Paolo Hospital, Università degli Studi di Milano, Milan, Italy
| | - Anna Maria Perilli
- Department of Clinical Neurosciences, Parkinson and Movement Disorders Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | | | - Arianna Ciullini
- Unit of Neurology 5 and Neuropathology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Ilaria Linda Dellarole
- Unit of Neurology 5 and Neuropathology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Roberta Telese
- Department of Clinical Neurosciences, Parkinson and Movement Disorders Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Barbara Garavaglia
- Medical Genetics and Neurogenetics Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Fabio Moda
- Department of Diagnostic and Technology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Roberto Eleopra
- Department of Clinical Neurosciences, Parkinson and Movement Disorders Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| |
Collapse
|
6
|
Chinchihualpa Paredes N, Pecoraro PM, Zaidi SA, Duque KR, Mahajan A, Mohammad ME, Duker AP, Kauffman MA, Espay AJ, Marsili L. Clinical Reasoning: Juvenile-Onset Dopa-Responsive Dystonia-Until It Isn't. Neurology 2025; 104:e213436. [PMID: 40014835 DOI: 10.1212/wnl.0000000000213436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Accepted: 01/06/2025] [Indexed: 03/01/2025] Open
Abstract
Dopa-responsive dystonia (DRD) encompasses a broad spectrum of phenotypically diverse conditions, offering unique diagnostic challenges. We describe the case of a 24-year-old woman with dystonic gait and jerky position-sensitive limb tremor that initially responded exquisitely well to low-dose levodopa, suggesting DRD. Two years later, she developed parkinsonian features, ultimately leading to a diagnostic reclassification. Despite extensive investigations, the underlying etiology remained elusive until 10 years after symptom onset. We discuss this DRD mimic and briefly review the disorders from which it must be distinguished.
Collapse
Affiliation(s)
- Nathaly Chinchihualpa Paredes
- James J. and Joan A. Gardner Center for Parkinson's Disease and Movement Disorders, Department of Neurology, University of Cincinnati, OH
| | - Pasquale Maria Pecoraro
- James J. and Joan A. Gardner Center for Parkinson's Disease and Movement Disorders, Department of Neurology, University of Cincinnati, OH
- Research Unit of Neurology, Neurophysiology and Neurobiology, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Italy
- Operative Research Unit of Neurology, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
| | - Syed A Zaidi
- James J. and Joan A. Gardner Center for Parkinson's Disease and Movement Disorders, Department of Neurology, University of Cincinnati, OH
| | - Kevin R Duque
- James J. and Joan A. Gardner Center for Parkinson's Disease and Movement Disorders, Department of Neurology, University of Cincinnati, OH
| | - Abhimanyu Mahajan
- James J. and Joan A. Gardner Center for Parkinson's Disease and Movement Disorders, Department of Neurology, University of Cincinnati, OH
| | - Mohammad Edrees Mohammad
- James J. and Joan A. Gardner Center for Parkinson's Disease and Movement Disorders, Department of Neurology, University of Cincinnati, OH
- Neurology Department, Cairo University, Egypt; and
| | - Andrew P Duker
- James J. and Joan A. Gardner Center for Parkinson's Disease and Movement Disorders, Department of Neurology, University of Cincinnati, OH
| | - Marcelo A Kauffman
- Consultorio y Laboratorio de Neurogenética, Centro Universitario de Neurología José María Ramos Mejía, Buenos Aires, Argentina
| | - Alberto J Espay
- James J. and Joan A. Gardner Center for Parkinson's Disease and Movement Disorders, Department of Neurology, University of Cincinnati, OH
| | - Luca Marsili
- James J. and Joan A. Gardner Center for Parkinson's Disease and Movement Disorders, Department of Neurology, University of Cincinnati, OH
| |
Collapse
|
7
|
Francisco M, Grau R. Biofilm proficient Bacillus subtilis prevents neurodegeneration in Caenorhabditis elegans Parkinson's disease models via PMK-1/p38 MAPK and SKN-1/Nrf2 signaling. Sci Rep 2025; 15:9864. [PMID: 40118903 PMCID: PMC11928646 DOI: 10.1038/s41598-025-93737-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 03/10/2025] [Indexed: 03/24/2025] Open
Abstract
Parkinson's disease (PD) is a no-curable neurodegenerative disease of pandemic distribution for which only palliative treatments are available. A hallmark of PD is injury to dopaminergic neurons in the substantia nigra pars compacta. Here, we report that Caenorhabditis elegans colonized by biofilm-forming Bacillus subtilis is resistant to injury of dopaminergic neurons caused by treatment with the PD-related neurotoxin 6-hydroxydopamine (6-OHDA). Biofilm-forming B. subtilis-colonized C. elegans display dopamine-dependent behaviors indistinguishable from those of 6-OHDA-untreated worms colonized by gut commensal E. coli OP50. In C. elegans PD model strains with early dopaminergic neuron decay or overexpressing human alpha-synuclein, biofilm-forming B. subtilis colonization had neuroprotective effects and prevents alpha-synulcein aggregation, respectively. The B. subtilis-controlled insulin/IGF-1 signaling (ILS), whose downregulation prevents aging-related PD, is not involved in protecting against 6-OHDA-related injury. We demonstrate that biofilm-forming B. subtilis activates PMK-1 (p38 MAPK)/SKN-1 (Nrf2) signaling, which protects C. elegans from 6-OHDA-induced dopaminergic neuron injury.
Collapse
Affiliation(s)
- Marcos Francisco
- Departamento de Microbiología, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, CONICET-Argentina, Kyojin Laboratories S.A. Castellanos 1335, 2000, Rosario, Santa Fe, Argentina
| | - Roberto Grau
- Departamento de Microbiología, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, CONICET-Argentina, Kyojin Laboratories S.A. Castellanos 1335, 2000, Rosario, Santa Fe, Argentina.
| |
Collapse
|
8
|
Chen L, Wang C, Qin L, Zhang H. Parkinson's disease and glucose metabolism impairment. Transl Neurodegener 2025; 14:10. [PMID: 39962629 PMCID: PMC11831814 DOI: 10.1186/s40035-025-00467-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Accepted: 01/02/2025] [Indexed: 02/21/2025] Open
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disorder. PD patients exhibit varying degrees of abnormal glucose metabolism throughout disease stages. Abnormal glucose metabolism is closely linked to the PD pathogenesis and progression. Key glucose metabolism processes involved in PD include glucose transport, glycolysis, the tricarboxylic acid cycle, oxidative phosphorylation, the pentose phosphate pathway, and gluconeogenesis. Recent studies suggest that glucose metabolism is a potential therapeutic target for PD. In this review, we explore the connection between PD and abnormal glucose metabolism, focusing on the underlying pathophysiological mechanisms. We also summarize potential therapeutic drugs related to glucose metabolism based on results from current cellular and animal model studies.
Collapse
Affiliation(s)
- Liangjing Chen
- Department of Neurology, Second Xiangya Hospital, Central South University, Changsha, 410011, China
- Clinical Medical Research Center for Stroke Prevention and Treatment of Hunan Province, Department of Neurology, Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Chunyu Wang
- Department of Neurology, Second Xiangya Hospital, Central South University, Changsha, 410011, China
- Clinical Medical Research Center for Stroke Prevention and Treatment of Hunan Province, Department of Neurology, Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Lixia Qin
- Department of Neurology, Second Xiangya Hospital, Central South University, Changsha, 410011, China.
- Clinical Medical Research Center for Stroke Prevention and Treatment of Hunan Province, Department of Neurology, Second Xiangya Hospital, Central South University, Changsha, 410011, China.
| | - Hainan Zhang
- Department of Neurology, Second Xiangya Hospital, Central South University, Changsha, 410011, China.
- Clinical Medical Research Center for Stroke Prevention and Treatment of Hunan Province, Department of Neurology, Second Xiangya Hospital, Central South University, Changsha, 410011, China.
| |
Collapse
|
9
|
Hung LY, Wu CS, Chang CJ, Li P, Hicks K, Dibble JJ, Morrison B, Smith CL, Davis RW, Xiao W. A network medicine approach to investigating ME/CFS pathogenesis in severely ill patients: a pilot study. Front Hum Neurosci 2025; 19:1509346. [PMID: 39996021 PMCID: PMC11847890 DOI: 10.3389/fnhum.2025.1509346] [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: 10/10/2024] [Accepted: 01/06/2025] [Indexed: 02/26/2025] Open
Abstract
This pilot study harnessed the power of network medicine to unravel the complex pathogenesis of Myalgic Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS). By utilizing a network analysis on whole genome sequencing (WGS) data from the Severely Ill Patient Study (SIPS), we identified ME/CFS-associated proteins and delineated the corresponding network-level module, termed the SIPS disease module, together with its relevant pathways. This module demonstrated significant overlap with genes implicated in fatigue, cognitive disorders, and neurodegenerative diseases. Our pathway analysis revealed potential associations between ME/CFS and conditions such as COVID-19, Epstein-Barr virus (EBV) infection, neurodegenerative diseases, and pathways involved in cortisol synthesis and secretion, supporting the hypothesis that ME/CFS is a neuroimmune disorder. Additionally, our findings underscore a potential link between ME/CFS and estrogen signaling pathways, which may elucidate the higher prevalence of ME/CFS in females. These findings provide insights into the pathogenesis of ME/CFS from a network medicine perspective and highlight potential therapeutic targets. Further research is needed to validate these findings and explore their implications for improving diagnosis and treatment.
Collapse
Affiliation(s)
- Li-Yuan Hung
- Computational Research Center for Complex Chronic Diseases, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Chan-Shuo Wu
- Computational Research Center for Complex Chronic Diseases, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Chia-Jung Chang
- ME/CFS Collaborative Research Center, Stanford University School of Medicine, Palo Alto, CA, United States
| | - Peng Li
- Computational Research Center for Complex Chronic Diseases, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Kimberly Hicks
- Open Medicine Foundation, Agoura Hills, CA, United States
| | - Joshua J. Dibble
- Computational Research Center for Complex Chronic Diseases, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Braxton Morrison
- Computational Research Center for Complex Chronic Diseases, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Chimere L. Smith
- Patient Led Research Collaborative, Washington, DC, United States
| | - Ronald W. Davis
- ME/CFS Collaborative Research Center, Stanford University School of Medicine, Palo Alto, CA, United States
| | - Wenzhong Xiao
- Computational Research Center for Complex Chronic Diseases, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
10
|
Azarfarin M, Moradikor N, Salatin S, Sarailoo M, Dadkhah M. Stress-related neurodegenerative diseases: Molecular mechanisms implicated in neurodegeneration and therapeutic strategies. PROGRESS IN BRAIN RESEARCH 2025; 291:253-288. [PMID: 40222783 DOI: 10.1016/bs.pbr.2025.01.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/15/2025]
Abstract
Chronic stress is a striking cause of major neurodegenerative diseases disorders (NDDs). These diseases share several common mechanisms regarding to disease pathology, in spite of they have various properties and clinical manifestations. NDDs are defined by progressive cognitive decline, and stress contribute to the promotion and progression of disease. In addition, various pathways such as production of reactive oxygen species (ROS), mitochondrial dysfunction, and neurodegeneration are the main crucial hallmarks to develop common NDDs, resulting in neuronal cell death. Although the exact mechanisms of NDDs are underexplored, the potential neuroprotective critical role of such therapies in neuronal loss the treatment of NDDs are not clear. In this regard, researchers investigate the neuroprotective effects of targeting underlying cascade to introduce a promising therapeutic option to NDDs. Herein, we provide an overview of the role of non-pharmacological treatments against oxidative stress, mitochondrial symbiosis, and neuroinflammation in NDDs, mainly discussing the music, diet, and exercise effects of targeting pathways.
Collapse
Affiliation(s)
- Maryam Azarfarin
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran; Neurosciences Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nasrollah Moradikor
- International Center for Neuroscience Research, Institute for Intelligent Research, Tbilisi, Georgia
| | - Sara Salatin
- Neurosciences Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mehdi Sarailoo
- Pharmaceutical Sciences Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Masoomeh Dadkhah
- Pharmaceutical Sciences Research Center, Ardabil University of Medical Sciences, Ardabil, Iran; Neuroscience Research Group, Pharmaceutical Sciences Research Center, Ardabil University of Medical Sciences, Ardabil, Iran.
| |
Collapse
|
11
|
Tenchov R, Sasso JM, Zhou QA. Evolving Landscape of Parkinson's Disease Research: Challenges and Perspectives. ACS OMEGA 2025; 10:1864-1892. [PMID: 39866628 PMCID: PMC11755173 DOI: 10.1021/acsomega.4c09114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Revised: 12/22/2024] [Accepted: 12/30/2024] [Indexed: 01/28/2025]
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disorder that primarily affects movement. It occurs due to a gradual deficit of dopamine-producing brain cells, particularly in the substantia nigra. The precise etiology of PD is not fully understood, but it likely involves a combination of genetic and environmental factors. The therapies available at present alleviate symptoms but do not stop the disease's advancement. Research endeavors are currently directed at inventing disease-controlling therapies that aim at the inherent mechanisms of PD. PD biomarker breakthroughs hold enormous potential: earlier diagnosis, better monitoring, and targeted treatment based on individual response could significantly improve patient outcomes and ease the burden of this disease. PD research is an active and evolving field, focusing on understanding disease mechanisms, identifying biomarkers, developing new treatments, and improving care. In this report, we explore data from the CAS Content Collection to outline the research progress in PD. We analyze the publication landscape to offer perspective into the latest expertise advancements. Key emerging concepts are reviewed and strategies to fight disease evaluated. Pharmacological targets, genetic risk factors, as well as comorbid diseases are explored, and clinical usage of products against PD with their production pipelines and trials for drug repurposing are examined. This review aims to offer a comprehensive overview of the advancing landscape of the current understanding about PD, to define challenges, and to assess growth prospects to stimulate efforts in battling the disease.
Collapse
Affiliation(s)
- Rumiana Tenchov
- CAS, a division of the American Chemical
Society, Columbus, Ohio 43210, United States
| | - Janet M. Sasso
- CAS, a division of the American Chemical
Society, Columbus, Ohio 43210, United States
| | | |
Collapse
|
12
|
Zheng C, Nguyen KK, Vishnivetskiy SA, Gurevich VV, Gurevich EV. Arrestin-3 binds parkin and enhances parkin-dependent mitophagy. J Neurochem 2025; 169:e16043. [PMID: 38196269 PMCID: PMC11231064 DOI: 10.1111/jnc.16043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 12/12/2023] [Accepted: 12/14/2023] [Indexed: 01/11/2024]
Abstract
Arrestins were discovered for their role in homologous desensitization of G-protein-coupled receptors (GPCRs). Later non-visual arrestins were shown to regulate several signaling pathways. Some of these pathways require arrestin binding to GPCRs, the regulation of others is receptor independent. Here, we demonstrate that arrestin-3 binds the E3 ubiquitin ligase parkin via multiple sites, preferentially interacting with its RING0 domain. Identification of the parkin domains involved suggests that arrestin-3 likely relieves parkin autoinhibition and/or stabilizes the enzymatically active "open" conformation of parkin. Arrestin-3 binding enhances ubiquitination by parkin of the mitochondrial protein mitofusin-1 and facilitates parkin-mediated mitophagy in HeLa cells. Furthermore, arrestin-3 and its mutant with enhanced parkin binding rescue mitofusin-1 ubiquitination and mitophagy in the presence of the Parkinson's disease-associated R275W parkin mutant, which is defective in both functions. Thus, modulation of parkin activity via arrestin-3 might be a novel strategy of anti-parkinsonian therapy.
Collapse
Affiliation(s)
- Chen Zheng
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA
| | - Kevin K. Nguyen
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA
| | | | | | | |
Collapse
|
13
|
Sharma R, Kour A, Dewangan HK. Enhancements in Parkinson's Disease Management: Leveraging Levodopa Optimization and Surgical Breakthroughs. Curr Drug Targets 2025; 26:17-32. [PMID: 39350551 DOI: 10.2174/0113894501319817240919103802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 08/02/2024] [Accepted: 08/13/2024] [Indexed: 02/19/2025]
Abstract
Parkinson's disease (PD) is a complex neurological condition caused due to inheritance, environment, and behavior among various other parameters. The onset, diagnosis, course of therapy, and future of PD are thoroughly examined in this comprehensive review. This review also presents insights into pathogenic mechanisms of reactive microgliosis, Lewy bodies, and their functions in the evolution of PD. It addresses interaction complexity with genetic mutations, especially in genes such as UCH-L1, parkin, and α-synuclein, which illuminates changes in the manner dopaminergic cells handle proteins and use proteases. This raises the improved outcomes and life quality for those with PD. Potential treatments for severe PD include new surgical methods like Deep Brain Stimulation (DBS). Further, exploration of non-motor manifestations, such as cognitive impairment, autonomic dysfunction, and others, is covered in this review article. These symptoms have a significant impact on patients' quality of life. Furthermore, one of the emerging therapeutic routes that are being investigated is neuroprotective medicines that aim to prevent the aggregation of α-synuclein and interventions that modify the progression of diseases. The review concludes by stressing the dynamic nature of PD research and the potential game-changing impact of precision medicines on current approaches to therapy.
Collapse
Affiliation(s)
- Ritika Sharma
- University Institute of Pharma Sciences (UIPS), Chandigarh University NH-95, Chandigarh Ludhiana Highway, Mohali, Punjab, India
| | - Avneet Kour
- Chitkara College of Pharmacy, Chitkara University, Punjab-140401, India
| | - Hitesh Kumar Dewangan
- University Institute of Pharma Sciences (UIPS), Chandigarh University NH-95, Chandigarh Ludhiana Highway, Mohali, Punjab, India
| |
Collapse
|
14
|
Lim SY, Tan AH, Ahmad-Annuar A, Okubadejo NU, Lohmann K, Morris HR, Toh TS, Tay YW, Lange LM, Bandres-Ciga S, Mata I, Foo JN, Sammler E, Ooi JCE, Noyce AJ, Bahr N, Luo W, Ojha R, Singleton AB, Blauwendraat C, Klein C. Uncovering the genetic basis of Parkinson's disease globally: from discoveries to the clinic. Lancet Neurol 2024; 23:1267-1280. [PMID: 39447588 DOI: 10.1016/s1474-4422(24)00378-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 08/29/2024] [Accepted: 09/04/2024] [Indexed: 10/26/2024]
Abstract
Knowledge on the genetic basis of Parkinson's disease has grown tremendously since the discovery of the first monogenic form, caused by a mutation in α-synuclein, and with the subsequent identification of multiple other causative genes and associated loci. Genetic studies provide insights into the phenotypic heterogeneity and global distribution of Parkinson's disease. By shedding light on the underlying biological mechanisms, genetics facilitates the identification of new biomarkers and therapeutic targets. Several clinical trials of genetics-informed therapies are ongoing or imminent. International programmes in populations who have been under-represented in Parkinson's disease genetics research are fostering collaboration and capacity-building, and have already generated novel findings. Many challenges remain for genetics research in these populations, but addressing them provides opportunities to obtain a more complete and equitable understanding of Parkinson's disease globally. These advances facilitate the integration of genetics into the clinic, to improve patient management and personalised medicine.
Collapse
Affiliation(s)
- Shen-Yang Lim
- Division of Neurology, Department of Medicine, and The Mah Pooi Soo & Tan Chin Nam Centre for Parkinson's & Related Disorders, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia.
| | - Ai Huey Tan
- Division of Neurology, Department of Medicine, and The Mah Pooi Soo & Tan Chin Nam Centre for Parkinson's & Related Disorders, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Azlina Ahmad-Annuar
- Department of Biomedical Science, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Njideka Ulunma Okubadejo
- College of Medicine, University of Lagos and Lagos University Teaching Hospital, Idi Araba, Lagos State, Nigeria
| | - Katja Lohmann
- Institute of Neurogenetics, University of Luebeck, Luebeck, Germany
| | - Huw R Morris
- Department of Clinical and Movement Neurosciences, University College London, Institute of Neurology, London, UK
| | - Tzi Shin Toh
- Division of Neurology, Department of Medicine, and The Mah Pooi Soo & Tan Chin Nam Centre for Parkinson's & Related Disorders, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Yi Wen Tay
- Department of Biomedical Science, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Lara M Lange
- Institute of Neurogenetics, University of Luebeck, Luebeck, Germany; Department of Neurology, University of Luebeck and University Hospital Schleswig-Holstein, Luebeck, Germany
| | - Sara Bandres-Ciga
- Center for Alzheimer's and Related Dementias, National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
| | - Ignacio Mata
- Lerner Research Institute, Genomic Medicine, Cleveland Clinic, Cleveland, OH, USA
| | - Jia Nee Foo
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Singapore; Laboratory of Neurogenetics, Genome Institute of Singapore, A*STAR, Singapore
| | - Esther Sammler
- Molecular and Clinical Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK; Medical Research Council Protein Phosphorylation and Ubiquitylation Unit, University of Dundee, Dundee, UK
| | - Joshua Chin Ern Ooi
- Department of Neurology, Queen Elizabeth Hospital, Kota Kinabalu, Sabah, Malaysia
| | - Alastair J Noyce
- Centre for Preventive Neurology, Wolfson Institute of Population Health, Queen Mary University of London, London, UK
| | - Natascha Bahr
- Institute of Neurogenetics, University of Luebeck, Luebeck, Germany; Department of Neurology, University of Luebeck and University Hospital Schleswig-Holstein, Luebeck, Germany
| | - Wei Luo
- Department of Neurology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, People's Republic of China
| | - Rajeev Ojha
- Department of Neurology, Tribhuvan University Teaching Hospital, Kathmandu, Nepal
| | - Andrew B Singleton
- Center for Alzheimer's and Related Dementias, National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA; Laboratory of Neurogenetics, National Institute on Aging, Bethesda, Maryland, USA
| | - Cornelis Blauwendraat
- Center for Alzheimer's and Related Dementias, National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA; Laboratory of Neurogenetics, National Institute on Aging, Bethesda, Maryland, USA
| | - Christine Klein
- Institute of Neurogenetics, University of Luebeck, Luebeck, Germany; Department of Neurology, University of Luebeck and University Hospital Schleswig-Holstein, Luebeck, Germany
| |
Collapse
|
15
|
Poplawska-Domaszewicz K, Qamar MA, Falup Pecurariu C, Chaudhuri KR. Recognition and characterising non-motor profile in early onset Parkinson's disease (EOPD). Parkinsonism Relat Disord 2024; 129:107123. [PMID: 39489627 DOI: 10.1016/j.parkreldis.2024.107123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 08/15/2024] [Accepted: 08/31/2024] [Indexed: 11/05/2024]
Abstract
Early onset Parkinson's disease (EOPD) has been recently defined as a clinical entity with subjects presenting with Parkinson's disease (PD) between the ages of 21-50 and replaces the term Young Onset PD (YOPD). Presentations in this age group are somewhat different to the typical Late Onset sporadic PD (LOPD) and genetic basis may play an important role. The presentations are however, to be differentiated from other causes of juvenile onset or early onset parkinsonism, which are often driven by rare genetic, brain metal deposition, or metabolic progressive disorders with a levolevodopa unresponsive or poorly responsive phenotype. Specific genetic mutations can also underpin EOPD and include nonmotor symptoms of EOPD, which have not been studied extensively. However, some real-life comparator studies with LOPD suggest a nonmotor profile in EOPD dominated by neuropsychiatric symptoms (anxiety), pain, sexual dysfunction, and a higher risk of impulse control disorders and segregation to the recently described noradrenergic and Park-sleep nonmotor endophenotypes may occur. Awareness of the phenotypic variants and nonmotor expression will pave the way for future precision and personalised medicine.
