1
|
Wang L, Sheng J, Zhang Q, Song Y, Zhang Q, Wang B, Zhang R. Diagnosis of Alzheimer's disease using FusionNet with improved secretary bird optimization algorithm for optimal MK-SVM based on imaging genetic data. Cereb Cortex 2025:bhae498. [PMID: 39756421 DOI: 10.1093/cercor/bhae498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Revised: 11/19/2024] [Accepted: 12/22/2024] [Indexed: 01/07/2025] Open
Abstract
Alzheimer's disease is an irreversible central neurodegenerative disease, and early diagnosis of Alzheimer's disease is beneficial for its prevention and early intervention treatment. In this study, we propose a novel framework, FusionNet-ISBOA-MK-SVM, which integrates a fusion network (FusionNet) and improved secretary bird optimization algorithm to optimize multikernel support vector machine for Alzheimer's disease diagnosis. The model leverages multimodality data, including functional magnetic resonance imaging and genetic information (single-nucleotide polymorphisms). Specifically, FusionNet employs U-shaped hierarchical graph convolutional networks and sparse graph attention networks to select feature effectively. Extensive validation using the Alzheimer's Disease Neuroimaging Initiative dataset demonstrates the model's superior interpretability and classification performance. Compared to other state-of-the-art machine learning methods, FusionNet-ISBOA-MK-SVM achieves classification accuracies of 98.6%, 95.7%, 93.0%, 91.8%, 93.1%, and 95.4% for HC vs. AD, EMCI vs. AD, LMCI vs. AD, EMCI vs. AD, HC vs. EMCI, and HC vs. LMCI, respectively. Moreover, the proposed model identifies affected brain regions and pathogenic genes, offering deeper insights into the mechanisms and progression of Alzheimer's disease. These findings provide valuable scientific evidence to support early diagnosis and preventive strategies for Alzheimer's disease.
Collapse
Affiliation(s)
- Luyun Wang
- School of Computer Science and Technology, Hangzhou Dianzi University, 1158 2nd Street, Hangzhou, Zhejiang 310018, China
- Key Laboratory of Intelligent Image Analysis for Sensory and Cognitive Health, Ministry of Industry and Information Technology of China, 215 6th Street, Hangzhou, Zhejiang 310018, China
- Hangzhou Vocational & Technical College, 68 Xueyuan Street, Hangzhou, Zhejiang 310018, China
| | - Jinhua Sheng
- School of Computer Science and Technology, Hangzhou Dianzi University, 1158 2nd Street, Hangzhou, Zhejiang 310018, China
- Key Laboratory of Intelligent Image Analysis for Sensory and Cognitive Health, Ministry of Industry and Information Technology of China, 215 6th Street, Hangzhou, Zhejiang 310018, China
| | - Qiao Zhang
- Beijing Hospital, 1 Dahua Road, Beijing 100730, China
- National Center of Gerontology, 1 Dahua Road, Beijing 100730, China
- Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, 1 Dahua Road, Beijing 100730, China
| | - Yan Song
- Beijing Hospital, 1 Dahua Road, Beijing 100730, China
- National Center of Gerontology, 1 Dahua Road, Beijing 100730, China
- Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, 1 Dahua Road, Beijing 100730, China
| | - Qian Zhang
- School of Computer Science and Technology, Hangzhou Dianzi University, 1158 2nd Street, Hangzhou, Zhejiang 310018, China
- Key Laboratory of Intelligent Image Analysis for Sensory and Cognitive Health, Ministry of Industry and Information Technology of China, 215 6th Street, Hangzhou, Zhejiang 310018, China
| | - Binbing Wang
- School of Computer Science and Technology, Hangzhou Dianzi University, 1158 2nd Street, Hangzhou, Zhejiang 310018, China
- Key Laboratory of Intelligent Image Analysis for Sensory and Cognitive Health, Ministry of Industry and Information Technology of China, 215 6th Street, Hangzhou, Zhejiang 310018, China
| | - Rong Zhang
- School of Computer Science and Technology, Hangzhou Dianzi University, 1158 2nd Street, Hangzhou, Zhejiang 310018, China
- Key Laboratory of Intelligent Image Analysis for Sensory and Cognitive Health, Ministry of Industry and Information Technology of China, 215 6th Street, Hangzhou, Zhejiang 310018, China
| |
Collapse
|
2
|
Escarcega RD, M J VK, Kyriakopoulos VE, Ortiz GJ, Gusdon AM, Fan H, Peesh P, Blasco Conesa MP, Colpo GD, Ahnstedt HW, Couture L, Kim SH, Hinojosa M, Farrell CM, Marrelli SP, Urayama A, Ganesh BP, Schulz PE, McCullough LD, Tsvetkov AS. Serum metabolome profiling in patients with mild cognitive impairment reveals sex differences in lipid metabolism. Neurobiol Dis 2025; 204:106747. [PMID: 39617329 DOI: 10.1016/j.nbd.2024.106747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Revised: 11/20/2024] [Accepted: 11/21/2024] [Indexed: 12/10/2024] Open
