1
|
Zhu Y, Cao B, Huang K, Liu J. Systematic identification of SNCA as a key gene in ischemic cardiomyopathy via integrated weighted gene co-expression network analysis and experimental validation. Biochem Biophys Res Commun 2025; 772:152063. [PMID: 40414013 DOI: 10.1016/j.bbrc.2025.152063] [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: 04/11/2025] [Revised: 05/09/2025] [Accepted: 05/20/2025] [Indexed: 05/27/2025]
Abstract
Ischemic cardiomyopathy (ICM) is associated with high mortality and hospitalization rates, and current treatments are suboptimal. The aim of this research was to identify novel therapeutic targets for ICM. The datasets GSE57338 and GSE5406 were obtained from the Gene Expression Omnibus (GEO) database. Gene modules were constructed using weighted gene co-expression network analysis (WGCNA), and the three relevant modules associated with ICM were identified. The biological functions and signaling pathways of the genes in these modules were further explored through Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses. Protein-protein interaction (PPI) networks were employed to identify hub genes within these modules. Least Absolute Shrinkage and Selection Operator (LASSO) logistic regression was performed to establish gene models, and 16 genes associated with left ventricular ejection fraction (LVEF) in ICM were identified. The genes were validated in heart tissues from human using quantitative real-time polymerase chain reaction (qRT-PCR). Among them, growth factor receptor-bound protein 14 (GRB14), ubiquitin C-terminal hydrolase L1 (UCHL1), and synuclein alpha (SNCA) were identified as key genes significantly associated with ICM. Additionally, key genes correlated with immune and stromal cell-related types were screened using xCell. In vivo and in vitro experiments demonstrated that inhibiting SNCA could improve cardiac dysfunction, inflammatory infiltration, and fibrosis in ICM. In conclusion, this study identified 16 genes closely related to LVEF and revealed that SNCA was a key gene in ICM, providing potential biomarkers and therapeutic targets for ICM.
Collapse
Affiliation(s)
- Yaoxi Zhu
- Department of Cardiovascular Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China; Hubei Provincial Engineering Research Center of Minimally Invasive Cardiovascular Surgery, Wuhan, 430071, China; Wuhan Clinical Research Center for Minimally Invasive Treatment of Structural Heart Disease, Wuhan, 430071, China
| | - Bingxin Cao
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kun Huang
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Jinping Liu
- Department of Cardiovascular Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China; Hubei Provincial Engineering Research Center of Minimally Invasive Cardiovascular Surgery, Wuhan, 430071, China; Wuhan Clinical Research Center for Minimally Invasive Treatment of Structural Heart Disease, Wuhan, 430071, China.
| |
Collapse
|
2
|
Trompeta C, Gasca-Salas C, Rodríguez-Rojas R, Fernández-Rodríguez B, Clarimón J, García-Cañamaque L, Guida P, Hernández-Fernández F, Sánchez-Juan P, Olazarán J, Vela-Desojo L. Apolipoprotein E genotypes and amyloid burden associated with cognitive decline in non-demented Parkinson's disease patients: A preliminary analysis. J Neurol Sci 2025; 473:123510. [PMID: 40300362 DOI: 10.1016/j.jns.2025.123510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 02/07/2025] [Accepted: 04/14/2025] [Indexed: 05/01/2025]
Abstract
BACKGROUND Cognitive impairment is a common non-motor manifestation in Parkinson's disease (PD). Amyloidosis seems to accelerate the onset of dementia in PD. The Apolipoprotein (APOE) ε4 genotype is linked to amyloid aggregation and has been suggested to be a candidate for development of PD dementia, while other APOE alleles have been less investigated. METHODS Forty-eight non-demented PD patients were included (ε2/ε3 carriers: n = 8; ε3/ε3 carriers: n = 35; and ε3/ε4 carriers: n = 5). Patients underwent APOE genotyping, a neuropsychological exam and a [18F]-Flutemetamol PET. Statistical Parametric Mapping 12 was used to analyze regional amyloid differences by APOE genotypes. Cognitive assessment was re-administered after follow-up. RESULTS APOE ε3/ε4 carriers showed significant amyloid load in left middle occipital areas in comparison with APOE ε3/ε3, whereas APOE ε3/ε4 exhibited several areas of amyloid accumulation in comparison with APOE ε2/ε3 that disappeared after family-wise error cluster level correction, but which hold in nonparametric analyses. No amyloid load differences were found between APOE ε2/ε3 and APOE ε3/ε3. The frequency of progression of cognitive decline was 80 % in ε3/ε4, 50 % in ε2/ε3, and 22.86 % in ε3/ε3 carriers. CONCLUSIONS This compared regional amyloid deposition and cognitive evolution among APOE genotypes. Our preliminary findings suggest an association between APOE ε4 and amyloid burden in posterior cortical areas in non-demented PD and a tendency to a higher frequency of cognitive decline. Larger studies should explore cognition and amyloid burden in APOE ε2 carriers.
Collapse
Affiliation(s)
- Clara Trompeta
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain; Instituto de Investigación Sanitaria HM Hospitales, Madrid, Spain; PhD Program in Health Sciences, University of Alcala de Henares, Alcala de Henares, Spain
| | - Carmen Gasca-Salas
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain; Instituto de Investigación Sanitaria HM Hospitales, Madrid, Spain; University CEU-San Pablo, Madrid, Spain; Network Center for Biomedical Research on Neurodegenerative Diseases (CIBERNED), Instituto Carlos III, Madrid, Spain.
| | - Rafael Rodríguez-Rojas
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain; Instituto de Investigación Sanitaria HM Hospitales, Madrid, Spain; Network Center for Biomedical Research on Neurodegenerative Diseases (CIBERNED), Instituto Carlos III, Madrid, Spain
| | - Beatriz Fernández-Rodríguez
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain; Instituto de Investigación Sanitaria HM Hospitales, Madrid, Spain; Department of Neurology, Hospital Regional Universitario de Málaga, Málaga, Spain
| | | | - Lina García-Cañamaque
- Nuclear Medicine Department, Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain
| | - Pasqualina Guida
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain; Instituto de Investigación Sanitaria HM Hospitales, Madrid, Spain; PhD Program in Neuroscience, Autonoma de Madrid University-Cajal Institute, Madrid, Spain
| | - Frida Hernández-Fernández
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain; Department of Nursing and Nutrition, Faculty of Biomedical and Health Sciences, Universidad Europea de Madrid,Madrid, Spain
| | - Pascual Sánchez-Juan
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain; Instituto de Investigación Sanitaria HM Hospitales, Madrid, Spain; Alzheimer's Centre Reina Sofia-CIEN Foundation-ISCIII, 28031 Madrid, Spain
| | - Javier Olazarán
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain; Instituto de Investigación Sanitaria HM Hospitales, Madrid, Spain; Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | | |
Collapse
|
3
|
Brennan SO, Tinworth AC. Genetically Proxied Phosphodiesterase Type 5 (PDE5) Inhibition and Risk of Dementia: A Drug Target Mendelian Randomization Study. Mol Neurobiol 2025; 62:7864-7874. [PMID: 39951190 PMCID: PMC12078358 DOI: 10.1007/s12035-025-04732-9] [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: 02/26/2024] [Accepted: 01/29/2025] [Indexed: 05/15/2025]
Abstract
Phosphodiesterase-5 (PDE5) inhibitors have gained interest as a potential treatment for dementia. However, current evidence is limited to observational and pre-clinical studies. We conducted a drug-target Mendelian randomization (MR) analysis to investigate the on-target effects of pharmacological PDE5 inhibition on dementia subtypes and related phenotypes. We selected variants from around the PDE5A locus associated with diastolic and systolic blood pressure, as well as circulating PDE5A levels, to create three instruments for genetically proxied PDE5A inhibition. Using two-sample MR, we validated the instruments against erectile dysfunction and pulmonary arterial hypertension before assessing their associations with dementia subtypes, dementia-related proteins, and neuroimaging traits. After correcting for multiple comparisons, genetically proxied PDE5 inhibition, per one SD lower in diastolic blood pressure, was associated with higher odds of Alzheimer's disease (OR 1.09, 95% CI 1.07-1.11) and Lewy body dementia (OR 1.32, 95% CI 1.23-1.41), but a trend towards lower odds of vascular dementia across all instruments. Genetically proxied PDE5 inhibition was associated with both beneficial and adverse effects on brain MRI traits. This included lower volumes of white matter hyperintensities (SD change - 0.035, 95% CI - 0.025, - 0.045), indicating potential benefits, but also reduced volumes of other structures, including the thalamus, suggesting potential adverse effects. PDE5 inhibition was associated with the concentrations of several proteins implicated in dementia pathophysiology. Our findings suggest that while PDE5 inhibition may be associated with a lower risk of vascular dementia, possibly by preventing white matter hyperintensities, it may increase risk of Alzheimer's disease and Lewy body dementia, warranting further investigation before clinical trials.
Collapse
Affiliation(s)
- Stephen O Brennan
- University of Galway, Galway, Ireland.
- Mater Misericordiae University Hospital, Dublin, Ireland.
| | - Alexander C Tinworth
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK.
| |
Collapse
|
4
|
Hao X, Xiao X, Weng L, Lin Z, Xu Q, Du J, Yang Q, Zhu Y, Liu Y, Xu T, Zhou Y, Liao X, Guo J, Luo S, Wang J, Yan X, Tang B, Li J, Jiao B, Shen L. Genetic risk and plasma biomarkers of dementia with Lewy bodies in a Chinese population. NPJ Parkinsons Dis 2025; 11:128. [PMID: 40374660 PMCID: PMC12081849 DOI: 10.1038/s41531-025-00988-z] [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: 11/26/2024] [Accepted: 05/02/2025] [Indexed: 05/17/2025] Open
Abstract
Genetic investigations and associations with plasma biomarkers of dementia with Lewy bodies (DLB) in East Asian populations are lacking. The aim of our study is to identify candidate pathogenic variants and assess the diagnostic performance of plasma biomarkers among DLB patients in the Chinese population. This cohort included 151 DLB patients and 2010 controls, all of whom underwent whole genome sequencing. Plasma glial fibrillary acidic protein (GFAP), α-synuclein(αSyn), neurofilament light (NfL), and phosphorylated tau 217 (p-tau217) were detected in a subgroup. As a result, the APOE ε4 allele significantly increased DLB risk (p = 1.84E-11), while rare missense variants of USP13 gene were first found to be suggestively associated with DLB risk (p = 1.31E-5). Higher levels of plasma GFAP, αSyn, NfL, and p-tau217 were detected in DLB patients compared to controls (p < 0.001), which combined with polygenic risk scores (PRS) achieving an AUC of 0.927 for DLB diagnosis. Besides, significant correlations were observed between PRS of DLB and age at onset, the cumulative incidence rate, as well as plasma GFAP levels. In conclusion, this is the first study to simultaneously investigate the genetics and plasma biomarkers of DLB, highlighting the discriminative ability for DLB using PRS and plasma biomarker assay.
Collapse
Affiliation(s)
- Xiaoli Hao
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Xuewen Xiao
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Engineering Research Center of Hunan Province in Cognitive Impairment Disorders, Central South University, Changsha, China
- Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, China
| | - Ling Weng
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Engineering Research Center of Hunan Province in Cognitive Impairment Disorders, Central South University, Changsha, China
- Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, China
| | - Zhuojie Lin
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Qian Xu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Engineering Research Center of Hunan Province in Cognitive Impairment Disorders, Central South University, Changsha, China
- Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, China
| | - Juan Du
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Engineering Research Center of Hunan Province in Cognitive Impairment Disorders, Central South University, Changsha, China
- Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, China
| | - Qijie Yang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Yuan Zhu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Yiliang Liu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Tianyan Xu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Yafang Zhou
- Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, China
| | - Xinxin Liao
- Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, China
| | - Jifeng Guo
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Engineering Research Center of Hunan Province in Cognitive Impairment Disorders, Central South University, Changsha, China
- Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, China
| | - Shilin Luo
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Engineering Research Center of Hunan Province in Cognitive Impairment Disorders, Central South University, Changsha, China
- Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, China
| | - Junling Wang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Engineering Research Center of Hunan Province in Cognitive Impairment Disorders, Central South University, Changsha, China
- Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, China
| | - Xinxiang Yan
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Engineering Research Center of Hunan Province in Cognitive Impairment Disorders, Central South University, Changsha, China
- Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, China
| | - Beisha Tang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Engineering Research Center of Hunan Province in Cognitive Impairment Disorders, Central South University, Changsha, China
- Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, China
- Brain Research Center, Central South University, Changsha, Hunan, China
- FuRong Laboratory, Central South University, Changsha, Hunan, China
| | - Jinchen Li
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Bin Jiao
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
- Engineering Research Center of Hunan Province in Cognitive Impairment Disorders, Central South University, Changsha, China.
- Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Xiangya Hospital, Central South University, Changsha, China.
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, China.
| | - Lu Shen
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
- Engineering Research Center of Hunan Province in Cognitive Impairment Disorders, Central South University, Changsha, China.
- Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Xiangya Hospital, Central South University, Changsha, China.
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, China.
- Brain Research Center, Central South University, Changsha, Hunan, China.
- FuRong Laboratory, Central South University, Changsha, Hunan, China.
| |
Collapse
|
5
|
Alghamdi A, Koen E, Helmantel M, Rafie K, Dolga AM, van Munster BC, Hak E. The Association Between Herpes Zoster, Antiherpetic Therapies, and Alzheimer's Disease: A Comprehensive Systematic Review. Pharmaceuticals (Basel) 2025; 18:722. [PMID: 40430540 PMCID: PMC12115117 DOI: 10.3390/ph18050722] [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: 03/25/2025] [Revised: 05/09/2025] [Accepted: 05/12/2025] [Indexed: 05/29/2025] Open
Abstract
Background: Alzheimer's Disease (AD) represents a significant public health challenge with a rising global burden. Emerging evidence suggests a potential link between herpes zoster (HZ) and an increased risk of AD. These findings indicate that HZ may contribute to neuroinflammation and other pathophysiological processes associated with AD, pointing out the potential role of the virus infection in the initiation and/or progression of AD pathology. Objective: This systematic review evaluates the relationships between HZ and the risk of AD, highlighting the potential neuroprotective role of HZ vaccination and antiherpetic drug (AHD) use. Methods: Adhering to the (Preferred Reporting Items for Systematic Reviews and Meta-Analyses) PRISMA guidelines, a comprehensive search of databases (e.g., PubMed) until October 2024 was conducted. Observational studies reporting risk estimates were included. Extracted data were synthesized and analyzed narratively due to study heterogeneity. Results: Eleven studies (out of two hundred) met the inclusion criteria, offering preliminary insights that could form a foundation for further investigation. Reports on HZ showed mixed outcomes, with some studies suggesting a potential increased risk of AD, while others showed no clear correlation. Interestingly, HZ vaccination led to a potential preventive role in reducing the risk of AD, with risk estimates ranging from 0.57 to 0.99. Additionally, the use of AHDs was linked to a reduced risk of AD, with risk estimates ranging from 0.65 to 0.96. Conclusions: Published findings suggest that HZ vaccination and AHD use could represent potential interventions to reduce the risk of AD; however, further research is necessary to validate these findings and better understand the underlying protective mechanisms.
Collapse
Affiliation(s)
- Ali Alghamdi
- Groningen Research Institute of Pharmacy, PharmacoTherapy, -Epidemiology & -Economics, University of Groningen, 9713 AV Groningen, The Netherlands
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Emma Koen
- Groningen Research Institute of Pharmacy, PharmacoTherapy, -Epidemiology & -Economics, University of Groningen, 9713 AV Groningen, The Netherlands
| | - Manon Helmantel
- Groningen Research Institute of Pharmacy, PharmacoTherapy, -Epidemiology & -Economics, University of Groningen, 9713 AV Groningen, The Netherlands
| | - Karim Rafie
- Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy, University of Groningen, 9712 CP Groningen, The Netherlands
| | - Amalia M. Dolga
- Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy, University of Groningen, 9712 CP Groningen, The Netherlands
| | - Barbara C. van Munster
- Department of Geriatric Medicine, University Medical Center Groningen, University of Groningen, 9700 AB Groningen, The Netherlands
- Alzheimer Center Groningen, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands
| | - Eelko Hak
- Groningen Research Institute of Pharmacy, PharmacoTherapy, -Epidemiology & -Economics, University of Groningen, 9713 AV Groningen, The Netherlands
- Department of Epidemiology, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands
| |
Collapse
|
6
|
Fu Y, Halliday GM. Dementia with Lewy bodies and Parkinson disease dementia - the same or different and is it important? Nat Rev Neurol 2025:10.1038/s41582-025-01090-x. [PMID: 40355531 DOI: 10.1038/s41582-025-01090-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/11/2025] [Indexed: 05/14/2025]
Abstract
Biological definitions of neurological diseases are now becoming a reality, although still in the research phase. This development will recategorize neurological diseases, providing objective diagnostics and the promise of therapeutics that target biological mechanisms - similar to the strategy that has proven successful in tumours and other conditions. In this Perspective article, we discuss this development for dementias with dominant Lewy pathology, as the availability of biological assays for this pathology has sparked new interest in a single disease diagnosis for all individuals positive for α-synuclein. On the basis of current evidence, we argue that an α-synuclein assay alone is unlikely to be a specific criterion for a spectrum of clinical syndromes with Lewy pathology or a definitive diagnostic marker for Lewy body dementia. We advocate that one biological assay will not reflect the complex spatiotemporal features of brain pathology. Diverse sequential mechanisms underpin the highly heterogeneous phenotypes and clinicopathological processes of Lewy body dementias. Disease modification, if possible, will be most effective when it targets the early underlying mechanisms, especially those leading to aggressive phenotypes.
Collapse
Affiliation(s)
- YuHong Fu
- Brain and Mind Centre & Faculty of Medicine and Health School of Medical Sciences, The University of Sydney, Sydney, New South Wales, Australia
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Glenda M Halliday
- Brain and Mind Centre & Faculty of Medicine and Health School of Medical Sciences, The University of Sydney, Sydney, New South Wales, Australia.
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA.
| |
Collapse
|
7
|
Tao F, Deng S, Zhuo B, Liu J, Liang X, Shi J, Meng Z. Causal relationship between schizophrenia and five types of dementia: A bidirectional two-sample Mendelian randomization study. PLoS One 2025; 20:e0322752. [PMID: 40338973 PMCID: PMC12061177 DOI: 10.1371/journal.pone.0322752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 03/27/2025] [Indexed: 05/10/2025] Open
Abstract
BACKGROUND Although observational research indicates an association between schizophrenia and dementia, it is unclear whether the two are causally related. In order to examine the causal relationship between schizophrenia and five types of dementia (all-cause dementia, Alzheimer's disease, vascular dementia, frontotemporal dementia, and dementia with Lewy bodies), we performed a bidirectional two-sample Mendelian randomization analysis. METHODS In this study, pooled statistics of schizophrenia and dementia were obtained from the Large-scale genome-wide association study (GWAS) in individuals of European ancestry. Inverse variance weighted (IVW) was the primary statistical approach used in this Mendelian randomization, to further support our findings, we also used MR-Egger, weighted median, and cML-MA. We also used a number of sensitivity analyses to evaluate pleiotropy and heterogeneity. RESULTS In the study of the effect of schizophrenia on dementia, findings from the IVW analysis suggested that schizophrenia is associated with an increased risk of all-cause dementia (OR = 1.065, 95%CI: 1.027 ~ 1.104, P = 0.001, FDR-corrected P = 0.003), Alzheimer's disease (OR = 1.029, 95%CI: 1.003 ~ 1.054, P = 0.027, FDR-corrected P = 0.045), and vascular dementia (OR = 1.106, 95%CI: 1.023 ~ 1.197, P = 0.012, FDR-corrected P = 0.029). In the study of the effect of dementia on schizophrenia, no form of dementia assessed in this study was found to be a risk factor for schizophrenia. CONCLUSION Our findings suggest that schizophrenia may be a risk factor for all-cause dementia, Alzheimer's disease, and vascular dementia, but no dementia of any kind was found to be a risk factor for schizophrenia. Our study provides insights into the potential genetic relationship between schizophrenia and dementia.