Collapse
Affiliation(s)
- Karolina Poplawska-Domaszewicz
- Department of Neurology, Poznan University of Medical Sciences, 60-355, Poznan, Poland; Parkinson's Foundation Centre of Excellence, King's College Hospital, Denmark Hill, London, SE5 9RS, UK
| | - Mubasher A Qamar
- Parkinson's Foundation Centre of Excellence, King's College Hospital, Denmark Hill, London, SE5 9RS, UK; Basic and Clinical Neuroscience Department, The Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King's College London, 5 Cutcombe Road, London SE5 9RX, UK
| | - Cristian Falup Pecurariu
- Parkinson's Foundation Centre of Excellence, King's College Hospital, Denmark Hill, London, SE5 9RS, UK; Department of Neurology, County Clinic Hospital, Faculty of Medicine, Transilvania University Brasov, 500019, Brasov, Romania; Department of Neurology, Transilvania University Brasov, Brașov, Romania
| | - K Ray Chaudhuri
- Parkinson's Foundation Centre of Excellence, King's College Hospital, Denmark Hill, London, SE5 9RS, UK; Basic and Clinical Neuroscience Department, The Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King's College London, 5 Cutcombe Road, London SE5 9RX, UK.
| |
Collapse
|
16
|
Tan AH, Saffie-Awad P, Schumacher Schuh AF, Lim SY, Madoev H, Ahmad-Annuar A, Solle J, Wegel CE, Doquenia ML, Dey S, Perinan MT, Makarious MB, Fiske B, Morris HR, Noyce AJ, Alcalay RN, Kumar KR, Klein C. Global Perspectives on Returning Genetic Research Results in Parkinson Disease. Neurol Genet 2024; 10:e200213. [PMID: 39807215 PMCID: PMC11727605 DOI: 10.1212/nxg.0000000000200213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Accepted: 10/14/2024] [Indexed: 01/16/2025]
Abstract
Background and Objectives In the era of precision medicine, genetic test results have become increasingly relevant in the care of patients with Parkinson disease (PD). While large research consortia are performing widespread research genetic testing to accelerate discoveries, debate continues about whether, and to what extent, the results should be returned to patients. Ethically, it is imperative to keep participants informed, especially when findings are potentially actionable. However, research testing may not hold the same standards required from clinical diagnostic laboratories and hold significant psychosocial implications. The absence of universally recognized protocols complicates the establishment of appropriate guidelines. Methods Aiming to develop recommendations on return of research results (RoR) practice within the Global Parkinson's Genetics Program (GP2), we conducted a global survey to gain insight on GP2 members' perceptions, practice, readiness, and needs surrounding RoR. Results GP2 members (n = 191), representing 147 institutions and 60 countries across 6 continents, completed the survey. Access to clinical genetic testing services was significantly higher in high-income countries compared with low- and middle-income countries (96.6% vs 58.4%), where funding was predominantly covered by patients themselves. While 92.7% of the respondents agreed that genetic research results should be returned, levels of agreement were higher for clinically relevant results relating to pathogenic or likely pathogenic variants in genes known to cause PD or other neurodegenerative diseases. Less than 10% offered separate clinically accredited genetic testing before returning genetic research results. A total of 48.7% reported having a specific statement on RoR policy in their ethics consent form, while 53.9% collected data on participants' preferences on RoR prospectively. 24.1% had formal genetic counselling training. Notably, the comfort level in returning incidental genetic findings or returning results to unaffected individuals remains low. Discussion Given the differences in resources and training for RoR, as well as ethical and regulatory considerations, tailored approaches are required to ensure equitable access to RoR. Several identified strategies to enhance RoR practices include improving informed consent processes, increasing capacity for genetic counselling including providing counselling toolkits for common genetic variants, broadening access to sustainable clinically accredited testing, building logistical infrastructure for RoR processes, and continuing public and health care education efforts on the important role of genetics in PD.
Collapse
Affiliation(s)
- Ai Huey Tan
- From the Division of Neurology (A.H.T., S.-Y.L.), Department of Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia; Programa de Pós-Graduação em Ciências Médicas da Universidade Federal do Rio Grande do Sul (P.S.-A.), Clínica Santa María, Santiago, Chile; Departamento de Farmacologia (A.F.S.S.), Universidade Federal do Rio Grande do Sul; Serviço de Neurologia (A.F.S.S.), Hospital de Clínicas de Porto Alegre, Brazil; Institute of Neurogenetics (H.M., M.L.D., C.K.), University of Lübeck, Germany; Department of Biomedical Science (A.A.-A.), Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia; The Michael J. Fox Foundation for Parkinson's Research (J.S., B.F.), New York; Department of Medical and Molecular Genetics (C.E.W.), Indiana University, Indianapolis; Department of Neuroscience and Brain Health (M.L.D.), Metropolitan Medical Center, Manila, Philippines; Centre for Preventive Neurology (S.D., M.T.P., A.J.N.), Wolfson Institute of Population Health, Queen Mary University of London, United Kingdom; Unidad de Trastornos del Movimiento (M.T.P.), Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Spain; Laboratory of Neurogenetics (M.B.M.), National Institute on Aging, National Institutes of Health, Bethesda, MD; Department of Clinical and Movement Neurosciences (M.B.M., H.R.M.), UCL Queen Square Institute of Neurology, University College London, United Kingdom; Department of Neurology (R.N.A.), Columbia University Irving Medical Center, New York; Movement Disorders Division (R.N.A.), Neurological Institute, Tel Aviv Sourasky Medical Center and Tel Aviv School of Medicine, Tel Aviv University, Israel; Molecular Medicine Laboratory and Neurology Department (K.R.K.), Concord Clinical School, Concord Repatriation General Hospital, The University of Sydney; Translational Neurogenomics Group (K.R.K.), Genomic and Inherited Disease Program, Garvan Institute of Medical Research; and St Vincent's Healthcare Campus (K.R.K.), Faculty of Medicine, UNSW Sydney, Darlinghurst, New South Wales, Australia
| | - Paula Saffie-Awad
- From the Division of Neurology (A.H.T., S.-Y.L.), Department of Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia; Programa de Pós-Graduação em Ciências Médicas da Universidade Federal do Rio Grande do Sul (P.S.-A.), Clínica Santa María, Santiago, Chile; Departamento de Farmacologia (A.F.S.S.), Universidade Federal do Rio Grande do Sul; Serviço de Neurologia (A.F.S.S.), Hospital de Clínicas de Porto Alegre, Brazil; Institute of Neurogenetics (H.M., M.L.D., C.K.), University of Lübeck, Germany; Department of Biomedical Science (A.A.-A.), Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia; The Michael J. Fox Foundation for Parkinson's Research (J.S., B.F.), New York; Department of Medical and Molecular Genetics (C.E.W.), Indiana University, Indianapolis; Department of Neuroscience and Brain Health (M.L.D.), Metropolitan Medical Center, Manila, Philippines; Centre for Preventive Neurology (S.D., M.T.P., A.J.N.), Wolfson Institute of Population Health, Queen Mary University of London, United Kingdom; Unidad de Trastornos del Movimiento (M.T.P.), Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Spain; Laboratory of Neurogenetics (M.B.M.), National Institute on Aging, National Institutes of Health, Bethesda, MD; Department of Clinical and Movement Neurosciences (M.B.M., H.R.M.), UCL Queen Square Institute of Neurology, University College London, United Kingdom; Department of Neurology (R.N.A.), Columbia University Irving Medical Center, New York; Movement Disorders Division (R.N.A.), Neurological Institute, Tel Aviv Sourasky Medical Center and Tel Aviv School of Medicine, Tel Aviv University, Israel; Molecular Medicine Laboratory and Neurology Department (K.R.K.), Concord Clinical School, Concord Repatriation General Hospital, The University of Sydney; Translational Neurogenomics Group (K.R.K.), Genomic and Inherited Disease Program, Garvan Institute of Medical Research; and St Vincent's Healthcare Campus (K.R.K.), Faculty of Medicine, UNSW Sydney, Darlinghurst, New South Wales, Australia
| | - Artur F Schumacher Schuh
- From the Division of Neurology (A.H.T., S.-Y.L.), Department of Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia; Programa de Pós-Graduação em Ciências Médicas da Universidade Federal do Rio Grande do Sul (P.S.-A.), Clínica Santa María, Santiago, Chile; Departamento de Farmacologia (A.F.S.S.), Universidade Federal do Rio Grande do Sul; Serviço de Neurologia (A.F.S.S.), Hospital de Clínicas de Porto Alegre, Brazil; Institute of Neurogenetics (H.M., M.L.D., C.K.), University of Lübeck, Germany; Department of Biomedical Science (A.A.-A.), Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia; The Michael J. Fox Foundation for Parkinson's Research (J.S., B.F.), New York; Department of Medical and Molecular Genetics (C.E.W.), Indiana University, Indianapolis; Department of Neuroscience and Brain Health (M.L.D.), Metropolitan Medical Center, Manila, Philippines; Centre for Preventive Neurology (S.D., M.T.P., A.J.N.), Wolfson Institute of Population Health, Queen Mary University of London, United Kingdom; Unidad de Trastornos del Movimiento (M.T.P.), Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Spain; Laboratory of Neurogenetics (M.B.M.), National Institute on Aging, National Institutes of Health, Bethesda, MD; Department of Clinical and Movement Neurosciences (M.B.M., H.R.M.), UCL Queen Square Institute of Neurology, University College London, United Kingdom; Department of Neurology (R.N.A.), Columbia University Irving Medical Center, New York; Movement Disorders Division (R.N.A.), Neurological Institute, Tel Aviv Sourasky Medical Center and Tel Aviv School of Medicine, Tel Aviv University, Israel; Molecular Medicine Laboratory and Neurology Department (K.R.K.), Concord Clinical School, Concord Repatriation General Hospital, The University of Sydney; Translational Neurogenomics Group (K.R.K.), Genomic and Inherited Disease Program, Garvan Institute of Medical Research; and St Vincent's Healthcare Campus (K.R.K.), Faculty of Medicine, UNSW Sydney, Darlinghurst, New South Wales, Australia
| | - Shen-Yang Lim
- From the Division of Neurology (A.H.T., S.-Y.L.), Department of Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia; Programa de Pós-Graduação em Ciências Médicas da Universidade Federal do Rio Grande do Sul (P.S.-A.), Clínica Santa María, Santiago, Chile; Departamento de Farmacologia (A.F.S.S.), Universidade Federal do Rio Grande do Sul; Serviço de Neurologia (A.F.S.S.), Hospital de Clínicas de Porto Alegre, Brazil; Institute of Neurogenetics (H.M., M.L.D., C.K.), University of Lübeck, Germany; Department of Biomedical Science (A.A.-A.), Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia; The Michael J. Fox Foundation for Parkinson's Research (J.S., B.F.), New York; Department of Medical and Molecular Genetics (C.E.W.), Indiana University, Indianapolis; Department of Neuroscience and Brain Health (M.L.D.), Metropolitan Medical Center, Manila, Philippines; Centre for Preventive Neurology (S.D., M.T.P., A.J.N.), Wolfson Institute of Population Health, Queen Mary University of London, United Kingdom; Unidad de Trastornos del Movimiento (M.T.P.), Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Spain; Laboratory of Neurogenetics (M.B.M.), National Institute on Aging, National Institutes of Health, Bethesda, MD; Department of Clinical and Movement Neurosciences (M.B.M., H.R.M.), UCL Queen Square Institute of Neurology, University College London, United Kingdom; Department of Neurology (R.N.A.), Columbia University Irving Medical Center, New York; Movement Disorders Division (R.N.A.), Neurological Institute, Tel Aviv Sourasky Medical Center and Tel Aviv School of Medicine, Tel Aviv University, Israel; Molecular Medicine Laboratory and Neurology Department (K.R.K.), Concord Clinical School, Concord Repatriation General Hospital, The University of Sydney; Translational Neurogenomics Group (K.R.K.), Genomic and Inherited Disease Program, Garvan Institute of Medical Research; and St Vincent's Healthcare Campus (K.R.K.), Faculty of Medicine, UNSW Sydney, Darlinghurst, New South Wales, Australia
| | - Harutyun Madoev
- From the Division of Neurology (A.H.T., S.-Y.L.), Department of Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia; Programa de Pós-Graduação em Ciências Médicas da Universidade Federal do Rio Grande do Sul (P.S.-A.), Clínica Santa María, Santiago, Chile; Departamento de Farmacologia (A.F.S.S.), Universidade Federal do Rio Grande do Sul; Serviço de Neurologia (A.F.S.S.), Hospital de Clínicas de Porto Alegre, Brazil; Institute of Neurogenetics (H.M., M.L.D., C.K.), University of Lübeck, Germany; Department of Biomedical Science (A.A.-A.), Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia; The Michael J. Fox Foundation for Parkinson's Research (J.S., B.F.), New York; Department of Medical and Molecular Genetics (C.E.W.), Indiana University, Indianapolis; Department of Neuroscience and Brain Health (M.L.D.), Metropolitan Medical Center, Manila, Philippines; Centre for Preventive Neurology (S.D., M.T.P., A.J.N.), Wolfson Institute of Population Health, Queen Mary University of London, United Kingdom; Unidad de Trastornos del Movimiento (M.T.P.), Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Spain; Laboratory of Neurogenetics (M.B.M.), National Institute on Aging, National Institutes of Health, Bethesda, MD; Department of Clinical and Movement Neurosciences (M.B.M., H.R.M.), UCL Queen Square Institute of Neurology, University College London, United Kingdom; Department of Neurology (R.N.A.), Columbia University Irving Medical Center, New York; Movement Disorders Division (R.N.A.), Neurological Institute, Tel Aviv Sourasky Medical Center and Tel Aviv School of Medicine, Tel Aviv University, Israel; Molecular Medicine Laboratory and Neurology Department (K.R.K.), Concord Clinical School, Concord Repatriation General Hospital, The University of Sydney; Translational Neurogenomics Group (K.R.K.), Genomic and Inherited Disease Program, Garvan Institute of Medical Research; and St Vincent's Healthcare Campus (K.R.K.), Faculty of Medicine, UNSW Sydney, Darlinghurst, New South Wales, Australia
| | - Azlina Ahmad-Annuar
- From the Division of Neurology (A.H.T., S.-Y.L.), Department of Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia; Programa de Pós-Graduação em Ciências Médicas da Universidade Federal do Rio Grande do Sul (P.S.-A.), Clínica Santa María, Santiago, Chile; Departamento de Farmacologia (A.F.S.S.), Universidade Federal do Rio Grande do Sul; Serviço de Neurologia (A.F.S.S.), Hospital de Clínicas de Porto Alegre, Brazil; Institute of Neurogenetics (H.M., M.L.D., C.K.), University of Lübeck, Germany; Department of Biomedical Science (A.A.-A.), Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia; The Michael J. Fox Foundation for Parkinson's Research (J.S., B.F.), New York; Department of Medical and Molecular Genetics (C.E.W.), Indiana University, Indianapolis; Department of Neuroscience and Brain Health (M.L.D.), Metropolitan Medical Center, Manila, Philippines; Centre for Preventive Neurology (S.D., M.T.P., A.J.N.), Wolfson Institute of Population Health, Queen Mary University of London, United Kingdom; Unidad de Trastornos del Movimiento (M.T.P.), Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Spain; Laboratory of Neurogenetics (M.B.M.), National Institute on Aging, National Institutes of Health, Bethesda, MD; Department of Clinical and Movement Neurosciences (M.B.M., H.R.M.), UCL Queen Square Institute of Neurology, University College London, United Kingdom; Department of Neurology (R.N.A.), Columbia University Irving Medical Center, New York; Movement Disorders Division (R.N.A.), Neurological Institute, Tel Aviv Sourasky Medical Center and Tel Aviv School of Medicine, Tel Aviv University, Israel; Molecular Medicine Laboratory and Neurology Department (K.R.K.), Concord Clinical School, Concord Repatriation General Hospital, The University of Sydney; Translational Neurogenomics Group (K.R.K.), Genomic and Inherited Disease Program, Garvan Institute of Medical Research; and St Vincent's Healthcare Campus (K.R.K.), Faculty of Medicine, UNSW Sydney, Darlinghurst, New South Wales, Australia
| | - J Solle
- From the Division of Neurology (A.H.T., S.-Y.L.), Department of Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia; Programa de Pós-Graduação em Ciências Médicas da Universidade Federal do Rio Grande do Sul (P.S.-A.), Clínica Santa María, Santiago, Chile; Departamento de Farmacologia (A.F.S.S.), Universidade Federal do Rio Grande do Sul; Serviço de Neurologia (A.F.S.S.), Hospital de Clínicas de Porto Alegre, Brazil; Institute of Neurogenetics (H.M., M.L.D., C.K.), University of Lübeck, Germany; Department of Biomedical Science (A.A.-A.), Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia; The Michael J. Fox Foundation for Parkinson's Research (J.S., B.F.), New York; Department of Medical and Molecular Genetics (C.E.W.), Indiana University, Indianapolis; Department of Neuroscience and Brain Health (M.L.D.), Metropolitan Medical Center, Manila, Philippines; Centre for Preventive Neurology (S.D., M.T.P., A.J.N.), Wolfson Institute of Population Health, Queen Mary University of London, United Kingdom; Unidad de Trastornos del Movimiento (M.T.P.), Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Spain; Laboratory of Neurogenetics (M.B.M.), National Institute on Aging, National Institutes of Health, Bethesda, MD; Department of Clinical and Movement Neurosciences (M.B.M., H.R.M.), UCL Queen Square Institute of Neurology, University College London, United Kingdom; Department of Neurology (R.N.A.), Columbia University Irving Medical Center, New York; Movement Disorders Division (R.N.A.), Neurological Institute, Tel Aviv Sourasky Medical Center and Tel Aviv School of Medicine, Tel Aviv University, Israel; Molecular Medicine Laboratory and Neurology Department (K.R.K.), Concord Clinical School, Concord Repatriation General Hospital, The University of Sydney; Translational Neurogenomics Group (K.R.K.), Genomic and Inherited Disease Program, Garvan Institute of Medical Research; and St Vincent's Healthcare Campus (K.R.K.), Faculty of Medicine, UNSW Sydney, Darlinghurst, New South Wales, Australia
| | - Claire E Wegel
- From the Division of Neurology (A.H.T., S.-Y.L.), Department of Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia; Programa de Pós-Graduação em Ciências Médicas da Universidade Federal do Rio Grande do Sul (P.S.-A.), Clínica Santa María, Santiago, Chile; Departamento de Farmacologia (A.F.S.S.), Universidade Federal do Rio Grande do Sul; Serviço de Neurologia (A.F.S.S.), Hospital de Clínicas de Porto Alegre, Brazil; Institute of Neurogenetics (H.M., M.L.D., C.K.), University of Lübeck, Germany; Department of Biomedical Science (A.A.-A.), Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia; The Michael J. Fox Foundation for Parkinson's Research (J.S., B.F.), New York; Department of Medical and Molecular Genetics (C.E.W.), Indiana University, Indianapolis; Department of Neuroscience and Brain Health (M.L.D.), Metropolitan Medical Center, Manila, Philippines; Centre for Preventive Neurology (S.D., M.T.P., A.J.N.), Wolfson Institute of Population Health, Queen Mary University of London, United Kingdom; Unidad de Trastornos del Movimiento (M.T.P.), Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Spain; Laboratory of Neurogenetics (M.B.M.), National Institute on Aging, National Institutes of Health, Bethesda, MD; Department of Clinical and Movement Neurosciences (M.B.M., H.R.M.), UCL Queen Square Institute of Neurology, University College London, United Kingdom; Department of Neurology (R.N.A.), Columbia University Irving Medical Center, New York; Movement Disorders Division (R.N.A.), Neurological Institute, Tel Aviv Sourasky Medical Center and Tel Aviv School of Medicine, Tel Aviv University, Israel; Molecular Medicine Laboratory and Neurology Department (K.R.K.), Concord Clinical School, Concord Repatriation General Hospital, The University of Sydney; Translational Neurogenomics Group (K.R.K.), Genomic and Inherited Disease Program, Garvan Institute of Medical Research; and St Vincent's Healthcare Campus (K.R.K.), Faculty of Medicine, UNSW Sydney, Darlinghurst, New South Wales, Australia
| | - Maria Leila Doquenia