Abstract
Alzheimer's disease (AD) affects more women than men. Although women live longer than men, it is not longevity alone, but other factors, including metabolic changes, that contribute to the higher risk of AD in women. Metabolic pathways have been implicated in AD progression, but studies to date examined targeted pathways, leaving many metabolites unmeasured. Sex is often a neglected biological variable, and most metabolomic studies were not designed to investigate sex differences in metabolomic profiles. Here, we performed untargeted metabolomic profiling of sera from male and female patients with mild cognitive impairment (MCI), a common precursor to AD, and matched controls. We discovered significant metabolic changes in individuals with MCI, and found several pathways that were strongly associated with sex. Peptide energy metabolism demonstrated sexual dimorphism. Lipid pathways exhibited the strongest differences between female and male MCI patients, including specific phosphatidylcholine lipids, lysophospholipids, long-chain fatty acids, and monoacylglycerols. 1-palmitoleoyl glycerol and 1-arachidonoyl glycerol were higher in female MCI subjects than in male MCI subjects with no differences between control males and females. Conversely, specific dicarboxylic fatty acids were lower in female MCI subjects than male MCI subjects. In cultured astrocytes, 1-arachidonoyl glycerol promoted phosphorylation of the transcriptional regulator sphingosine kinase 2, which was inhibited by the transient receptor potential vanilloid 1 receptor antagonists, as well as chromatin remodelling. Overall, we identified novel sex-specific metabolites in MCI patients that could serve as biomarkers of MCI in both sexes, help further define AD etiology, and reveal new potential prevention strategies for AD.
Collapse
Affiliation(s)
- Rocio Diaz Escarcega
- Department of Neurology, the University of Texas McGovern Medical School at Houston, TX, USA
| | - Vijay Kumar M J
- Department of Neurology, the University of Texas McGovern Medical School at Houston, TX, USA
| | - Vasilia E Kyriakopoulos
- Department of Neurology, the University of Texas McGovern Medical School at Houston, TX, USA
| | - Guadalupe J Ortiz
- Department of Neurology, the University of Texas McGovern Medical School at Houston, TX, USA
| | - Aaron M Gusdon
- Department of Neurosurgery, the University of Texas McGovern Medical School at Houston, TX, USA
| | - Huihui Fan
- Department of Neurology, the University of Texas McGovern Medical School at Houston, TX, USA
| | - Pedram Peesh
- Department of Neurology, the University of Texas McGovern Medical School at Houston, TX, USA
| | - Maria P Blasco Conesa
- Department of Neurology, the University of Texas McGovern Medical School at Houston, TX, USA
| | - Gabriela Delevati Colpo
- Department of Neurology, the University of Texas McGovern Medical School at Houston, TX, USA
| | - Hilda W Ahnstedt
- Department of Neurology, the University of Texas McGovern Medical School at Houston, TX, USA
| | - Lucy Couture
- Department of Neurology, the University of Texas McGovern Medical School at Houston, TX, USA
| | - Stella H Kim
- Department of Neurology, the University of Texas McGovern Medical School at Houston, TX, USA; The University of Texas Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Miriam Hinojosa
- Department of Neurology, the University of Texas McGovern Medical School at Houston, TX, USA
| | - Christine M Farrell
- Department of Neurology, the University of Texas McGovern Medical School at Houston, TX, USA
| | - Sean P Marrelli
- Department of Neurology, the University of Texas McGovern Medical School at Houston, TX, USA
| | - Akihiko Urayama
- Department of Neurology, the University of Texas McGovern Medical School at Houston, TX, USA
| | - Bhanu P Ganesh
- Department of Neurology, the University of Texas McGovern Medical School at Houston, TX, USA
| | - Paul E Schulz
- Department of Neurology, the University of Texas McGovern Medical School at Houston, TX, USA
| | - Louise D McCullough
- Department of Neurology, the University of Texas McGovern Medical School at Houston, TX, USA; The University of Texas Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Andrey S Tsvetkov
- Department of Neurology, the University of Texas McGovern Medical School at Houston, TX, USA; The University of Texas Graduate School of Biomedical Sciences, Houston, TX, USA; UTHealth Consortium on Aging, the University of Texas McGovern Medical School, Houston, TX, USA.