Collapse
Affiliation(s)
- Feng Tao
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Shizhe Deng
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Bifang Zhuo
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Jingyue Liu
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Xuan Liang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Jiangwei Shi
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Zhihong Meng
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| |
Collapse
|
8
|
Lu Z, Wang X, Mao T, Liu L, Zhang J. Evidence from a Mendelian randomization study: Delirium's noncausal role in dementia onset. Neuroscience 2025; 573:247-253. [PMID: 40132793 DOI: 10.1016/j.neuroscience.2025.03.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 03/06/2025] [Accepted: 03/13/2025] [Indexed: 03/27/2025]
Abstract
BACKGROUND Previous observational studies have suggested a possible association between dementia and delirium. However, these findings might be influenced by confounding variables. Thus, our study aimed to investigate the causal relationship between dementia and delirium using a bidirectional Mendelian randomization (MR) analysis. METHODS In our investigation, bidirectional MR analysis was conducted by summary statistics from genome-wide association studies (GWAS). This enabled us to evaluate the causal impact of delirium and different types of dementia, such as Alzheimer's dementia (AD), vascular dementia (VaD) and Lewy body dementia (LD). RESULTS According to MR analysis, there was a significant positive correlation between delirium risk and AD (odds ratio [OR] = 1.363; 95 % confidence interval [CI], 1.223-1.519; p = 2.140E-08) and LD (OR = 1.403; 95 % CI, 1.151-1.711; p = 8.226E-04). However, the analysis also revealed that there was no causal relationship between VaD (OR = 1.044; 95 % CI = 1.136-1.027; p = 0.316) and the risk of delirium. Additionally, our study does not provide evidence to support a causal correlation between delirium and the risk of developing any form of dementia. CONCLUSION The results of the MR analysis suggest a potential causal link between dementia and an increased risk of delirium. Nevertheless, it should be emphasized that the existing evidence does not provide support for a causal connection in the reverse direction, implying that delirium may not play a causative role in the onset of dementia.
Collapse
Affiliation(s)
- Zhongyuan Lu
- Department of Anesthesiology and Perioperative Medicine, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, Henan Province 450000, China; Laboratory Animal Center, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province 450000, China
| | - Xiaoling Wang
- Department of Anesthesiology and Perioperative Medicine, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, Henan Province 450000, China; Laboratory Animal Center, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province 450000, China
| | - Tian Mao
- Department of Anesthesiology and Perioperative Medicine, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, Henan Province 450000, China; Laboratory Animal Center, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province 450000, China
| | - Lu Liu
- Department of Anesthesiology and Perioperative Medicine, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, Henan Province 450000, China; Laboratory Animal Center, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province 450000, China
| | - Jiaqiang Zhang
- Department of Anesthesiology and Perioperative Medicine, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, Henan Province 450000, China; Laboratory Animal Center, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province 450000, China.
| |
Collapse
|
9
|
Shwab EK, Man Z, Gingerich DC, Gamache J, Garrett ME, Serrano GE, Beach TG, Crawford GE, Ashley‐Koch AE, Chiba‐Falek O. Comparative mapping of single-cell transcriptomic landscapes in neurodegenerative diseases. Alzheimers Dement 2025; 21:e70012. [PMID: 40344336 PMCID: PMC12061851 DOI: 10.1002/alz.70012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 01/15/2025] [Accepted: 01/25/2025] [Indexed: 05/11/2025]
Abstract
INTRODUCTION Alzheimer's disease (AD), dementia with Lewy bodies (DLB), and Parkinson's disease (PD) represent a spectrum of neurodegenerative diseases (NDDs). Here, we performed the first direct comparison of their transcriptomic landscapes. METHODS We profiled whole transcriptomes of NDD cortical tissue by single-nucleus RNA sequencing, using computational analyses to identify common and distinct differentially expressed genes (DEGs), pathways, vulnerable and disease-driver cell subtypes, and altered cell-to-cell interactions. RESULTS The same inhibitory neuron subtype was depleted in both AD and DLB. Potentially disease-driving neuronal cell subtypes were identified in both PD and DLB. Cell-cell communication was predicted to be increased in AD but decreased in DLB and PD. DEGs were most commonly shared across NDDs within inhibitory neuron subtypes. Overall, AD and PD showed greatest transcriptomic divergence, while DLB exhibited an intermediate signature. DISCUSSION These results may help explain the clinicopathological spectrum of these NDDs and provide unique insights into shared and distinct molecular mechanisms underlying pathogenesis. HIGHLIGHTS The same vulnerable inhibitory neuron subtype population was depleted in both Alzheimer's disease (AD) and dementia with Lewy bodies (DLB). Potentially disease-driving neuronal cell subtypes were discovered in both Parkinson's disease (PD) and DLB. Cell-cell communication was predicted to be increased in AD but decreased in DLB and PD. Differentially expressed genes were most commonly shared across neurodegenerative diseases in inhibitory neuron types. AD and PD had the greatest transcriptomic divergence, with DLB showing an intermediate signature.
Collapse
Affiliation(s)
- E. Keats Shwab
- Division of Translational Brain SciencesDepartment of NeurologyDuke University Medical CenterDurhamNorth CarolinaUSA
- Center for Genomic and Computational BiologyDuke University Medical CenterDurhamNorth CarolinaUSA
| | - Zhaohui Man
- Division of Translational Brain SciencesDepartment of NeurologyDuke University Medical CenterDurhamNorth CarolinaUSA
- Center for Genomic and Computational BiologyDuke University Medical CenterDurhamNorth CarolinaUSA
| | - Daniel C. Gingerich
- Division of Translational Brain SciencesDepartment of NeurologyDuke University Medical CenterDurhamNorth CarolinaUSA
- Center for Genomic and Computational BiologyDuke University Medical CenterDurhamNorth CarolinaUSA
| | - Julia Gamache
- Division of Translational Brain SciencesDepartment of NeurologyDuke University Medical CenterDurhamNorth CarolinaUSA
- Center for Genomic and Computational BiologyDuke University Medical CenterDurhamNorth CarolinaUSA
| | - Melanie E. Garrett
- Duke Molecular Physiology InstituteDuke University Medical CenterDurhamNorth CarolinaUSA
| | | | | | - Gregory E. Crawford
- Center for Genomic and Computational BiologyDuke University Medical CenterDurhamNorth CarolinaUSA
- Department of PediatricsDivision of Medical GeneticsDuke University Medical CenterDurhamNorth CarolinaUSA
- Center for Advanced Genomic TechnologiesDuke University Medical CenterDurhamNorth CarolinaUSA
| | - Allison E. Ashley‐Koch
- Duke Molecular Physiology InstituteDuke University Medical CenterDurhamNorth CarolinaUSA
- Department of MedicineDuke University Medical CenterDurhamNorth CarolinaUSA
| | - Ornit Chiba‐Falek
- Division of Translational Brain SciencesDepartment of NeurologyDuke University Medical CenterDurhamNorth CarolinaUSA
- Center for Genomic and Computational BiologyDuke University Medical CenterDurhamNorth CarolinaUSA
| |
Collapse
|
10
|
Ye J, Dai X, Zhang C, Duan Z, Zhou G, Wang J. Investigating the causal relationships between mitochondrial proteins and dementia with Lewy bodies. J Alzheimers Dis 2025; 105:378-386. [PMID: 40111912 DOI: 10.1177/13872877251328882] [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] [Indexed: 03/22/2025]
Abstract
BackgroundDisruptions in mitochondrial function have been implicated in various neurodegenerative diseases. However, the specific role of mitochondrial proteins in the pathogenesis of dementia with Lewy bodies (DLB) remains poorly understood.ObjectiveThis study aims to investigate potential causal relationships between mitochondrial proteins and DLB risk using Mendelian randomization (MR) analysis.MethodsCausal associations between 66 mitochondrial proteins (MPs) and DLB were assessed by MR analysis, utilizing data from comprehensive genome-wide association studies (GWAS), with various analytical methods, including the inverse variance weighted, MR-Egger, and weighted median. Cochran's Q statistics assessed the heterogeneity of instrumental variables.ResultsGenetic predispositions to increased levels of ES1 protein homolog and apoptosis-inducing factor 1 (AIF-1) were associated with an elevated risk of DLB. Conversely, genetic predispositions to increased levels of glutaredoxin-2 (GLRX-2), complement component 1 Q subcomponent-binding protein (C1QBP), and mitochondrial glutamate carrier 2 (GC2) were found to be protective against DLB. Sensitivity analyses revealed no heterogeneity or horizontal pleiotropy among the selected instrumental variables.ConclusionsOur MR study identifies specific MPs potentially causally linked to DLB risk. These findings offer new insights into the MP-related mechanisms underlying DLB pathogenesis and highlight potential therapeutic targets.
Collapse
Affiliation(s)
- Jingna Ye
- Department of Neurology, Luoyang Center Hospital Affiliated to Zhengzhou University, Luoyang, China
| | - Xuelian Dai
- Department of Neurology, Luoyang Center Hospital Affiliated to Zhengzhou University, Luoyang, China
| | - Canwen Zhang
- Department of Neurology, The Second Affiliated Hospital of Shandong First Medical University, Taian, China
| | - Zhihui Duan
- Department of Neurology, Luoyang Center Hospital Affiliated to Zhengzhou University, Luoyang, China
| | - Guoqing Zhou
- Department of Neurology, The Second Affiliated Hospital of Shandong First Medical University, Taian, China
| | - Juan Wang
- Department of Neurology, The Second Affiliated Hospital of Shandong First Medical University, Taian, China
| |
Collapse
|
11
|
Dourlen P, Kilinc D, Landrieu I, Chapuis J, Lambert JC. BIN1 and Alzheimer's disease: the tau connection. Trends Neurosci 2025; 48:349-361. [PMID: 40268578 DOI: 10.1016/j.tins.2025.03.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 03/06/2025] [Accepted: 03/16/2025] [Indexed: 04/25/2025]
Abstract
Bridging integrator 1 (BIN1) is a ubiquitously expressed protein that plays a critical role in endocytosis, trafficking and cytoskeletal dynamics. In 2010, BIN1 gene was reported as a major genetic risk factor for Alzheimer's disease (AD), which shifted the focus on its physiological and pathophysiological roles in the brain (at a time when data available were scarce). In this review, we discuss the multiple cerebral roles of BIN1, especially in regulating synaptic function, and the strong link between BIN1 and tau pathology, supported by recent evidence ranging from genetic and clinical/postmortem observations to molecular interactions.
Collapse
Affiliation(s)
- Pierre Dourlen
- Univ Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE - Risk factors and molecular determinants of aging-related diseases, Institut Pasteur de Lille, Université de Lille, Lille, France
| | - Devrim Kilinc
- Univ Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE - Risk factors and molecular determinants of aging-related diseases, Institut Pasteur de Lille, Université de Lille, Lille, France
| | - Isabelle Landrieu
- Univ Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE - Risk factors and molecular determinants of aging-related diseases, Institut Pasteur de Lille, Université de Lille, Lille, France; CNRS EMR9002-BSI-Integrative Structural Biology, Lille, France
| | - Julien Chapuis
- Univ Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE - Risk factors and molecular determinants of aging-related diseases, Institut Pasteur de Lille, Université de Lille, Lille, France
| | - Jean-Charles Lambert
- Univ Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE - Risk factors and molecular determinants of aging-related diseases, Institut Pasteur de Lille, Université de Lille, Lille, France.
| |
Collapse
|
12
|
Zhao Y, Fei L, Duan Y. Combining GWAS Summary Data and Proteomics Identified Potential Drug Targets in Dementia. Mol Neurobiol 2025:10.1007/s12035-025-04967-6. [PMID: 40266545 DOI: 10.1007/s12035-025-04967-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 04/14/2025] [Indexed: 04/24/2025]
Abstract
Due to progressive cognitive loss and subsequent incapability of daily life, the development of novel therapeutics is urgently needed for dementia patients. We performed a two-sample bi-directional Mendelian randomization (MR) analysis using summary-level statistics to identify causality between peripheral and cerebrospinal fluid (CSF) proteins and the risk of dementia. Genetic variants were subtracted from the Genome-Wide Association Studies (GWAS) results. Wald ratio (WR) and inverse-variance weighted (IVW) ratio were utilized to estimate the causal effects of plasma and CSF proteins on dementia. Reverse MR, Steiger filtering, Bayesian co-localization phenotype scanning, and external validation were integrated to strengthen the robustness of primary MR results. After sensitivity analysis, six circulating proteins were identified in three dementia classifications, whereas no causality was found in frontotemporal dementia (FTD). Elevated levels of circulating C1R protein increased the odds of developing Alzheimer's disease (AD), while PILRA and CELA2A were estimated to protect against the pathogenesis of AD; genetically predicted increase of α-synuclein and APOE elevated the occurrence of Dementia of Lewy Bodies (DLB); elevated level of circulating CRP was assessed to increase the onset of vascular dementia (VD). Our MR analyses identified a genetically predicted association between circulating C1R, PILRA, and CELA2A and the risk of AD, causal estimates between α-syn, APOE protein, and the onset of DLB, and a robust correlation between CRP and the etiology of VD. This study might guide the discovery of disease etiology and build up a novel disease-modifying paradigm of dementia.
Collapse
Affiliation(s)
- Yingjie Zhao
- Department of Cardiology, The First Hospital of Jilin University, Jilin University, Changchun, 130021, Jilin Province, China
| | - Lu Fei
- Department of Neurology, The First Hospital of Jilin University, Jilin University, Changchun, 130021, Jilin Province, China.
| | - Yongtao Duan
- Henan Provincial Key Laboratory of Pediatric Hematology, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou, 450053, Henan Province, China
| |
Collapse
|
13
|
Babalola DO, Adewale BA, Okwunze KF, Mohammed TT, Oduguwa IO, Igwe HA, Farombi T, Akinyemi RO. Knowledge and attitudes about dementia and dementia genetics in a cohort of geriatric clinic attendees in Nigeria. J Alzheimers Dis 2025:13872877251333816. [PMID: 40261356 DOI: 10.1177/13872877251333816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/24/2025]
Abstract
BackgroundPopulation ageing in Africa will increase the burden of Alzheimer's disease and related dementias within the next few decades. Despite the potential for discovery of novel genetic risks in African populations, there is still a paucity of dementia genetic research among indigenous Africans.ObjectiveWe aimed to investigate the knowledge and attitudes of elderly population in Nigeria about dementia and dementia genetics.MethodsOne hundred clinic attendees (aged ≥60 years) recruited at the University College Hospital, Ibadan, Nigeria were surveyed using an interviewer-administered questionnaire consisting of the Dementia Knowledge Assessment Scale (DKAS) and other items assessing knowledge and attitudes about dementia genetics.ResultsThe mean age (±SD) of participants was 71.0 (±7.1) years, and the mean (±SD) DKAS score was 8.87 (±10.84). Only 10% were considered to have good knowledge of dementia (i.e., DKAS score ≥26). Attempts by participants to translate "dementia" in their local languages revealed misleading themes in their perception of the condition. Of the 42 participants who claimed to know what dementia is, 32 (76.2%) of them had poor knowledge (i.e., DKAS <26). Twenty-one participants were aware of the existence of genetic risk factors for dementia, but none could name a dementia risk gene. Seventy participants expressed willingness to undergo genetic testing to assess their risk of dementia.ConclusionsThere is a poor level of knowledge about dementia and dementia genetics among the elderly population in Nigeria. Public health education and community engagement is important for maximizing the impact of dementia genetic studies in Africa.
Collapse
Affiliation(s)
- David Oluwasayo Babalola
- Neuroscience and Ageing Research Unit, Institute for Advanced Medical Research and Training, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Boluwatife Adeleye Adewale
- Neuroscience and Ageing Research Unit, Institute for Advanced Medical Research and Training, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Kenechukwu Franklin Okwunze
- Neuroscience and Ageing Research Unit, Institute for Advanced Medical Research and Training, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Teslim Timilehin Mohammed
- Neuroscience and Ageing Research Unit, Institute for Advanced Medical Research and Training, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Ifeoluwa Oluwasegun Oduguwa
- Neuroscience and Ageing Research Unit, Institute for Advanced Medical Research and Training, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Hilda Amauche Igwe
- Neuroscience and Ageing Research Unit, Institute for Advanced Medical Research and Training, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Temitope Farombi
- Chief Tony Anenih Geriatric Centre, University College Hospital, Ibadan, Nigeria
| | - Rufus Olusola Akinyemi
- Neuroscience and Ageing Research Unit, Institute for Advanced Medical Research and Training, College of Medicine, University of Ibadan, Ibadan, Nigeria
- Chief Tony Anenih Geriatric Centre, University College Hospital, Ibadan, Nigeria
- Centre for Genomic and Precision Medicine, College of Medicine, University of Ibadan, Ibadan, Nigeria
| |
Collapse
|
14
|
Du K, Zuo YL, Zhang ZM, Li A, Zuo QH, Zhang CY, Guo R, Ping C, Du WS, Li SM. The role of the hippocampus and SLC39A8 in chronic musculoskeletal pain-induced dementia: a Mendelian randomization study. Sci Rep 2025; 15:13211. [PMID: 40240578 PMCID: PMC12003831 DOI: 10.1038/s41598-025-97428-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Accepted: 04/04/2025] [Indexed: 04/18/2025] Open
Abstract
Despite observational studies suggesting a link between chronic musculoskeletal pain (CMP) and increased risk of cognitive decline and dementia, the causal nature of this relationship remains uncertain due to potential confounding factors and reverse causality. We employed two-sample Mendelian Randomization (TSMR), bidirectional MR, mediation MR, drug-target MR, and colocalization analysis, along with gene set enrichment and protein-protein interaction (PPI) analyses. TSMR assessed the causal associations between CMP and the risk of dementia and its subtypes, including Alzheimer's disease (AD), vascular dementia (VaD), Lewy body dementia (LBD), frontotemporal dementia (FTD), and Parkinson's disease (PD). Bidirectional MR evaluated reverse causality, while mediation analyses identified potential mediators, focusing on neuroimaging and cognitive phenotypes. Drug-target MR investigated the role of the SLC39A8 gene, and colocalization analysis determined shared causal genetic variants. Gene set enrichment and PPI analyses elucidated the biological pathways implicated in the CMP-dementia relationship. Robust evidence established a causal relationship between chronic low back pain (LBP) and increased risk of PD, with knee osteoarthritis identified as a partial mediator, suggesting a pathway involving chronic inflammation. Bidirectional MR analysis revealed no evidence of reverse causality, further supporting the unidirectional causal link from LBP to PD. Colocalization analysis confirmed distinct genetic architectures for LBP and PD, while drug-target MR implicated the SLC39A8 gene as a potential mediator. Gene set enrichment and PPI analyses highlighted critical biological pathways, such as purine metabolism and glutamate receptor signaling. Suggestive evidence indicated potential causal links between limb pain and overall dementia, myalgia and VaD, as well as potential protective effects of Polymyalgia Rheumatica (PMR) against AD and rheumatism against PD. This study reveals a complex causal relationship between CMP and neurodegenerative diseases, particularly the robust link between LBP and PD. The findings underscore the need for further research to elucidate the underlying mechanisms and inform targeted prevention and treatment strategies.
Collapse
Affiliation(s)
- Kai Du
- Department of Pain Medicine, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Yong-Li Zuo
- Department of Pain Medicine, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Zi-Meng Zhang
- Department of Pain Medicine, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Ao Li
- Department of Pain Medicine, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Qi-Heng Zuo
- Department of Pain Medicine, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Chen-Yu Zhang
- Department of Pain Medicine, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Ren Guo
- Department of Pain Medicine, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Chen Ping
- Department of Pain Medicine, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Wei-Shuai Du
- Department of Pain Medicine, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Shu-Ming Li
- Department of Pain Medicine, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China.
| |
Collapse
|
15
|
Saeed U, Piracha ZZ, Tariq MN, Syed S, Rauf M, Razaq L, Iftikhar MK, Maqsood A, Ahsan SM. Decoding the genetic blueprints of neurological disorders: disease mechanisms and breakthrough gene therapies. Front Neurol 2025; 16:1422707. [PMID: 40291849 PMCID: PMC12022314 DOI: 10.3389/fneur.2025.1422707] [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: 04/24/2024] [Accepted: 02/13/2025] [Indexed: 04/30/2025] Open
Abstract
Neurological disorders pose a rapidly growing global health burden, significantly affecting cognitive and motor functions with profound societal repercussions. This comprehensive review probe into the genetic foundations of various neurological conditions while exploring innovative RNA-based therapeutics particularly gene therapies as cutting edge treatment strategies. Through an in-depth analysis of existing literature, the study examines the genetic landscape, disease mechanisms, and gene-based intervention possibilities across a range of neurological disorders, including Cerebellar Ataxias, Autosomal Recessive Ataxia, Mitochondrial Cerebellar Ataxia, Multiple System Atrophy (MSA), Idiopathic Late-Onset Cerebellar Ataxia, Hereditary Spastic Paraplegias, Alzheimer's Disease, Vascular Dementia, Lewy Body Dementia, Frontotemporal Dementias, Inherited Prion Diseases, and Huntington's Disease. It uncovers the intricate network of genetic mutations driving these disorders, shedding light on their mechanisms and uncovering promising therapeutic targets. The review also highlights the remarkable potential of RNA-based therapeutics, with gene therapies standing at the forefront of precision treatment approaches. By offering an up-to-date understanding of the genetic intricacies and emerging therapeutic possibilities in neurological disorders, this study significantly contributes to the advancement of precision medicine in neurology. It also paves the way for future research and clinical applications aimed at improving patient care and outcomes.