- From the Division of Neurology (A.H.T., S.-Y.L.), Department of Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia; Programa de Pós-Graduação em Ciências Médicas da Universidade Federal do Rio Grande do Sul (P.S.-A.), Clínica Santa María, Santiago, Chile; Departamento de Farmacologia (A.F.S.S.), Universidade Federal do Rio Grande do Sul; Serviço de Neurologia (A.F.S.S.), Hospital de Clínicas de Porto Alegre, Brazil; Institute of Neurogenetics (H.M., M.L.D., C.K.), University of Lübeck, Germany; Department of Biomedical Science (A.A.-A.), Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia; The Michael J. Fox Foundation for Parkinson's Research (J.S., B.F.), New York; Department of Medical and Molecular Genetics (C.E.W.), Indiana University, Indianapolis; Department of Neuroscience and Brain Health (M.L.D.), Metropolitan Medical Center, Manila, Philippines; Centre for Preventive Neurology (S.D., M.T.P., A.J.N.), Wolfson Institute of Population Health, Queen Mary University of London, United Kingdom; Unidad de Trastornos del Movimiento (M.T.P.), Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Spain; Laboratory of Neurogenetics (M.B.M.), National Institute on Aging, National Institutes of Health, Bethesda, MD; Department of Clinical and Movement Neurosciences (M.B.M., H.R.M.), UCL Queen Square Institute of Neurology, University College London, United Kingdom; Department of Neurology (R.N.A.), Columbia University Irving Medical Center, New York; Movement Disorders Division (R.N.A.), Neurological Institute, Tel Aviv Sourasky Medical Center and Tel Aviv School of Medicine, Tel Aviv University, Israel; Molecular Medicine Laboratory and Neurology Department (K.R.K.), Concord Clinical School, Concord Repatriation General Hospital, The University of Sydney; Translational Neurogenomics Group (K.R.K.), Genomic and Inherited Disease Program, Garvan Institute of Medical Research; and St Vincent's Healthcare Campus (K.R.K.), Faculty of Medicine, UNSW Sydney, Darlinghurst, New South Wales, Australia
| | - Sumit Dey
- From the Division of Neurology (A.H.T., S.-Y.L.), Department of Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia; Programa de Pós-Graduação em Ciências Médicas da Universidade Federal do Rio Grande do Sul (P.S.-A.), Clínica Santa María, Santiago, Chile; Departamento de Farmacologia (A.F.S.S.), Universidade Federal do Rio Grande do Sul; Serviço de Neurologia (A.F.S.S.), Hospital de Clínicas de Porto Alegre, Brazil; Institute of Neurogenetics (H.M., M.L.D., C.K.), University of Lübeck, Germany; Department of Biomedical Science (A.A.-A.), Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia; The Michael J. Fox Foundation for Parkinson's Research (J.S., B.F.), New York; Department of Medical and Molecular Genetics (C.E.W.), Indiana University, Indianapolis; Department of Neuroscience and Brain Health (M.L.D.), Metropolitan Medical Center, Manila, Philippines; Centre for Preventive Neurology (S.D., M.T.P., A.J.N.), Wolfson Institute of Population Health, Queen Mary University of London, United Kingdom; Unidad de Trastornos del Movimiento (M.T.P.), Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Spain; Laboratory of Neurogenetics (M.B.M.), National Institute on Aging, National Institutes of Health, Bethesda, MD; Department of Clinical and Movement Neurosciences (M.B.M., H.R.M.), UCL Queen Square Institute of Neurology, University College London, United Kingdom; Department of Neurology (R.N.A.), Columbia University Irving Medical Center, New York; Movement Disorders Division (R.N.A.), Neurological Institute, Tel Aviv Sourasky Medical Center and Tel Aviv School of Medicine, Tel Aviv University, Israel; Molecular Medicine Laboratory and Neurology Department (K.R.K.), Concord Clinical School, Concord Repatriation General Hospital, The University of Sydney; Translational Neurogenomics Group (K.R.K.), Genomic and Inherited Disease Program, Garvan Institute of Medical Research; and St Vincent's Healthcare Campus (K.R.K.), Faculty of Medicine, UNSW Sydney, Darlinghurst, New South Wales, Australia
| | - Maria Teresa Perinan
- From the Division of Neurology (A.H.T., S.-Y.L.), Department of Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia; Programa de Pós-Graduação em Ciências Médicas da Universidade Federal do Rio Grande do Sul (P.S.-A.), Clínica Santa María, Santiago, Chile; Departamento de Farmacologia (A.F.S.S.), Universidade Federal do Rio Grande do Sul; Serviço de Neurologia (A.F.S.S.), Hospital de Clínicas de Porto Alegre, Brazil; Institute of Neurogenetics (H.M., M.L.D., C.K.), University of Lübeck, Germany; Department of Biomedical Science (A.A.-A.), Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia; The Michael J. Fox Foundation for Parkinson's Research (J.S., B.F.), New York; Department of Medical and Molecular Genetics (C.E.W.), Indiana University, Indianapolis; Department of Neuroscience and Brain Health (M.L.D.), Metropolitan Medical Center, Manila, Philippines; Centre for Preventive Neurology (S.D., M.T.P., A.J.N.), Wolfson Institute of Population Health, Queen Mary University of London, United Kingdom; Unidad de Trastornos del Movimiento (M.T.P.), Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Spain; Laboratory of Neurogenetics (M.B.M.), National Institute on Aging, National Institutes of Health, Bethesda, MD; Department of Clinical and Movement Neurosciences (M.B.M., H.R.M.), UCL Queen Square Institute of Neurology, University College London, United Kingdom; Department of Neurology (R.N.A.), Columbia University Irving Medical Center, New York; Movement Disorders Division (R.N.A.), Neurological Institute, Tel Aviv Sourasky Medical Center and Tel Aviv School of Medicine, Tel Aviv University, Israel; Molecular Medicine Laboratory and Neurology Department (K.R.K.), Concord Clinical School, Concord Repatriation General Hospital, The University of Sydney; Translational Neurogenomics Group (K.R.K.), Genomic and Inherited Disease Program, Garvan Institute of Medical Research; and St Vincent's Healthcare Campus (K.R.K.), Faculty of Medicine, UNSW Sydney, Darlinghurst, New South Wales, Australia
| | - Mary B Makarious
- From the Division of Neurology (A.H.T., S.-Y.L.), Department of Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia; Programa de Pós-Graduação em Ciências Médicas da Universidade Federal do Rio Grande do Sul (P.S.-A.), Clínica Santa María, Santiago, Chile; Departamento de Farmacologia (A.F.S.S.), Universidade Federal do Rio Grande do Sul; Serviço de Neurologia (A.F.S.S.), Hospital de Clínicas de Porto Alegre, Brazil; Institute of Neurogenetics (H.M., M.L.D., C.K.), University of Lübeck, Germany; Department of Biomedical Science (A.A.-A.), Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia; The Michael J. Fox Foundation for Parkinson's Research (J.S., B.F.), New York; Department of Medical and Molecular Genetics (C.E.W.), Indiana University, Indianapolis; Department of Neuroscience and Brain Health (M.L.D.), Metropolitan Medical Center, Manila, Philippines; Centre for Preventive Neurology (S.D., M.T.P., A.J.N.), Wolfson Institute of Population Health, Queen Mary University of London, United Kingdom; Unidad de Trastornos del Movimiento (M.T.P.), Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Spain; Laboratory of Neurogenetics (M.B.M.), National Institute on Aging, National Institutes of Health, Bethesda, MD; Department of Clinical and Movement Neurosciences (M.B.M., H.R.M.), UCL Queen Square Institute of Neurology, University College London, United Kingdom; Department of Neurology (R.N.A.), Columbia University Irving Medical Center, New York; Movement Disorders Division (R.N.A.), Neurological Institute, Tel Aviv Sourasky Medical Center and Tel Aviv School of Medicine, Tel Aviv University, Israel; Molecular Medicine Laboratory and Neurology Department (K.R.K.), Concord Clinical School, Concord Repatriation General Hospital, The University of Sydney; Translational Neurogenomics Group (K.R.K.), Genomic and Inherited Disease Program, Garvan Institute of Medical Research; and St Vincent's Healthcare Campus (K.R.K.), Faculty of Medicine, UNSW Sydney, Darlinghurst, New South Wales, Australia
| | - Brian Fiske
- From the Division of Neurology (A.H.T., S.-Y.L.), Department of Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia; Programa de Pós-Graduação em Ciências Médicas da Universidade Federal do Rio Grande do Sul (P.S.-A.), Clínica Santa María, Santiago, Chile; Departamento de Farmacologia (A.F.S.S.), Universidade Federal do Rio Grande do Sul; Serviço de Neurologia (A.F.S.S.), Hospital de Clínicas de Porto Alegre, Brazil; Institute of Neurogenetics (H.M., M.L.D., C.K.), University of Lübeck, Germany; Department of Biomedical Science (A.A.-A.), Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia; The Michael J. Fox Foundation for Parkinson's Research (J.S., B.F.), New York; Department of Medical and Molecular Genetics (C.E.W.), Indiana University, Indianapolis; Department of Neuroscience and Brain Health (M.L.D.), Metropolitan Medical Center, Manila, Philippines; Centre for Preventive Neurology (S.D., M.T.P., A.J.N.), Wolfson Institute of Population Health, Queen Mary University of London, United Kingdom; Unidad de Trastornos del Movimiento (M.T.P.), Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Spain; Laboratory of Neurogenetics (M.B.M.), National Institute on Aging, National Institutes of Health, Bethesda, MD; Department of Clinical and Movement Neurosciences (M.B.M., H.R.M.), UCL Queen Square Institute of Neurology, University College London, United Kingdom; Department of Neurology (R.N.A.), Columbia University Irving Medical Center, New York; Movement Disorders Division (R.N.A.), Neurological Institute, Tel Aviv Sourasky Medical Center and Tel Aviv School of Medicine, Tel Aviv University, Israel; Molecular Medicine Laboratory and Neurology Department (K.R.K.), Concord Clinical School, Concord Repatriation General Hospital, The University of Sydney; Translational Neurogenomics Group (K.R.K.), Genomic and Inherited Disease Program, Garvan Institute of Medical Research; and St Vincent's Healthcare Campus (K.R.K.), Faculty of Medicine, UNSW Sydney, Darlinghurst, New South Wales, Australia
| | - Huw R Morris
- From the Division of Neurology (A.H.T., S.-Y.L.), Department of Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia; Programa de Pós-Graduação em Ciências Médicas da Universidade Federal do Rio Grande do Sul (P.S.-A.), Clínica Santa María, Santiago, Chile; Departamento de Farmacologia (A.F.S.S.), Universidade Federal do Rio Grande do Sul; Serviço de Neurologia (A.F.S.S.), Hospital de Clínicas de Porto Alegre, Brazil; Institute of Neurogenetics (H.M., M.L.D., C.K.), University of Lübeck, Germany; Department of Biomedical Science (A.A.-A.), Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia; The Michael J. Fox Foundation for Parkinson's Research (J.S., B.F.), New York; Department of Medical and Molecular Genetics (C.E.W.), Indiana University, Indianapolis; Department of Neuroscience and Brain Health (M.L.D.), Metropolitan Medical Center, Manila, Philippines; Centre for Preventive Neurology (S.D., M.T.P., A.J.N.), Wolfson Institute of Population Health, Queen Mary University of London, United Kingdom; Unidad de Trastornos del Movimiento (M.T.P.), Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Spain; Laboratory of Neurogenetics (M.B.M.), National Institute on Aging, National Institutes of Health, Bethesda, MD; Department of Clinical and Movement Neurosciences (M.B.M., H.R.M.), UCL Queen Square Institute of Neurology, University College London, United Kingdom; Department of Neurology (R.N.A.), Columbia University Irving Medical Center, New York; Movement Disorders Division (R.N.A.), Neurological Institute, Tel Aviv Sourasky Medical Center and Tel Aviv School of Medicine, Tel Aviv University, Israel; Molecular Medicine Laboratory and Neurology Department (K.R.K.), Concord Clinical School, Concord Repatriation General Hospital, The University of Sydney; Translational Neurogenomics Group (K.R.K.), Genomic and Inherited Disease Program, Garvan Institute of Medical Research; and St Vincent's Healthcare Campus (K.R.K.), Faculty of Medicine, UNSW Sydney, Darlinghurst, New South Wales, Australia
| | - Alastair J Noyce
- From the Division of Neurology (A.H.T., S.-Y.L.), Department of Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia; Programa de Pós-Graduação em Ciências Médicas da Universidade Federal do Rio Grande do Sul (P.S.-A.), Clínica Santa María, Santiago, Chile; Departamento de Farmacologia (A.F.S.S.), Universidade Federal do Rio Grande do Sul; Serviço de Neurologia (A.F.S.S.), Hospital de Clínicas de Porto Alegre, Brazil; Institute of Neurogenetics (H.M., M.L.D., C.K.), University of Lübeck, Germany; Department of Biomedical Science (A.A.-A.), Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia; The Michael J. Fox Foundation for Parkinson's Research (J.S., B.F.), New York; Department of Medical and Molecular Genetics (C.E.W.), Indiana University, Indianapolis; Department of Neuroscience and Brain Health (M.L.D.), Metropolitan Medical Center, Manila, Philippines; Centre for Preventive Neurology (S.D., M.T.P., A.J.N.), Wolfson Institute of Population Health, Queen Mary University of London, United Kingdom; Unidad de Trastornos del Movimiento (M.T.P.), Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Spain; Laboratory of Neurogenetics (M.B.M.), National Institute on Aging, National Institutes of Health, Bethesda, MD; Department of Clinical and Movement Neurosciences (M.B.M., H.R.M.), UCL Queen Square Institute of Neurology, University College London, United Kingdom; Department of Neurology (R.N.A.), Columbia University Irving Medical Center, New York; Movement Disorders Division (R.N.A.), Neurological Institute, Tel Aviv Sourasky Medical Center and Tel Aviv School of Medicine, Tel Aviv University, Israel; Molecular Medicine Laboratory and Neurology Department (K.R.K.), Concord Clinical School, Concord Repatriation General Hospital, The University of Sydney; Translational Neurogenomics Group (K.R.K.), Genomic and Inherited Disease Program, Garvan Institute of Medical Research; and St Vincent's Healthcare Campus (K.R.K.), Faculty of Medicine, UNSW Sydney, Darlinghurst, New South Wales, Australia
| | - Roy N Alcalay
- From the Division of Neurology (A.H.T., S.-Y.L.), Department of Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia; Programa de Pós-Graduação em Ciências Médicas da Universidade Federal do Rio Grande do Sul (P.S.-A.), Clínica Santa María, Santiago, Chile; Departamento de Farmacologia (A.F.S.S.), Universidade Federal do Rio Grande do Sul; Serviço de Neurologia (A.F.S.S.), Hospital de Clínicas de Porto Alegre, Brazil; Institute of Neurogenetics (H.M., M.L.D., C.K.), University of Lübeck, Germany; Department of Biomedical Science (A.A.-A.), Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia; The Michael J. Fox Foundation for Parkinson's Research (J.S., B.F.), New York; Department of Medical and Molecular Genetics (C.E.W.), Indiana University, Indianapolis; Department of Neuroscience and Brain Health (M.L.D.), Metropolitan Medical Center, Manila, Philippines; Centre for Preventive Neurology (S.D., M.T.P., A.J.N.), Wolfson Institute of Population Health, Queen Mary University of London, United Kingdom; Unidad de Trastornos del Movimiento (M.T.P.), Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Spain; Laboratory of Neurogenetics (M.B.M.), National Institute on Aging, National Institutes of Health, Bethesda, MD; Department of Clinical and Movement Neurosciences (M.B.M., H.R.M.), UCL Queen Square Institute of Neurology, University College London, United Kingdom; Department of Neurology (R.N.A.), Columbia University Irving Medical Center, New York; Movement Disorders Division (R.N.A.), Neurological Institute, Tel Aviv Sourasky Medical Center and Tel Aviv School of Medicine, Tel Aviv University, Israel; Molecular Medicine Laboratory and Neurology Department (K.R.K.), Concord Clinical School, Concord Repatriation General Hospital, The University of Sydney; Translational Neurogenomics Group (K.R.K.), Genomic and Inherited Disease Program, Garvan Institute of Medical Research; and St Vincent's Healthcare Campus (K.R.K.), Faculty of Medicine, UNSW Sydney, Darlinghurst, New South Wales, Australia
| | - Kishore Raj Kumar
- From the Division of Neurology (A.H.T., S.-Y.L.), Department of Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia; Programa de Pós-Graduação em Ciências Médicas da Universidade Federal do Rio Grande do Sul (P.S.-A.), Clínica Santa María, Santiago, Chile; Departamento de Farmacologia (A.F.S.S.), Universidade Federal do Rio Grande do Sul; Serviço de Neurologia (A.F.S.S.), Hospital de Clínicas de Porto Alegre, Brazil; Institute of Neurogenetics (H.M., M.L.D., C.K.), University of Lübeck, Germany; Department of Biomedical Science (A.A.-A.), Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia; The Michael J. Fox Foundation for Parkinson's Research (J.S., B.F.), New York; Department of Medical and Molecular Genetics (C.E.W.), Indiana University, Indianapolis; Department of Neuroscience and Brain Health (M.L.D.), Metropolitan Medical Center, Manila, Philippines; Centre for Preventive Neurology (S.D., M.T.P., A.J.N.), Wolfson Institute of Population Health, Queen Mary University of London, United Kingdom; Unidad de Trastornos del Movimiento (M.T.P.), Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Spain; Laboratory of Neurogenetics (M.B.M.), National Institute on Aging, National Institutes of Health, Bethesda, MD; Department of Clinical and Movement Neurosciences (M.B.M., H.R.M.), UCL Queen Square Institute of Neurology, University College London, United Kingdom; Department of Neurology (R.N.A.), Columbia University Irving Medical Center, New York; Movement Disorders Division (R.N.A.), Neurological Institute, Tel Aviv Sourasky Medical Center and Tel Aviv School of Medicine, Tel Aviv University, Israel; Molecular Medicine Laboratory and Neurology Department (K.R.K.), Concord Clinical School, Concord Repatriation General Hospital, The University of Sydney; Translational Neurogenomics Group (K.R.K.), Genomic and Inherited Disease Program, Garvan Institute of Medical Research; and St Vincent's Healthcare Campus (K.R.K.), Faculty of Medicine, UNSW Sydney, Darlinghurst, New South Wales, Australia
| | - Christine Klein
- From the Division of Neurology (A.H.T., S.-Y.L.), Department of Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia; Programa de Pós-Graduação em Ciências Médicas da Universidade Federal do Rio Grande do Sul (P.S.-A.), Clínica Santa María, Santiago, Chile; Departamento de Farmacologia (A.F.S.S.), Universidade Federal do Rio Grande do Sul; Serviço de Neurologia (A.F.S.S.), Hospital de Clínicas de Porto Alegre, Brazil; Institute of Neurogenetics (H.M., M.L.D., C.K.), University of Lübeck, Germany; Department of Biomedical Science (A.A.-A.), Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia; The Michael J. Fox Foundation for Parkinson's Research (J.S., B.F.), New York; Department of Medical and Molecular Genetics (C.E.W.), Indiana University, Indianapolis; Department of Neuroscience and Brain Health (M.L.D.), Metropolitan Medical Center, Manila, Philippines; Centre for Preventive Neurology (S.D., M.T.P., A.J.N.), Wolfson Institute of Population Health, Queen Mary University of London, United Kingdom; Unidad de Trastornos del Movimiento (M.T.P.), Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Spain; Laboratory of Neurogenetics (M.B.M.), National Institute on Aging, National Institutes of Health, Bethesda, MD; Department of Clinical and Movement Neurosciences (M.B.M., H.R.M.), UCL Queen Square Institute of Neurology, University College London, United Kingdom; Department of Neurology (R.N.A.), Columbia University Irving Medical Center, New York; Movement Disorders Division (R.N.A.), Neurological Institute, Tel Aviv Sourasky Medical Center and Tel Aviv School of Medicine, Tel Aviv University, Israel; Molecular Medicine Laboratory and Neurology Department (K.R.K.), Concord Clinical School, Concord Repatriation General Hospital, The University of Sydney; Translational Neurogenomics Group (K.R.K.), Genomic and Inherited Disease Program, Garvan Institute of Medical Research; and St Vincent's Healthcare Campus (K.R.K.), Faculty of Medicine, UNSW Sydney, Darlinghurst, New South Wales, Australia
| |
Collapse
|
17
|
Gonul Oner O, Biboulet Bruneau C, Fraix V, Bourg V, Defebvre L, Mutez E, Roze E, Laroche C, Béreau M, Nguyen-Morel MA, Moro E. Pediatric-onset PRKN disease: New insights into an understudied population. JOURNAL OF PARKINSON'S DISEASE 2024; 14:1631-1641. [PMID: 39957199 DOI: 10.1177/1877718x241296153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/18/2025]
Abstract
BACKGROUND In pediatric age, the PRKN mutation is reported as one of the most common genetic causes of Parkinson's disease. However, detailed clinical data on PRKN patients with pediatric onset are scarce. OBJECTIVE To describe clinical characteristics, disease progression, and management of PRKN patients with pediatric onset. METHODS PRKN patients with onset of clinical signs before the age of 18 years were included in this retrospective multicenter study. Collected data included detailed clinical characteristics, progression, and disease management. Data presentation is descriptive due to the sample size. RESULTS Nine patients (five females) were included from five French movement disorders centers. The mean age at symptom onset was 10.78 ± 2.22 years (median, 11; range, 7-14). Dystonia was the first most common motor symptom (six patients). The mean time from symptom onset to genetic diagnosis was 13.33 ± 9.21 years (median, 11; range, 3-32). The most commonly reported non-motor symptoms were sleep disorders (seven patients), anxiety (six patients), and depression (five patients). The first treatment was L-dopa in four patients, dopamine agonist in two, carbamazepine in two, and rasagiline in one. Dyskinesia and impulse control disorders were the most common treatment-related side effects (nine and six patients, respectively). Four patients underwent deep brain stimulation surgery. The last available follow-up was at 27.22 ± 14.05 years (median, 28; range, 6-56) after the diagnosis. CONCLUSIONS This is the first study reporting detailed clinical features and long-term management of PRKN patients with pediatric onset. Prompt diagnosis and appropriate treatment strategies are important to optimize disease management.
Collapse
Affiliation(s)
- Ozge Gonul Oner
- Université Grenoble Alpes, Inserm, Grenoble Institute Neurosciences, Grenoble, France
- Movement Disorders Unit, Division of Neurology, Centre Hospitalier Universitaire de Grenoble, Grenoble, France
| | - Céline Biboulet Bruneau
- Service de Neurologie Pédiatrique, Hôpital Couple Mère Enfant, Centre Hospitalier Universitaire de Grenoble, Grenoble, France
| | - Valérie Fraix
- Movement Disorders Unit, Division of Neurology, Centre Hospitalier Universitaire de Grenoble, Grenoble, France
| | - Véronique Bourg
- Service De Médecine Physique Et Réadaptation Pédiatrique, Hôpital Couple-Enfant, Centre Hospitalier Universitaire de Grenoble, Grenoble, France
| | - Luc Defebvre
- Université de Lille, Inserm, CHU Lille, U1172 - LilNCog - Lille Neuroscience & Cognition, Lille, France
- Department of Neurology and Movement Disorders, CHU de Lille, Lille, France
| | - Eugénie Mutez
- Université de Lille, Inserm, CHU Lille, U1172 - LilNCog - Lille Neuroscience & Cognition, Lille, France
- Department of Neurology and Movement Disorders, CHU de Lille, Lille, France
| | - Emmanuel Roze
- Neurology Department, Pitié-Salpêtrière Hospital, Paris, France
- Paris Brain Institute, Sorbonne University, Paris, France
| | - Cécile Laroche
- Department of Pediatric, Hôpital mère enfant, Limoges, France
| | - Matthieu Béreau
- Department of Neurology, Centre Hospitalier Universitaire de Besançon, France
| | - Marie-Ange Nguyen-Morel
- Service de Neurologie Pédiatrique, Hôpital Couple Mère Enfant, Centre Hospitalier Universitaire de Grenoble, Grenoble, France
| | - Elena Moro
- Université Grenoble Alpes, Inserm, Grenoble Institute Neurosciences, Grenoble, France
- Movement Disorders Unit, Division of Neurology, Centre Hospitalier Universitaire de Grenoble, Grenoble, France
| |
Collapse
|
18
|
Ye X, Kotaru S, Lopes R, Cravens S, Lasagna M, Wand AJ. Cooperative Substructure and Energetics of Allosteric Regulation of the Catalytic Core of the E3 Ubiquitin Ligase Parkin by Phosphorylated Ubiquitin. Biomolecules 2024; 14:1338. [PMID: 39456270 PMCID: PMC11506642 DOI: 10.3390/biom14101338] [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: 08/20/2024] [Revised: 10/13/2024] [Accepted: 10/14/2024] [Indexed: 10/28/2024] Open
Abstract
Mutations in the parkin gene product Parkin give rise to autosomal recessive juvenile parkinsonism. Parkin is an E3 ubiquitin ligase that is a critical participant in the process of mitophagy. Parkin has a complex structure that integrates several allosteric signals to maintain precise control of its catalytic activity. Though its allosterically controlled structural reorganization has been extensively characterized by crystallography, the energetics and mechanisms of allosteric regulation of Parkin are much less well understood. Allostery is fundamentally linked to the energetics of the cooperative (sub)structure of the protein. Herein, we examine the mechanism of allosteric activation by phosphorylated ubiquitin binding to the enzymatic core of Parkin, which lacks the antagonistic Ubl domain. In this way, the allosteric effects of the agonist phosphorylated ubiquitin can be isolated. Using native-state hydrogen exchange monitored by mass spectrometry, we find that the five structural domains of the core of Parkin are energetically distinct. Nevertheless, association of phosphorylated ubiquitin destabilizes structural elements that bind the ubiquitin-like domain antagonist while promoting the dissociation of the catalytic domain and energetically poises the protein for transition to the fully activated structure.
Collapse
Affiliation(s)
- Xiang Ye
- Department of Biochemistry & Biophysics, Texas A&M University, College Station, TX 77843, USA
- Department of Biochemistry & Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Sravya Kotaru
- Department of Biochemistry & Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Rosana Lopes
- Department of Biochemistry & Biophysics, Texas A&M University, College Station, TX 77843, USA
| | - Shannen Cravens
- Department of Biochemistry & Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Chemistry and Biochemistry, Gonzaga University, Spokane, WA 99258, USA
| | - Mauricio Lasagna
- Department of Biochemistry & Biophysics, Texas A&M University, College Station, TX 77843, USA
| | - A. Joshua Wand
- Department of Biochemistry & Biophysics, Texas A&M University, College Station, TX 77843, USA
- Department of Biochemistry & Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Chemistry, Texas A&M University, College Station, TX 77843, USA
| |
Collapse
|
19
|
Shih JY, Hsu YHH. Peptide Activator Stabilizes DJ-1 Structure and Enhances Its Activity. Int J Mol Sci 2024; 25:11075. [PMID: 39456860 PMCID: PMC11508141 DOI: 10.3390/ijms252011075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Revised: 10/09/2024] [Accepted: 10/11/2024] [Indexed: 10/28/2024] Open
Abstract
DJ-1 is a vital enzyme involved in the maintenance of mitochondrial health, and its mutation has been associated with an increased risk of Parkinson's disease (PD). Effective regulation of DJ-1 activity is essential for the well-being of mitochondria, and DJ-1 is thus a potential target for PD drug development. In this study, two peptides (15EEMETIIPVDVMRRA29 and 47SRDVVICPDA56) were utilized with the aim of enhancing the activity of DJ-1. The mechanisms underlying the activity enhancement by these two peptides were investigated using hydrogen/deuterium exchange mass spectrometry (HDXMS). The HDXMS results revealed distinct mechanisms. Peptide 1 obstructs the access of solvent to the dimer interface and stabilizes the α/β hydrolase structure, facilitating substrate binding to a stabilized active site. Conversely, peptide 2 induces a destabilization of the α/β hydrolase core, enhancing substrate accessibility and subsequently increasing DJ-1 activity. The binding of these two peptides optimizes the activity site within the dimeric structure. These findings offer valuable insights into the mechanisms underlying the activity enhancement of DJ-1 by the two peptides, potentially aiding the development of new drugs that can enhance the activity of DJ-1 and, consequently, advance PD treatment.
Collapse
Affiliation(s)
| | - Yuan-Hao Howard Hsu
- Department of Chemistry, Tunghai University, No. 1727, Sec. 4, Taiwan Boulevard, Xitun District, Taichung 40704, Taiwan;
| |
Collapse
|
20
|
Liu Y, Zhou X, Chen C, Li X, Pan T, Liu Z, Wu D, Chen X. Association between osteoarthritis with Parkinson's disease in the US (NHANES 2011-2020). Front Neurosci 2024; 18:1393740. [PMID: 39234184 PMCID: PMC11371570 DOI: 10.3389/fnins.2024.1393740] [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: 02/29/2024] [Accepted: 08/06/2024] [Indexed: 09/06/2024] Open
Abstract
Objected To evaluate the association between osteoarthritis (OA) and Parkinson's disease (PD) in adults in the United States. Methods Using 2011-2020 NHANES data, a cross-sectional study of 11,117 adults over the age of 40 was conducted. Univariate logistic regression and multivariate logistic regression were used to analyze the relationship between arthritis and PD. In addition, stratified analysis was used to examine whether the relationship between arthritis and PD was interactive with age, gender, race, education, BMI. Results In this study, a total of 11,117 participants were included, and we found that osteoarthritis was positively correlated with the development of PD compared with non-arthritis patients [1.95 (1.44 ~ 2.62)] (p < 0.001). After adjusting the covariates, the results are still stable. Conclusion PD patients were positively correlated with OA. Among people with OA, there was a 95% increased risk of PD compared to people without arthritis. Therefore, when treating OA, attention should be paid to the increased risk of PD. In the meantime, further studies are needed to explore the link between OA and PD patients.
Collapse
Affiliation(s)
- Yang Liu
- Changchun University of Chinese Medicine, Changchun, China
| | - Xue Zhou
- Changchun University of Chinese Medicine, Changchun, China
| | - Chunhai Chen
- The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, China
| | - Xuefeng Li
- Changchun University of Chinese Medicine, Changchun, China
| | - Ting Pan
- The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, China
| | - Ziqi Liu
- Changchun University of Chinese Medicine, Changchun, China
| | - Dalong Wu
- The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, China
| | - Xinhua Chen
- The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, China
| |
Collapse
|
21
|
Rubilar JC, Outeiro TF, Klein AD. The lysosomal β-glucocerebrosidase strikes mitochondria: implications for Parkinson's therapeutics. Brain 2024; 147:2610-2620. [PMID: 38437875 DOI: 10.1093/brain/awae070] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 01/31/2024] [Accepted: 02/14/2024] [Indexed: 03/06/2024] Open
Abstract
Parkinson's disease is a neurodegenerative disorder primarily known for typical motor features that arise due to the loss of dopaminergic neurons in the substantia nigra. However, the precise molecular aetiology of the disease is still unclear. Several cellular pathways have been linked to Parkinson's disease, including the autophagy-lysosome pathway, α-synuclein aggregation and mitochondrial function. Interestingly, the mechanistic link between GBA1, the gene that encodes for lysosomal β-glucocerebrosidase (GCase), and Parkinson's disease lies in the interplay between GCase functions in the lysosome and mitochondria. GCase mutations alter mitochondria-lysosome contact sites. In the lysosome, reduced GCase activity leads to glycosphingolipid build-up, disrupting lysosomal function and autophagy, thereby triggering α-synuclein accumulation. Additionally, α-synuclein aggregates reduce GCase activity, creating a self-perpetuating cycle of lysosomal dysfunction and α-synuclein accumulation. GCase can also be imported into the mitochondria, where it promotes the integrity and function of mitochondrial complex I. Thus, GCase mutations that impair its normal function increase oxidative stress in mitochondria, the compartment where dopamine is oxidized. In turn, the accumulation of oxidized dopamine adducts further impairs GCase activity, creating a second cycle of GCase dysfunction. The oxidative state triggered by GCase dysfunction can also induce mitochondrial DNA damage which, in turn, can cause dopaminergic cell death. In this review, we highlight the pivotal role of GCase in Parkinson's disease pathogenesis and discuss promising examples of GCase-based therapeutics, such as gene and enzyme replacement therapies, small molecule chaperones and substrate reduction therapies, among others, as potential therapeutic interventions.
Collapse
Affiliation(s)
- Juan Carlos Rubilar
- Centro de Genética y Genómica, Facultad de Medicina, Clínica Alemana Universidad del Desarrollo, Santiago 7780272, Chile
| | - Tiago Fleming Outeiro
- Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, 37073, Göttingen, Germany
- Max Planck Institute for Natural Sciences, 37073, Göttingen, Germany
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Framlington Place, Newcastle Upon Tyne, NE2 4HH, UK
- Scientific Employee with an Honorary Contract at Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), 37075, Göttingen, Germany
| | - Andrés D Klein
- Centro de Genética y Genómica, Facultad de Medicina, Clínica Alemana Universidad del Desarrollo, Santiago 7780272, Chile
| |
Collapse
|
22
|
Daida K, Yoshino H, Malik L, Baker B, Ishiguro M, Genner R, Paquette K, Li Y, Nishioka K, Masuzugawa S, Hirano M, Takahashi K, Kolmogolv M, Billingsley KJ, Funayama M, Blauwendraat C, Hattori N. The Utility of Long-Read Sequencing in Diagnosing Genetic Autosomal Recessive Parkinson's Disease: a genetic screening study. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.06.14.24308784. [PMID: 39108517 PMCID: PMC11302705 DOI: 10.1101/2024.06.14.24308784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 08/10/2024]
Abstract
Background Mutations within the genes PRKN and PINK1 are the leading cause of early onset autosomal recessive Parkinson's disease (PD). However, the genetic cause of most early-onset PD (EOPD) cases still remains unresolved. Long-read sequencing has successfully identified many pathogenic structural variants that cause disease, but this technology has not been widely applied to PD. We recently identified the genetic cause of EOPD in a pair of monozygotic twins by uncovering a complex structural variant that spans over 7 Mb, utilizing Oxford Nanopore Technologies (ONT) long-read sequencing. In this study, we aimed to expand on this and assess whether a second variant could be detected with ONT long-read sequencing in other unresolved EOPD cases reported to carry one heterozygous variant in PRKN or PINK1. Methods ONT long-read sequencing was performed on patients with one reported PRKN/PINK1 pathogenic variant. EOPD patients with an age at onset younger than 50 were included in this study. As a positive control, we also included EOPD patients who had already been identified to carry two known PRKN pathogenic variants. Initial genetic testing was performed using either short-read targeted panel sequencing for single nucleotide variants and multiplex ligation-dependent probe amplification (MLPA) for copy number variants. Results 48 patients were included in this study (PRKN "one-variant" n = 24, PINK1 "one-variant" n = 12, PRKN "two-variants" n = 12). Using ONT long-read sequencing, we detected a second pathogenic variant in six PRKN "one-variant" patients (26%, 6/23) but none in the PINK1 "one-variant" patients (0%, 0/12). Long-read sequencing identified one case with a complex inversion, two instances of structural variant overlap, and three cases of duplication. In addition, in the positive control PRKN "two-variants" group, we were able to identify both pathogenic variants in PRKN in all the patients (100%, 12/12). Conclusions This data highlights that ONT long-read sequencing is a powerful tool to identify a pathogenic structural variant at the PRKN locus that is often missed by conventional methods. Therefore, for cases where conventional methods fail to detect a second variant for EOPD, long-read sequencing should be considered as an alternative and complementary approach.
Collapse
Affiliation(s)
- Kensuke Daida
- Integrative Neurogenomics Unit, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
- Center for Alzheimer's and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
- Department of Neurology, Faculty of Medicine, Juntendo University, Tokyo, Japan
| | - Hiroyo Yoshino
- Research Institute for Diseases of Old Age, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Laksh Malik
- Center for Alzheimer's and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Breeana Baker
- Center for Alzheimer's and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Mayu Ishiguro
- Department of Neurology, Faculty of Medicine, Juntendo University, Tokyo, Japan
| | - Rylee Genner
- Center for Alzheimer's and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Kimberly Paquette
- Center for Alzheimer's and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Yuanzhe Li
- Department of Neurology, Faculty of Medicine, Juntendo University, Tokyo, Japan
- Department of Diagnosis, Prevention and Treatment of Dementia, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Kenya Nishioka
- Department of Neurology, Juntendo Tokyo Koto Geriatric Medical Center, Koto-ku, Tokyo, Japan
| | | | - Makito Hirano
- Department of Neurology, Kindai University Faculty of Medicine, Osaka, Japan
| | - Kenta Takahashi
- Division of Neurology and Gerontology, Department of Internal Medicine, School of Medicine, Iwate Medical University, Morioka, Japan
| | - Mikhail Kolmogolv
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Kimberley J Billingsley
- Molecular Genetics Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - Manabu Funayama
- Department of Neurology, Faculty of Medicine, Juntendo University, Tokyo, Japan
- Research Institute for Diseases of Old Age, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Cornelis Blauwendraat
- Integrative Neurogenomics Unit, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
- Center for Alzheimer's and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Nobutaka Hattori
- Department of Neurology, Faculty of Medicine, Juntendo University, Tokyo, Japan
- Research Institute for Diseases of Old Age, Graduate School of Medicine, Juntendo University, Tokyo, Japan
- Neurodegenerative Disorders Collaborative Laboratory, RIKEN Center for Brain Science, Wako, Saitama, Japan
| |
Collapse
|
23
|
Zhao Y, Lin M, Zhai F, Chen J, Jin X. Exploring the Role of Ubiquitin-Proteasome System in the Pathogenesis of Parkinson's Disease. Pharmaceuticals (Basel) 2024; 17:782. [PMID: 38931449 PMCID: PMC11207014 DOI: 10.3390/ph17060782] [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: 04/24/2024] [Revised: 05/23/2024] [Accepted: 06/08/2024] [Indexed: 06/28/2024] Open
Abstract
Parkinson's disease (PD) is a prevalent neurodegenerative disorder among the elderly population. The pathogenesis of PD encompasses genetic alterations, environmental factors, and age-related neurodegenerative processes. Numerous studies have demonstrated that aberrant functioning of the ubiquitin-proteasome system (UPS) plays a crucial role in the initiation and progression of PD. Notably, E3 ubiquitin ligases serve as pivotal components determining substrate specificity within UPS and are intimately associated with the regulation of various proteins implicated in PD pathology. This review comprehensively summarizes the mechanisms by which E3 ubiquitin ligases and deubiquitinating enzymes modulate PD-associated proteins and signaling pathways, while exploring the intricate relationship between UPS dysfunctions and PD etiology. Furthermore, this article discusses recent research advancements regarding inhibitors targeting PD-related E3 ubiquitin ligases.
Collapse
Affiliation(s)
- Yiting Zhao
- Department of Chemoradiotherapy, The Affiliated People’s Hospital of Ningbo University, Ningbo 315040, China; (Y.Z.); (M.L.)
- Department of Ultrasound Medicine, The Affiliated People’s Hospital of Ningbo University, Ningbo 315040, China
- Zhejiang Key Laboratory of Pathophysiology, Department of Biochemistry and Molecular Biology, Health Science Center of Ningbo University, Ningbo 315211, China;
| | - Man Lin
- Department of Chemoradiotherapy, The Affiliated People’s Hospital of Ningbo University, Ningbo 315040, China; (Y.Z.); (M.L.)
- Zhejiang Key Laboratory of Pathophysiology, Department of Biochemistry and Molecular Biology, Health Science Center of Ningbo University, Ningbo 315211, China;
| | - Fengguang Zhai
- Zhejiang Key Laboratory of Pathophysiology, Department of Biochemistry and Molecular Biology, Health Science Center of Ningbo University, Ningbo 315211, China;
| | - Jun Chen
- Department of Chemoradiotherapy, The Affiliated People’s Hospital of Ningbo University, Ningbo 315040, China; (Y.Z.); (M.L.)
- Zhejiang Key Laboratory of Pathophysiology, Department of Biochemistry and Molecular Biology, Health Science Center of Ningbo University, Ningbo 315211, China;
| | - Xiaofeng Jin
- Department of Chemoradiotherapy, The Affiliated People’s Hospital of Ningbo University, Ningbo 315040, China; (Y.Z.); (M.L.)
- Zhejiang Key Laboratory of Pathophysiology, Department of Biochemistry and Molecular Biology, Health Science Center of Ningbo University, Ningbo 315211, China;
| |
Collapse
|
24
|
Cucinotta L, Mannino D, Filippone A, Romano A, Esposito E, Paterniti I. The role of autophagy in Parkinson's disease: a gender difference overview. Front Pharmacol 2024; 15:1408152. [PMID: 38933683 PMCID: PMC11199695 DOI: 10.3389/fphar.2024.1408152] [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: 03/27/2024] [Accepted: 05/21/2024] [Indexed: 06/28/2024] Open
Abstract
Recent studies have demonstrated dysregulation of the autophagy pathway in patients with Parkinson's disease (PD) and in animal models of PD, highlighting its emerging role in disease. In particular, several studies indicate that autophagy, which is an essential degradative process for the damaged protein homeostasis and the management of cell balance, can manifest significant variations according to gender. While some evidence suggests increased autophagic activation in men with PD, women may have distinct regulatory patterns. In this review, we examined the existing literature on gender differences in PD-associated autophagic processes, focusing on the autophagy related proteins (ATGs) and leucine rich repeat kinase 2 (LRRK2) genes. Also, this review would suggest that an in-depth understanding of these gender differences in autophagic processes could open new perspectives for personalized therapeutic strategies, promoting more effective and targeted management of PD.
Collapse
Affiliation(s)
- Laura Cucinotta
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| | - Deborah Mannino
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| | - Alessia Filippone
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| | - Adele Romano
- Department of Physiology and Pharmacology “V. Erspamer”, Sapienza University of Rome, Rome, Italy
| | - Emanuela Esposito
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| | - Irene Paterniti
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| |
Collapse
|
25
|
Trease AJ, Totusek S, Lichter EZ, Stauch KL, Fox HS. Mitochondrial DNA Instability Supersedes Parkin Mutations in Driving Mitochondrial Proteomic Alterations and Functional Deficits in Polg Mutator Mice. Int J Mol Sci 2024; 25:6441. [PMID: 38928146 PMCID: PMC11203920 DOI: 10.3390/ijms25126441] [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: 04/19/2024] [Revised: 05/30/2024] [Accepted: 06/07/2024] [Indexed: 06/28/2024] Open
Abstract
Mitochondrial quality control is essential in mitochondrial function. To examine the importance of Parkin-dependent mechanisms in mitochondrial quality control, we assessed the impact of modulating Parkin on proteome flux and mitochondrial function in a context of reduced mtDNA fidelity. To accomplish this, we crossed either the Parkin knockout mouse or ParkinW402A knock-in mouse lines to the Polg mitochondrial mutator line to generate homozygous double mutants. In vivo longitudinal isotopic metabolic labeling was followed by isolation of liver mitochondria and synaptic terminals from the brain, which are rich in mitochondria. Mass spectrometry and bioenergetics analysis were assessed. We demonstrate that slower mitochondrial protein turnover is associated with loss of mtDNA fidelity in liver mitochondria but not synaptic terminals, and bioenergetic function in both tissues is impaired. Pathway analysis revealed loss of mtDNA fidelity is associated with disturbances of key metabolic pathways, consistent with its association with metabolic disorders and neurodegeneration. Furthermore, we find that loss of Parkin leads to exacerbation of Polg-driven proteomic consequences, though it may be bioenergetically protective in tissues exhibiting rapid mitochondrial turnover. Finally, we provide evidence that, surprisingly, dis-autoinhibition of Parkin (ParkinW402A) functionally resembles Parkin knockout and fails to rescue deleterious Polg-driven effects. Our study accomplishes three main outcomes: (1) it supports recent studies suggesting that Parkin dependence is low in response to an increased mtDNA mutational load, (2) it provides evidence of a potential protective role of Parkin insufficiency, and (3) it draws into question the therapeutic attractiveness of enhancing Parkin function.