Collapse
Affiliation(s)
- Umar Saeed
- Operational Research Center in Healthcare, Near East University, Nicosia, Türkiye
- Foundation University School of Health Sciences (FUSH), Foundation University Islamabad, Islamabad, Pakistan
| | - Zahra Zahid Piracha
- International Center of Medical Sciences Research (ICMSR), Islamabad, Pakistan
- International Center of Medical Sciences Research (ICMSR), Austin, TX, United States
- International Center of Medical Sciences Research (ICMSR), Essex, United Kingdom
| | | | - Shayan Syed
- Akhtar Saeed Medical & Dental College, Lahore, Pakistan
| | - Maria Rauf
- University College of Medicine and Dentistry, Lahore, Pakistan
| | - Laiba Razaq
- Akhtar Saeed Medical & Dental College, Lahore, Pakistan
| | | | - Amna Maqsood
- Akhtar Saeed Medical & Dental College, Lahore, Pakistan
| | | |
Collapse
|
16
|
Minakaki G, Safren N, Bustos BI, Lubbe SJ, Mencacci NE, Krainc D. Commander complex regulates lysosomal function and is implicated in Parkinson's disease risk. Science 2025; 388:204-211. [PMID: 40209002 DOI: 10.1126/science.adq6650] [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: 05/24/2024] [Revised: 12/02/2024] [Accepted: 02/12/2025] [Indexed: 04/12/2025]
Abstract
Variants in GBA1 resulting in decreased lysosomal glucocerebrosidase (GCase) activity are a common risk factor for Parkinson's disease (PD) and dementia with Lewy bodies (DLB). Incomplete penetrance of GBA1 variants suggests that additional genes contribute to PD and DLB manifestation. By using a pooled genome-wide CRISPR interference screen, we identified copper metabolism MURR1 domain-containing 3 (COMMD3) protein, a component of the COMMD/coiled-coil domain-containing protein 22 (CCDC22)/CCDC93 (CCC) and Commander complexes, as a modifier of GCase and lysosomal activity. Loss of COMMD3 increased the release of lysosomal proteins through extracellular vesicles, leading to their impaired delivery to endolysosomes and consequent lysosomal dysfunction. Rare variants in the Commander gene family were associated with increased PD risk. Thus, COMMD genes and related complexes regulate lysosomal homeostasis and may represent modifiers in PD and other neurodegenerative diseases associated with lysosomal dysfunction.
Collapse
Affiliation(s)
- Georgia Minakaki
- Davee Department of Neurology, Northwestern University, Feinberg School of Medicine, Chicago, IL, USA
| | - Nathaniel Safren
- Davee Department of Neurology, Northwestern University, Feinberg School of Medicine, Chicago, IL, USA
| | - Bernabe I Bustos
- Davee Department of Neurology, Northwestern University, Feinberg School of Medicine, Chicago, IL, USA
| | - Steven J Lubbe
- Davee Department of Neurology, Northwestern University, Feinberg School of Medicine, Chicago, IL, USA
| | - Niccolò E Mencacci
- Davee Department of Neurology, Northwestern University, Feinberg School of Medicine, Chicago, IL, USA
| | - Dimitri Krainc
- Davee Department of Neurology, Northwestern University, Feinberg School of Medicine, Chicago, IL, USA
| |
Collapse
|
17
|
Gaudio A, Gotta F, Ponti C, Geroldi A, La Barbera A, Mandich P. GWAS by Subtraction to Disentangle RBD Genetic Background from α-Synucleinopathies. Int J Mol Sci 2025; 26:3578. [PMID: 40332088 PMCID: PMC12026788 DOI: 10.3390/ijms26083578] [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: 03/17/2025] [Revised: 04/04/2025] [Accepted: 04/07/2025] [Indexed: 05/08/2025] Open
Abstract
Rapid eye movement (REM) sleep behavior disorder (RBD) is a parasomnia characterized by loss of muscle atonia and abnormal behaviors occurring during REM sleep. Idiopathic RBD (iRBD) is recognized as the strongest prodromal hallmark of α-synucleinopathies, with an established conversion rate to a neurodegenerative condition that reaches up to 96.6% at 15 years of follow-up. Moreover, RBD-converters display a more severe clinical trajectory compared to those that do not present with RBD. However, the extent to which iRBD represents a distinct genetic entity or an early manifestation of neurodegeneration remains unclear. To address this, we applied Genomic Structural Equation Modeling (GenomicSEM) using a GWAS-by-subtraction approach to disentangle the genetic architecture of iRBD from the shared genomic liability across α-synucleinopathies. Our findings highlight the SNCA locus as a key genetic regulator of iRBD susceptibility. While iRBD exhibits a partially distinct genetic signature, residual genomic overlap with neurodegenerative traits suggests that its genetic architecture exists along a continuum of α-synucleinopathy risk. In this scenario, the associations with neuroanatomical correlates may serve as early indicators of a trajectory toward future neurodegeneration. These findings provide a framework for identifying biomarkers that could aid in disease stratification and risk prediction, potentially improving early intervention strategies.
Collapse
Affiliation(s)
- Andrea Gaudio
- IRCCS Ospedale Policlinico San Martino–UOC Genetica Medica, Largo R. Benzi 10, 16132 Genova, Italy; (F.G.); (A.L.B.); (P.M.)
| | - Fabio Gotta
- IRCCS Ospedale Policlinico San Martino–UOC Genetica Medica, Largo R. Benzi 10, 16132 Genova, Italy; (F.G.); (A.L.B.); (P.M.)
| | - Clarissa Ponti
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics and Maternal and Child Health, University of Genova, Largo P. Daneo 3, 16132 Genova, Italy; (C.P.); (A.G.)
| | - Alessandro Geroldi
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics and Maternal and Child Health, University of Genova, Largo P. Daneo 3, 16132 Genova, Italy; (C.P.); (A.G.)
| | - Andrea La Barbera
- IRCCS Ospedale Policlinico San Martino–UOC Genetica Medica, Largo R. Benzi 10, 16132 Genova, Italy; (F.G.); (A.L.B.); (P.M.)
| | - Paola Mandich
- IRCCS Ospedale Policlinico San Martino–UOC Genetica Medica, Largo R. Benzi 10, 16132 Genova, Italy; (F.G.); (A.L.B.); (P.M.)
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics and Maternal and Child Health, University of Genova, Largo P. Daneo 3, 16132 Genova, Italy; (C.P.); (A.G.)
| |
Collapse
|
18
|
Jia T, Yang F, Qin F, He Y, Han F, Zhang C. Identification of Common Brain Protein and Genetic Loci Between Parkinson's Disease and Lewy Body Dementia. CNS Neurosci Ther 2025; 31:e70370. [PMID: 40202048 PMCID: PMC11979625 DOI: 10.1111/cns.70370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 02/25/2025] [Accepted: 03/17/2025] [Indexed: 04/10/2025] Open
Abstract
BACKGROUND Parkinson's disease (PD) and Lewy body dementia (LBD) have many common features, including clinical manifestations, neurochemistry, and pathology, but little is known about their shared brain proteins and genetic factors. METHODS To identify susceptibility-related brain proteins that are shared between PD and LBD patients, proteome-wide association studies (PWASs) were conducted by integrating human brain protein quantitative trait loci (pQTLs) with large-scale genome-wide association studies (GWASs) of both diseases. Subsequently, pleiotropy-informed conditional false discovery rate (pleioFDR) analysis was performed to identify common risk genetic loci between PD and LBD. Finally, the downregulation of these risk genes in different disease states was validated by differential gene expression analysis. RESULTS PWASs identified 12 PD risk proteins and nine LBD risk proteins, among which TMEM175 (zPD = -7.25, PPD = 4.12E-13; zLBD = -6.02, PLBD = 1.75E-09) and DOC2A (zPD = -4.13, PPD = 3.71E-05; zLBD = -3.91, PLBD = 9.08E-05) were shared. PleioFDR analysis revealed that five genetic risk loci mapped to eight genes associated with PD and LBD, including the proteome-wide significant risk gene TMEM175 (ConjFDR = 5.74E-03). Differential expression analysis verified that TMEM175 was significantly downregulated in the midbrains of PD patients (p = 1.19E-02), and further exploration revealed that TMEM175 was also dramatically downregulated in the substantia nigra of PD patients (p = 1.16E-02) and incidental Lewy body disease patients (p = 7.52E-03). Moreover, TMEM175 was significantly downregulated in induced pluripotent stem cell-derived dopaminergic neurons from PD patients (p = 4.60E-02). CONCLUSION Dysregulation of TMEM175 may confer PD and LBD risk and may be partly responsible for their comorbidity. Our results revealed the common genetic risk factors between PD and LBD, which elucidated the shared genetic basis of these diseases.
Collapse
Affiliation(s)
- Tingting Jia
- Mental Health Center and Psychiatric Laboratory, the State Key Laboratory of BiotherapyWest China Hospital of Sichuan UniversityChengduSichuanChina
- Department of Gastroenterology and Hepatology and Sichuan University‐University of Oxford Huaxi Joint Centre for Gastrointestinal CancerWest China Hospital of Sichuan UniversityChengduSichuanChina
| | - Fuhua Yang
- Department of NephrologyThe Sixth People's Hospital of ChengduChengduSichuanChina
| | - Fengqin Qin
- Department of NeurologyThe 3rd Affiliated Hospital of Chengdu Medical CollegeChengduSichuanChina
| | - Yongji He
- Clinical Trial Center, National Medical Products Administration key Laboratory for Clinical Research and Evaluation of Innovative DrugsWest China Hospital Sichuan UniversityChengduChina
| | - Feng Han
- Department of Emergency MedicineHainan General Hospital, Hainan Affiliated Hospital of Hainan Medical UniversityHaikouChina
| | - Chengcheng Zhang
- Mental Health Center and Psychiatric Laboratory, the State Key Laboratory of BiotherapyWest China Hospital of Sichuan UniversityChengduSichuanChina
| |
Collapse
|
19
|
Hou X, Heckman MG, Fiesel FC, Koga S, Soto‐Beasley AI, Watzlawik JO, Zhao J, Valentino RR, Johnson PW, White LJ, Quicksall ZS, Reddy JS, Bras J, Guerreiro R, Zhao N, Bu G, Dickson DW, Ross OA, Springer W. Genome-wide association analysis identifies APOE as a mitophagy modifier in Lewy body disease. Alzheimers Dement 2025; 21:e70198. [PMID: 40309932 PMCID: PMC12044520 DOI: 10.1002/alz.70198] [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: 12/13/2024] [Revised: 03/17/2025] [Accepted: 03/24/2025] [Indexed: 05/02/2025]
Abstract
INTRODUCTION Phosphorylated ubiquitin (p-S65-Ub) is generated during PINK1-PRKN mitophagy as a specific marker of mitochondrial damage. Despite the widespread deposition of p-S65-Ub in aged and diseased human brain, the genetic contribution to its accumulation remains unclear. METHODS To identify novel mitophagy regulators, we performed a genome-wide association study using p-S65-Ub level as a quantitative trait in 1012 autopsy-confirmed Lewy body disease (LBD) samples. RESULTS We identified a significant genome-wide association with p-S65-Ub for rs429358 (apolipoprotein E ε4 [APOE4]) and a suggestive association for rs6480922 (ZMIZ1). APOE4 was associated with higher p-S65-Ub levels and greater neuropathological burden. Functional validation in mouse and human induced pluripotent stem cell (iPSC) models confirmed APOE4-mediated mitophagy alterations. Intriguingly, ZMIZ1 rs6480922 was associated with lower p-S65-Ub levels, reduced neuropathological load, and increased brain weight, indicating a potential protective role. DISCUSSION Our findings underscore the importance of mitochondrial quality control in LBD pathogenesis and nominate regulators that may contribute to disease risk or resilience. HIGHLIGHTS p-S65-Ub levels were used as a quantitative marker of mitochondrial damage. A GWAS identified two genetic variants that modify mitophagy in LBD autopsy brain. APOE4 was associated with increased p-S65-Ub accumulation and neuropathology. APOE4 altered mitophagy via pathology-dependent and pathology-independent mechanisms. ZMIZ1 was linked to reduced p-S65-Ub and neuropathology indicative of protection.
Collapse
Affiliation(s)
- Xu Hou
- Department of NeuroscienceMayo ClinicJacksonvilleFloridaUSA
| | - Michael G. Heckman
- Division of Clinical Trials and BiostatisticsMayo ClinicJacksonvilleFloridaUSA
| | - Fabienne C. Fiesel
- Department of NeuroscienceMayo ClinicJacksonvilleFloridaUSA
- Neuroscience PhD ProgramMayo Clinic Graduate School of Biomedical SciencesJacksonvilleFloridaUSA
| | - Shunsuke Koga
- Department of NeuroscienceMayo ClinicJacksonvilleFloridaUSA
| | | | | | - Jing Zhao
- Department of NeuroscienceMayo ClinicJacksonvilleFloridaUSA
| | | | - Patrick W. Johnson
- Division of Clinical Trials and BiostatisticsMayo ClinicJacksonvilleFloridaUSA
| | - Launia J. White
- Division of Clinical Trials and BiostatisticsMayo ClinicJacksonvilleFloridaUSA
| | | | - Joseph S. Reddy
- Department of Quantitative Health SciencesMayo ClinicJacksonvilleFloridaUSA
| | - Jose Bras
- Department of Neurodegenerative ScienceVan Andel InstituteGrand RapidsMichiganUSA
- Division of Psychiatry and Behavioral MedicineMichigan State University College of Human MedicineGrand RapidsMichiganUSA
| | - Rita Guerreiro
- Department of Neurodegenerative ScienceVan Andel InstituteGrand RapidsMichiganUSA
- Division of Psychiatry and Behavioral MedicineMichigan State University College of Human MedicineGrand RapidsMichiganUSA
| | - Na Zhao
- Department of NeuroscienceMayo ClinicJacksonvilleFloridaUSA
- Neuroscience PhD ProgramMayo Clinic Graduate School of Biomedical SciencesJacksonvilleFloridaUSA
| | - Guojun Bu
- Department of NeuroscienceMayo ClinicJacksonvilleFloridaUSA
- Neuroscience PhD ProgramMayo Clinic Graduate School of Biomedical SciencesJacksonvilleFloridaUSA
| | - Dennis W. Dickson
- Department of NeuroscienceMayo ClinicJacksonvilleFloridaUSA
- Neuroscience PhD ProgramMayo Clinic Graduate School of Biomedical SciencesJacksonvilleFloridaUSA
| | - Owen A. Ross
- Department of NeuroscienceMayo ClinicJacksonvilleFloridaUSA
- Neuroscience PhD ProgramMayo Clinic Graduate School of Biomedical SciencesJacksonvilleFloridaUSA
| | - Wolfdieter Springer
- Department of NeuroscienceMayo ClinicJacksonvilleFloridaUSA
- Neuroscience PhD ProgramMayo Clinic Graduate School of Biomedical SciencesJacksonvilleFloridaUSA
| |
Collapse
|
20
|
Bhalala OG, Beamish J, Eratne D, Summerell P, Porter T, Laws SM, Kang MJY, Huq AJ, Chiu WH, Cadwallader C, Walterfang M, Farrand S, Evans AH, Kelso W, Churilov L, Watson R, Yassi N, Velakoulis D, Loi SM. Blood biomarker profiles in young-onset neurocognitive disorders: A cohort study. Aust N Z J Psychiatry 2025; 59:378-388. [PMID: 39825484 PMCID: PMC11924289 DOI: 10.1177/00048674241312805] [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] [Indexed: 01/20/2025]
Abstract
INTRODUCTION Young-onset neurocognitive symptoms result from a heterogeneous group of neurological and psychiatric disorders which present a diagnostic challenge. To identify such factors, we analysed the Biomarkers in Younger-Onset Neurocognitive Disorders cohort, a study of individuals <65 years old presenting with neurocognitive symptoms for a diagnosis and who have undergone cognitive and biomarker analyses. METHODS Sixty-five participants (median age at assessment of 56 years, 45% female) were recruited during their index presentation to the Royal Melbourne Hospital Neuropsychiatry Centre, a tertiary specialist service in Melbourne, Australia, and categorized as either early-onset Alzheimer's disease (n = 18), non-Alzheimer's disease neurodegeneration (n = 23) or primary psychiatric disorders (n = 24). Levels of neurofilament light chain, glial fibrillary acidic protein and phosphorylated-tau 181, apolipoprotein E genotype and late-onset Alzheimer's disease polygenic risk scores were determined. Information-theoretic model selection identified discriminatory factors. RESULTS Neurofilament light chain, glial fibrillary acidic protein and phosphorylated-tau 181 levels were elevated in early-onset Alzheimer's disease compared with other diagnostic categories. A multi-omic model selection identified that a combination of cognitive and blood biomarkers, but not the polygenic risk score, discriminated between early-onset Alzheimer's disease and primary psychiatric disorders (area under the curve ⩾ 0.975, 95% confidence interval: 0.825-1.000). Phosphorylated-tau 181 alone significantly discriminated between early-onset Alzheimer's disease and non-Alzheimer's disease neurodegeneration causes (area under the curve = 0.950, 95% confidence interval: 0.877-1.00). DISCUSSION Discriminating between early-onset Alzheimer's disease, non-Alzheimer's disease neurodegeneration and primary psychiatric disorders causes of young-onset neurocognitive symptoms is possible by combining cognitive profiles with blood biomarkers. These results support utilizing blood biomarkers for the work-up of young-onset neurocognitive symptoms and highlight the need for the development of a young-onset Alzheimer's disease-specific polygenic risk score.
Collapse
Affiliation(s)
- Oneil G Bhalala
- Department of Medicine, Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, Australia
- Population Health and Immunity Division, Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
| | - Jessica Beamish
- Neuropsychiatry Centre, The Royal Melbourne Hospital, Parkville, VIC, Australia
| | - Dhamidhu Eratne
- Neuropsychiatry Centre, The Royal Melbourne Hospital, Parkville, VIC, Australia
- Department of Psychiatry, The University of Melbourne, Parkville, VIC, Australia
| | - Patrick Summerell
- Neuropsychiatry Centre, The Royal Melbourne Hospital, Parkville, VIC, Australia
| | - Tenielle Porter
- Centre for Precision Health, Edith Cowan University, Joondalup, WA, Australia
| | - Simon M Laws
- Centre for Precision Health, Edith Cowan University, Joondalup, WA, Australia
| | - Matthew JY Kang
- Neuropsychiatry Centre, The Royal Melbourne Hospital, Parkville, VIC, Australia
- Department of Psychiatry, The University of Melbourne, Parkville, VIC, Australia
| | - Aamira J Huq
- Department of Medicine, Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, Australia
| | - Wei-Hsuan Chiu
- Department of Psychiatry, The University of Melbourne, Parkville, VIC, Australia
| | - Claire Cadwallader
- Neuropsychiatry Centre, The Royal Melbourne Hospital, Parkville, VIC, Australia
- Memory and Aging Center, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Mark Walterfang
- Neuropsychiatry Centre, The Royal Melbourne Hospital, Parkville, VIC, Australia
- Department of Psychiatry, The University of Melbourne, Parkville, VIC, Australia
- The Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia
- School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia
| | - Sarah Farrand
- Neuropsychiatry Centre, The Royal Melbourne Hospital, Parkville, VIC, Australia
- Department of Psychiatry, The University of Melbourne, Parkville, VIC, Australia
| | - Andrew H Evans
- Department of Medicine, Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, Australia
| | - Wendy Kelso
- Neuropsychiatry Centre, The Royal Melbourne Hospital, Parkville, VIC, Australia
| | - Leonid Churilov
- Department of Medicine, Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, Australia
| | - Rosie Watson
- Department of Medicine, Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, Australia
- Population Health and Immunity Division, Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
| | - Nawaf Yassi
- Department of Medicine, Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, Australia
- Population Health and Immunity Division, Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
| | - Dennis Velakoulis
- Neuropsychiatry Centre, The Royal Melbourne Hospital, Parkville, VIC, Australia
- Department of Psychiatry, The University of Melbourne, Parkville, VIC, Australia
| | - Samantha M Loi
- Neuropsychiatry Centre, The Royal Melbourne Hospital, Parkville, VIC, Australia
- Department of Psychiatry, The University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
21
|
Cheng F, Feng Y, Yang X, Flanagan M, Chen X, Bonakdarpour B, Jamshidi P, Castellani R, Mao Q, Chu X, Gao H, Liu Y, Dou L, Xu J, Hou Y, Martin W, Nelson P, Leverenz J, Hu M, Li Y, Pieper A, Cummings J. Genomic and epigenomic insights into purkinje and granule neurons in Alzheimer's disease and related dementia using single-nucleus multiome analysis. RESEARCH SQUARE 2025:rs.3.rs-6264481. [PMID: 40235507 PMCID: PMC11998783 DOI: 10.21203/rs.3.rs-6264481/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Although the human cerebellum is known to be neuropathologically impaired in Alzheimer's disease (AD) and AD-related dementias (ADRD), the cell type-specific transcriptional and epigenomic changes that contribute to this pathology are not well understood. Here, we report single-nucleus multiome (snRNA-seq and snATAC-seq) analysis of 103,861 nuclei isolated from both cerebellum and frontal cortex of AD/ADRD patients and normal controls. Using peak-to-gene linkage analysis, we identified 431,834 significant linkages between gene expression and cell subtype-specific chromatin accessibility regions enriched for candidate cis-regulatory elements (cCREs). These cCREs were associated with AD/ADRD-specific transcriptomic changes and disease-related gene regulatory networks, especially for RAR Related Orphan Receptor A (RORA) and E74 Like ETS Transcription Factor 1 (ELF1) in cerebellar Purkinje cells and granule cells, respectively. Trajectory analysis of granule cell populations further identified disease-relevant transcription factors, such as RORA, and their regulatory targets. Finally, we pinpointed two likely causal genes, Seizure Related 6 Homolog Like 2 (SEZ6L2) in Purkinje cells and KAT8 Regulatory NSL Complex Subunit 1 (KANSL1) in granule cells, through integrative analysis of cCREs derived from snATAC-seq, genome-wide AD/ADRD loci, and three-dimensional (3D) genome data. Via CRISPRi experiments, we found that perturbation of rs4788201 and rs62056801 significantly inhibited the expression of their target genes, SEZ6L2 and KANSL1, in human iPSC-derived neurons. This cell subtype-specific regulatory landscape in the human cerebellum identified here offers novel genomic and epigenomic insights into the neuropathology and pathobiology of AD/ADRD and other neurological disorders if broadly applied.