Collapse
Affiliation(s)
- Andrew J. Trease
- Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, NE 68198, USA; (A.J.T.); (S.T.); (K.L.S.)
| | - Steven Totusek
- Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, NE 68198, USA; (A.J.T.); (S.T.); (K.L.S.)
| | - Eliezer Z. Lichter
- Computational Biomedicine Section, Department of Medicine, Boston University School of Medicine, Boston, MA 02118, USA;
| | - Kelly L. Stauch
- Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, NE 68198, USA; (A.J.T.); (S.T.); (K.L.S.)
| | - Howard S. Fox
- Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, NE 68198, USA; (A.J.T.); (S.T.); (K.L.S.)
| |
Collapse
|
26
|
Clausen L, Okarmus J, Voutsinos V, Meyer M, Lindorff-Larsen K, Hartmann-Petersen R. PRKN-linked familial Parkinson's disease: cellular and molecular mechanisms of disease-linked variants. Cell Mol Life Sci 2024; 81:223. [PMID: 38767677 PMCID: PMC11106057 DOI: 10.1007/s00018-024-05262-8] [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: 02/27/2024] [Revised: 04/25/2024] [Accepted: 05/02/2024] [Indexed: 05/22/2024]
Abstract
Parkinson's disease (PD) is a common and incurable neurodegenerative disorder that arises from the loss of dopaminergic neurons in the substantia nigra and is mainly characterized by progressive loss of motor function. Monogenic familial PD is associated with highly penetrant variants in specific genes, notably the PRKN gene, where homozygous or compound heterozygous loss-of-function variants predominate. PRKN encodes Parkin, an E3 ubiquitin-protein ligase important for protein ubiquitination and mitophagy of damaged mitochondria. Accordingly, Parkin plays a central role in mitochondrial quality control but is itself also subject to a strict protein quality control system that rapidly eliminates certain disease-linked Parkin variants. Here, we summarize the cellular and molecular functions of Parkin, highlighting the various mechanisms by which PRKN gene variants result in loss-of-function. We emphasize the importance of high-throughput assays and computational tools for the clinical classification of PRKN gene variants and how detailed insights into the pathogenic mechanisms of PRKN gene variants may impact the development of personalized therapeutics.
Collapse
Affiliation(s)
- Lene Clausen
- Department of Biology, Linderstrøm-Lang Centre for Protein Science, University of Copenhagen, 2200, Copenhagen, Denmark
| | - Justyna Okarmus
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, 5230, Odense, Denmark
| | - Vasileios Voutsinos
- Department of Biology, Linderstrøm-Lang Centre for Protein Science, University of Copenhagen, 2200, Copenhagen, Denmark
| | - Morten Meyer
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, 5230, Odense, Denmark
- Department of Neurology, Odense University Hospital, 5000, Odense, Denmark
- Department of Clinical Research, BRIDGE, Brain Research Inter Disciplinary Guided Excellence, University of Southern Denmark, 5230, Odense, Denmark
| | - Kresten Lindorff-Larsen
- Department of Biology, Linderstrøm-Lang Centre for Protein Science, University of Copenhagen, 2200, Copenhagen, Denmark
| | - Rasmus Hartmann-Petersen
- Department of Biology, Linderstrøm-Lang Centre for Protein Science, University of Copenhagen, 2200, Copenhagen, Denmark.
| |
Collapse
|
27
|
Cogan G, Daida K, Billingsley KJ, Tesson C, Forlani S, Jornea L, Arnaud L, Tissier L, LeGuern E, Singleton AB, Ferrien M, Gervais Bernard H, Lesage S, Blauwendraat C, Brice A. Long-read sequencing unravels the complexity of structural variants in PRKN in two individuals with early-onset Parkinson's disease. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.05.02.24306523. [PMID: 38746197 PMCID: PMC11092742 DOI: 10.1101/2024.05.02.24306523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Background PRKN biallelic pathogenic variants are the most common cause of autosomal recessive early-onset Parkinson's disease (PD). However, the variants responsible for suspected PRKN- PD individuals are not always identified with standard genetic testing. Objectives Identify the genetic cause in two siblings with a PRKN -PD phenotype using long-read sequencing (LRS). Methods The genetic investigation involved standard testing using successively multiple ligation probe amplification (MLPA), Sanger sequencing, targeted sequencing, whole-exome sequencing and LRS. Results MLPA and targeted sequencing identified one copy of exon four in PRKN but no other variants were identified. Subsequently, LRS unveiled a large deletion encompassing exon 3 to 4 on one allele and a duplication of exon 3 on the second allele; explaining the siblings' phenotype. MLPA could not identify the balanced rearrangement of exon 3. Conclusions This study highlights the potential utility of long-read sequencing in the context of unsolved typical PRKN- PD individuals.
Collapse
|
28
|
Slade L, Deane CS, Szewczyk NJ, Etheridge T, Whiteman M. Hydrogen sulfide supplementation as a potential treatment for primary mitochondrial diseases. Pharmacol Res 2024; 203:107180. [PMID: 38599468 DOI: 10.1016/j.phrs.2024.107180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 04/06/2024] [Accepted: 04/06/2024] [Indexed: 04/12/2024]
Abstract
Primary mitochondrial diseases (PMD) are amongst the most common inborn errors of metabolism causing fatal outcomes within the first decade of life. With marked heterogeneity in both inheritance patterns and physiological manifestations, these conditions present distinct challenges for targeted drug therapy, where effective therapeutic countermeasures remain elusive within the clinic. Hydrogen sulfide (H2S)-based therapeutics may offer a new option for patient treatment, having been proposed as a conserved mitochondrial substrate and post-translational regulator across species, displaying therapeutic effects in age-related mitochondrial dysfunction and neurodegenerative models of mitochondrial disease. H2S can stimulate mitochondrial respiration at sites downstream of common PMD-defective subunits, augmenting energy production, mitochondrial function and reducing cell death. Here, we highlight the primary signalling mechanisms of H2S in mitochondria relevant for PMD and outline key cytoprotective proteins/pathways amenable to post-translational restoration via H2S-mediated persulfidation. The mechanisms proposed here, combined with the advent of potent mitochondria-targeted sulfide delivery molecules, could provide a framework for H2S as a countermeasure for PMD disease progression.
Collapse
Affiliation(s)
- Luke Slade
- University of Exeter Medical School, University of Exeter, St. Luke's Campus, Exeter EX1 2LU, UK; Leibniz-Institut für Analytische Wissenschaften-ISAS-e.V, Dortmund, Germany
| | - Colleen S Deane
- Human Development & Health, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, UK
| | - Nathaniel J Szewczyk
- Medical Research Council Versus Arthritis Centre for Musculoskeletal Ageing Research, Royal Derby Hospital, University of Nottingham, Derby DE22 3DT, United Kingdom; Ohio Musculoskeletal and Neurologic Institute, Heritage College of Osteopathic Medicine, Ohio University, Athens, Ohio 45701, Greece
| | - Timothy Etheridge
- Public Health and Sport Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter EX1 2LU, United Kingdom.
| | - Matthew Whiteman
- University of Exeter Medical School, University of Exeter, St. Luke's Campus, Exeter EX1 2LU, UK.
| |
Collapse
|
29
|
Asimakidou E, Xiromerisiou G, Sidiropoulos C. Motor and Non-motor Outcomes of Deep Brain Stimulation across the Genetic Panorama of Parkinson's Disease: A Multi-Scale Meta-Analysis. Mov Disord Clin Pract 2024; 11:465-477. [PMID: 38318989 PMCID: PMC11078493 DOI: 10.1002/mdc3.13994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 01/12/2024] [Accepted: 01/21/2024] [Indexed: 02/07/2024] Open
Abstract
BACKGROUND In the era of modern medicine, where high-throughput sequencing techniques are readily available, it is desirable to elucidate the role of genetic background in patients with Parkinson's Disease (PD) undergoing Deep Brain Stimulation (DBS). Genetic stratification of PD patients undergoing DBS may assist in patient selection and prediction of clinical outcomes and complement existing selection procedures such as levodopa challenge testing. OBJECTIVE To capture a broad spectrum of motor and non-motor DBS outcomes in genetic PD patients with data from the recently updated literature. METHODS A multi-scale meta-analysis with 380 genetic PD cases was conducted using the Cochrane Review Manager, JASP software and R. RESULTS This meta-analysis revealed that overall, patients with genetic PD are good candidates for DBS but the outcomes might differ depending on the presence of specific mutations. PRKN carriers benefited the most regarding motor function, daily dose medication and motor complications. However, GBA carriers appeared to be more prone to cognitive decline after subthalamic nucleus DBS accompanied by a low quality of life with variable severity depending on genetic variants and concomitant alterations in other genes. Apart from GBA, cognitive worsening was also observed in SNCA carriers. Pre-operative levodopa responsiveness and a younger age of onset are associated with a favorable motor outcome. CONCLUSION A personalized approach with a variant-based risk stratification within the emerging field of surgicogenomics is needed. Integration of polygenic risk scores in clinical-decision making should be encouraged.
Collapse
|
30
|
Yan Y, Zhang M, Ren W, Zheng X, Chang Y. Neuromelanin-sensitive magnetic resonance imaging: Possibilities and promises as an imaging biomarker for Parkinson's disease. Eur J Neurosci 2024; 59:2616-2627. [PMID: 38441250 DOI: 10.1111/ejn.16296] [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: 09/23/2023] [Revised: 02/03/2024] [Accepted: 02/07/2024] [Indexed: 05/22/2024]
Abstract
Parkinson's disease (PD) is an age-related progressive neurodegenerative disorder characterized by both motor and non-motor symptoms resulting from the death of dopaminergic neurons in the substantia nigra pars compacta (SNpc) and noradrenergic neurons in the locus coeruleus (LC). The current diagnosis of PD primarily relies on motor symptoms, often leading to diagnoses in advanced stages, where a significant portion of SNpc dopamine neurons has already succumbed. Therefore, the identification of imaging biomarkers for early-stage PD diagnosis and disease progression monitoring is imperative. Recent studies propose that neuromelanin-sensitive magnetic resonance imaging (NM-MRI) holds promise as an imaging biomarker. In this review, we summarize the latest findings concerning NM-MRI characteristics at various stages in patients with PD and those with atypical parkinsonism. In conclusion, alterations in neuromelanin within the LC are associated with non-motor symptoms and prove to be a reliable imaging biomarker in the prodromal phase of PD. Furthermore, NM-MRI demonstrates efficacy in differentiating progressive supranuclear palsy (PSP) from PD and multiple system atrophy with predominant parkinsonism. The spatial patterns of changes in the SNpc can be indicative of PD progression and aid in distinguishing between PSP and synucleinopathies. We recommend that patients with PD and individuals at risk for PD undergo regular NM-MRI examinations. This technology holds the potential for widespread use in PD diagnosis.
Collapse
Affiliation(s)
- Yayun Yan
- Department of Neurology, China-Japan Union Hospital, Jilin University, Changchun, China
| | - Mengchao Zhang
- Department of Radiology, China-Japan Union Hospital, Jilin University, Changchun, China
| | - Wenhua Ren
- Department of Neurology, China-Japan Union Hospital, Jilin University, Changchun, China
| | - Xiaoqi Zheng
- Department of Neurology, China-Japan Union Hospital, Jilin University, Changchun, China
| | - Ying Chang
- Department of Neurology, China-Japan Union Hospital, Jilin University, Changchun, China
| |
Collapse
|
31
|
Filograna R, Gerlach J, Choi HN, Rigoni G, Barbaro M, Oscarson M, Lee S, Tiklova K, Ringnér M, Koolmeister C, Wibom R, Riggare S, Nennesmo I, Perlmann T, Wredenberg A, Wedell A, Motori E, Svenningsson P, Larsson NG. PARKIN is not required to sustain OXPHOS function in adult mammalian tissues. NPJ Parkinsons Dis 2024; 10:93. [PMID: 38684669 PMCID: PMC11058849 DOI: 10.1038/s41531-024-00707-0] [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: 09/12/2023] [Accepted: 04/11/2024] [Indexed: 05/02/2024] Open
Abstract
Loss-of-function variants in the PRKN gene encoding the ubiquitin E3 ligase PARKIN cause autosomal recessive early-onset Parkinson's disease (PD). Extensive in vitro and in vivo studies have reported that PARKIN is involved in multiple pathways of mitochondrial quality control, including mitochondrial degradation and biogenesis. However, these findings are surrounded by substantial controversy due to conflicting experimental data. In addition, the existing PARKIN-deficient mouse models have failed to faithfully recapitulate PD phenotypes. Therefore, we have investigated the mitochondrial role of PARKIN during ageing and in response to stress by employing a series of conditional Parkin knockout mice. We report that PARKIN loss does not affect oxidative phosphorylation (OXPHOS) capacity and mitochondrial DNA (mtDNA) levels in the brain, heart, and skeletal muscle of aged mice. We also demonstrate that PARKIN deficiency does not exacerbate the brain defects and the pro-inflammatory phenotype observed in mice carrying high levels of mtDNA mutations. To rule out compensatory mechanisms activated during embryonic development of Parkin-deficient mice, we generated a mouse model where loss of PARKIN was induced in adult dopaminergic (DA) neurons. Surprisingly, also these mice did not show motor impairment or neurodegeneration, and no major transcriptional changes were found in isolated midbrain DA neurons. Finally, we report a patient with compound heterozygous PRKN pathogenic variants that lacks PARKIN and has developed PD. The PARKIN deficiency did not impair OXPHOS activities or induce mitochondrial pathology in skeletal muscle from the patient. Altogether, our results argue that PARKIN is dispensable for OXPHOS function in adult mammalian tissues.
Collapse
Affiliation(s)
- Roberta Filograna
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden.
| | - Jule Gerlach
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Hae-Na Choi
- Institute for Biochemistry, University of Cologne, Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Giovanni Rigoni
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Michela Barbaro
- Centre for Inherited Metabolic Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Mikael Oscarson
- Centre for Inherited Metabolic Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Seungmin Lee
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Katarina Tiklova
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Markus Ringnér
- Department of Biology, National Bioinformatics Infrastructure Sweden, Science for Life Laboratory, Lund University, Lund, Sweden
| | - Camilla Koolmeister
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Rolf Wibom
- Centre for Inherited Metabolic Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Sara Riggare
- Department of Women's and Children's Health, Uppsala University, Uppsala, Sweden
| | - Inger Nennesmo
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Thomas Perlmann
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Anna Wredenberg
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
- Centre for Inherited Metabolic Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Anna Wedell
- Centre for Inherited Metabolic Diseases, Karolinska University Hospital, Stockholm, Sweden
- Department of Molecular Medicine and Surgery, Karolinska Institute, Stockholm, Sweden
| | - Elisa Motori
- Institute for Biochemistry, University of Cologne, Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Per Svenningsson
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Department of Neurology, Karolinska University Hospital, Stockholm, Sweden
| | - Nils-Göran Larsson
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
32
|
Bej E, Cesare P, Volpe AR, d’Angelo M, Castelli V. Oxidative Stress and Neurodegeneration: Insights and Therapeutic Strategies for Parkinson's Disease. Neurol Int 2024; 16:502-517. [PMID: 38804477 PMCID: PMC11130796 DOI: 10.3390/neurolint16030037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 04/25/2024] [Accepted: 04/26/2024] [Indexed: 05/29/2024] Open
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative condition marked by the gradual deterioration of dopaminergic neurons in the substantia nigra. Oxidative stress has been identified as a key player in the development of PD in recent studies. In the first part, we discuss the sources of oxidative stress in PD, including mitochondrial dysfunction, dopamine metabolism, and neuroinflammation. This paper delves into the possibility of mitigating oxidative stress as a potential treatment approach for PD. In addition, we examine the hurdles and potential of antioxidant therapy, including the challenge of delivering antioxidants to the brain and the requirement for biomarkers to track oxidative stress in PD patients. However, even if antioxidant therapy holds promise, further investigation is needed to determine its efficacy and safety in PD treatment.
Collapse
Affiliation(s)
| | | | | | | | - Vanessa Castelli
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (E.B.); (P.C.); (A.R.V.); (M.d.)
| |
Collapse
|
33
|
Papagiannakis N, Liu H, Koros C, Simitsi AM, Stamelou M, Maniati M, Buena-Atienza E, Kartanou C, Karadima G, Makrythanasis P, Vatsellas G, Valente EM, Gasser T, Stefanis L. Parkin mRNA Expression Levels in Peripheral Blood Mononuclear Cells in Parkin-Related Parkinson's Disease. Mov Disord 2024; 39:715-722. [PMID: 38357851 DOI: 10.1002/mds.29739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 01/17/2024] [Accepted: 01/22/2024] [Indexed: 02/16/2024] Open
Abstract
INTRODUCTION Pathogenic variants in parkin (PRKN gene) are the second most prevalent known monogenic cause of Parkinson's disease (PD). How monoallelic or biallelic pathogenic variants in the PRKN gene may affect its transcription in patient-derived biological material has not been systematically studied. METHODS PRKN mRNA expression levels were measured with real-time polymerase chain reaction (RT-PCR) in peripheral blood mononuclear cells (PBMCs). PBMCs were derived from PRKN-mutated PD patients (PRKN-PD) (n = 12), sporadic PD (sPD) (n = 21) and healthy controls (n = 21). Six of the PRKN-PD patients were heterozygous, four were compound heterozygous, and two were homozygous for PRKN variants. RESULTS A statistically significant decrease in PRKN expression levels was present, compared to healthy controls and sPD, in heterozygous (P = 0.019 and 0.031 respectively) and biallelic (P < 0.001 for both) PRKN-PD. PRKN expression levels in biallelic PD patients were uniformly very low and were reduced, albeit not significantly, compared to heterozygotes. Based on receiver operating characteristic analysis, low PRKN expression levels were a sensitive and extremely specific indicator for the presence of PRKN pathogenic variants. CONCLUSIONS Assessment of PRKN mRNA levels in PBMCs may be a useful way to screen for biallelic pathogenic variants in the PRKN gene. Suspicion for certain variants in a heterozygous state may also be raised based on low PRKN mRNA levels. © 2024 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Nikolaos Papagiannakis
- Center of Clinical Research, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
- 1st Department of Neurology, Eginitio Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Hui Liu
- Department of Neurodegenerative Diseases, Hertie Center for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Christos Koros
- 1st Department of Neurology, Eginitio Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Athina-Maria Simitsi
- 1st Department of Neurology, Eginitio Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Maria Stamelou
- 1st Department of Neurology, Eginitio Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Matina Maniati
- Center of Clinical Research, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Elena Buena-Atienza
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany
- NGS Competence Center Tübingen, Tübingen, Germany
| | - Chrysoula Kartanou
- 1st Department of Neurology, Eginitio Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Georgia Karadima
- 1st Department of Neurology, Eginitio Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Periklis Makrythanasis
- Laboratory of Medical Genetics, Medical School, National and Kapodistrian University of Athens, Athens, Greece
- Department of Genetic Medicine and Development, Medical School, University of Geneva, Geneva, Switzerland
- Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Giannis Vatsellas
- Center of Basic Research, Biomedical Research Foundation, Academy of Athens, Athens, Greece
| | - Enza Maria Valente
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
- Neurogenetics Research Center, IRCCS Mondino Foundation, Pavia, Italy
| | - Thomas Gasser
- Department of Neurodegenerative Diseases, Hertie Center for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Leonidas Stefanis
- Center of Clinical Research, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
- 1st Department of Neurology, Eginitio Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
34
|
Menon PJ, Sambin S, Criniere-Boizet B, Courtin T, Tesson C, Casse F, Ferrien M, Mariani LL, Carvalho S, Lejeune FX, Rebbah S, Martet G, Houot M, Lanore A, Mangone G, Roze E, Vidailhet M, Aasly J, Gan Or Z, Yu E, Dauvilliers Y, Zimprich A, Tomantschger V, Pirker W, Álvarez I, Pastor P, Di Fonzo A, Bhatia KP, Magrinelli F, Houlden H, Real R, Quattrone A, Limousin P, Korlipara P, Foltynie T, Grosset D, Williams N, Narendra D, Lin HP, Jovanovic C, Svetel M, Lynch T, Gallagher A, Vandenberghe W, Gasser T, Brockmann K, Morris HR, Borsche M, Klein C, Corti O, Brice A, Lesage S, Corvol JC. Genotype-phenotype correlation in PRKN-associated Parkinson's disease. NPJ Parkinsons Dis 2024; 10:72. [PMID: 38553467 PMCID: PMC10980707 DOI: 10.1038/s41531-024-00677-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 03/07/2024] [Indexed: 04/02/2024] Open
Abstract
Bi-allelic pathogenic variants in PRKN are the most common cause of autosomal recessive Parkinson's disease (PD). 647 patients with PRKN-PD were included in this international study. The pathogenic variants present were characterised and investigated for their effect on phenotype. Clinical features and progression of PRKN-PD was also assessed. Among 133 variants in index cases (n = 582), there were 58 (43.6%) structural variants, 34 (25.6%) missense, 20 (15%) frameshift, 10 splice site (7.5%%), 9 (6.8%) nonsense and 2 (1.5%) indels. The most frequent variant overall was an exon 3 deletion (n = 145, 12.3%), followed by the p.R275W substitution (n = 117, 10%). Exon3, RING0 protein domain and the ubiquitin-like protein domain were mutational hotspots with 31%, 35.4% and 31.7% of index cases presenting mutations in these regions respectively. The presence of a frameshift or structural variant was associated with a 3.4 ± 1.6 years or a 4.7 ± 1.6 years earlier age at onset of PRKN-PD respectively (p < 0.05). Furthermore, variants located in the N-terminus of the protein, a region enriched with frameshift variants, were associated with an earlier age at onset. The phenotype of PRKN-PD was characterised by slow motor progression, preserved cognition, an excellent motor response to levodopa therapy and later development of motor complications compared to early-onset PD. Non-motor symptoms were however common in PRKN-PD. Our findings on the relationship between the type of variant in PRKN and the phenotype of the disease may have implications for both genetic counselling and the design of precision clinical trials.