Collapse
|
22
|
Zhang H, Zhou Z, Gu J, Lin Y, Yan Y, Chen X, Fan M, Huang Y. Genetic insights of lipid metabolism and lipid-lowering drugs with Lewy body dementia risk: Evidence from Mendelian randomization. Prog Neuropsychopharmacol Biol Psychiatry 2025; 137:111282. [PMID: 39929371 DOI: 10.1016/j.pnpbp.2025.111282] [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: 10/24/2024] [Revised: 01/29/2025] [Accepted: 02/03/2025] [Indexed: 02/13/2025]
Abstract
BACKGROUND Lewy body dementia (LBD) is the second common dementia, with unclear mechanisms and limited treatment options. Dyslipidemia has been implicated in LBD, but the role of lipid-lowering drugs remains underexplored. This study aims to investigate the association between lipid traits, drug targets, and LBD risk using Mendelian Randomization (MR) analysis. METHODS We performed univariable and multivariable MR analyses to evaluate the causal effects of lipid traits on the risk of LBD. Then, drug-target MR analysis and subtype analysis were conducted to evaluate the effects of lipid-lowering therapies on LBD. RESULTS In univariable MR, genetically predicted low-density lipoprotein cholesterol (LDL-C) and remnant cholesterol (RC) levels were associated with an increased risk of LBD. Mediation analysis suggested a potential interaction between LDL-C and RC in influencing LBD risk. Drug-target MR analysis identified significant associations between genetically proxied inhibition of ANGPTL3, CETP, and HMGCR and LBD risk. CONCLUSION This MR analysis provided evidence that elevated LDL-C and RC may increase the risk of LBD. Additionally, targeting ANGPTL3, CETP, and HMGCR may represent potential therapeutic strategies for the prevention or treatment of LBD.
Collapse
Affiliation(s)
- Hanyu Zhang
- Department of General Medicine, Huashan Hospital, Fudan University, Shanghai, PR China
| | - Zengyuan Zhou
- Department of Nutrition, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Onclogy, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jie Gu
- Department of General Medicine, Huashan Hospital, Fudan University, Shanghai, PR China
| | - Yingnan Lin
- Department of General Medicine, Huashan Hospital, Fudan University, Shanghai, PR China
| | - Yunyun Yan
- Department of General Medicine, Huashan Hospital, Fudan University, Shanghai, PR China
| | - Xiaonan Chen
- Department of General Medicine, Huashan Hospital, Fudan University, Shanghai, PR China
| | - Meixiang Fan
- Department of General Medicine, Huashan Hospital, Fudan University, Shanghai, PR China
| | - Yanyan Huang
- Department of General Medicine, Huashan Hospital, Fudan University, Shanghai, PR China; National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan Universiy, Shanghai, PR China; Tianqiao and Chrissy Chen Institute Clinic Translational Research Center, Shanghai, PR China; Department of Geriatrics, Huashan Hospital, Fudan University, Shanghai, PR China.
| |
Collapse
|
23
|
Wang P, Luan H, Li S, Han X, Sun W, Gong J, Xu C, Chen R, Wei C. Extensive perivascular spaces burden causally affects neurodegenerative diseases and brain structure: A two-sample bidirectional Mendelian randomization study. Prog Neuropsychopharmacol Biol Psychiatry 2025; 137:111284. [PMID: 39921030 DOI: 10.1016/j.pnpbp.2025.111284] [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: 07/01/2024] [Revised: 01/30/2025] [Accepted: 02/04/2025] [Indexed: 02/10/2025]
Abstract
BACKGROUND Extensive perivascular spaces (PVS) burden has been reported to be associated with neurodegenerative diseases and brain structure; however, the causal effects has not been determined yet. Therefore, this study aimed to investigate the causal effect of extensive PVS burden on neurodegenerative diseases and brain structure through Mendelian randomization (MR) analysis. METHODS Two-sample bidirectional MR was conducted based on publicly available genome-wide association studies (GWAS) summary statistics. Causal estimates of extensive PVS burden on neurodegenerative diseases and brain structure were primarily assessed using the inverse-variance weighted (IVW) method, supplemented by additional methods, including MR-Egger, weighted median, simple mode, and weighted mode. Sensitivity analyses were performed to assess heterogeneity and pleiotropy. In addition, we explored whether brain structure act as a mediating factor in the pathway from extensive PVS burden to neurodegenerative diseases. RESULTS Our MR study found that extensive PVS burden in white matter (WM-PVS) burden was associated with lower Alzheimer's disease (AD) risk (IVW OR (95 % CI) = 0.963(0.929 to 0.999), P = 0.0428), with no heterogeneity and pleiotropy detected. In addition, following FDR correction, we found bidirectional causal relationships between extensive PVS burden and brain structure. Moreover, our results of the mediated analysis showed that the surface area of parahippocampal, as a mediating variable, plays an important role in the causal relationship between WM-PVS and AD. The mediation effect is 18 %. CONCLUSIONS Our study provides evidence for the causal associations of different extensive PVS burden phenotypes with neurodegenerative diseases and brain structures, improving our understanding of the complex relationships between different brain injuries.
Collapse
Affiliation(s)
- Pin Wang
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - Heya Luan
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - Shaoqi Li
- College of Integrated Traditional Chinese and Western Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Xiaodong Han
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - Wenxian Sun
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - Jin Gong
- College of Integrated Traditional Chinese and Western Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Chang Xu
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - Runqi Chen
- School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Cuibai Wei
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, China.
| |
Collapse
|
24
|
Harvey J, Imm J, Kouhsar M, Smith AR, Creese B, Smith RG, Wheildon G, Chouliaras L, Shireby G, Jaunmuktane Z, De Pablo-Fernández E, Warner T, Lett D, Gveric D, Brooks H, Attems J, Thomas A, Dempster E, Ballard C, O’Brien JT, Aarsland D, Mill J, Pihlstrøm L, Pishva E, Lunnon K. Interrogating DNA methylation associated with Lewy body pathology in a cross brain-region and multi-cohort study. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.03.13.25323837. [PMID: 40162278 PMCID: PMC11952592 DOI: 10.1101/2025.03.13.25323837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Lewy body (LB) diseases are an umbrella term encompassing a range of neurodegenerative conditions all characterized by the hallmark of intra-neuronal α-synuclein associated with the development of motor and cognitive dysfunction. In this study, we have conducted a large meta-analysis of DNA methylation across multiple cortical brain regions, in relation to increasing burden of LB pathology. Utilizing a combined dataset of 1239 samples across 855 unique donors, we identified a set of 30 false discovery rate (FDR) significant loci that are differentially methylated in association with LB pathology, the most significant of which were located in UBASH3B and PTAFR, as well as an intergenic locus. Ontological enrichment analysis of our meta-analysis results highlights several neurologically relevant traits, including synaptic, inflammatory and vascular alterations. We leverage our summary statistics to compare DNA methylation signatures between different neurodegenerative pathologies and highlight a shared epigenetic profile across LB diseases, Alzheimer's disease and Huntington's disease, although the top-ranked loci show disease specificity. Finally, utilizing summary statistics from previous large-scale genome-wide association studies we report FDR significant enrichment of DNA methylation differences with respect to increasing LB pathology in the SNCA genomic region, a gene previously associated with Parkinson's disease and dementia with Lewy bodies.
Collapse
Affiliation(s)
- Joshua Harvey
- Department of Clinical and Biomedical Science, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| | - Jennifer Imm
- Department of Clinical and Biomedical Science, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| | - Morteza Kouhsar
- Department of Clinical and Biomedical Science, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| | - Adam R. Smith
- Department of Clinical and Biomedical Science, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| | - Byron Creese
- Department of Clinical and Biomedical Science, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
- Department of Life Sciences, College of Health, Medicine and Life Sciences, Brunel University of London, London, UK
| | - Rebecca G. Smith
- Department of Clinical and Biomedical Science, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| | - Gregory Wheildon
- Department of Clinical and Biomedical Science, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| | - Leonidas Chouliaras
- Department of Psychiatry, University of Cambridge School of Clinical Medicine, Cambridge, UK
- Specialty Dementia and Frailty Service, Essex Partnership University NHS Foundation Trust, St Margaret’s Hospital. Epping, UK
| | - Gemma Shireby
- Great Ormond Street Hospital, University College London, London, UK
| | - Zane Jaunmuktane
- Queen Square Brain Bank for Neurological Disorders, University College London Queen Square Institute of Neurology, University College London, London, UK
| | - Eduardo De Pablo-Fernández
- Queen Square Brain Bank for Neurological Disorders, University College London Queen Square Institute of Neurology, University College London, London, UK
| | - Thomas Warner
- Queen Square Brain Bank for Neurological Disorders, University College London Queen Square Institute of Neurology, University College London, London, UK
| | - Debbie Lett
- Newcastle Brain Tissue Resource, Newcastle University, Newcastle, UK
| | - Djordje Gveric
- MS and Parkinson’s Tissue Bank, Department of Brain Sciences, Imperial College London, London, UK
| | - Hannah Brooks
- The Oxford Brain Bank, University of Oxford, Oxford, UK
| | - Johannes Attems
- Newcastle Brain Tissue Resource, Newcastle University, Newcastle, UK
| | - Alan Thomas
- Newcastle Brain Tissue Resource, Newcastle University, Newcastle, UK
| | - Emma Dempster
- Department of Clinical and Biomedical Science, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| | - Clive Ballard
- Department of Clinical and Biomedical Science, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| | - John T O’Brien
- Department of Psychiatry, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - Dag Aarsland
- Institute of Psychiatry, Psychology & Neuroscience (IoPPN), Kings College London, UK
| | - Jonathan Mill
- Department of Clinical and Biomedical Science, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| | - Lasse Pihlstrøm
- Institute of Clinical Medicine, Oslo University Hospital, Oslo, Norway
| | - Ehsan Pishva
- Department of Clinical and Biomedical Science, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNS), Maastricht University, Maastricht, The Netherlands
| | - Katie Lunnon
- Department of Clinical and Biomedical Science, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| |
Collapse
|
25
|
Gao YL, Xu QH. No causal link between smoking and dementia with Lewy bodies. Parkinsonism Relat Disord 2025:107775. [PMID: 40089453 DOI: 10.1016/j.parkreldis.2025.107775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Accepted: 03/06/2025] [Indexed: 03/17/2025]
Abstract
This letter addresses the findings of Goodheart and Gomperts (2024), which observed an association between smoking and reduced odds of dementia with Lewy bodies (DLB). In contrast, our Mendelian randomization analysis found no causal link, suggesting that further research using genetically informed methods is needed to clarify these results.
Collapse
Affiliation(s)
- Yu-Liang Gao
- Department of Neurology, Quzhou Hospital of Traditional Chinese Medicine, Quzhou, Zhejiang, PR China
| | - Qiu-Han Xu
- School of Medicine, Zhejiang University, Hangzhou, Zhejiang, PR China.
| |
Collapse
|
26
|
Mitsumori R, Asanomi Y, Morizono T, Shigemizu D, Niida S, Ozaki K. A genome-wide association study identifies a novel East Asian-specific locus for dementia with Lewy bodies in Japanese subjects. Mol Med 2025; 31:87. [PMID: 40045203 PMCID: PMC11884146 DOI: 10.1186/s10020-025-01115-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 02/04/2025] [Indexed: 03/09/2025] Open
Abstract
BACKGROUND Dementia with Lewy bodies (DLB) is the second most common type of degenerative dementia in older patients. As with other multifactorial diseases, the pathogenesis results from interactions of environmental and genetic factors. The genetic basis of DLB is not yet fully understood. Recent genomic analyses of DLB in Caucasian cohorts identified genetic susceptibility loci for DLB, but the comprehensive genomic analysis in Asians was still not performed. METHODS We conducted a genome-wide association study (GWAS) in Japanese subjects (211 DLB cases and 6113 controls) to clarify the genetic architecture of DLB pathogenesis. RESULTS We identified the East Asian-specific DHTKD1 locus (rs138587229) on chromosome 10 with genome-wide significance (GWS; P = 3.27 × 10-8) and the ICOS/PARD3B locus on chromosome 2 with suggestive significance (P = 3.95 × 10-7) as novel DLB genetic risk loci. We also confirmed the APOE locus (rs429358, P < 5.0 × 10-8), a known risk locus for DLB and Alzheimer's disease in Caucasians. The DHTKD1 locus was associated with the gene expression of SEC61A2 and showed a causal relationship with cholinesterase levels. In a trans-ethnic meta-analysis that included Japanese, UK Biobank, and other Caucasian GWAS, we confirmed the risk for DLB at APOE and SNCA loci with GWS. Transcriptome-wide association analysis identified ZNF155 and ZNF284 in the brain cortex and GPRIN3 in the substantia nigra as putative causal genes for DLB. CONCLUSIONS This is the first GWAS for DLB in East Asians, and our findings provide new biological and clinical insights into the pathogenesis of DLB.
Collapse
Affiliation(s)
- Risa Mitsumori
- Medical Genome Center, National Center for Geriatrics and Gerontology, Research Institute, 7-430 Morioka-Cho, Obu, Aichi, 474-8511, Japan
| | - Yuya Asanomi
- Medical Genome Center, National Center for Geriatrics and Gerontology, Research Institute, 7-430 Morioka-Cho, Obu, Aichi, 474-8511, Japan
| | - Takashi Morizono
- Medical Genome Center, National Center for Geriatrics and Gerontology, Research Institute, 7-430 Morioka-Cho, Obu, Aichi, 474-8511, Japan
| | - Daichi Shigemizu
- Medical Genome Center, National Center for Geriatrics and Gerontology, Research Institute, 7-430 Morioka-Cho, Obu, Aichi, 474-8511, Japan
- Department of Cardiovascular Medicine, Hiroshima University Graduate School of Biomedical and Health Sciences, 1-2-3 Kasumi, Minami-Ku, Hiroshima, 734-8553, Japan
| | - Shumpei Niida
- National Center for Geriatrics and Gerontology, Research Institute, 7-430 Morioka-Cho, Obu, Aichi, 474-8511, Japan
| | - Kouichi Ozaki
- Medical Genome Center, National Center for Geriatrics and Gerontology, Research Institute, 7-430 Morioka-Cho, Obu, Aichi, 474-8511, Japan.
- Department of Cardiovascular Medicine, Hiroshima University Graduate School of Biomedical and Health Sciences, 1-2-3 Kasumi, Minami-Ku, Hiroshima, 734-8553, Japan.
- RIKEN Center for Integrative Medical Sciences, 1-7-22 Suehiro-Cho, Tsurumi-Ku, Yokohama, Kanagawa, 230-0045, Japan.
| |
Collapse
|
27
|
Vialle RA, de Paiva Lopes K, Li Y, Ng B, Schneider JA, Buchman AS, Wang Y, Farfel JM, Barnes LL, Wingo AP, Wingo TS, Seyfried NT, De Jager PL, Gaiteri C, Tasaki S, Bennett DA. Structural variants linked to Alzheimer's disease and other common age-related clinical and neuropathologic traits. Genome Med 2025; 17:20. [PMID: 40038788 PMCID: PMC11881306 DOI: 10.1186/s13073-025-01444-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 02/24/2025] [Indexed: 03/06/2025] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is a complex neurodegenerative disorder with substantial genetic influence. While genome-wide association studies (GWAS) have identified numerous risk loci for late-onset AD (LOAD), the functional mechanisms underlying most of these associations remain unresolved. Large genomic rearrangements, known as structural variants (SVs), represent a promising avenue for elucidating such mechanisms within some of these loci. METHODS By leveraging data from two ongoing cohort studies of aging and dementia, the Religious Orders Study and Rush Memory and Aging Project (ROS/MAP), we performed genome-wide association analysis testing 20,205 common SVs from 1088 participants with whole genome sequencing (WGS) data. A range of Alzheimer's disease and other common age-related clinical and neuropathologic traits were examined. RESULTS First, we mapped SVs across 81 AD risk loci and discovered 22 SVs in linkage disequilibrium (LD) with GWAS lead variants and directly associated with the phenotypes tested. The strongest association was a deletion of an Alu element in the 3'UTR of the TMEM106B gene, in high LD with the respective AD GWAS locus and associated with multiple AD and AD-related disorders (ADRD) phenotypes, including tangles density, TDP-43, and cognitive resilience. The deletion of this element was also linked to lower TMEM106B protein abundance. We also found a 22-kb deletion associated with depression in ROS/MAP and bearing similar association patterns as GWAS SNPs at the IQCK locus. In addition, we leveraged our catalog of SV-GWAS to replicate and characterize independent findings in SV-based GWAS for AD and five other neurodegenerative diseases. Among these findings, we highlight the replication of genome-wide significant SVs for progressive supranuclear palsy (PSP), including markers for the 17q21.31 MAPT locus inversion and a 1483-bp deletion at the CYP2A13 locus, along with other suggestive associations, such as a 994-bp duplication in the LMNTD1 locus, suggestively linked to AD and a 3958-bp deletion at the DOCK5 locus linked to Lewy body disease (LBD) (P = 3.36 × 10-4). CONCLUSIONS While still limited in sample size, this study highlights the utility of including analysis of SVs for elucidating mechanisms underlying GWAS loci and provides a valuable resource for the characterization of the effects of SVs in neurodegenerative disease pathogenesis.