Collapse
Affiliation(s)
- Poornima Jayadev Menon
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, Paris, France.
- Assistance Publique Hôpitaux de Paris, Department of Neurology, CIC Neurosciences, Hôpital Pitié-Salpêtrière, Paris, France.
- School of Postgraduate Studies, Royal College of Surgeons in Ireland, Dublin, Ireland.
| | - Sara Sambin
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, Paris, France
- Assistance Publique Hôpitaux de Paris, Department of Neurology, CIC Neurosciences, Hôpital Pitié-Salpêtrière, Paris, France
| | - Baptiste Criniere-Boizet
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, Paris, France
| | - Thomas Courtin
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, Paris, France
- Assistance Publique Hôpitaux de Paris, Department of Genetics, Hôpital Pitié-Salpêtrière, Paris, France
| | - Christelle Tesson
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, Paris, France
| | - Fanny Casse
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, Paris, France
| | - Melanie Ferrien
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, Paris, France
| | - Louise-Laure Mariani
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, Paris, France
- Assistance Publique Hôpitaux de Paris, Department of Neurology, CIC Neurosciences, Hôpital Pitié-Salpêtrière, Paris, France
| | - Stephanie Carvalho
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, Paris, France
| | - Francois-Xavier Lejeune
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, Paris, France
| | - Sana Rebbah
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, Paris, France
| | - Gaspard Martet
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, Paris, France
| | - Marion Houot
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, Paris, France
- Assistance Publique Hôpitaux de Paris, Department of Neurology, CIC Neurosciences, Hôpital Pitié-Salpêtrière, Paris, France
- Centre of Excellence of Neurodegenerative Disease (CoEN), AP-HP, Pitié-Salpêtrière Hospital, Paris, France
- Institute of Memory and Alzheimer's Disease (IM2A), Department of Neurology, AP-HP, Pitié-Salpêtrière Hospital, Paris, France
| | - Aymeric Lanore
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, Paris, France
- Assistance Publique Hôpitaux de Paris, Department of Neurology, CIC Neurosciences, Hôpital Pitié-Salpêtrière, Paris, France
| | - Graziella Mangone
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, Paris, France
- Assistance Publique Hôpitaux de Paris, Department of Neurology, CIC Neurosciences, Hôpital Pitié-Salpêtrière, Paris, France
- Department of Neurology, Movement Disorder Division, Rush University Medical Center, 1725 W. Harrison Street, Chicago, IL, USA
| | - Emmanuel Roze
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, Paris, France
- Assistance Publique Hôpitaux de Paris, Department of Neurology, CIC Neurosciences, Hôpital Pitié-Salpêtrière, Paris, France
| | - Marie Vidailhet
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, Paris, France
- Assistance Publique Hôpitaux de Paris, Department of Neurology, CIC Neurosciences, Hôpital Pitié-Salpêtrière, Paris, France
| | - Jan Aasly
- Department of Neurology, Norwegian University of Science and Technology, Trondheim, Norway
| | - Ziv Gan Or
- The Neuro (Montreal Neurological Institute-Hospital), McGill University, Montreal, QC, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
- Department of Human Genetics, McGill University, Montreal, QC, Canada
| | - Eric Yu
- The Neuro (Montreal Neurological Institute-Hospital), McGill University, Montreal, QC, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
- Department of Human Genetics, McGill University, Montreal, QC, Canada
| | - Yves Dauvilliers
- Department of Neurology, Gui-de-Chauliac Hospital, CHU Montpellier, University of Montpellier, Institute for Neurosciences of Montpellier (INM), INSERM, Montpellier, France
| | | | | | - Walter Pirker
- Department of Neurology, Ottakring Clinic, Vienna, Austria
| | - Ignacio Álvarez
- Department of Neurology, Hospital Universitari Mutua de Terrassa, and Fundació per a la Recerca Biomèdica i Social Mútua de Terrassa, Terrassa, Barcelona, Spain
| | - Pau Pastor
- Unit of Neurodegenerative diseases, Department of Neurology, University Hospital Germans Trias i Pujol and The Germans Trias i Pujol Research Institute (IGTP) Badalona, Barcelona, Spain
| | - Alessio Di Fonzo
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Kailash P Bhatia
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Francesca Magrinelli
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Henry Houlden
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, UK
| | - Raquel Real
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, University College London, London, UK
- UCL Movement Disorders Centre, University College London, London, UK
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
| | - Andrea Quattrone
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, University College London, London, UK
- Institute of Neurology, Department of Medical and Surgical Sciences, Magna Graecia University of Catanzaro, Catanzaro, Italy
| | - Patricia Limousin
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Prasad Korlipara
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Thomas Foltynie
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Donald Grosset
- Institute of Neurological Sciences, University of Glasgow, Glasgow, UK
| | - Nigel Williams
- Department of Psychological Medicine and Neurology, Cardiff University, Cardiff, UK
| | - Derek Narendra
- Inherited Disorders Unit, Neurogenetics Branch, Division of Intramural Research, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Hsin-Pin Lin
- Inherited Disorders Unit, Neurogenetics Branch, Division of Intramural Research, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Carna Jovanovic
- University Clinical Center of Serbia, Neurology Clinic, Belgrade, Serbia
| | - Marina Svetel
- University Clinical Center of Serbia, Neurology Clinic, Belgrade, Serbia
| | - Timothy Lynch
- The Dublin Neurological Institute at the Mater Misericordiae University Hospital, Dublin Ireland and University College Dublin, Dublin, Ireland
| | - Amy Gallagher
- The Dublin Neurological Institute at the Mater Misericordiae University Hospital, Dublin Ireland and University College Dublin, Dublin, Ireland
| | - Wim Vandenberghe
- Department of Neurology, University Hospitals Leuven; Department of Neurosciences, KU Leuven; Leuven Brain Institute, Leuven, Belgium
| | - Thomas Gasser
- Department of Neurodegenerative Diseases, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
- DZNE, German Center for Neurodegenerative Diseases, Tübingen, Germany
| | - Kathrin Brockmann
- Department of Neurodegenerative Diseases, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
- DZNE, German Center for Neurodegenerative Diseases, Tübingen, Germany
| | - Huw R Morris
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Max Borsche
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
| | - Christine Klein
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
| | - Olga Corti
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, Paris, France
| | - Alexis Brice
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, Paris, France
- Assistance Publique Hôpitaux de Paris, Department of Genetics, Hôpital Pitié-Salpêtrière, Paris, France
| | - Suzanne Lesage
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, Paris, France
| | - Jean Christophe Corvol
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, Paris, France
- Assistance Publique Hôpitaux de Paris, Department of Neurology, CIC Neurosciences, Hôpital Pitié-Salpêtrière, Paris, France
| |
Collapse
|
35
|
Trevisan L, Gaudio A, Monfrini E, Avanzino L, Di Fonzo A, Mandich P. Genetics in Parkinson's disease, state-of-the-art and future perspectives. Br Med Bull 2024; 149:60-71. [PMID: 38282031 PMCID: PMC10938543 DOI: 10.1093/bmb/ldad035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Revised: 11/30/2023] [Accepted: 12/12/2023] [Indexed: 01/30/2024]
Abstract
BACKGROUND Parkinson's disease (PD) is the second most common neurodegenerative disorder and is clinically characterized by the presence of motor (bradykinesia, rigidity, rest tremor and postural instability) and non-motor symptoms (cognitive impairment, autonomic dysfunction, sleep disorders, depression and hyposmia). The aetiology of PD is unknown except for a small but significant contribution of monogenic forms. SOURCES OF DATA No new data were generated or analyzed in support of this review. AREAS OF AGREEMENT Up to 15% of PD patients carry pathogenic variants in PD-associated genes. Some of these genes are associated with mendelian inheritance, while others act as risk factors. Genetic background influences age of onset, disease course, prognosis and therapeutic response. AREAS OF CONTROVERSY Genetic testing is not routinely offered in the clinical setting, but it may have relevant implications, especially in terms of prognosis, response to therapies and inclusion in clinical trials. Widely adopted clinical guidelines on genetic testing are still lacking and open to debate. Some new genetic associations are still awaiting confirmation, and selecting the appropriate genes to be included in diagnostic panels represents a difficult task. Finally, it is still under study whether (and to which degree) specific genetic forms may influence the outcome of PD therapies. GROWING POINTS Polygenic Risk Scores (PRS) may represent a useful tool to genetically stratify the population in terms of disease risk, prognosis and therapeutic outcomes. AREAS TIMELY FOR DEVELOPING RESEARCH The application of PRS and integrated multi-omics in PD promises to improve the personalized care of patients.
Collapse
Affiliation(s)
- L Trevisan
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics and Maternal and Child Health, University of Genoa, Largo P. Daneo 3, Genova, 16132, Italy
- IRCCS Ospedale Policlinico San Martino – SS Centro Tumori Ereditari, Largo R. Benzi 10, Genova, 16132, Italy
| | - A Gaudio
- IRCCS Ospedale Policlinico San Martino- UOC Genetica Medica, Largo R. Benzi 10, Genova, 16132, Italy
| | - E Monfrini
- Dino Ferrari Center, Neuroscience Section, Department of Pathophysiology and Transplantation, University of Milan, Via Francesco Sforza 35, Milan, 20122, Italy
- Neurology Unit, Foundation IRCCS Ca’Granda Ospedale Maggiore Policlinico, Via Festa del Perdono 7, Milan, 20122, Italy
| | - L Avanzino
- Department of Experimental Medicine, Section of Human Physiology, University of Genoa, Viale Benedetto XV/3, Genova, 16132, Italy
- IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 3, Genova, 16132, Italy
| | - A Di Fonzo
- Dino Ferrari Center, Neuroscience Section, Department of Pathophysiology and Transplantation, University of Milan, Via Francesco Sforza 35, Milan, 20122, Italy
- Neurology Unit, Foundation IRCCS Ca’Granda Ospedale Maggiore Policlinico, Via Festa del Perdono 7, Milan, 20122, Italy
| | - P Mandich
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics and Maternal and Child Health, University of Genoa, Largo P. Daneo 3, Genova, 16132, Italy
- IRCCS Ospedale Policlinico San Martino- UOC Genetica Medica, Largo R. Benzi 10, Genova, 16132, Italy
| |
Collapse
|
36
|
Lo T, Kushima I, Kimura H, Aleksic B, Okada T, Kato H, Inada T, Nawa Y, Torii Y, Yamamoto M, Kimura R, Funabiki Y, Kosaka H, Numata S, Kasai K, Sasaki T, Yokoyama S, Munesue T, Hashimoto R, Yasuda Y, Fujimoto M, Usami M, Itokawa M, Arai M, Ohi K, Someya T, Watanabe Y, Egawa J, Takahashi T, Suzuki M, Yamasue H, Iwata N, Ikeda M, Ozaki N. Association between copy number variations in parkin (PRKN) and schizophrenia and autism spectrum disorder: A case-control study. Neuropsychopharmacol Rep 2024; 44:42-50. [PMID: 37915257 PMCID: PMC10932780 DOI: 10.1002/npr2.12370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Accepted: 07/19/2023] [Indexed: 11/03/2023] Open
Abstract
AIM The present study aimed to examine the association between copy number variations (CNVs) in parkin (PRKN) and schizophrenia (SCZ) and autism spectrum disorder (ASD) in a large case-control sample. METHOD Array comparative genomic hybridization was performed on 3111 cases with SCZ, 1236 cases with ASD, and 2713 controls. We systematically prioritized likely pathogenic CNVs (LP-CNVs) in PRKN and examined their association with SCZ and ASD. RESULTS In total, 3014 SCZ cases (96.9%), 1205 ASD cases (97.5%), and 2671 controls (98.5%) passed quality control. We found that monoallelic carriers of LP-CNVs in PRKN were common (70/6890, 1.02%) and were not at higher risk of SCZ (p = 0.29) or ASD (p = 0.72). We observed that the distribution pattern of LP-CNVs in the Japanese population was consistent with those in other populations. We also identified a patient diagnosed with SCZ and early-onset Parkinson's disease carrying biallelic pathogenic CNVs in PRKN. The absence of Parkinson's symptoms in 10 other monoallelic carriers of the same pathogenic CNV further reflects the lack of effect of monoallelic pathogenic variants in PRKN in the absence of a second hit. CONCLUSION The present findings suggest that monoallelic CNVs in PRKN do not confer a significant risk for SCZ or ASD. However, further studies to investigate the association between biallelic CNVs in PRKN and SCZ and ASD are warranted.
Collapse
Grants
- JP15K19720 Japan Society for the Promotion of Science
- JP17H05090 Japan Society for the Promotion of Science
- JP18H04040 Japan Society for the Promotion of Science
- JP18K19511 Japan Society for the Promotion of Science
- JP19K17087 Japan Society for the Promotion of Science
- JP20H03608 Japan Society for the Promotion of Science
- JP20K07942 Japan Society for the Promotion of Science
- JP20K20602 Japan Society for the Promotion of Science
- JP21H00194 Japan Society for the Promotion of Science
- JP21H04815 Japan Society for the Promotion of Science
- JP21H05326 Japan Society for the Promotion of Science
- JP21K07543 Japan Society for the Promotion of Science
- JP22H00986 Japan Society for the Promotion of Science
- JP16dm0107134 Ministry of Education, Culture, Sports, Science and Technology
- JP19ak0101113 Ministry of Education, Culture, Sports, Science and Technology
- JP19dm0207075 Ministry of Education, Culture, Sports, Science and Technology
- JP19ek0109411 Ministry of Education, Culture, Sports, Science and Technology
- JP19km0405216 Ministry of Education, Culture, Sports, Science and Technology
- JP20ek0109488 Ministry of Education, Culture, Sports, Science and Technology
- JP21dk0307103 Ministry of Education, Culture, Sports, Science and Technology
- JP21tm0424220 Ministry of Education, Culture, Sports, Science and Technology
- JP21wm0425007 Ministry of Education, Culture, Sports, Science and Technology
- JP21wm0425008 Ministry of Education, Culture, Sports, Science and Technology
- JP21wm0425012 Ministry of Education, Culture, Sports, Science and Technology
- JP21wm0425019 Ministry of Education, Culture, Sports, Science and Technology
- JP21wm0525024 Ministry of Education, Culture, Sports, Science and Technology
- JP22tm0424222 Ministry of Education, Culture, Sports, Science and Technology
- Otsuka Toshimi Scholarship Foundation
- SENSHIN Medical Research Foundation
- Uehara Memorial Foundation
- Japan Society for the Promotion of Science
- Ministry of Education, Culture, Sports, Science and Technology
- Otsuka Toshimi Scholarship Foundation
- SENSHIN Medical Research Foundation
- Uehara Memorial Foundation
Collapse
Affiliation(s)
- Tzuyao Lo
- Department of PsychiatryNagoya University Graduate School of MedicineNagoyaJapan
| | - Itaru Kushima
- Department of PsychiatryNagoya University Graduate School of MedicineNagoyaJapan
- Medical Genomics CenterNagoya University HospitalNagoyaJapan
| | - Hiroki Kimura
- Department of PsychiatryNagoya University Graduate School of MedicineNagoyaJapan
| | - Branko Aleksic
- Department of PsychiatryNagoya University Graduate School of MedicineNagoyaJapan
| | - Takashi Okada
- Department of Developmental Disorders, National Institute of Mental HealthNational Center of Neurology and PsychiatryNagoyaJapan
| | - Hidekazu Kato
- Department of PsychiatryNagoya University Graduate School of MedicineNagoyaJapan
| | - Toshiya Inada
- Department of PsychiatryNagoya University Graduate School of MedicineNagoyaJapan
| | - Yoshihiro Nawa
- Department of PsychiatryNagoya University Graduate School of MedicineNagoyaJapan
| | - Youta Torii
- Department of PsychiatryNagoya University Graduate School of MedicineNagoyaJapan
| | - Maeri Yamamoto
- Department of PsychiatryNagoya University Graduate School of MedicineNagoyaJapan
| | - Ryo Kimura
- Department of Anatomy and Developmental BiologyGraduate School of MedicineKyoto UniversityKyotoJapan
| | - Yasuko Funabiki
- Department of Cognitive, Behavioral and Health Sciences, Graduate School of Human and Environmental StudiesKyoto UniversityKyotoJapan
| | - Hirotaka Kosaka
- Department of Neuropsychiatry, Faculty of Medical SciencesUniversity of FukuiFukuiJapan
| | - Shusuke Numata
- Department of Psychiatry, Graduate School of Biomedical ScienceTokushima UniversityTokushimaJapan
| | - Kiyoto Kasai
- Department of Neuropsychiatry, Graduate School of MedicineUniversity of TokyoTokyoJapan
- International Research Center for Neurointelligence at University of Tokyo Institutes for Advanced StudyTokyoJapan
| | - Tsukasa Sasaki
- Laboratory of Health Education, Graduate School of EducationUniversity of TokyoTokyoJapan
| | - Shigeru Yokoyama
- Research Center for Child Mental DevelopmentKanazawa UniversityIshikawaJapan
| | - Toshio Munesue
- Research Center for Child Mental DevelopmentKanazawa UniversityIshikawaJapan
| | - Ryota Hashimoto
- Department of Pathology of Mental DiseasesNational Institute of Mental HealthNational Center of Neurology and PsychiatryTokyoJapan
| | - Yuka Yasuda
- Department of Pathology of Mental DiseasesNational Institute of Mental HealthNational Center of Neurology and PsychiatryTokyoJapan
| | - Michiko Fujimoto
- Department of PsychiatryOsaka University Graduate School of MedicineOsakaJapan
| | - Masahide Usami
- Department of Child and Adolescent PsychiatryKohnodai Hospital, National Center for Global Health and MedicineChibaJapan
| | - Masanari Itokawa
- Schizophrenia Research Project, Department of Psychiatry and Behavioral SciencesTokyo Metropolitan Institute of Medical ScienceTokyoJapan
- Department of PsychiatryTokyo Metropolitan Matsuzawa HospitalTokyoJapan
| | - Makoto Arai
- Schizophrenia Research Project, Department of Psychiatry and Behavioral SciencesTokyo Metropolitan Institute of Medical ScienceTokyoJapan
| | - Kazutaka Ohi
- Department of PsychiatryGifu University Graduate School of MedicineGifuJapan
- Department of General Internal MedicineKanazawa Medical UniversityIshikawaJapan
| | - Toshiyuki Someya
- Department of PsychiatryNiigata University Graduate School of Medical and Dental SciencesNiigataJapan
| | - Yuichiro Watanabe
- Department of PsychiatryNiigata University Graduate School of Medical and Dental SciencesNiigataJapan
| | - Jun Egawa
- Department of PsychiatryNiigata University Graduate School of Medical and Dental SciencesNiigataJapan
| | - Tsutomu Takahashi
- Department of NeuropsychiatryUniversity of Toyama Graduate School of Medicine and Pharmaceutical SciencesToyamaJapan
- Research Center for Idling Brain ScienceUniversity of ToyamaToyamaJapan
| | - Michio Suzuki
- Department of NeuropsychiatryUniversity of Toyama Graduate School of Medicine and Pharmaceutical SciencesToyamaJapan
- Research Center for Idling Brain ScienceUniversity of ToyamaToyamaJapan
| | - Hidenori Yamasue
- Department of PsychiatryHamamatsu University School of MedicineHamamatsuJapan
| | - Nakao Iwata
- Department of PsychiatryFujita Health University School of MedicineToyoakeJapan
| | - Masashi Ikeda
- Department of PsychiatryNagoya University Graduate School of MedicineNagoyaJapan
- Department of PsychiatryFujita Health University School of MedicineToyoakeJapan
| | - Norio Ozaki
- Department of PsychiatryNagoya University Graduate School of MedicineNagoyaJapan
- Institute for Glyco‐core ResearchNagoya UniversityNagoyaJapan
| |
Collapse
|
37
|
Esteca MV, Divino IA, Vieira da Silva AL, Severino MB, Braga RR, Ropelle ER, Simabuco FM, Baptista IL. Parkin is a critical player in the effects of caffeine over mitochondrial quality control pathways during skeletal muscle regeneration in mice. Acta Physiol (Oxf) 2024; 240:e14111. [PMID: 38314948 DOI: 10.1111/apha.14111] [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: 08/16/2023] [Revised: 01/04/2024] [Accepted: 01/12/2024] [Indexed: 02/07/2024]
Abstract
AIM This study aimed to investigate the effects of caffeine on pathways associated with mitochondrial quality control and mitochondrial capacity during skeletal muscle regeneration, focusing on the role of Parkin, a key protein involved in mitophagy. METHODS We used in vitro C2C12 myoblast during differentiation with and without caffeine in the medium, and we evaluated several markers of mitochondrial quality control pathways and myotube growth. In vivo experiments, we used C57BL/6J (WT) and Parkintm 1Shn lineage (Parkin-/- ) mice and injured tibial anterior muscle. The mice regenerated TA muscle for 3, 10, and 21 days with or without caffeine ingestion. TA muscle was used to analyze the protein content of several markers of mitochondrial quality pathways, muscle satellite cell differentiation, and protein synthesis. Furthermore, it analyzed mtDNA, mitochondrial respiration, and myofiber growth. RESULTS C2C12 differentiation experiments showed that caffeine decreased Parkin content, potentially leading to increased DRP1 and PGC-1α content and altered mitochondrial population, thereby enhancing growth capacity. Using Parkin-/- mice, we found that caffeine intake during the regenerative process induces an increase in AMPKα phosphorylation and PGC-1α and TFAM content, changes that were partly Parkin-dependent. In addition, the absence of Parkin potentiates the ergogenic effect of caffeine by increasing mitochondrial capacity and myotube growth. Those effects are related to increased ATF4 content and activation of protein synthesis pathways, such as increased 4E-BP1 phosphorylation. CONCLUSION These findings demonstrate that caffeine ingestion changes mitochondrial quality control during skeletal muscle regeneration, and Parkin is a central player in those mechanisms.