Collapse
Affiliation(s)
- Ricardo A Vialle
- Rush Alzheimer's Disease Center, Rush University Medical Center, 1750 W Harrison St, Chicago, IL, 60612, USA.
| | - Katia de Paiva Lopes
- Rush Alzheimer's Disease Center, Rush University Medical Center, 1750 W Harrison St, Chicago, IL, 60612, USA
| | - Yan Li
- Rush Alzheimer's Disease Center, Rush University Medical Center, 1750 W Harrison St, Chicago, IL, 60612, USA
| | - Bernard Ng
- Rush Alzheimer's Disease Center, Rush University Medical Center, 1750 W Harrison St, Chicago, IL, 60612, USA
| | - Julie A Schneider
- Rush Alzheimer's Disease Center, Rush University Medical Center, 1750 W Harrison St, Chicago, IL, 60612, USA
| | - Aron S Buchman
- Rush Alzheimer's Disease Center, Rush University Medical Center, 1750 W Harrison St, Chicago, IL, 60612, USA
| | - Yanling Wang
- Rush Alzheimer's Disease Center, Rush University Medical Center, 1750 W Harrison St, Chicago, IL, 60612, USA
| | - Jose M Farfel
- Rush Alzheimer's Disease Center, Rush University Medical Center, 1750 W Harrison St, Chicago, IL, 60612, USA
| | - Lisa L Barnes
- Rush Alzheimer's Disease Center, Rush University Medical Center, 1750 W Harrison St, Chicago, IL, 60612, USA
| | - Aliza P Wingo
- Department of Psychiatry, University of California, Davis, Davis, CA, USA
- VA Northern California Health Care System, Davis, CA, USA
| | - Thomas S Wingo
- Department of Neurology, University of California, Davis, Davis, CA, USA
| | - Nicholas T Seyfried
- Department of Neurology and Department of Biochemistry, Goizueta Alzheimer's Disease Research Center, Emory University School of Medicine, Atlanta, GA, USA
| | - Philip L De Jager
- Center for Translational and Computational Neuroimmunology, Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
| | - Chris Gaiteri
- Rush Alzheimer's Disease Center, Rush University Medical Center, 1750 W Harrison St, Chicago, IL, 60612, USA
- Department of Psychiatry, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Shinya Tasaki
- Rush Alzheimer's Disease Center, Rush University Medical Center, 1750 W Harrison St, Chicago, IL, 60612, USA
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, 1750 W Harrison St, Chicago, IL, 60612, USA
| |
Collapse
|
28
|
Nassan M, Daghlas I, Diamond BR, Martersteck A, Rogalski E. The causal association between resting state intrinsic functional networks and neurodegeneration. Brain Commun 2025; 7:fcaf098. [PMID: 40103583 PMCID: PMC11913654 DOI: 10.1093/braincomms/fcaf098] [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: 05/31/2024] [Revised: 11/30/2024] [Accepted: 03/02/2025] [Indexed: 03/20/2025] Open
Abstract
Alterations of resting state intrinsic functional networks have been associated with neurodegenerative diseases even before the onset of cognitive symptoms. Emerging hypotheses propose a role of resting state intrinsic functional networks alterations in the risk or vulnerability to neurodegeneration. It is unknown whether intrinsic functional network alterations can be causal for neurodegenerative diseases. We sought to answer this question using two-sample Mendelian randomization. Using the largest genome-wide association study of resting state intrinsic functional connectivity (n = 47 276), we generated genetic instruments (at the significance level 2.8 ×10-11) to proxy resting state intrinsic functional network features. Based on the known brain regions implicated in different neurodegenerative diseases, we generated genetically proxied resting state intrinsic functional features and tested their association with their paired neurodegenerative outcomes: features in parieto-temporal regions and Alzheimer dementia (111 326 cases, 677 663 controls); frontal region and frontotemporal dementia (2154 cases, 4308 controls); temporal pole region and semantic dementia (308 cases, 616 controls), and occipital region with Lewy body dementia (LBD) (2591 cases, 4027 controls). Major depressive disorder outcome (170 756 cases, 329 443 controls) was included as a positive control and tested for its association with genetically proxied default mode network (DMN) exposure. Inverse-variance weighted analysis was used to estimate the association between the exposures (standard deviation units) and outcomes. Power and sensitivity analyses were completed to assess the robustness of the results. None of the genetically proxied functional network features were significantly associated with neurodegenerative outcomes (adjusted P value >0.05), despite sufficient calculated power. Two resting state features in the visual cortex showed a nominal level of association with LBD (P = 0.01), a finding that was replicated using a different instrument (P = 0.03). The genetically proxied DMN connectivity was associated with the risk of depression (P = 0.024), supporting the validity of the genetic instruments. Sensitivity analyses were supportive of the main results. This is the first study to comprehensively assess the potential causal effect of resting state intrinsic functional network features on the risk of neurodegeneration. Overall, the results do not support a causal role for the tested associations. However, we report a nominal association between visual network connectivity and Lewy body dementia that requires further evaluation.
Collapse
Affiliation(s)
- Malik Nassan
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Northwestern University, Chicago, IL 60611, USA
| | - Iyas Daghlas
- Department of Neurology, University of California San Francisco, San Francisco, CA 94143, USA
| | - Bram R Diamond
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Northwestern University, Chicago, IL 60611, USA
| | - Adam Martersteck
- Healthy Aging and Alzheimer's Research Care (HAARC) Center, University of Chicago, Chicago, IL 60637, USA
| | - Emily Rogalski
- Healthy Aging and Alzheimer's Research Care (HAARC) Center, University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
29
|
Yin KF, Chen T, Gu XJ, Jiang Z, Su WM, Duan QQ, Wen XJ, Cao B, Li JR, Chi LY, Chen YP. Identification of Potential Causal Genes for Neurodegenerative Diseases by Mitochondria-Related Genome-Wide Mendelian Randomization. Mol Neurobiol 2025; 62:3892-3902. [PMID: 39347895 DOI: 10.1007/s12035-024-04528-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Accepted: 09/23/2024] [Indexed: 10/01/2024]
Abstract
Current research lacks comprehensive investigations into the potential causal link between mitochondrial-related genes and the risk of neurodegenerative diseases (NDDs). We aimed to identify potential causative genes for five NDDs through an examination of mitochondrial-related gene expression levels. Through the integration of summary statistics from expression quantitative trait loci (eQTL) datasets (human blood and brain tissue), mitochondrial DNA copy number (mtDNA-CN), and genome-wide association studies (GWAS) datasets of five NDDs from European ancestry, we conducted a Mendelian randomization (MR) analysis to explore the potential causal relationship between mitochondrial-related genes and Alzheimer's disease (AD), Parkinson's disease (PD), amyotrophic lateral sclerosis (ALS), frontotemporal dementia (FTD), and Lewy body dementia (LBD). Sensitivity analysis and Bayesian colocalization were employed to validate this causal relationship. Through MR analysis, we have identified potential causal relationships between 12 mitochondria-related genes and AD, PD, ALS, and FTD overlapping with motor neuron disease (FTD_MND) in human blood or brain tissue. Bayesian colocalization analysis further confirms 9 causal genes, including NDUFS2, EARS2, and MRPL41 for AD; NDUFAF2, MALSU1, and METTL8 for PD; MYO19 and MRM1 for ALS; and FASTKD1 for FTD_MND. Importantly, in both human blood and brain tissue, NDUFS2 exhibits a significant pathogenic effect on AD, while NDUFAF2 demonstrates a robust protective effect on PD. Additionally, the mtDNA-CN plays a protected role in LBD (OR = 0.62, p = 0.031). This study presents evidence establishing a causal relationship between mitochondrial dysfunction and NDDs. Furthermore, the identified candidate genes may serve as potential targets for drug development aimed at preventing NDDs.
Collapse
Affiliation(s)
- Kang-Fu Yin
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
- Institute of Brain Science and Brain-Inspired Technology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Ting Chen
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
- Institute of Brain Science and Brain-Inspired Technology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Xiao-Jing Gu
- Mental Health Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Zheng Jiang
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
- Institute of Brain Science and Brain-Inspired Technology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Wei-Ming Su
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
- Institute of Brain Science and Brain-Inspired Technology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Qing-Qing Duan
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
- Institute of Brain Science and Brain-Inspired Technology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Xiang-Jin Wen
- West China School of Medicine, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Bei Cao
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
- Institute of Brain Science and Brain-Inspired Technology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Ju-Rong Li
- Department of Geriatrics, Dazhou Central Hospital, Dazhou, 635000, Sichuan, China
| | - Li-Yi Chi
- Department of Neurology, First Affiliated Hospital of Air Force Military Medical University, Xi'an, 710072, Shaanxi, China
| | - Yong-Ping Chen
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
- Institute of Brain Science and Brain-Inspired Technology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
30
|
Reus LM, Boltz T, Francia M, Bot M, Ramesh N, Koromina M, Pijnenburg YAL, den Braber A, van der Flier WM, Visser PJ, van der Lee SJ, Tijms BM, Teunissen CE, Loohuis LO, Ophoff RA. Quantitative trait loci mapping of circulating metabolites in cerebrospinal fluid to uncover biological mechanisms involved in brain-related phenotypes. Mol Psychiatry 2025:10.1038/s41380-025-02934-0. [PMID: 40021830 DOI: 10.1038/s41380-025-02934-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 12/16/2024] [Accepted: 02/12/2025] [Indexed: 03/03/2025]
Abstract
Genomic studies of molecular traits have provided mechanistic insights into complex disease, though these lag behind for brain-related traits due to the inaccessibility of brain tissue. We leveraged cerebrospinal fluid (CSF) to study neurobiological mechanisms in vivo, measuring 5543 CSF metabolites, the largest panel in CSF to date, in 977 individuals of European ancestry. Individuals originated from two separate cohorts including cognitively healthy subjects (n = 490) and a well-characterized memory clinic sample, the Amsterdam Dementia Cohort (ADC, n = 487). We performed metabolite quantitative trait loci (mQTL) mapping on CSF metabolomics and found 126 significant mQTLs, representing 65 unique CSF metabolites across 51 independent loci. To better understand the role of CSF mQTLs in brain-related disorders we integrated our CSF mQTL results with pre-existing summary statistics on brain traits, identifying 34 genetic associations between CSF metabolites and brain traits. Over 90% of significant mQTLs demonstrated colocalized associations with brain-specific gene expression, unveiling potential neurobiological pathways.
Collapse
Affiliation(s)
- Lianne M Reus
- Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, CA, USA.
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands.
- Amsterdam Neuroscience, Neurodegeneration, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands.
| | - Toni Boltz
- Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, CA, USA.
- Department of Human Genetics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA.
| | - Marcelo Francia
- Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, CA, USA
| | - Merel Bot
- Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, CA, USA
| | - Naren Ramesh
- Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, CA, USA
| | - Maria Koromina
- Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, NY, USA
| | - Yolande A L Pijnenburg
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
| | - Anouk den Braber
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
- Department of Biological Psychology, Amsterdam Public Health Research Institute, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Wiesje M van der Flier
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
- Epidemiology and Data Science, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
| | - Pieter Jelle Visser
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
- Department of Psychiatry, Maastricht University, Maastricht, The Netherlands
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Karolinska Institutet, Stockholm, Sweden
| | - Sven J van der Lee
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
- Section Genomics of Neurodegenerative Diseases and Aging, Department of Human Genetics, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
| | - Betty M Tijms
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
| | - Charlotte E Teunissen
- Neurochemistry Lab, Department of Laboratory Medicine, Amsterdam Neuroscience, Neurodegeneration, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Loes Olde Loohuis
- Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, CA, USA
- Department of Human Genetics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
- Department of Computational Medicine, University of California, Los Angeles, CA, USA
| | - Roel A Ophoff
- Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, CA, USA.
- Department of Human Genetics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA.
- Department of Psychiatry, Erasmus University Medical Center, Rotterdam, The Netherlands.
| |
Collapse
|
31
|
Rosal AE, Martin SL, Strafella AP. The role of Apolipoprotein E4 on cognitive impairment in Parkinson's disease and Parkinsonisms. Front Neurosci 2025; 19:1515374. [PMID: 40052092 PMCID: PMC11882537 DOI: 10.3389/fnins.2025.1515374] [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: 10/22/2024] [Accepted: 02/03/2025] [Indexed: 03/09/2025] Open
Abstract
Cognitive impairment is a prevalent non-motor symptom of Parkinson's disease (PD), increasing the risk of dementia as the disease progresses. Despite its clinical significance, the etiology of cognitive impairment in PD remains unclear. Apolipoprotein E4 (APOE4), a well-known genetic risk factor of Alzheimer's disease, has been studied for its potential role in PD-related cognitive impairment. However, findings have been conflicting and thus inconclusive, highlighting a need to critically evaluate the current research. Several studies using neuroimaging modalities have explored the brains of individuals with PD and atypical parkinsonian disorders who have APOE4. Some of these studies have identified distinct neuropathological changes that have been previously reported to be associated with cognitive impairments in those with Parkinsonisms. Here, we review the role of APOE4 on cognitive impairment in PD and atypical Parkinsonisms using neuroimaging evidence. We will examine how APOE4 may contribute to pathological changes within the brain and its association with cognitive impairment.
Collapse
Affiliation(s)
- Angenelle Eve Rosal
- Brain Health Imaging Centre, Centre for Addiction and Mental Health, Toronto, ON, Canada
- Institute of Medical Sciences, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Sarah L. Martin
- Brain Health Imaging Centre, Centre for Addiction and Mental Health, Toronto, ON, Canada
- Translation and Computational Neurosciences Unit (TCNU), Faculty of Health and Education, Manchester Metropolitan University, Manchester, United Kingdom
| | - Antonio P. Strafella
- Brain Health Imaging Centre, Centre for Addiction and Mental Health, Toronto, ON, Canada
- Institute of Medical Sciences, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Edmond J. Safra Parkinson Disease Program, Neurology Division, Toronto Western Hospital and Krembil Brain Institute, University Health Network, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
32
|
Stefani A, Antelmi E, Arnaldi D, Arnulf I, During E, Högl B, Hu MMT, Iranzo A, Luke R, Peever J, Postuma RB, Videnovic A, Gan-Or Z. From mechanisms to future therapy: a synopsis of isolated REM sleep behavior disorder as early synuclein-related disease. Mol Neurodegener 2025; 20:19. [PMID: 39934903 PMCID: PMC11817540 DOI: 10.1186/s13024-025-00809-0] [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: 10/14/2024] [Accepted: 02/05/2025] [Indexed: 02/13/2025] Open
Abstract
Parkinson disease (PD), dementia with Lewy bodies (DLB) and multiple system atrophy are synucleinopathies, characterized by neuronal loss, gliosis and the abnormal deposition of α-synuclein in vulnerable areas of the nervous system. Neurodegeneration begins however several years before clinical onset of motor, cognitive or autonomic symptoms. The isolated form of REM sleep behavior disorder (RBD), a parasomnia with dream enactment behaviors and excessive muscle activity during REM sleep, is an early stage synucleinopathy. The neurophysiological hallmark of RBD is REM sleep without atonia (RWSA), i.e. the loss of physiological muscle atonia during REM sleep. RBD pathophysiology is not fully clarified yet, but clinical and basic science suggest that ɑ-syn pathology begins in the lower brainstem where REM atonia circuits are located, including the sublaterodorsal tegmental/subcoeruleus nucleus and the ventral medulla, then propagates rostrally to brain regions such as the substantia nigra, limbic system, cortex. Genetically, there is only a partial overlap between RBD, PD and DLB, and individuals with iRBD may represent a specific subpopulation. A genome-wide association study identified five loci, which all seem to revolve around the GBA1 pathway. iRBD patients often show subtle motor, cognitive, autonomic and/or sensory signs, neuroimaging alterations as well as biofluid and tissue markers of neurodegeneration (in particular pathologic α-synuclein aggregates), which can be useful for risk stratification. Patients with iRBD represent thus the ideal population for neuroprotective/neuromodulating trials. This review provides insights into these aspects, highlighting and substantiating the central role of iRBD in treatment development strategies for synucleinopathies.
Collapse
Affiliation(s)
| | - Elena Antelmi
- DIMI Department of Engineering and Medicine of Innovation, University of Verona, Verona, Italy
| | - Dario Arnaldi
- Clinical Neurophysiology, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
- DINOGMI, University of Genoa, Genoa, Italy
| | - Isabelle Arnulf
- Sleep Clinic, Pitié-Salpêtrière Hospital, APHP - Sorbonne University, Paris, France
- Paris Brain Institute, Paris, France
| | - Emmanuel During
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Birgit Högl
- Medical University Innsbruck, Innsbruck, Austria
| | - Michele M T Hu
- Division of Neurology, Nuffield Department of Clinical Neurosciences, Oxford University, Oxford, UK
| | - Alex Iranzo
- Sleep Unit, Neurology Service, Hospital Clínic de Barcelona, IDIBAPS, CIBERNED: CB06/05/0018-ISCIII, Universitat de Barcelona,, Barcelona, Spain
| | - Russell Luke
- Department of Cell and System Biology, University of Toronto, Toronto, ON, Canada
| | - John Peever
- Department of Cell and System Biology, University of Toronto, Toronto, ON, Canada
| | - Ronald B Postuma
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
- The Neuro (Montreal Neurological Institute-Hospital), Montreal, QC, Canada
| | - Aleksandar Videnovic
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, USA
| | - Ziv Gan-Or
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada.
- The Neuro (Montreal Neurological Institute-Hospital), Montreal, QC, Canada.
- Department of Human Genetics, McGill University, Montreal, QC, Canada.
| |
Collapse
|
33
|
Piga I, Sajic T. Editorial: Next generation of omics analysis to study lipid-rich tissues. Front Endocrinol (Lausanne) 2025; 16:1561447. [PMID: 39980851 PMCID: PMC11839433 DOI: 10.3389/fendo.2025.1561447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Accepted: 01/24/2025] [Indexed: 02/22/2025] Open
Affiliation(s)
| | - Tatjana Sajic
- Faculty Unit of Toxicology, University Center of Legal Medicine, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
- Lausanne University Hospital and Geneva University Hospital, Lausanne, Switzerland
| |
Collapse
|
34
|
Kouhsar M, Weymouth L, Smith AR, Imm J, Bredemeyer C, Wedatilake Y, Torkamani A, Bergh S, Selbæk G, Mill J, Ballard C, Sweet RA, Kofler J, Creese B, Pishva E, Lunnon K. A brain DNA co-methylation network analysis of psychosis in Alzheimer's disease. Alzheimers Dement 2025; 21:e14501. [PMID: 39936280 PMCID: PMC11815327 DOI: 10.1002/alz.14501] [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: 05/28/2024] [Revised: 11/22/2024] [Accepted: 12/03/2024] [Indexed: 02/13/2025]
Abstract
INTRODUCTION The presence of psychosis in Alzheimer's disease (AD) is suggested to be associated with distinct molecular and neuropathological profiles in the brain. METHODS We assessed brain DNA methylation in AD donors with psychosis (AD+P) and without psychosis (AD-P) using the EPIC array. Weighted gene correlation network analysis identified modules of co-methylated genes in a discovery cohort (PITT-ADRC: N = 113 AD+P, N = 40 AD-P), with validation in an independent cohort (BDR: N = 79 AD+P, N = 117 AD-P), with Gene Ontology and cell-type enrichment analysis. Genetic data were integrated to identify methylation quantitative trait loci (mQTLs), which were co-localized with GWAS for related traits. RESULTS We replicated one AD+P associated module, which was enriched for synaptic pathways and in excitatory and inhibitory neurons. mQTLs in this module co-localized with variants associated with schizophrenia and educational attainment. DISCUSSION This represents the largest epigenetic study of AD+P to date, identifying pleiotropic relationships between AD+P and related traits. HIGHLIGHTS DNA methylation was assessed in the prefrontal cortex in subjects with AD+P and AD-P. WGCNA identified six modules of co-methylated loci associated with AD+P in a discovery cohort. One of the modules was replicated in an independent cohort. This module was enriched for synaptic genes and in excitatory and inhibitory neurons. mQTLs mapping to genes in the module co-localized with GWAS loci for schizophrenia and educational attainment.