Collapse
Affiliation(s)
- M V Esteca
- Laboratory of Cell and Tissue Biology, School of Applied Sciences, University of Campinas, Limeira, Brazil
| | - I A Divino
- Laboratory of Cell and Tissue Biology, School of Applied Sciences, University of Campinas, Limeira, Brazil
| | - A L Vieira da Silva
- Laboratory of Cell and Tissue Biology, School of Applied Sciences, University of Campinas, Limeira, Brazil
| | - M B Severino
- Laboratory of Cell and Tissue Biology, School of Applied Sciences, University of Campinas, Limeira, Brazil
- Multidisciplinarity Laboratory of Food and Health, School of Applied Sciences, University of Campinas, Limeira, Brazil
| | - R R Braga
- Laboratory of Molecular Biology of Exercise, School of Applied Sciences, University of Campinas, Limeira, Brazil
| | - E R Ropelle
- Laboratory of Molecular Biology of Exercise, School of Applied Sciences, University of Campinas, Limeira, Brazil
| | - F M Simabuco
- Multidisciplinarity Laboratory of Food and Health, School of Applied Sciences, University of Campinas, Limeira, Brazil
- Department of Biochemistry, Federal University of São Paulo, São Paulo, Brazil
| | - I L Baptista
- Laboratory of Cell and Tissue Biology, School of Applied Sciences, University of Campinas, Limeira, Brazil
| |
Collapse
|
38
|
Clausen L, Voutsinos V, Cagiada M, Johansson KE, Grønbæk-Thygesen M, Nariya S, Powell RL, Have MKN, Oestergaard VH, Stein A, Fowler DM, Lindorff-Larsen K, Hartmann-Petersen R. A mutational atlas for Parkin proteostasis. Nat Commun 2024; 15:1541. [PMID: 38378758 PMCID: PMC10879094 DOI: 10.1038/s41467-024-45829-4] [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: 07/05/2023] [Accepted: 02/01/2024] [Indexed: 02/22/2024] Open
Abstract
Proteostasis can be disturbed by mutations affecting folding and stability of the encoded protein. An example is the ubiquitin ligase Parkin, where gene variants result in autosomal recessive Parkinsonism. To uncover the pathological mechanism and provide comprehensive genotype-phenotype information, variant abundance by massively parallel sequencing (VAMP-seq) is leveraged to quantify the abundance of Parkin variants in cultured human cells. The resulting mutational map, covering 9219 out of the 9300 possible single-site amino acid substitutions and nonsense Parkin variants, shows that most low abundance variants are proteasome targets and are located within the structured domains of the protein. Half of the known disease-linked variants are found at low abundance. Systematic mapping of degradation signals (degrons) reveals an exposed degron region proximal to the so-called "activation element". This work provides examples of how missense variants may cause degradation either via destabilization of the native protein, or by introducing local signals for degradation.
Collapse
Affiliation(s)
- Lene Clausen
- Linderstrøm-Lang Centre for Protein Science, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Vasileios Voutsinos
- Linderstrøm-Lang Centre for Protein Science, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Matteo Cagiada
- Linderstrøm-Lang Centre for Protein Science, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Kristoffer E Johansson
- Linderstrøm-Lang Centre for Protein Science, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Martin Grønbæk-Thygesen
- Linderstrøm-Lang Centre for Protein Science, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Snehal Nariya
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - Rachel L Powell
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - Magnus K N Have
- Linderstrøm-Lang Centre for Protein Science, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | | | - Amelie Stein
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Douglas M Fowler
- Department of Genome Sciences, University of Washington, Seattle, WA, USA.
- Department of Bioengineering, University of Washington, Seattle, WA, USA.
| | - Kresten Lindorff-Larsen
- Linderstrøm-Lang Centre for Protein Science, Department of Biology, University of Copenhagen, Copenhagen, Denmark.
| | - Rasmus Hartmann-Petersen
- Linderstrøm-Lang Centre for Protein Science, Department of Biology, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
39
|
Bovenzi R, Conti M, Degoli GR, Cerroni R, Artusi CA, Pierantozzi M, Stefani A, Mercuri NB, Schirinzi T. Pregnancy, fertile life factors, and associated clinical course in PRKN early-onset Parkinson's disease. Neurol Sci 2024; 45:591-599. [PMID: 37658959 PMCID: PMC10791711 DOI: 10.1007/s10072-023-07029-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 08/20/2023] [Indexed: 09/05/2023]
Abstract
INTRODUCTION As the most common cause of autosomal recessive early onset Parkinson's disease (EOPD), parkin type Parkinson's disease (PRKN-PD) may affect female patients in childbearing age. Accordingly, issues related to fertility must be adequately addressed. Here, we landscaped fertile life factors and pregnancy course of a PRKN-PD cohort, including both novel cases directly observed at our center and published ones. METHODS Six patients with confirmed PRKN-PD were examined by a structured interview on reproductive factors and associated modifications of PD disturbances, including one case followed up throughout pregnancy which was described in greater detail. Six studies reporting fertile life factors of nine PRKN-PD patients were reviewed collecting homogeneous data on fertile life and pregnancy course. RESULTS PRKN-PD female patients experienced motor fluctuations with the menstrual cycle, pregnancy, and puerperium, which suggests a role for sex hormones in PD clinical burden. In some cases, abortion and miscarriages occurred during the organogenesis phase in patients receiving oral antiparkinsonian therapy; however, levodopa/benserazide monotherapy resulted to be the safest choice in pregnancy. CONCLUSION Collectively these data disclose the importance of pre-conception counseling in childbearing age PRKN-PD patients and EOPD in general.
Collapse
Affiliation(s)
- Roberta Bovenzi
- Unit of Neurology, Department of Systems Medicine, University of Rome "Tor Vergata", Via Montpellier, 00133, Rome, Italy
| | - Matteo Conti
- Unit of Neurology, Department of Systems Medicine, University of Rome "Tor Vergata", Via Montpellier, 00133, Rome, Italy
| | - Giulia Rebecca Degoli
- Unit of Neurology, Department of Systems Medicine, University of Rome "Tor Vergata", Via Montpellier, 00133, Rome, Italy
| | - Rocco Cerroni
- Unit of Neurology, Department of Systems Medicine, University of Rome "Tor Vergata", Via Montpellier, 00133, Rome, Italy
| | - Carlo Alberto Artusi
- Department of Neuroscience "Rita Levi Montalcini", University of Turin, Turin, Italy
| | - Mariangela Pierantozzi
- Unit of Neurology, Department of Systems Medicine, University of Rome "Tor Vergata", Via Montpellier, 00133, Rome, Italy
| | - Alessandro Stefani
- Unit of Neurology, Department of Systems Medicine, University of Rome "Tor Vergata", Via Montpellier, 00133, Rome, Italy
- UOSD Parkinson Centre, Tor Vergata University Hospital, Rome, Italy
| | - Nicola Biagio Mercuri
- Unit of Neurology, Department of Systems Medicine, University of Rome "Tor Vergata", Via Montpellier, 00133, Rome, Italy
| | - Tommaso Schirinzi
- Unit of Neurology, Department of Systems Medicine, University of Rome "Tor Vergata", Via Montpellier, 00133, Rome, Italy.
| |
Collapse
|
40
|
Zhang W, Ju Y, Ren Y, Miao Y, Wang Y. Exploring the Efficient Natural Products for the Therapy of Parkinson's Disease via Drosophila Melanogaster (Fruit Fly) Models. Curr Drug Targets 2024; 25:77-93. [PMID: 38213160 DOI: 10.2174/0113894501281402231218071641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 10/23/2023] [Accepted: 10/25/2023] [Indexed: 01/13/2024]
Abstract
Parkinson's disease (PD) is a severe neurodegenerative disorder, partly attributed to mutations, environmental toxins, oxidative stress, abnormal protein aggregation, and mitochondrial dysfunction. However, the precise pathogenesis of PD and its treatment strategy still require investigation. Fortunately, natural products have demonstrated potential as therapeutic agents for alleviating PD symptoms due to their neuroprotective properties. To identify promising lead compounds from herbal medicines' natural products for PD management and understand their modes of action, suitable animal models are necessary. Drosophila melanogaster (fruit fly) serves as an essential model for studying genetic and cellular pathways in complex biological processes. Diverse Drosophila PD models have been extensively utilized in PD research, particularly for discovering neuroprotective natural products. This review emphasizes the research progress of natural products in PD using the fruit fly PD model, offering valuable insights into utilizing invertebrate models for developing novel anti-PD drugs.
Collapse
Affiliation(s)
- Wen Zhang
- School of Pharmaceutical Science and Technology, Tianjin University, 300072, Tianjin, China
| | - Yingjie Ju
- School of Pharmaceutical Science and Technology, Tianjin University, 300072, Tianjin, China
| | - Yunuo Ren
- School of Pharmaceutical Science and Technology, Tianjin University, 300072, Tianjin, China
| | - Yaodong Miao
- Second Affiliated Hospital of Tianjin University of Traditional Chinese Medicine, 300250, Tianjin, China
| | - Yiwen Wang
- School of Pharmaceutical Science and Technology, Tianjin University, 300072, Tianjin, China
| |
Collapse
|
41
|
Xu X, Li M, Su Y, Wang Q, Qin P, Huang H, Zhang Y, Zhou Y, Yan J. Cdk5 phosphorylation-dependent C9orf72 degradation promotes neuronal death in Parkinson's disease models. CNS Neurosci Ther 2023; 29:3952-3966. [PMID: 37353944 PMCID: PMC10651984 DOI: 10.1111/cns.14319] [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: 09/02/2022] [Revised: 04/08/2023] [Accepted: 05/22/2023] [Indexed: 06/25/2023] Open
Abstract
AIMS Chromosome 9 open reading frame 72 (C9orf72) is one of the most dazzling molecules in neurodegenerative diseases, albeit that its role in Parkinson's disease (PD) remains unknown. This article aimed to explore the potential mechanism of C9orf72 involved in the pathogenesis of PD. METHODS The expression and phosphorylation levels of C9orf72 were examined by Western blotting, RT-PCR, and immunoprecipitation using PD models. Multiple bioinformatics software was used to predict the potential phosphorylation sites of C9orf72 by Cdk5, followed by verification of whether Cdk5-inhibitor ROSCOVITINE could reverse the degradation of C9orf72 in PD. By constructing the sh-C9orf72-knockdown adenovirus and overexpressing the FLAG-C9orf72 plasmid, the effects of C9orf72 knockdown and overexpression, respectively, were determined. A short peptide termed Myr-C9orf72 was used to verify whether interfering with Cdk5 phosphorylation at the Ser9 site of the C9orf72 protein could alleviate autophagy disorder, neuronal death, and movement disorder in PD models. RESULTS The expression level of the C9orf72 protein was significantly reduced, albeit the mRNA expression was not changed in the PD models. Moreover, the phosphorylation level was enhanced, and its reduction was mainly degraded by the ubiquitin-proteasome pathway. The key nervous system kinase Cdk5 directly phosphorylated the S9 site of the C9orf72 protein, which promoted the degradation of the C9orf72 protein. The knockdown of C9orf72 aggravated autophagy dysfunction and increased neuronal loss and motor dysfunction in substantia nigra neurons of PD mice. The overexpression of C9orf72 alleviated autophagy dysfunction in PD neurons. Specifically, interference with Cdk5 phosphorylation at the S9 site of C9orf72 alleviated autophagy dysfunction, neuronal death, and motor dysfunction mediated by C9orf72 protein degradation during PD. CONCLUSIONS Cumulatively, our findings illustrate the importance of the role of C9orf72 in the regulation of neuronal death during PD progression via the Cdk5-dependent degradation.
Collapse
Affiliation(s)
- Xingfeng Xu
- Department of PhysiologyGuilin Medical UniversityGuilinGuangxiChina
- Guangxi Key Laboratory of Brain and Cognitive NeuroscienceGuilin Medical UniversityGuilinGuangxiChina
| | - Mao Li
- Department of PhysiologyGuilin Medical UniversityGuilinGuangxiChina
- Guangxi Key Laboratory of Brain and Cognitive NeuroscienceGuilin Medical UniversityGuilinGuangxiChina
| | - Yan Su
- Department of PhysiologyGuilin Medical UniversityGuilinGuangxiChina
- Guangxi Key Laboratory of Brain and Cognitive NeuroscienceGuilin Medical UniversityGuilinGuangxiChina
| | - Qi Wang
- Department of PhysiologyGuilin Medical UniversityGuilinGuangxiChina
- Guangxi Key Laboratory of Brain and Cognitive NeuroscienceGuilin Medical UniversityGuilinGuangxiChina
| | - Peifang Qin
- Department of MicrobiologyGuilin Medical UniversityGuilinGuangxiChina
| | - Haitao Huang
- Department of MicrobiologyGuilin Medical UniversityGuilinGuangxiChina
| | - Yuting Zhang
- Department of MicrobiologyGuilin Medical UniversityGuilinGuangxiChina
| | - Yali Zhou
- Department of MicrobiologyGuilin Medical UniversityGuilinGuangxiChina
| | - Jianguo Yan
- Department of PhysiologyGuilin Medical UniversityGuilinGuangxiChina
- Guangxi Key Laboratory of Brain and Cognitive NeuroscienceGuilin Medical UniversityGuilinGuangxiChina
| |
Collapse
|
42
|
Salemi M, Lanza G, Salluzzo MG, Schillaci FA, Di Blasi FD, Cordella A, Caniglia S, Lanuzza B, Morreale M, Marano P, Tripodi M, Ferri R. A Next-Generation Sequencing Study in a Cohort of Sicilian Patients with Parkinson's Disease. Biomedicines 2023; 11:3118. [PMID: 38137339 PMCID: PMC10740523 DOI: 10.3390/biomedicines11123118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 11/20/2023] [Accepted: 11/21/2023] [Indexed: 12/24/2023] Open
Abstract
Parkinson's disease (PD) is a multisystem and multifactorial disorder and, therefore, the application of modern genetic techniques may assist in unraveling its complex pathophysiology. We conducted a clinical-demographic evaluation of 126 patients with PD, all of whom were Caucasian and of Sicilian ancestry. DNA was extracted from the peripheral blood for each patient, followed by sequencing using a Next-Generation Sequencing system. This system was based on a custom gene panel comprising 162 genes. The sample underwent further filtering, taking into account the allele frequencies of genetic variants, their presence in the Human Gene Mutation Database, and their association in the literature with PD or other movement/neurodegenerative disorders. The largest number of variants was identified in the leucine-rich repeat kinase 2 (LRRK2) gene. However, variants in other genes, such as acid beta-glucosidase (GBA), DNA polymerase gamma catalytic subunit (POLG), and parkin RBR E3 ubiquitin protein ligase (PRKN), were also discovered. Interestingly, some of these variants had not been previously associated with PD. Enhancing our understanding of the genetic basis of PD and identifying new variants possibly linked to the disease will contribute to improved diagnostic accuracy, therapeutic developments, and prognostic insights for affected individuals.
Collapse
Affiliation(s)
- Michele Salemi
- Oasi Research Institute—IRCCS, 94018 Troina, EN, Italy; (M.S.); (M.G.S.); (F.A.S.); (F.D.D.B.); (S.C.); (B.L.); (M.M.); (P.M.); (M.T.); (R.F.)
| | - Giuseppe Lanza
- Oasi Research Institute—IRCCS, 94018 Troina, EN, Italy; (M.S.); (M.G.S.); (F.A.S.); (F.D.D.B.); (S.C.); (B.L.); (M.M.); (P.M.); (M.T.); (R.F.)
- Department of Surgery and Medical-Surgical Specialties, University of Catania, 95123 Catania, CT, Italy
| | - Maria Grazia Salluzzo
- Oasi Research Institute—IRCCS, 94018 Troina, EN, Italy; (M.S.); (M.G.S.); (F.A.S.); (F.D.D.B.); (S.C.); (B.L.); (M.M.); (P.M.); (M.T.); (R.F.)
| | - Francesca A. Schillaci
- Oasi Research Institute—IRCCS, 94018 Troina, EN, Italy; (M.S.); (M.G.S.); (F.A.S.); (F.D.D.B.); (S.C.); (B.L.); (M.M.); (P.M.); (M.T.); (R.F.)
| | - Francesco Domenico Di Blasi
- Oasi Research Institute—IRCCS, 94018 Troina, EN, Italy; (M.S.); (M.G.S.); (F.A.S.); (F.D.D.B.); (S.C.); (B.L.); (M.M.); (P.M.); (M.T.); (R.F.)
| | - Angela Cordella
- Genomix4Life Srl, 84081 Baronissi, SA, Italy;
- Genome Research Center for Health—CRGS, 84081 Baronissi, SA, Italy
| | - Salvatore Caniglia
- Oasi Research Institute—IRCCS, 94018 Troina, EN, Italy; (M.S.); (M.G.S.); (F.A.S.); (F.D.D.B.); (S.C.); (B.L.); (M.M.); (P.M.); (M.T.); (R.F.)
| | - Bartolo Lanuzza
- Oasi Research Institute—IRCCS, 94018 Troina, EN, Italy; (M.S.); (M.G.S.); (F.A.S.); (F.D.D.B.); (S.C.); (B.L.); (M.M.); (P.M.); (M.T.); (R.F.)
| | - Manuela Morreale
- Oasi Research Institute—IRCCS, 94018 Troina, EN, Italy; (M.S.); (M.G.S.); (F.A.S.); (F.D.D.B.); (S.C.); (B.L.); (M.M.); (P.M.); (M.T.); (R.F.)
| | - Pietro Marano
- Oasi Research Institute—IRCCS, 94018 Troina, EN, Italy; (M.S.); (M.G.S.); (F.A.S.); (F.D.D.B.); (S.C.); (B.L.); (M.M.); (P.M.); (M.T.); (R.F.)
| | - Mariangela Tripodi
- Oasi Research Institute—IRCCS, 94018 Troina, EN, Italy; (M.S.); (M.G.S.); (F.A.S.); (F.D.D.B.); (S.C.); (B.L.); (M.M.); (P.M.); (M.T.); (R.F.)
| | - Raffaele Ferri
- Oasi Research Institute—IRCCS, 94018 Troina, EN, Italy; (M.S.); (M.G.S.); (F.A.S.); (F.D.D.B.); (S.C.); (B.L.); (M.M.); (P.M.); (M.T.); (R.F.)