Collapse
Affiliation(s)
- Morteza Kouhsar
- Department of Clinical and Biomedical SciencesFaculty of Health and Life SciencesUniversity of ExeterExeterDevonUK
| | - Luke Weymouth
- Department of Clinical and Biomedical SciencesFaculty of Health and Life SciencesUniversity of ExeterExeterDevonUK
| | - Adam R. Smith
- Department of Clinical and Biomedical SciencesFaculty of Health and Life SciencesUniversity of ExeterExeterDevonUK
| | - Jennifer Imm
- Department of Clinical and Biomedical SciencesFaculty of Health and Life SciencesUniversity of ExeterExeterDevonUK
| | - Claudia Bredemeyer
- Department of Clinical and Biomedical SciencesFaculty of Health and Life SciencesUniversity of ExeterExeterDevonUK
| | - Yehani Wedatilake
- Norwegian National Centre for Aging and HealthVestfold Hospital TrustTønsbergNorway
- Research Centre for Age‐related Functional Decline and DiseaseInnlandet Hospital TrustOttestadNorway
| | | | - Sverre Bergh
- Norwegian National Centre for Aging and HealthVestfold Hospital TrustTønsbergNorway
- Research Centre for Age‐related Functional Decline and DiseaseInnlandet Hospital TrustOttestadNorway
| | - Geir Selbæk
- Norwegian National Centre for Aging and HealthVestfold Hospital TrustTønsbergNorway
- Department of Geriatric MedicineOslo University HospitalNydalenOsloNorway
| | - Jonathan Mill
- Department of Clinical and Biomedical SciencesFaculty of Health and Life SciencesUniversity of ExeterExeterDevonUK
| | - Clive Ballard
- Department of Clinical and Biomedical SciencesFaculty of Health and Life SciencesUniversity of ExeterExeterDevonUK
| | - Robert A. Sweet
- Department of PsychiatryUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Julia Kofler
- Department of PathologyUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Byron Creese
- Department of Clinical and Biomedical SciencesFaculty of Health and Life SciencesUniversity of ExeterExeterDevonUK
- Division of PsychologyDepartment of Life SciencesBrunel University LondonUxbridgeUK
| | - Ehsan Pishva
- Department of Clinical and Biomedical SciencesFaculty of Health and Life SciencesUniversity of ExeterExeterDevonUK
- Department of Psychiatry and NeuropsychologySchool for Mental Health and Neuroscience (MHeNs)Faculty of HealthMedicine and Life Sciences (FHML)Maastricht UniversityMaastrichtThe Netherlands
| | - Katie Lunnon
- Department of Clinical and Biomedical SciencesFaculty of Health and Life SciencesUniversity of ExeterExeterDevonUK
| |
Collapse
|
35
|
Vattathil SM, Gerasimov ES, Canon SM, Lori A, Tan SSM, Kim PJ, Liu Y, Lai EC, Bennett DA, Wingo TS, Wingo AP. Mapping the microRNA landscape in the older adult brain and its genetic contribution to neuropsychiatric conditions. NATURE AGING 2025; 5:306-319. [PMID: 39643657 PMCID: PMC11839474 DOI: 10.1038/s43587-024-00778-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 11/07/2024] [Indexed: 12/09/2024]
Abstract
MicroRNAs (miRNAs) play a crucial role in regulating gene expression and influence many biological processes. Despite their importance, understanding of how genetic variation affects miRNA expression in the brain and how this relates to brain disorders remains limited. Here we investigated these questions by identifying microRNA expression quantitative trait loci (miR-QTLs), or genetic variants associated with brain miRNA levels, using genome-wide small RNA sequencing profiles from dorsolateral prefrontal cortex samples of 604 older adult donors of European ancestry. Here we show that nearly half (224 of 470) of the analyzed miRNAs have associated miR-QTLs, many of which fall in regulatory regions such as brain promoters and enhancers. We also demonstrate that intragenic miRNAs often have genetic regulation independent from their host genes. Furthermore, by integrating our findings with 16 genome-wide association studies of psychiatric and neurodegenerative disorders, we identified miRNAs that likely contribute to bipolar disorder, depression, schizophrenia and Parkinson's disease. These findings advance understanding of the genetic regulation of miRNAs and their role in brain health and disease.
Collapse
Affiliation(s)
- Selina M Vattathil
- Department of Neurology, University of California, Davis, Sacramento, CA, USA
| | | | - Se Min Canon
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA
| | - Adriana Lori
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA
| | - Sarah Sze Min Tan
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA
| | - Paul J Kim
- Department of Psychiatry, Emory University School of Medicine, Atlanta, GA, USA
| | - Yue Liu
- Department of Neurology, University of California, Davis, Sacramento, CA, USA
| | - Eric C Lai
- Developmental Biology Program, Sloan Kettering Institute, New York, NY, USA
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Thomas S Wingo
- Department of Neurology, University of California, Davis, Sacramento, CA, USA.
- Alzheimer's Disease Research Center, University of California, Davis, Sacramento, CA, USA.
| | - Aliza P Wingo
- Department of Psychiatry, University of California, Davis, Sacramento, CA, USA.
- Veterans Affairs Northern California Health Care System, Sacramento, CA, USA.
| |
Collapse
|
36
|
Zhang C, Liu X, Wang J, Zhou G, Geng B, Hu M, Tang Q. Investigating the causal relationships between lipid traits and dementia with lewy bodies: A mendelian randomization study. Parkinsonism Relat Disord 2025:107312. [PMID: 39909750 DOI: 10.1016/j.parkreldis.2025.107312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Revised: 01/25/2025] [Accepted: 01/29/2025] [Indexed: 02/07/2025]
Abstract
BACKGROUND Disruptions in lipid metabolism have been implicated in various neurodegenerative diseases. However, the specific role of lipid species in the pathogenesis of dementia with Lewy bodies (DLB) remains poorly understood. This study aims to investigate potential causal relationships between lipid traits and DLB risk using Mendelian randomization (MR). METHODS We employed MR analysis to assess causal associations between 179 lipid traits and DLB, utilizing data from comprehensive genome-wide association studies (GWAS). The lipid-related GWAS included 7174 participants, and the DLB-related GWAS included 2981 DLB cases and 4391 healthy controls. RESULTS Genetic predispositions to increased levels of phosphatidylinositol (PI) (18:1_20:4) were associated with an elevated risk of DLB. Conversely, genetic predispositions to increased levels of specific phosphatidylcholine (PC) species, including PC (O-18:1_20:4), PC (O-16:0_20:4) and PC (O-18:0_20:4), were found to be protective against DLB. Sensitivity analyses revealed no evidence of heterogeneity or horizontal pleiotropy among the selected instrumental variables. CONCLUSIONS Our MR study identifies specific lipid species potentially causally linked to DLB risk. Elevated levels of PI (18:1_20:4) were associated with increased DLB risk, while higher levels of certain PC species were found to be protective. These findings offer new insights into the lipid-related mechanisms underlying DLB pathogenesis and highlight potential therapeutic targets.
Collapse
Affiliation(s)
- Canwen Zhang
- Department of Neurology, The Second Affiliated Hospital of Shandong First Medical University, Taian, 271000, China.
| | - Xiaoshuang Liu
- Department of Nursing, Tai an Hospital of Traditional Chinese Medicine, Taian, 271000, China.
| | - Juan Wang
- Department of Neurology, The Second Affiliated Hospital of Shandong First Medical University, Taian, 271000, China.
| | - Guoqing Zhou
- Department of Neurology, The Second Affiliated Hospital of Shandong First Medical University, Taian, 271000, China.
| | - Biao Geng
- Department of Pharmacy, The Second Affiliated Hospital of Shandong First Medical University, Taian, 271000, China.
| | - Ming Hu
- Department of Neurology, The Second Affiliated Hospital of Shandong First Medical University, Taian, 271000, China.
| | - Qin Tang
- Department of Neurology, The Second Affiliated Hospital of Shandong First Medical University, Taian, 271000, China.
| |
Collapse
|
37
|
Liu Z, Song SY. Genomic and Transcriptomic Approaches Advance the Diagnosis and Prognosis of Neurodegenerative Diseases. Genes (Basel) 2025; 16:135. [PMID: 40004464 PMCID: PMC11855287 DOI: 10.3390/genes16020135] [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: 12/23/2024] [Revised: 01/16/2025] [Accepted: 01/22/2025] [Indexed: 02/27/2025] Open
Abstract
Neurodegenerative diseases, such as Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD), and amyotrophic lateral sclerosis (ALS), represent a growing societal challenge due to their irreversible progression and significant impact on patients, caregivers, and healthcare systems. Despite advances in clinical and imaging-based diagnostics, these diseases are often detected at advanced stages, limiting the effectiveness of therapeutic interventions. Recent breakthroughs in genomic and transcriptomic technologies, including whole-genome sequencing, single-cell RNA sequencing (scRNA-seq), and CRISPR-based screens, have revolutionized the field, offering new avenues for early diagnosis and personalized prognosis. Genomic approaches have elucidated disease-specific genetic risk factors and molecular pathways, while transcriptomic studies have identified stage-specific biomarkers that correlate with disease progression and severity. Furthermore, genome-wide association studies (GWAS), polygenic risk scores (PRS), and spatial transcriptomics are enabling the stratification of patients based on their risk profiles and prognostic trajectories. Advances in functional genomics have uncovered actionable targets, such as ATXN2 in ALS and TREM2 in AD, paving the way for tailored therapeutic strategies. Despite these achievements, challenges remain in translating genomic discoveries into clinical practice due to disease heterogeneity and the complexity of neurodegenerative pathophysiology. Future integration of genetic technologies holds promise for transforming diagnostic and prognostic paradigms, offering hope for improved patient outcomes and precision medicine approaches.
Collapse
Affiliation(s)
- Zheng Liu
- Pathology Department, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
| | - Si-Yuan Song
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
38
|
Zhao K, Nie L, Zhao J, Dong Y, Jin K, Wang S, Ye X. Association between osteoarthritis and cognitive function: results from the NHANES 2011-2014 and Mendelian randomization study. Ther Adv Musculoskelet Dis 2025; 17:1759720X241304189. [PMID: 39839677 PMCID: PMC11748087 DOI: 10.1177/1759720x241304189] [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: 02/07/2024] [Accepted: 11/14/2024] [Indexed: 01/23/2025] Open
Abstract
Background Previous meta-analyses have demonstrated osteoarthritis (OA) is associated with an increased risk of dementia, but these studies were prone to bias based on residual confounding factors and reverse causality. Objectives We aimed to investigate associations between OA and cognitive function using data from the National Health and Nutrition Examination Survey (NHANES) and to investigate the causality using Mendelian randomization (MR). Design This is a cross-sectional study and MR study. Methods Data from the NHANES 2011-2014 were used. Multiple linear, logistic regressions and stratified analyses were used to determine the association between OA status and cognitive function. Sample weights were used to ensure result generalizability. Two-sample MR analysis was conducted to examine the association between OA and dementia. Mediation analyses were performed to investigate the mediating effects of depression. Results We did not demonstrate a significant association between OA and cognitive performance after adjusting for relevant covariates (p > 0.05), and the population of individuals with both OA and depression was associated with higher odds of low total word recall cognitive performance (odds ratio (OR) = 4.74, 95% confidence interval (CI): 1.09-20.63; p = 0.04). Genetically predicted specific-site OA was not significantly associated with the risk of dementia (OR = 1.12; 95% CI: 0.96-1.32; p = 0.16), Alzheimer's disease (OR = 0.95, 95% CI: 0.68-1.31, p = 0.74), vascular dementia (OR = 1.32, 95% CI: 0.82-2.13, p = 0.25) with accepted heterogeneity and no evidence of directional pleiotropy. Furthermore, major depression was found to mediate the pathway between OA and vascular dementia (β = 0.044, 95% CI: -0.391 to 0.479, p < 0.05). Conclusion Our findings indicate that there is no significant association or causal relationship between OA and cognitive decline. However, depression may serve as an important factor influencing cognitive outcomes. Future research should further explore the bidirectional causal relationship and underlying mechanisms.
Collapse
Affiliation(s)
- Kun Zhao
- Center for Rehabilitation Medicine, Rehabilitation & Sports Medicine Research Institute of Zhejiang Province, Department of Rehabilitation Medicine, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Liuyan Nie
- Department of Rheumatology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Jingting Zhao
- Center for Rehabilitation Medicine, Rehabilitation & Sports Medicine Research Institute of Zhejiang Province, Department of Rehabilitation Medicine, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Yankai Dong
- College of Life Science, Northwest University, Xian, Shaanxi, China
| | - Kaixiu Jin
- Big Data Research and Biostatistics Center, Hangzhou Muyomu Healthcare Data Co. Ltd, Hangzhou, Zhejiang, China
| | - Song Wang
- Department of Urology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China
| | - Xiangming Ye
- Center for Rehabilitation Medicine, Rehabilitation & Sports Medicine Research Institute of Zhejiang Province, Department of Rehabilitation Medicine, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, 158 Shangtang Road, Hangzhou, Zhejiang 310014, China
| |
Collapse
|
39
|
Su B, He Z, Mao L, Huang X. The causal role of lipids in dementia: A Mendelian randomization study. J Alzheimers Dis Rep 2025; 9:25424823241312106. [PMID: 40034502 PMCID: PMC11864250 DOI: 10.1177/25424823241312106] [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: 07/15/2024] [Accepted: 11/26/2024] [Indexed: 03/05/2025] Open
Abstract
Background Increasing evidence suggests that abnormal lipid metabolism is one of the pathogeneses of dementia. It is necessary to reveal the relationship between lipids and dementia. Objective This study used bidirectional two-sample Mendelian randomization to explore the causal relationship between 179 lipid species and the risk of dementia. Methods We assessed the causal effects of 179 lipid species and four subtypes of dementia including Alzheimer's disease (AD), vascular dementia (VaD), frontotemporal dementia (FTD), and dementia with Lewy bodies (DLB). Inverse variance weighting, MR-Egger method, weighted median, simple mode, and weighted mode were used to analyze the relationship between lipids and dementia. Cochran's Q, MR-Egger intercept test, and MR-PRESSO test were used to test the heterogeneity and pleiotropy of the results. In addition, we performed an inverse MR analysis testing the causal effects of dementia on lipids. Results Our study revealed causal effects of glycerophospholipid, glycerolipid, and sterol on the risk of dementia. Phosphatidylcholine, phosphatidylinositol, and triglycerides play significant roles in AD. Notably, phosphatidylcholine played a protective role in both FTD and DLB. However, this study did not observe a significant effect of phosphatidylinositol on FTD. In the case of VaD, not only glycerophospholipid, but also glycerolipid, exerted an influence, but sterol was also a risk factor. Conclusions Our study provided new evidence supporting the causal role of genetically predicted lipid species in dementia. Future clinical trials are necessary to evaluate the potential role of lipid levels in dementia prevention.
Collapse
Affiliation(s)
- Boyang Su
- Medical School of Chinese PLA, Beijing, China
- Neurological Department of the First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Zhengqing He
- Department of Neurology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Li Mao
- Neurological Department of the First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Xusheng Huang
- Medical School of Chinese PLA, Beijing, China
- Neurological Department of the First Medical Center, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
40
|
Cocoș R, Popescu BO. Scrutinizing neurodegenerative diseases: decoding the complex genetic architectures through a multi-omics lens. Hum Genomics 2024; 18:141. [PMID: 39736681 DOI: 10.1186/s40246-024-00704-7] [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: 10/05/2024] [Accepted: 12/10/2024] [Indexed: 01/01/2025] Open
Abstract
Neurodegenerative diseases present complex genetic architectures, reflecting a continuum from monogenic to oligogenic and polygenic models. Recent advances in multi-omics data, coupled with systems genetics, have significantly refined our understanding of how these data impact neurodegenerative disease mechanisms. To contextualize these genetic discoveries, we provide a comprehensive critical overview of genetic architecture concepts, from Mendelian inheritance to the latest insights from oligogenic and omnigenic models. We explore the roles of common and rare genetic variants, gene-gene and gene-environment interactions, and epigenetic influences in shaping disease phenotypes. Additionally, we emphasize the importance of multi-omics layers including genomic, transcriptomic, proteomic, epigenetic, and metabolomic data in elucidating the molecular mechanisms underlying neurodegeneration. Special attention is given to missing heritability and the contribution of rare variants, particularly in the context of pleiotropy and network pleiotropy. We examine the application of single-cell omics technologies, transcriptome-wide association studies, and epigenome-wide association studies as key approaches for dissecting disease mechanisms at tissue- and cell-type levels. Our review introduces the OmicPeak Disease Trajectory Model, a conceptual framework for understanding the genetic architecture of neurodegenerative disease progression, which integrates multi-omics data across biological layers and time points. This review highlights the critical importance of adopting a systems genetics approach to unravel the complex genetic architecture of neurodegenerative diseases. Finally, this emerging holistic understanding of multi-omics data and the exploration of the intricate genetic landscape aim to provide a foundation for establishing more refined genetic architectures of these diseases, enhancing diagnostic precision, predicting disease progression, elucidating pathogenic mechanisms, and refining therapeutic strategies for neurodegenerative conditions.
Collapse
Affiliation(s)
- Relu Cocoș
- Department of Medical Genetics, 'Carol Davila' University of Medicine and Pharmacy, Bucharest, Romania.
- Genomics Research and Development Institute, Bucharest, Romania.
| | - Bogdan Ovidiu Popescu
- Department of Clinical Neurosciences, 'Carol Davila' University of Medicine and Pharmacy, Bucharest, Romania.
| |
Collapse
|
41
|
Shwab EK, Man Z, Gingerich DC, Gamache J, Garrett ME, Serrano GE, Beach TG, Crawford GE, Ashley-Koch AE, Chiba-Falek O. Comparative mapping of single-cell transcriptomic landscapes in neurodegenerative diseases. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.13.628436. [PMID: 39764045 PMCID: PMC11702568 DOI: 10.1101/2024.12.13.628436] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/11/2025]
Abstract
INTRODUCTION Alzheimer's disease (AD), Dementia with Lewy bodies (DLB), and Parkinson's disease (PD) represent a spectrum of neurodegenerative disorders (NDDs). Here, we performed the first direct comparison of their transcriptomic landscapes. METHODS We profiled the whole transcriptomes of NDD cortical tissue by snRNA-seq. We used computational analyses to identify common and distinct differentially expressed genes (DEGs), biological pathways, vulnerable and disease-driver cell subtypes, and alteration in cell-to-cell interactions. RESULTS The same vulnerable inhibitory neuron subtype was depleted in both AD and DLB. Potentially disease-driving neuronal cell subtypes were present in both PD and DLB. Cell-cell communication was predicted to be increased in AD but decreased in DLB and PD. DEGs were most commonly shared across NDDs within inhibitory neuron subtypes. Overall, we observed the greatest transcriptomic divergence between AD and PD, while DLB exhibited an intermediate transcriptomic signature. DISCUSSION These results help explain the clinicopathological spectrum of this group of NDDs and provide unique insights into the shared and distinct molecular mechanisms underlying the pathogenesis of NDDs.
Collapse
Affiliation(s)
- E. Keats Shwab
- Division of Translational Brain Sciences, Department of Neurology, Duke University Medical Center, Durham, NC, 27710, USA
- Center for Genomic and Computational Biology, Duke University Medical Center, Durham, NC, 27708, USA
| | - Zhaohui Man
- Division of Translational Brain Sciences, Department of Neurology, Duke University Medical Center, Durham, NC, 27710, USA
- Center for Genomic and Computational Biology, Duke University Medical Center, Durham, NC, 27708, USA
| | - Daniel C. Gingerich
- Division of Translational Brain Sciences, Department of Neurology, Duke University Medical Center, Durham, NC, 27710, USA
- Center for Genomic and Computational Biology, Duke University Medical Center, Durham, NC, 27708, USA
| | - Julia Gamache
- Division of Translational Brain Sciences, Department of Neurology, Duke University Medical Center, Durham, NC, 27710, USA
- Center for Genomic and Computational Biology, Duke University Medical Center, Durham, NC, 27708, USA
| | - Melanie E. Garrett
- Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC, 27701, USA
| | - Geidy E. Serrano
- Banner Sun Health Research Institute, Sun City, Arizona, 85351, USA
| | - Thomas G. Beach
- Banner Sun Health Research Institute, Sun City, Arizona, 85351, USA
| | - Gregory E. Crawford
- Center for Genomic and Computational Biology, Duke University Medical Center, Durham, NC, 27708, USA
- Department of Pediatrics, Division of Medical Genetics, Duke University Medical Center, Durham, NC, 27708, USA
- Center for Advanced Genomic Technologies, Duke University Medical Center, Durham, NC, 27708, USA
| | - Allison E. Ashley-Koch
- Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC, 27701, USA
- Department of Medicine, Duke University Medical Center, Durham, NC, 27708, USA
| | - Ornit Chiba-Falek
- Division of Translational Brain Sciences, Department of Neurology, Duke University Medical Center, Durham, NC, 27710, USA
- Center for Genomic and Computational Biology, Duke University Medical Center, Durham, NC, 27708, USA
| |
Collapse
|
42
|
Palushaj B, Lewis SJG, Abdelnour C. What is the future for dementia with Lewy bodies? J Neurol 2024; 272:43. [PMID: 39666092 DOI: 10.1007/s00415-024-12734-1] [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: 10/07/2024] [Revised: 11/13/2024] [Accepted: 11/15/2024] [Indexed: 12/13/2024]
Abstract
Dementia with Lewy bodies (DLB) is the second most common neurodegenerative dementia after Alzheimer's disease (AD), yet it remains under-recognized and frequently misdiagnosed due to heterogenous clinical presentations, the presence of co-pathology, and the lack of specific diagnostic tools. Pathologically, DLB is characterized by the accumulation of misfolded alpha-synuclein (aSyn) aggregates, known as Lewy bodies. Recent advancements have improved in vivo detection of aSyn pathology through techniques such as seed amplification assays, monoclonal antibodies, and positron emission tomography using novel small-molecule ligands. The ability to detect aSyn in vivo has sparked dialogue about using biomarkers to identify individuals with aSyn, similar to the approach influencing the field of AD. Proponents argue that biological staging could facilitate the detection of preclinical disease stages, allowing for earlier intervention and targets for disease modification, and could improve diagnostic sensitivity and accuracy in selecting patients for clinical trials. However, critics caution that this method may oversimplify the complexity of DLB and overlook its clinical heterogeneity, also highlighting practical challenges related to implementation, cost, and global access to advanced diagnostic technologies. Importantly, although significant progress has been made in detecting aSyn for diagnostic purposes, disease-modifying therapies targeting aSyn have yet to demonstrate clear efficacy in slowing disease progression. Elucidating the physiological and pathophysiological roles of aSyn remains an urgent priority in neurodegenerative research. Other experimental research priorities for DLB include developing improved cellular and animal models that reflect epigenetic and environmental factors, mapping post-translational modifications, and systematically characterizing neurons that are vulnerable and resistant to lewy pathology using a multi-omic approach. Clinically, there is an urgent need for international, prospective, longitudinal studies and for validated, disease-specific outcome measures. Addressing these priorities is essential for advancing our understanding of DLB and developing effective therapies.