| |
Collapse
|
43
|
Grossmann D, Malburg N, Glaß H, Weeren V, Sondermann V, Pfeiffer JF, Petters J, Lukas J, Seibler P, Klein C, Grünewald A, Hermann A. Mitochondria-Endoplasmic Reticulum Contact Sites Dynamics and Calcium Homeostasis Are Differentially Disrupted in PINK1-PD or PRKN-PD Neurons. Mov Disord 2023; 38:1822-1836. [PMID: 37449534 DOI: 10.1002/mds.29525] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 06/16/2023] [Indexed: 07/18/2023] Open
Abstract
BACKGROUND It is generally believed that the pathogenesis of PINK1/parkin-related Parkinson's disease (PD) is due to a disturbance in mitochondrial quality control. However, recent studies have found that PINK1 and Parkin play a significant role in mitochondrial calcium homeostasis and are involved in the regulation of mitochondria-endoplasmic reticulum contact sites (MERCSs). OBJECTIVE The aim of our study was to perform an in-depth analysis of the role of MERCSs and impaired calcium homeostasis in PINK1/Parkin-linked PD. METHODS In our study, we used induced pluripotent stem cell-derived dopaminergic neurons from patients with PD with loss-of-function mutations in PINK1 or PRKN. We employed a split-GFP-based contact site sensor in combination with the calcium-sensitive dye Rhod-2 AM and applied Airyscan live-cell super-resolution microscopy to determine how MERCSs are involved in the regulation of mitochondrial calcium homeostasis. RESULTS Our results showed that thapsigargin-induced calcium stress leads to an increase of the abundance of narrow MERCSs in wild-type neurons. Intriguingly, calcium levels at the MERCSs remained stable, whereas the increased net calcium influx resulted in elevated mitochondrial calcium levels. However, PINK1-PD or PRKN-PD neurons showed an increased abundance of MERCSs at baseline, accompanied by an inability to further increase MERCSs upon thapsigargin-induced calcium stress. Consequently, calcium distribution at MERCSs and within mitochondria was disrupted. CONCLUSIONS Our results demonstrated how the endoplasmic reticulum and mitochondria work together to cope with calcium stress in wild-type neurons. In addition, our results suggests that PRKN deficiency affects the dynamics and composition of MERCSs differently from PINK1 deficiency, resulting in differentially affected calcium homeostasis. © 2023 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Dajana Grossmann
- Translational Neurodegeneration Section "Albrecht Kossel," Department of Neurology, University Medical Center Rostock, University of Rostock, Rostock, Germany
| | - Nina Malburg
- Translational Neurodegeneration Section "Albrecht Kossel," Department of Neurology, University Medical Center Rostock, University of Rostock, Rostock, Germany
| | - Hannes Glaß
- Translational Neurodegeneration Section "Albrecht Kossel," Department of Neurology, University Medical Center Rostock, University of Rostock, Rostock, Germany
| | - Veronika Weeren
- Translational Neurodegeneration Section "Albrecht Kossel," Department of Neurology, University Medical Center Rostock, University of Rostock, Rostock, Germany
| | - Verena Sondermann
- Translational Neurodegeneration Section "Albrecht Kossel," Department of Neurology, University Medical Center Rostock, University of Rostock, Rostock, Germany
| | - Julia F Pfeiffer
- Translational Neurodegeneration Section "Albrecht Kossel," Department of Neurology, University Medical Center Rostock, University of Rostock, Rostock, Germany
| | - Janine Petters
- Translational Neurodegeneration Section "Albrecht Kossel," Department of Neurology, University Medical Center Rostock, University of Rostock, Rostock, Germany
| | - Jan Lukas
- Translational Neurodegeneration Section "Albrecht Kossel," Department of Neurology, University Medical Center Rostock, University of Rostock, Rostock, Germany
- Center for Transdisciplinary Neurosciences Rostock, University Medical Center Rostock, University of Rostock, Rostock, Germany
| | - Philip Seibler
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
| | - Christine Klein
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
| | - Anne Grünewald
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belvaux, Luxembourg
| | - Andreas Hermann
- Translational Neurodegeneration Section "Albrecht Kossel," Department of Neurology, University Medical Center Rostock, University of Rostock, Rostock, Germany
- Center for Transdisciplinary Neurosciences Rostock, University Medical Center Rostock, University of Rostock, Rostock, Germany
- Deutsches Zentrum für Neurodegenerative Erkrankungen Rostock/Greifswald, Rostock, Germany
| |
Collapse
|
44
|
Ibrahim A, Ipinloju N, Atasie NH, Babalola RM, Muhammad SA, Oyeneyin OE. Discovery of Small Molecule PARKIN Activator from Antipsychotic/Anti-neuropsychiatric Drugs as Therapeutics for PD: an In Silico Repurposing Approach. Appl Biochem Biotechnol 2023; 195:5980-6002. [PMID: 36735144 DOI: 10.1007/s12010-023-04376-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/12/2023] [Indexed: 02/04/2023]
Abstract
Although there is presently no cure for Parkinson's disease (PD), the available therapies are only able to lessen symptoms and preserve the quality of life. Around 10 million people globally had PD as of 2020. The widely used standard drug has recently been revealed to have several negative effects. Additionally, there is a dearth of innovative compounds entering the market as a result of subpar ADMET characteristics. Drug repurposing provides a chance to reenergize the sluggish drug discovery process by identifying new applications for already-approved medications. As this strategy offers a practical way to speed up the process of developing alternative medications for PD. This study used a computer-aided technique to select therapeutic agent(s) from FDA-approved neuropsychiatric/psychotic drugs that can be adopted in the treatment of Parkinson's disease. In the current work, a computational approach via molecular docking, density functional theory (DFT), and pharmacokinetics were used to identify possible (anti)neuropsychiatric/psychotic medications for the treatment of PD. By using molecular docking, about eight (anti)neuropsychiatric/psychotic medications were tested against PARKIN, a key protein in PD. Based on the docking score, the best ligand in the trial was determined. The top hits were compared to the reference ligand levodopa (L-DOPA). A large proportion of the drugs displayed binding affinity that was relatively higher than L-DOPA. Also, DFT analysis confirms the ligand-receptor interactions and the molecular charge transfer. All the compounds were found to obey Lipinski's rule with acceptable pharmacokinetic properties. The current study has revealed the effectiveness of antineuropsychiatric/antipsychotic drugs against PARKIN in the treatment of PD and lumateperone was revealed to be the most promising candidate interacting with PARKIN.
Collapse
Affiliation(s)
- Abdulwasiu Ibrahim
- Drosophila Laboratory, Department of Biochemistry, University of Ibadan, Ibadan, Oyo State, Nigeria.
- Department of Biochemistry and Molecular Biology, Usmanu Danfodiyo University, Sokoto, Nigeria.
- Drosophila Research and Training Centre, Ibadan, Oyo State, Nigeria.
| | - Nureni Ipinloju
- Theoretical and Computational Chemistry Unit, Department of Chemical Sciences, Adekunle Ajasin University, Akungba Akoko, Ondo State, Nigeria.
| | | | | | | | - Oluwatoba Emmanuel Oyeneyin
- Theoretical and Computational Chemistry Unit, Department of Chemical Sciences, Adekunle Ajasin University, Akungba Akoko, Ondo State, Nigeria
| |
Collapse
|
45
|
Bevilacqua A, Ho PC, Franco F. Metabolic reprogramming in inflammaging and aging in T cells. LIFE METABOLISM 2023; 2:load028. [PMID: 39872627 PMCID: PMC11749375 DOI: 10.1093/lifemeta/load028] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 06/23/2023] [Accepted: 06/26/2023] [Indexed: 01/30/2025]
Abstract
Aging represents an emerging challenge for public health due to the declined immune responses against pathogens, weakened vaccination efficacy, and disturbed tissue homeostasis. Metabolic alterations in cellular and systemic levels are also known to be cardinal features of aging. Moreover, cellular metabolism has emerged to provide regulations to guide immune cell behavior via modulations on signaling cascades and epigenetic landscape, and the aberrant aging process in immune cells can lead to inflammaging, a chronic and low-grade inflammation that facilitates aging by perturbing homeostasis in tissues and organs. Here, we review how the metabolic program in T cells is influenced by the aging process and how aged T cells modulate inflammaging. In addition, we discuss the potential approaches to reverse or ameliorate aging by rewiring the metabolic programming of immune cells.
Collapse
Affiliation(s)
- Alessio Bevilacqua
- Department of Fundamental Oncology, University of Lausanne, 1007 Lausanne, Switzerland
- Ludwig Institute for Cancer Research, University of Lausanne, 1066 Epalinges, Switzerland
| | - Ping-Chih Ho
- Department of Fundamental Oncology, University of Lausanne, 1007 Lausanne, Switzerland
- Ludwig Institute for Cancer Research, University of Lausanne, 1066 Epalinges, Switzerland
| | - Fabien Franco
- Department of Fundamental Oncology, University of Lausanne, 1007 Lausanne, Switzerland
- Ludwig Institute for Cancer Research, University of Lausanne, 1066 Epalinges, Switzerland
| |
Collapse
|
46
|
Franco F, Bevilacqua A, Wu RM, Kao KC, Lin CP, Rousseau L, Peng FT, Chuang YM, Peng JJ, Park J, Xu Y, Cassotta A, Yu YR, Speiser DE, Sallusto F, Ho PC. Regulatory circuits of mitophagy restrict distinct modes of cell death during memory CD8 + T cell formation. Sci Immunol 2023; 8:eadf7579. [PMID: 37738363 DOI: 10.1126/sciimmunol.adf7579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 08/11/2023] [Indexed: 09/24/2023]
Abstract
Mitophagy, a central process guarding mitochondrial quality, is commonly impaired in human diseases such as Parkinson's disease, but its impact in adaptive immunity remains unclear. The differentiation and survival of memory CD8+ T cells rely on oxidative metabolism, a process that requires robust mitochondrial quality control. Here, we found that Parkinson's disease patients have a reduced frequency of CD8+ memory T cells compared with healthy donors and failed to form memory T cells upon vaccination against COVID-19, highlighting the importance of mitochondrial quality control for memory CD8+ T cell formation. We further uncovered that regulators of mitophagy, including Parkin and NIX, were up-regulated in response to interleukin-15 (IL-15) for supporting memory T cell formation. Mechanistically, Parkin suppressed VDAC1-dependent apoptosis in memory T cells. In contrast, NIX expression in T cells counteracted ferroptosis by preventing metabolic dysfunction resulting from impaired mitophagy. Together, our results indicate that the mitophagy machinery orchestrates survival and metabolic dynamics required for memory T cell formation, as well as highlight a deficit in T cell-mediated antiviral responses in Parkinson's disease patients.
Collapse
Affiliation(s)
- Fabien Franco
- Department of Fundamental Oncology, University of Lausanne, Epalinges, Switzerland
- Ludwig Institute for Cancer Research, Lausanne Branch, Epalinges, Switzerland
| | - Alessio Bevilacqua
- Department of Fundamental Oncology, University of Lausanne, Epalinges, Switzerland
- Ludwig Institute for Cancer Research, Lausanne Branch, Epalinges, Switzerland
| | - Ruey-Mei Wu
- Neurology Department, National Taiwan University Hospital, Taipei, Taiwan
| | - Kung-Chi Kao
- Department of Fundamental Oncology, University of Lausanne, Epalinges, Switzerland
- Ludwig Institute for Cancer Research, Lausanne Branch, Epalinges, Switzerland
| | - Chun-Pu Lin
- Department of Fundamental Oncology, University of Lausanne, Epalinges, Switzerland
- Ludwig Institute for Cancer Research, Lausanne Branch, Epalinges, Switzerland
| | - Lorène Rousseau
- Department of Fundamental Oncology, University of Lausanne, Epalinges, Switzerland
- Ludwig Institute for Cancer Research, Lausanne Branch, Epalinges, Switzerland
| | - Fu-Ti Peng
- Neurology Department, National Taiwan University Hospital, Taipei, Taiwan
| | - Yu-Ming Chuang
- Department of Fundamental Oncology, University of Lausanne, Epalinges, Switzerland
- Ludwig Institute for Cancer Research, Lausanne Branch, Epalinges, Switzerland
| | - Jhan-Jie Peng
- Department of Fundamental Oncology, University of Lausanne, Epalinges, Switzerland
- Ludwig Institute for Cancer Research, Lausanne Branch, Epalinges, Switzerland
- Center for Molecular and Clinical Immunology, Chang Gung University, Taoyuan City, Taiwan
| | - Jaeoh Park
- Department of Fundamental Oncology, University of Lausanne, Epalinges, Switzerland
- Ludwig Institute for Cancer Research, Lausanne Branch, Epalinges, Switzerland
| | - Yingxi Xu
- Department of Fundamental Oncology, University of Lausanne, Epalinges, Switzerland
- Ludwig Institute for Cancer Research, Lausanne Branch, Epalinges, Switzerland
| | - Antonino Cassotta
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
| | - Yi-Ru Yu
- Department of Fundamental Oncology, University of Lausanne, Epalinges, Switzerland
- Ludwig Institute for Cancer Research, Lausanne Branch, Epalinges, Switzerland
| | - Daniel E Speiser
- Department of Fundamental Oncology, University of Lausanne, Epalinges, Switzerland
| | - Federica Sallusto
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
- Institute of Microbiology, ETH Zürich, Zürich, Switzerland
| | - Ping-Chih Ho
- Department of Fundamental Oncology, University of Lausanne, Epalinges, Switzerland
- Ludwig Institute for Cancer Research, Lausanne Branch, Epalinges, Switzerland
| |
Collapse
|
47
|
Tesson C, Bouchetara MS, Ferrien M, Lesage S, Brice A. Identification of a DAGLB Mutation in a Non-Chinese Patient with Parkinson's Disease. Mov Disord 2023; 38:1756-1757. [PMID: 37431851 DOI: 10.1002/mds.29533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 06/13/2023] [Accepted: 06/20/2023] [Indexed: 07/12/2023] Open
Affiliation(s)
- Christelle Tesson
- Sorbonne Université, Institut du Cerveau-Paris Brain Institute-ICM, Inserm, CNRS, Paris, France
| | | | - Mélanie Ferrien
- Sorbonne Université, Institut du Cerveau-Paris Brain Institute-ICM, Inserm, CNRS, Paris, France
| | - Suzanne Lesage
- Sorbonne Université, Institut du Cerveau-Paris Brain Institute-ICM, Inserm, CNRS, Paris, France
| | - Alexis Brice
- Sorbonne Université, Institut du Cerveau-Paris Brain Institute-ICM, Inserm, CNRS, Paris, France
- Assistance Publique Hôpitaux de Paris, Hôpital Pitié-Salpêtrière, Département de Neurologie, Centre d'Investigation Clinique Neurosciences, DMU Neuroscience, Paris, France
| |
Collapse
|
48
|
Stevenson-Hoare J, Schalkamp AK, Sandor C, Hardy J, Escott-Price V. New cases of dementia are rising in elderly populations in Wales, UK. J Neurol Sci 2023; 451:120715. [PMID: 37385025 PMCID: PMC7615574 DOI: 10.1016/j.jns.2023.120715] [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: 04/09/2023] [Revised: 06/02/2023] [Accepted: 06/15/2023] [Indexed: 07/01/2023]
Abstract
Dementia is one of the most common diseases in elderly populations, and older populations are one of the fastest growing groups globally. Consequently, the number of people developing and living with dementia is likely to grow. Using longitudinal medical records from Wales, UK between 1999 and 2018, diagnoses of overall dementia and common subtypes were combined with demographic data to assess numbers of new and existing cases per year. Data extraction resulted in 161,186 diagnoses from 116,645 individuals. Mean age at diagnosis of dementia increased over this period, resulting in fewer younger people with the disease. New cases of dementia have risen, as has the number of people living with dementia. Individuals with dementia are also living longer, even accounting for their older age. This may present a challenge for healthcare systems as the number of elderly people living with dementia is expected to continue to grow.
Collapse
Affiliation(s)
- Joshua Stevenson-Hoare
- Department of Psychological Medicine and Clinical Neuroscience, Cardiff University, United Kingdom; MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, United Kingdom
| | - Ann-Kathrin Schalkamp
- Department of Psychological Medicine and Clinical Neuroscience, Cardiff University, United Kingdom; UK Dementia Research Institute at Cardiff University, United Kingdom
| | - Cynthia Sandor
- Department of Psychological Medicine and Clinical Neuroscience, Cardiff University, United Kingdom; UK Dementia Research Institute at Cardiff University, United Kingdom
| | - John Hardy
- Department of Neurodegenerative Disease, UCL Institute of Neurology, United Kingdom; UK Dementia Research Institute at UCL, London, United Kingdom
| | - Valentina Escott-Price
- Department of Psychological Medicine and Clinical Neuroscience, Cardiff University, United Kingdom; MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, United Kingdom.
| |
Collapse
|
49
|
Bispo AG, Silva CS, Sena-dos-Santos C, Dalledone Moura D, Koshimoto BHB, Santos-Lobato BL, Ribeiro-dos-Santos Â, Cavalcante GC. Investigation of PRKN Mutations in Levodopa-Induced Dyskinesia in Parkinson's Disease Treatment. Biomedicines 2023; 11:2230. [PMID: 37626726 PMCID: PMC10452529 DOI: 10.3390/biomedicines11082230] [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: 06/26/2023] [Revised: 07/26/2023] [Accepted: 08/03/2023] [Indexed: 08/27/2023] Open
Abstract
Mitophagy is an important process that participates in mitochondrial quality control. Dysfunctions in this process can be caused by mutations in genes like PRKN and are associated with the development and progression of Parkinson's Disease (PD). The most used drug in the treatment of PD is levodopa (LD), but it can cause adverse effects, such as dyskinesia. Currently, few studies are searching for biomarkers for an effective use of lLD for this disease, especially regarding mitophagy genetics. Thus, this work investigates the association of 14 variants of the PRKN gene with LD in the treatment of PD. We recruited 70 patients with PD undergoing treatment with LD (39 without dyskinesia and 31 with dyskinesia). Genotyping was based on Sanger sequencing. Our results reinforce that age at onset of symptoms, duration of PD, and treatment and dosage of LD can influence the occurrence of dyskinesia but not the investigated PRKN variants. The perspective presented here of variants of mitophagy-related genes in the context of treatment with LD is still underexplored, although an association has been indicated in previous studies. We suggest that other variants in PRKN or in other mitophagy genes may participate in the development of levodopa-induced dyskinesia in PD treatment.
Collapse
Affiliation(s)
- Ana Gabrielle Bispo
- Laboratory of Human and Medical Genetics, Federal University of Pará, Belém 66075-110, Brazil; (A.G.B.); (C.S.S.); (C.S.-d.-S.); (Â.R.-d.-S.)
| | - Caio S. Silva
- Laboratory of Human and Medical Genetics, Federal University of Pará, Belém 66075-110, Brazil; (A.G.B.); (C.S.S.); (C.S.-d.-S.); (Â.R.-d.-S.)
| | - Camille Sena-dos-Santos
- Laboratory of Human and Medical Genetics, Federal University of Pará, Belém 66075-110, Brazil; (A.G.B.); (C.S.S.); (C.S.-d.-S.); (Â.R.-d.-S.)
| | - Dafne Dalledone Moura
- Laboratory of Experimental Neuropathology, Federal University of Pará, Belém 66075-110, Brazil; (D.D.M.); (B.H.B.K.); (B.L.S.-L.)
| | - Brenda Hanae Bentes Koshimoto
- Laboratory of Experimental Neuropathology, Federal University of Pará, Belém 66075-110, Brazil; (D.D.M.); (B.H.B.K.); (B.L.S.-L.)
| | - Bruno Lopes Santos-Lobato
- Laboratory of Experimental Neuropathology, Federal University of Pará, Belém 66075-110, Brazil; (D.D.M.); (B.H.B.K.); (B.L.S.-L.)
| | - Ândrea Ribeiro-dos-Santos
- Laboratory of Human and Medical Genetics, Federal University of Pará, Belém 66075-110, Brazil; (A.G.B.); (C.S.S.); (C.S.-d.-S.); (Â.R.-d.-S.)
| | - Giovanna C. Cavalcante
- Laboratory of Human and Medical Genetics, Federal University of Pará, Belém 66075-110, Brazil; (A.G.B.); (C.S.S.); (C.S.-d.-S.); (Â.R.-d.-S.)
| |
Collapse
|
50
|
Yu J, Shi J, Chen L, Wang Y, Cai G, Chen X, Hong W, Ye Q. Diffusion tensor imaging techniques show that parkin gene S/N167 polymorphism is responsible for extensive brain white matter damage in patients with Parkinson's disease. Heliyon 2023; 9:e18395. [PMID: 37600423 PMCID: PMC10432609 DOI: 10.1016/j.heliyon.2023.e18395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 07/12/2023] [Accepted: 07/17/2023] [Indexed: 08/22/2023] Open
Abstract
Objective To explore the influence of disease and genetic factors on the white matter microstructure in patients with PD. The white matter microstructural changes in the substantia nigra-striatum system were detected by diffusion tensor imaging (DTI) using the region of interest (ROI) and diffusion tensor tracer (DTT) methods. Methods Patients with primary Parkinson's disease (PD) without a family history of PD were selected and divided into PD-G/G and PD-G/A groups according to their parkin S/N167 polymorphism. Control groups matched for age, sex, and gene type (G/G and G/A) were also included. Three-dimensional brain volume imaging (3D-BRAVO) and DTI were performed. The microstructural changes in the substantia nigra-striatum system were evaluated by the ROI and DTT methods. The Mini-Mental State Examination (MMSE), Montreal Cognitive Assessment (MoCA), Hoehn-Yahr (H-Y) staging, and the third part of the Unified Parkinson's Disease Rating (UPDRS-III) scales evaluated the cognitive and motor function impairment in patients with PD. Independent samples t-test compared normally-distributed data, and the Wilcoxon rank sum test compared measurement or categorical non-normally distributed data. Multiple regression analysis was used to analyze the correlation between various DTI indicators and the MMSE, MoCA, UPDRS-III, and H-Y scores in the PD-G/G and PD-G/A groups. P < 0.05 was considered statistically significant. Results The white matter microstructural changes in the nigrostriatal pathway differed significantly between the PD or PD-G/A and the control group (P < 0.05)The ROI method showed that the left globus pallidus radial diffusivity (RD) value was negatively correlated with the MMSE score (r = -0.404, P = 0.040), and the left substantia nigra (LSN) fractional anisotropy (FA) value was positively correlated with the MoCA score (r = 0.405, P = 0.040) and negatively with the H-Y stage (r = -0.479, P = 0.013).The DTT method showed that the MMSE score was positively correlated with the right substantia nigra (RSN) FA value (r = 0.592, P = 0.001) and negatively with its RD value (r = -0.439, P = 0.025). The H-Y grade was negatively correlated with the number of fibers in the RSN (r = -0.406, P = 0.040). The UPDRS-Ⅲ score was positively correlated with the mean diffusivity (r = 0.420, P = 0.033) and RD (r = 0.396, P = 0.045) values of the LSN, and the AD value of the RSN (r = 0.439, P = 0.025). Conclusion The DTI technique detected extensive white matter fiber damage in patients with PD, primarily in those with the G/A genotype, that led to motor and cognitivesymptoms.
Collapse
Affiliation(s)
- Jinqiu Yu
- Department of Neurology, Fujian Institute of Geriatrics, Fujian Medical University Union Hospital, Fuzhou, China
- Department of Neurology, Affiliated Sanming First Hospital, Fujian Medical University, Sanming, China
- Institute of Neuroscience, Fujian Key Laboratory of Molecular Neurology, Fuzhou, China
| | - Jinying Shi
- Department of Neurology, Fujian Institute of Geriatrics, Fujian Medical University Union Hospital, Fuzhou, China
| | - Lina Chen
- Department of Neurology, Fujian Institute of Geriatrics, Fujian Medical University Union Hospital, Fuzhou, China
| | - Yingqing Wang
- Department of Neurology, Fujian Institute of Geriatrics, Fujian Medical University Union Hospital, Fuzhou, China
| | - Guoen Cai
- Department of Neurology, Fujian Institute of Geriatrics, Fujian Medical University Union Hospital, Fuzhou, China
| | - Xiaochun Chen
- Department of Neurology, Fujian Institute of Geriatrics, Fujian Medical University Union Hospital, Fuzhou, China
- Institute of Neuroscience, Fujian Key Laboratory of Molecular Neurology, Fuzhou, China
| | - Weiming Hong
- Department of Neurology, Affiliated Sanming First Hospital, Fujian Medical University, Sanming, China
| | - Qinyong Ye
- Department of Neurology, Fujian Institute of Geriatrics, Fujian Medical University Union Hospital, Fuzhou, China
- Institute of Neuroscience, Fujian Key Laboratory of Molecular Neurology, Fuzhou, China
- Institute of Clinical Neurology, Fujian Medical University, Fuzhou, China
| |
Collapse
|