Collapse
Affiliation(s)
- Bianca Palushaj
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA.
| | | | - Carla Abdelnour
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
43
|
Galvin JE. Lewy Body Dementia. Continuum (Minneap Minn) 2024; 30:1673-1698. [PMID: 39620839 DOI: 10.1212/con.0000000000001496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2024]
Abstract
OBJECTIVE Lewy body dementia (LBD) is an umbrella term describing two closely related conditions: Parkinson disease dementia (PDD) and dementia with Lewy bodies (DLB). LBD is the second most common cause of neurodegenerative dementia but is often underrecognized in clinical practice. This review covers the key epidemiologic, clinical, cognitive, behavioral, and biomarker features of LBD and discusses current treatment options. LATEST DEVELOPMENTS Indicative biomarkers of LBD improve the ability to make a diagnosis and include single-photon emission computed tomography (SPECT) of the dopamine system (brain) and the noradrenergic system (cardiac), and polysomnography. α-Synuclein-specific biomarkers in spinal fluid, skin, plasma, and brain imaging are in active development with some available for clinical use. Prodromal stages of PDD and DLB have been contextualized, and diagnostic criteria have been published. An emerging theme is whether an integrated staging system focusing on protein aggregation, rather than clinical symptoms, may advance research efforts. ESSENTIAL POINTS LBD is a common cause of cognitive impairment in older adults but is often subject to significant delays in diagnosis and treatment, increasing the burden on patients and family care partners. Understanding key features of disease and the use of biomarkers will improve recognition. Earlier detection may also facilitate the development of new therapeutics and enrollment in clinical trials.
Collapse
|
44
|
Álvarez Jerez P, Wild Crea P, Ramos DM, Gustavsson EK, Radefeldt M, Damianov A, Makarious MB, Ojo OO, Billingsley KJ, Malik L, Daida K, Bromberek S, Hu F, Schneider Z, Surapaneni AL, Stadler J, Rizig M, Morris HR, Pantazis CB, Leonard HL, Screven L, Qi YA, Nalls MA, Bandres-Ciga S, Hardy J, Houlden H, Eng C, Burchard EG, Kachuri L, Lin CH, Black DL, Singleton AB, Fischer S, Bauer P, Reed X, Ryten M, Beetz C, Ward M, Okubadejo NU, Blauwendraat C. African ancestry neurodegeneration risk variant disrupts an intronic branchpoint in GBA1. Nat Struct Mol Biol 2024; 31:1955-1963. [PMID: 39668204 PMCID: PMC11638064 DOI: 10.1038/s41594-024-01423-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 10/10/2024] [Indexed: 12/14/2024]
Abstract
Recently, an African ancestry-specific Parkinson disease (PD) risk signal was identified at the gene encoding glucocerebrosidase (GBA1). This variant ( rs3115534 -G) is carried by ~50% of West African PD cases and imparts a dose-dependent increase in risk for disease. The risk variant has varied frequencies across African ancestry groups but is almost absent in European and Asian ancestry populations. GBA1 is a gene of high clinical and therapeutic interest. Damaging biallelic protein-coding variants cause Gaucher disease and monoallelic variants confer risk for PD and dementia with Lewy bodies, likely by reducing the function of glucocerebrosidase. Interestingly, the African ancestry-specific GBA1 risk variant is a noncoding variant, suggesting a different mechanism of action. Using full-length RNA transcript sequencing, we identified partial intron 8 expression in risk variant carriers (G) but not in nonvariant carriers (T). Antibodies targeting the N terminus of glucocerebrosidase showed that this intron-retained isoform is likely not protein coding and subsequent proteomics did not identify a shorter protein isoform, suggesting that the disease mechanism is RNA based. Clustered regularly interspaced short palindromic repeats editing of the reported index variant ( rs3115534 ) revealed that this is the sequence alteration responsible for driving the production of these transcripts containing intron 8. Follow-up analysis of this variant showed that it is in a key intronic branchpoint sequence and, therefore, has important implications in splicing and disease. In addition, when measuring glucocerebrosidase activity, we identified a dose-dependent reduction in risk variant carriers. Overall, we report the functional effect of a GBA1 noncoding risk variant, which acts by interfering with the splicing of functional GBA1 transcripts, resulting in reduced protein levels and reduced glucocerebrosidase activity. This understanding reveals a potential therapeutic target in an underserved and underrepresented population.
Collapse
Affiliation(s)
- Pilar Álvarez Jerez
- Center for Alzheimer's and Related Dementias, National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
- Laboratory of Neurogenetics, National Institute on Aging, Bethesda, MD, USA
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Peter Wild Crea
- Laboratory of Neurogenetics, National Institute on Aging, Bethesda, MD, USA
| | - Daniel M Ramos
- Center for Alzheimer's and Related Dementias, National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Emil K Gustavsson
- Genetics and Genomic Medicine, Great Ormond Street Institute of Child Health, University College London, London, UK
| | | | - Andrey Damianov
- Department of Microbiology, Immunology and Molecular Genetics, The David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Mary B Makarious
- Center for Alzheimer's and Related Dementias, National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
- DataTecnica, Washington, DC, USA
| | - Oluwadamilola O Ojo
- College of Medicine, University of Lagos, Lagos, Nigeria
- Lagos University Teaching Hospital, Lagos, Nigeria
| | - Kimberley J Billingsley
- Center for Alzheimer's and Related Dementias, National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
- Laboratory of Neurogenetics, National Institute on Aging, Bethesda, MD, USA
| | - Laksh Malik
- Center for Alzheimer's and Related Dementias, National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Kensuke Daida
- Laboratory of Neurogenetics, National Institute on Aging, Bethesda, MD, USA
| | - Sarah Bromberek
- Center for Alzheimer's and Related Dementias, National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Fangle Hu
- Center for Alzheimer's and Related Dementias, National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Zachary Schneider
- Laboratory of Neurogenetics, National Institute on Aging, Bethesda, MD, USA
| | - Aditya L Surapaneni
- Department of Medicine, New York University Langone School of Medicine, New York, NY, USA
| | - Julia Stadler
- Center for Alzheimer's and Related Dementias, National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Mie Rizig
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Huw R Morris
- UCL Movement Disorders Centre, University College London, London, UK
| | - Caroline B Pantazis
- Center for Alzheimer's and Related Dementias, National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Hampton L Leonard
- Center for Alzheimer's and Related Dementias, National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
- DataTecnica, Washington, DC, USA
| | - Laurel Screven
- Center for Alzheimer's and Related Dementias, National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Yue A Qi
- Center for Alzheimer's and Related Dementias, National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Mike A Nalls
- Center for Alzheimer's and Related Dementias, National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
- DataTecnica, Washington, DC, USA
| | - 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, MD, USA
| | - John Hardy
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Henry Houlden
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Celeste Eng
- Department of Biotherapeutic Sciences and Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Esteban González Burchard
- Department of Biotherapeutic Sciences and Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Linda Kachuri
- Department of Epidemiology and Population Health, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Chia-Ho Lin
- Department of Microbiology, Immunology and Molecular Genetics, The David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Douglas L Black
- Department of Microbiology, Immunology and Molecular Genetics, The David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - 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, MD, USA
- Laboratory of Neurogenetics, National Institute on Aging, Bethesda, MD, USA
| | | | | | - Xylena Reed
- Center for Alzheimer's and Related Dementias, National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Mina Ryten
- Genetics and Genomic Medicine, Great Ormond Street Institute of Child Health, University College London, London, UK
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
- UK Dementia Research Institute and Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | | | - Michael Ward
- Center for Alzheimer's and Related Dementias, National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
- Neurogenetics Branch, National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA
| | - Njideka U Okubadejo
- College of Medicine, University of Lagos, Lagos, Nigeria
- Lagos University Teaching Hospital, Lagos, Nigeria
| | - 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, MD, USA.
- Laboratory of Neurogenetics, National Institute on Aging, Bethesda, MD, USA.
| |
Collapse
|
45
|
Wu Y, Sun Z, Zheng Q, Miao J, Dorn S, Mukherjee S, Fletcher JM, Lu Q. Pervasive biases in proxy genome-wide association studies based on parental history of Alzheimer's disease. Nat Genet 2024; 56:2696-2703. [PMID: 39496879 PMCID: PMC11929606 DOI: 10.1038/s41588-024-01963-9] [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: 10/26/2023] [Accepted: 09/27/2024] [Indexed: 11/06/2024]
Abstract
Almost every recent Alzheimer's disease (AD) genome-wide association study (GWAS) has performed meta-analysis to combine studies with clinical diagnosis of AD with studies that use proxy phenotypes based on parental disease history. Here, we report major limitations in current GWAS-by-proxy (GWAX) practices due to uncorrected survival bias and nonrandom participation in parental illness surveys, which cause substantial discrepancies between AD GWAS and GWAX results. We demonstrate that the current AD GWAX provide highly misleading genetic correlations between AD risk and higher education, which subsequently affects a variety of genetic epidemiological applications involving AD and cognition. Our study sheds light on potential issues in the design and analysis of middle-aged biobank cohorts and underscores the need for caution when interpreting genetic association results based on proxy-reported parental disease history.
Collapse
Affiliation(s)
- Yuchang Wu
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, WI, USA
| | - Zhongxuan Sun
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, WI, USA
| | - Qinwen Zheng
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, WI, USA
| | - Jiacheng Miao
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, WI, USA
| | - Stephen Dorn
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, WI, USA
| | | | - Jason M Fletcher
- Department of Population Health Sciences, University of Wisconsin-Madison, Madison, WI, USA
- La Follette School of Public Affairs, University of Wisconsin-Madison, Madison, WI, USA
| | - Qiongshi Lu
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
46
|
Scholz SW, Cobos I. Genetics and Neuropathology of Neurodegenerative Dementias. Continuum (Minneap Minn) 2024; 30:1801-1822. [PMID: 39620845 DOI: 10.1212/con.0000000000001505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2024]
Abstract
OBJECTIVE This article provides an overview of the current understanding of the genetic and pathologic features of neurodegenerative dementias, with an emphasis on Alzheimer disease and related dementias. LATEST DEVELOPMENTS In recent years, there has been substantial progress in genetic research, contributing significant knowledge to our understanding of the molecular risk factors involved in neurodegenerative dementia syndromes. Several genes have been linked to monogenic forms of dementia (eg, APP, PSEN1, PSEN2, SNCA, GRN, C9orf72, MAPT) and an even larger number of genetic variants are known to influence susceptibility for developing dementia. As anti-amyloid therapies for patients with early-stage Alzheimer disease have entered the clinical arena, screening for the apolipoprotein E ε4 high-risk allele has come into focus, emphasizing the importance of genetic counseling. Similarly, advances in the pathologic classifications of neurodegenerative dementia syndromes and molecular pathology highlight their heterogeneity and overlapping features and provide insights into the pathogenesis of these conditions. ESSENTIAL POINTS Recent progress in neurogenetics and molecular pathology has improved our understanding of the complex pathogenetic changes associated with neurodegenerative dementias, facilitating improved disease modeling, enhanced diagnostics, and individualized counseling. The hope is that this knowledge will ultimately pave the way for the development of novel therapeutics.
Collapse
|
47
|
Li T, Yi J, Hu X, Wu H, Wang K, Zhou B. Association between dementia, Alzheimer's disease, and liver cancer: A Mendelian randomization analysis. J Alzheimers Dis Rep 2024; 8:1587-1595. [PMID: 40034364 PMCID: PMC11863743 DOI: 10.1177/25424823241299661] [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: 05/31/2024] [Accepted: 10/21/2024] [Indexed: 03/05/2025] Open
Abstract
Background Epidemiological studies have indicated an inverse association between neurodegenerative diseases and cancer. However, the relationship between liver cancer and dementia, particularly Alzheimer's disease (AD), remains underexplored. Objective This study aimed to determine the association between dementia, specifically AD, and liver cancer using Mendelian randomization (MR) analysis. Methods We conducted a bidirectional, two-sample MR analysis using publicly available genome-wide association studies data. The inverse-variance weighted method was employed as the primary analytical approach. To detect and correct for the effects of horizontal pleiotropy, we applied three complementary methods: MR Egger, weighted median, and Maximum likelihood. Results The analysis indicated significant associations between dementia and a reduced risk of hepatocellular carcinoma (HCC) (OR: 0.87; 95%CI: 0.81-0.95; p < 0.001) and intrahepatic cholangiocarcinoma (ICC) (OR: 0.81; 95%CI: 0.72-0.92; p < 0.001). AD was significantly associated with a decreased risk of HCC (OR: 0.94; 95%CI: 0.88-0.99; p = 0.033), ICC (OR: 0.85; 95%CI: 0.78-0.93; p < 0.001), and combined hepatocellular-cholangiocarcinoma (CHC) (OR: 0.64; 95%CI: 0.43-0.93; p = 0.020). Conversely, inverse MR analyses indicated that ICC was associated with increased dementia risk (OR: 1.05; 95%CI: 1.01-1.09; p = 0.019) and CHC with increased AD risk (OR: 1.03; 95%CI: 1.00-1.04; p = 0.014). Conclusions This study suggests that dementia, particularly AD, is associated with a reduced risk of liver cancer. Conversely, liver cancer may be associated with a slightly increased risk of developing dementia and AD, although some observational studies have reported a lower risk of these conditions among cancer survivors.
Collapse
Affiliation(s)
- Tianze Li
- Hepato-Biliary-Pancreatic Surgery Division, Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Queen Mary School, Nanchang University, Nanchang, Jiangxi Province, P.R. China
| | - Jianwei Yi
- Hepato-Biliary-Pancreatic Surgery Division, Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Xuliang Hu
- Hepato-Biliary-Pancreatic Surgery Division, Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Huajun Wu
- Hepato-Biliary-Pancreatic Surgery Division, Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Kai Wang
- Hepato-Biliary-Pancreatic Surgery Division, Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Jiangxi Province Engineering Research Center of Hepatobiliary Disease, Nanchang, China
| | - Binghai Zhou
- Hepato-Biliary-Pancreatic Surgery Division, Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
48
|
Kimura T, Fujita K, Sakurai T, Niida S, Ozaki K, Shigemizu D. Whole-genome sequencing to identify rare variants in East Asian patients with dementia with Lewy bodies. NPJ AGING 2024; 10:52. [PMID: 39572598 PMCID: PMC11582613 DOI: 10.1038/s41514-024-00180-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 10/25/2024] [Indexed: 11/24/2024]
Abstract
Dementia with Lewy bodies (DLB) is the second most common form of age-related dementia, following Alzheimer's disease (AD). DLB is associated with a worse prognosis than AD and is characterized by a more rapid progression of cognitive impairment and a poorer quality of life. In addition, the pathogenesis of DLB is less understood than that of AD, and only three genes-SNCA (α-synuclein), APOE (apolipoprotein E), and GBA1 (glucosylceramidase beta 1)-have been convincingly demonstrated to be associated with DLB. In this study, we utilized whole-genome sequencing data from 1744 Japanese individuals, comprising 45 DLB patients and 1699 cognitively normal older adults, aiming to identify new genes associated with DLB. Our genome-wide association studies of genes with potentially deleterious mutations identified the CDH23 gene as being associated with DLB, reaching a Bonferroni-corrected significance (P = 7.43 × 10-4). The gene contained three ethnicity-specific heterozygous missense variants (rs181275139, rs563688802, and rs137937502). CDH23 has been linked to deafness syndromes, and DLB patients carrying these mutations displayed symptoms of subjective hearing loss, suggesting a potential association between DLB onset and auditory impairment. Additionally, we explored human leukocyte antigen (HLA) genotypes associated with DLB but found no significant associations. This result suggests that the pathology of DLB differs from that of Parkinson's disease, which has been reported to have an association with HLA. Although a limitation of this study is the lack of replication of our findings, which require further validation in independent cohorts, our study enhances the understanding of the etiology of DLB in the Japanese population and provides new insights into the underlying mechanisms of its pathogenesis.
Collapse
Affiliation(s)
- Tetsuaki Kimura
- Medical Genome Center, Research Institute, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan
| | - Kosuke Fujita
- Department of Prevention and Care Science, Research Institute, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan
| | - Takashi Sakurai
- Department of Prevention and Care Science, Research Institute, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan
| | - Shumpei Niida
- Research Institute, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan
| | - Kouichi Ozaki
- Medical Genome Center, Research Institute, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan
- RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa, Japan
- Department of Cardiovascular Medicine, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| | - Daichi Shigemizu
- Medical Genome Center, Research Institute, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan.
- Department of Cardiovascular Medicine, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, Japan.
| |
Collapse
|
49
|
Somerville EN, Gan-Or Z. Genetic-based diagnostics of Parkinson's disease and other Parkinsonian syndromes. Expert Rev Mol Diagn 2024:1-13. [PMID: 39545628 DOI: 10.1080/14737159.2024.2427625] [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: 08/20/2024] [Accepted: 11/06/2024] [Indexed: 11/17/2024]
Abstract
INTRODUCTION Parkinson's disease (PD) is a complex disorder with vast clinical heterogeneity. Recent genetic, imaging and clinical evidence suggest that there are multiple subtypes of PD, and perhaps even distinct clinical entities, which are being diagnosed under the umbrella of PD. These might have similar clinical presentation, but potentially different underlying mechanisms, which, in future, will require different treatments. Despite extensive genetic research progress, genetic testing is still not a common practice in clinical patient care. AREAS COVERED This review examines the numerous genes that have been discovered to affect the risk of, or cause, PD. We also outline genetic variants that affect PD age at onset, its progression, and the presence or severity of motor and non-motor symptoms. We differentiate between PD, other synucleinopathies, and atypical parkinsonism syndromes, and describe genes responsible for familial forms of typical PD and atypical parkinsonism. Lastly, we present current clinical trails that are underway for targeted therapies, particularly for GBA1-PD and LRRK2-PD which are the most significant subtypes. EXPERT OPINION While genetic studies alone cannot be diagnostic for PD, proper utilization of genetic screening for PD could improve diagnostic accuracy and predictions for prognosis, guide treatment, and identify individuals that qualify for clinical trials.
Collapse
Affiliation(s)
- Emma N Somerville
- The Neuro (Montréal Neurological Institute-Hospital), McGill University, Montréal, Canada
- Department of Human Genetics, McGill University, Montréal, Canada
| | - Ziv Gan-Or
- The Neuro (Montréal Neurological Institute-Hospital), McGill University, Montréal, Canada
- Department of Human Genetics, McGill University, Montréal, Canada
- Department of Neurology and Neurosurgery, McGill University, Montréal, Canada
| |
Collapse
|
50
|
Saez-Atienzar S, Souza CDS, Chia R, Beal SN, Lorenzini I, Huang R, Levy J, Burciu C, Ding J, Gibbs JR, Jones A, Dewan R, Pensato V, Peverelli S, Corrado L, van Vugt JJFA, van Rheenen W, Tunca C, Bayraktar E, Xia M, Iacoangeli A, Shatunov A, Tiloca C, Ticozzi N, Verde F, Mazzini L, Kenna K, Al Khleifat A, Opie-Martin S, Raggi F, Filosto M, Piccinelli SC, Padovani A, Gagliardi S, Inghilleri M, Ferlini A, Vasta R, Calvo A, Moglia C, Canosa A, Manera U, Grassano M, Mandrioli J, Mora G, Lunetta C, Tanel R, Trojsi F, Cardinali P, Gallone S, Brunetti M, Galimberti D, Serpente M, Fenoglio C, Scarpini E, Comi GP, Corti S, Del Bo R, Ceroni M, Pinter GL, Taroni F, Bella ED, Bersano E, Curtis CJ, Lee SH, Chung R, Patel H, Morrison KE, Cooper-Knock J, Shaw PJ, Breen G, Dobson RJB, Dalgard CL, Scholz SW, Al-Chalabi A, van den Berg LH, McLaughlin R, Hardiman O, Cereda C, Sorarù G, D'Alfonso S, Chandran S, Pal S, Ratti A, Gellera C, Johnson K, Doucet-O'Hare T, Pasternack N, Wang T, Nath A, Siciliano G, Silani V, Başak AN, Veldink JH, Camu W, Glass JD, Landers JE, Chiò A, Sattler R, Shaw CE, Ferraiuolo L, et alSaez-Atienzar S, Souza CDS, Chia R, Beal SN, Lorenzini I, Huang R, Levy J, Burciu C, Ding J, Gibbs JR, Jones A, Dewan R, Pensato V, Peverelli S, Corrado L, van Vugt JJFA, van Rheenen W, Tunca C, Bayraktar E, Xia M, Iacoangeli A, Shatunov A, Tiloca C, Ticozzi N, Verde F, Mazzini L, Kenna K, Al Khleifat A, Opie-Martin S, Raggi F, Filosto M, Piccinelli SC, Padovani A, Gagliardi S, Inghilleri M, Ferlini A, Vasta R, Calvo A, Moglia C, Canosa A, Manera U, Grassano M, Mandrioli J, Mora G, Lunetta C, Tanel R, Trojsi F, Cardinali P, Gallone S, Brunetti M, Galimberti D, Serpente M, Fenoglio C, Scarpini E, Comi GP, Corti S, Del Bo R, Ceroni M, Pinter GL, Taroni F, Bella ED, Bersano E, Curtis CJ, Lee SH, Chung R, Patel H, Morrison KE, Cooper-Knock J, Shaw PJ, Breen G, Dobson RJB, Dalgard CL, Scholz SW, Al-Chalabi A, van den Berg LH, McLaughlin R, Hardiman O, Cereda C, Sorarù G, D'Alfonso S, Chandran S, Pal S, Ratti A, Gellera C, Johnson K, Doucet-O'Hare T, Pasternack N, Wang T, Nath A, Siciliano G, Silani V, Başak AN, Veldink JH, Camu W, Glass JD, Landers JE, Chiò A, Sattler R, Shaw CE, Ferraiuolo L, Fogh I, Traynor BJ. Mechanism-free repurposing of drugs for C9orf72-related ALS/FTD using large-scale genomic data. CELL GENOMICS 2024; 4:100679. [PMID: 39437787 PMCID: PMC11605688 DOI: 10.1016/j.xgen.2024.100679] [Show More Authors] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 07/02/2024] [Accepted: 09/22/2024] [Indexed: 10/25/2024]
Abstract
Repeat expansions in the C9orf72 gene are the most common genetic cause of (ALS) and frontotemporal dementia (FTD). Like other genetic forms of neurodegeneration, pinpointing the precise mechanism(s) by which this mutation leads to neuronal death remains elusive, and this lack of knowledge hampers the development of therapy for C9orf72-related disease. We used an agnostic approach based on genomic data (n = 41,273 ALS and healthy samples, and n = 1,516 C9orf72 carriers) to overcome these bottlenecks. Our drug-repurposing screen, based on gene- and expression-pattern matching and information about the genetic variants influencing onset age among C9orf72 carriers, identified acamprosate, a γ-aminobutyric acid analog, as a potentially repurposable treatment for patients carrying C9orf72 repeat expansions. We validated its neuroprotective effect in cell models and showed comparable efficacy to riluzole, the current standard of care. Our work highlights the potential value of genomics in repurposing drugs in situations where the underlying pathomechanisms are inherently complex. VIDEO ABSTRACT.
Collapse
Affiliation(s)
- Sara Saez-Atienzar
- Neuromuscular Diseases Research Section, National Institute on Aging, National Institutes of Health (NIH), Bethesda, MD 20892, USA; Department of Neurology, Ohio State University, Columbus, OH 43210, USA.
| | - Cleide Dos Santos Souza
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield S10 2HQ, UK
| | - Ruth Chia
- Neuromuscular Diseases Research Section, National Institute on Aging, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Selina N Beal
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield S10 2HQ, UK
| | - Ileana Lorenzini
- Department of Translational Neuroscience, Barrow Neurological Institute, Phoenix, AZ 85013, USA
| | - Ruili Huang
- Division of Pre-clinical Innovation, National Center for Advancing Translational Sciences (NCATS), NIH, Rockville, MD 20850, USA
| | - Jennifer Levy
- Department of Translational Neuroscience, Barrow Neurological Institute, Phoenix, AZ 85013, USA
| | - Camelia Burciu
- Department of Translational Neuroscience, Barrow Neurological Institute, Phoenix, AZ 85013, USA
| | - Jinhui Ding
- Computational Biology Group, Laboratory of Neurogenetics, National Institute on Aging, Bethesda, MD 20892, USA
| | - J Raphael Gibbs
- Computational Biology Group, Laboratory of Neurogenetics, National Institute on Aging, Bethesda, MD 20892, USA
| | - Ashley Jones
- Maurice Wohl Clinical Neuroscience Institute, Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, UK
| | - Ramita Dewan
- Neuromuscular Diseases Research Section, National Institute on Aging, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Viviana Pensato
- Unit of Medical Genetics and Neurogenetics, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Silvia Peverelli
- Department of Neurology and Laboratory of Neuroscience, Istituto Auxologico Italiano IRCCS, Milan, Italy
| | - Lucia Corrado
- Department of Health Sciences, University of Eastern Piedmont, Novara, Italy
| | - Joke J F A van Vugt
- Department of Neurology, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Wouter van Rheenen
- Department of Neurology, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Ceren Tunca
- Neurodegeneration Research Laboratory (NDAL), Research Center for Translational Medicine (KUTTAM), Koç University School of Medicine, Istanbul, Turkey
| | - Elif Bayraktar
- Neurodegeneration Research Laboratory (NDAL), Research Center for Translational Medicine (KUTTAM), Koç University School of Medicine, Istanbul, Turkey
| | - Menghang Xia
- Division of Pre-clinical Innovation, National Center for Advancing Translational Sciences (NCATS), NIH, Rockville, MD 20850, USA
| | - Alfredo Iacoangeli
- Maurice Wohl Clinical Neuroscience Institute, Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, UK; Department of Biostatistics and Health Informatics, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, UK; National Institute for Health Research Biomedical Research Centre and Dementia Unit, South London and Maudsley NHS Foundation Trust and King's College London, London, UK
| | - Aleksey Shatunov
- Maurice Wohl Clinical Neuroscience Institute, Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, UK
| | - Cinzia Tiloca
- Department of Neurology and Laboratory of Neuroscience, Istituto Auxologico Italiano IRCCS, Milan, Italy
| | - Nicola Ticozzi
- Department of Neurology and Laboratory of Neuroscience, Istituto Auxologico Italiano IRCCS, Milan, Italy; Department of Pathophysiology and Transplantation, "Dino Ferrari" Center, Università degli Studi di Milano, Milan, Italy
| | - Federico Verde
- Department of Neurology and Laboratory of Neuroscience, Istituto Auxologico Italiano IRCCS, Milan, Italy; Department of Pathophysiology and Transplantation, "Dino Ferrari" Center, Università degli Studi di Milano, Milan, Italy
| | - Letizia Mazzini
- Amyotrophic Lateral Sclerosis Center, Department of Neurology "Maggiore della Carità" University Hospital, Novara, Italy
| | - Kevin Kenna
- Department of Neurology, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Ahmad Al Khleifat
- Maurice Wohl Clinical Neuroscience Institute, Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, UK
| | - Sarah Opie-Martin
- Maurice Wohl Clinical Neuroscience Institute, Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, UK
| | - Flavia Raggi
- Department of Neurosciences, University of Padova, Padova, Italy
| | - Massimiliano Filosto
- NeMO-Brescia Clinical Center for Neuromuscular Diseases, University of Brescia, Brescia, Italy; Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Stefano Cotti Piccinelli
- NeMO-Brescia Clinical Center for Neuromuscular Diseases, University of Brescia, Brescia, Italy; Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Alessandro Padovani
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Stella Gagliardi
- Genomic and Post-Genomic Center, IRCCS Mondino Foundation, Pavia, Italy
| | - Maurizio Inghilleri
- Department of Human Neurosciences, Rare Neuromuscular Diseases Centre, Sapienza University, 00185 Rome, Italy; IRCCS Neuromed, Pozzilli, Italy
| | - Alessandra Ferlini
- Unit of Medical Genetics, Department of Medical Science, University of Ferrara, Ferrara, Italy
| | - Rosario Vasta
- "Rita Levi Montalcini" Department of Neuroscience, Amyotrophic Lateral Sclerosis Center, University of Turin, Turin, Italy
| | - Andrea Calvo
- "Rita Levi Montalcini" Department of Neuroscience, Amyotrophic Lateral Sclerosis Center, University of Turin, Turin, Italy; Azienda Ospedaliero Universitaria Città della Salute e della Scienza, Turin, Italy
| | - Cristina Moglia
- "Rita Levi Montalcini" Department of Neuroscience, Amyotrophic Lateral Sclerosis Center, University of Turin, Turin, Italy; Azienda Ospedaliero Universitaria Città della Salute e della Scienza, Turin, Italy
| | - Antonio Canosa
- "Rita Levi Montalcini" Department of Neuroscience, Amyotrophic Lateral Sclerosis Center, University of Turin, Turin, Italy; Azienda Ospedaliero Universitaria Città della Salute e della Scienza, Turin, Italy; Institute of Cognitive Sciences and Technologies, C.N.R., Rome, Italy
| | - Umberto Manera
- "Rita Levi Montalcini" Department of Neuroscience, Amyotrophic Lateral Sclerosis Center, University of Turin, Turin, Italy; Azienda Ospedaliero Universitaria Città della Salute e della Scienza, Turin, Italy
| | - Maurizio Grassano
- "Rita Levi Montalcini" Department of Neuroscience, Amyotrophic Lateral Sclerosis Center, University of Turin, Turin, Italy
| | - Jessica Mandrioli
- Department of Biomedical, Metabolic, and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy; Department of Neurosciences, Azienda Ospedaliero Universitaria di Modena, Modena, Italy
| | - Gabriele Mora
- "Rita Levi Montalcini" Department of Neuroscience, Amyotrophic Lateral Sclerosis Center, University of Turin, Turin, Italy
| | - Christian Lunetta
- Department of Neurorehabilitation, Istituti Clinici Scientifici Maugeri IRCCS, Institute of Milan, Milan, Italy; NEMO Clinical Center Milano, Fondazione Serena Onlus, Milan, Italy
| | - Raffaella Tanel
- Operative Unit of Neurology, S. Chiara Hospital, Trento, Italy
| | - Francesca Trojsi
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli," Naples, Italy
| | | | - Salvatore Gallone
- "Rita Levi Montalcini" Department of Neuroscience, Amyotrophic Lateral Sclerosis Center, University of Turin, Turin, Italy
| | - Maura Brunetti
- "Rita Levi Montalcini" Department of Neuroscience, Amyotrophic Lateral Sclerosis Center, University of Turin, Turin, Italy
| | - Daniela Galimberti
- Neurodegenerative Diseases Unit, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy; Department of Biomedical, Surgical, and Dental Sciences, University of Milan, Milan, Italy
| | - Maria Serpente
- Neurodegenerative Diseases Unit, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Chiara Fenoglio
- Neurodegenerative Diseases Unit, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy; Department of Biomedical, Surgical, and Dental Sciences, University of Milan, Milan, Italy
| | - Elio Scarpini
- Neurodegenerative Diseases Unit, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Giacomo P Comi
- Department of Pathophysiology and Transplantation, "Dino Ferrari" Center, Università degli Studi di Milano, Milan, Italy; Neurology Unit, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Stefania Corti
- Department of Pathophysiology and Transplantation, "Dino Ferrari" Center, Università degli Studi di Milano, Milan, Italy; Neurology Unit, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Roberto Del Bo
- Department of Pathophysiology and Transplantation, "Dino Ferrari" Center, Università degli Studi di Milano, Milan, Italy; Neurology Unit, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Mauro Ceroni
- Genomic and Post-Genomic Center, IRCCS Mondino Foundation, Pavia, Italy; Department of Brain and Behavioural Sciences, University of Pavia, Pavia, Italy
| | - Giuseppe Lauria Pinter
- 3rd Neurology Unit, Motor Neuron Diseases Center, Fondazione IRCCS Istituto Neurologico "Carlo Besta," Milan, Italy; Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Franco Taroni
- Unit of Medical Genetics and Neurogenetics, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Eleonora Dalla Bella
- 3rd Neurology Unit, Motor Neuron Diseases Center, Fondazione IRCCS Istituto Neurologico "Carlo Besta," Milan, Italy
| | - Enrica Bersano
- 3rd Neurology Unit, Motor Neuron Diseases Center, Fondazione IRCCS Istituto Neurologico "Carlo Besta," Milan, Italy; "L. Sacco" Department of Biomedical and Clinical Sciences, Università degli Studi di Milano, Milan, Italy
| | - Charles J Curtis
- Social Genetic & Developmental Psychiatry Centre, Institute of Psychiatry, Psychology, and Neuroscience (IoPPN), King's College London, London, UK; NIHR BioResource Centre Maudsley, NIHR Maudsley Biomedical Research Centre (BRC) at South London and Maudsley NHS Foundation Trust (SLaM), London, UK
| | - Sang Hyuck Lee
- Social Genetic & Developmental Psychiatry Centre, Institute of Psychiatry, Psychology, and Neuroscience (IoPPN), King's College London, London, UK; NIHR BioResource Centre Maudsley, NIHR Maudsley Biomedical Research Centre (BRC) at South London and Maudsley NHS Foundation Trust (SLaM), London, UK
| | - Raymond Chung
- Social Genetic & Developmental Psychiatry Centre, Institute of Psychiatry, Psychology, and Neuroscience (IoPPN), King's College London, London, UK; NIHR BioResource Centre Maudsley, NIHR Maudsley Biomedical Research Centre (BRC) at South London and Maudsley NHS Foundation Trust (SLaM), London, UK
| | - Hamel Patel
- Department of Biostatistics and Health Informatics, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, UK; NIHR BioResource Centre Maudsley, NIHR Maudsley Biomedical Research Centre (BRC) at South London and Maudsley NHS Foundation Trust (SLaM), London, UK
| | - Karen E Morrison
- School of Medicine, Dentistry, and Biomedical Sciences, Faculty of Medicine Health and Life Sciences, Queen's University, Belfast, UK
| | - Johnathan Cooper-Knock
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, and the NIHR Sheffield Biomedical Research Centre, Sheffield, UK
| | - Pamela J Shaw
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, and the NIHR Sheffield Biomedical Research Centre, Sheffield, UK
| | - Gerome Breen
- Social Genetic & Developmental Psychiatry Centre, Institute of Psychiatry, Psychology, and Neuroscience (IoPPN), King's College London, London, UK; NIHR BioResource Centre Maudsley, NIHR Maudsley Biomedical Research Centre (BRC) at South London and Maudsley NHS Foundation Trust (SLaM), London, UK
| | - Richard J B Dobson
- Department of Biostatistics and Health Informatics, Institute of Psychiatry, Psychology, and Neuroscience (IoPPN), King's College London, London SE5 8AF, UK; NIHR Biomedical Research Centre at South London and Maudsley NHS Foundation Trust and King's College London, London, UK; Health Data Research UK London, University College London, London, UK; Institute of Health Informatics, University College London, London, UK; NIHR Biomedical Research Centre at University College London Hospitals NHS Foundation Trust, London, UK
| | - Clifton L Dalgard
- Department of Anatomy, Physiology, & Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA; The American Genome Center, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - Sonja W Scholz
- Neurodegenerative Diseases Research Section, National Institute of Neurological Disorders and Stroke (NINDS), NIH, Bethesda, MD 20892, USA; Department of Neurology, Johns Hopkins University Medical Center, Baltimore, MD 21287, USA
| | - Ammar Al-Chalabi
- Maurice Wohl Clinical Neuroscience Institute, Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, UK; Department of Clinical Neuroscience, King's College Hospital, London SE5 9RS, UK
| | - Leonard H van den Berg
- Department of Neurology, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Russell McLaughlin
- Complex Trait Genomics Laboratory, Smurfit Institute of Genetics, Trinity College Dublin, Dublin, Ireland
| | - Orla Hardiman
- Academic Unit of Neurology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland; Department of Neurology, Beaumont Hospital, Dublin, Ireland
| | - Cristina Cereda
- Genomic and Post-Genomic Center, IRCCS Mondino Foundation, Pavia, Italy
| | - Gianni Sorarù
- Department of Neurosciences, University of Padova, Padova, Italy
| | - Sandra D'Alfonso
- Department of Health Sciences, University of Eastern Piedmont, Novara, Italy
| | - Siddharthan Chandran
- Euan MacDonald Centre for Motor Neurone Disease Research, Edinburgh, UK; UK Dementia Research Institute, University of Edinburgh, Edinburgh, UK
| | - Suvankar Pal
- Euan MacDonald Centre for Motor Neurone Disease Research, Edinburgh, UK; Centre for Neuroregeneration and Medical Research Council Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, UK
| | - Antonia Ratti
- Department of Neurology and Laboratory of Neuroscience, Istituto Auxologico Italiano IRCCS, Milan, Italy; Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, Milan, Italy
| | - Cinzia Gellera
- Unit of Medical Genetics and Neurogenetics, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Kory Johnson
- Bioinformatics Section, Information Technology Program (ITP), Division of Intramural Research (DIR), National Institute of Neurological Disorders & Stroke, NIH, Bethesda, MD 20892, USA
| | - Tara Doucet-O'Hare
- Neuro-oncology Branch, Center for Cancer Research, National Cancer Institute (NCI), NIH, Bethesda, MD 20892, USA
| | - Nicholas Pasternack
- Translational Neuroscience Center, National Institute of Neurological Disorders and Stroke (NINDS), NIH, Bethesda, MD 20892, USA
| | - Tongguang Wang
- Translational Neuroscience Center, National Institute of Neurological Disorders and Stroke (NINDS), NIH, Bethesda, MD 20892, USA
| | - Avindra Nath
- Translational Neuroscience Center, National Institute of Neurological Disorders and Stroke (NINDS), NIH, Bethesda, MD 20892, USA
| | - Gabriele Siciliano
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Vincenzo Silani
- Department of Neurology and Laboratory of Neuroscience, Istituto Auxologico Italiano IRCCS, Milan, Italy; Department of Pathophysiology and Transplantation, "Dino Ferrari" Center, Università degli Studi di Milano, Milan, Italy
| | - Ayşe Nazlı Başak
- Neurodegeneration Research Laboratory (NDAL), Research Center for Translational Medicine (KUTTAM), Koç University School of Medicine, Istanbul, Turkey
| | - Jan H Veldink
- Department of Neurology, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - William Camu
- ALS Center, CHU Gui de Chauliac, University of Montpellier, Montpellier, France
| | - Jonathan D Glass
- Department of Neurology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - John E Landers
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA, USA
| | - Adriano Chiò
- "Rita Levi Montalcini" Department of Neuroscience, Amyotrophic Lateral Sclerosis Center, University of Turin, Turin, Italy; Azienda Ospedaliero Universitaria Città della Salute e della Scienza, Turin, Italy; Institute of Cognitive Sciences and Technologies, C.N.R., Rome, Italy
| | - Rita Sattler
- Department of Translational Neuroscience, Barrow Neurological Institute, Phoenix, AZ 85013, USA
| | - Christopher E Shaw
- United Kingdom Dementia Research Institute, Maurice Wohl Clinical Neuroscience Institute, Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, UK; Centre for Brain Research, University of Auckland, Auckland, New Zealand
| | - Laura Ferraiuolo
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield S10 2HQ, UK
| | - Isabella Fogh
- United Kingdom Dementia Research Institute, Maurice Wohl Clinical Neuroscience Institute, Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, UK
| | - Bryan J Traynor
- Neuromuscular Diseases Research Section, National Institute on Aging, National Institutes of Health (NIH), Bethesda, MD 20892, USA; Department of Neurology, Johns Hopkins University Medical Center, Baltimore, MD 21287, USA; Reta Lila Weston Institute, UCL Queen Square Institute of Neurology, University College London, London WC1N 1PJ, UK; National Institute of Neurological Disorders and Stroke (NINDS), NIH, Bethesda, MD 20892, USA; RNA Therapeutics Laboratory, National Center for Advancing Translational Sciences (NCATS), NIH, Rockville, MD 20850, USA.
| |
Collapse
|