| |
Collapse
|
3
|
Li P, Ye L, Sun S, Wang Y, Chen Y, Chang J, Yin R, Liu X, Zuo W, Xu H, Zhang X, Zhao RC, Han Q, Wei J. Molecular intersections of traumatic brain injury and Alzheimer's disease: the role of ADMSC-derived exosomes and hub genes in microglial polarization. Metab Brain Dis 2024; 40:77. [PMID: 39715972 DOI: 10.1007/s11011-024-01503-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 12/11/2024] [Indexed: 12/25/2024]
Abstract
Traumatic brain injury (TBI) is a significant contributor to global mortality and morbidity, with emerging evidence indicating a heightened risk of developing Alzheimer's disease (AD) following TBI. This study aimed to explore the molecular intersections between TBI and AD, focusing on the role of adipose mesenchymal stem cell (ADMSC)-derived exosomes and hub genes involved in microglial polarization. Transcriptome profiles from TBI (GSE58485) and AD (GSE74614) datasets were analyzed to identify differentially expressed genes (DEGs). The hub genes were validated in independent datasets (GSE180811 for TBI and GSE135999 for AD) and localized to specific cell types using single-cell RNA (scRNA) sequencing data (GSE160763 for TBI and GSE224398 for AD). Experimental validation was conducted to investigate the role of these genes in microglial polarization using cell culture and ADMSC-derived exosomes interventions. Our results identified three hub genes-Bst2, B2m, and Lgals3bp-that were upregulated in both TBI and AD, with strong associations to inflammation, neuronal apoptosis, and tissue repair processes. scRNA sequencing revealed that these genes are predominantly expressed in microglia, with increased expression during M1 polarization. Knockdown of these genes reduced M1 polarization and promoted M2 phenotype in microglia. Additionally, ADMSC-derived exosomes attenuated M1 polarization and downregulated the expression of hub genes. This study provides novel insights into the shared molecular pathways between TBI and AD, highlighting potential therapeutic targets for mitigating neuroinflammation and promoting recovery in both conditions.
Collapse
Affiliation(s)
- Pengtao Li
- Department of Neurosurgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Liguo Ye
- Department of Neurosurgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Sishuai Sun
- Department of Neurosurgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Yue Wang
- Department of Neurosurgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Yihao Chen
- Department of Neurosurgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Jianbo Chang
- Department of Neurosurgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Rui Yin
- Department of Neurosurgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Xiaoyu Liu
- Department of Neurosurgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Wei Zuo
- Department of Neurosurgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Houshi Xu
- Department of Neurosurgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Xiao Zhang
- Department of Neurosurgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Robert Chunhua Zhao
- Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
| | - Qin Han
- Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China.
| | - Junji Wei
- Department of Neurosurgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China.
| |
Collapse
|
4
|
Tsantzali I, Athanasaki A, Boufidou F, Constantinides VC, Stefanou MI, Moschovos C, Zompola C, Paraskevas SG, Bonakis A, Giannopoulos S, Tsivgoulis G, Kapaki E, Paraskevas GP. Cerebrospinal Fluid Classical Biomarker Levels in Mixed vs. Pure A +T + (A +T 1+) Alzheimer's Disease. Biomedicines 2024; 12:2904. [PMID: 39767810 PMCID: PMC11672946 DOI: 10.3390/biomedicines12122904] [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: 11/19/2024] [Revised: 12/15/2024] [Accepted: 12/18/2024] [Indexed: 01/11/2025] Open
Abstract
Background: Alzheimer's disease (AD) may present with pure (typical or atypical) and mixed phenotypes, sometimes causing difficulties in (differential) diagnosis. In order to achieve a diagnostic accuracy as high as possible, the diagnosis of AD during life depends on various biomarkers, including the cerebrospinal fluid (CSF) biomarkers. Methods: Classical CSF AD biomarkers were determined in a total of 61 patients, classified as both beta amyloid- and tau-positive A+T+ (or A+T1+ according to the recently revised Alzheimer Association criteria for diagnosis and staging of AD). Twenty one of these patients fulfilled the criteria for mixed AD (mixed with Lewy bodies, cerebrovascular disease, or normal pressure hydrocephalus), whilst 40 had pure AD. Results: Patients did not differ with respect to gender, education, disease duration, and cognitive status. After controlling for confounding factors, no difference was observed between mixed and pure AD groups in Aβ42 or Aβ42/Aβ40 levels. Although by definition, patients of both groups had abnormal (increased) levels of phospho-tau181, the mixed AD group presented with lower (less abnormal) levels of phospho-tau181 and total tau as compared to the pure group. Conclusions: In patients with AD of comparable cognitive status, mixed AD cases may present with lower levels of tau proteins and, if close to the cut-off values, diagnostic uncertainty may be increased.
Collapse
Affiliation(s)
- Ioanna Tsantzali
- 2nd Department of Neurology, “Attikon” General University Hospital, School of Medicine, National and Kapodistrian University of Athens, 12462 Athens, Greece; (I.T.); (A.A.); (M.-I.S.); (C.M.); (C.Z.); (S.G.P.); (A.B.); (S.G.); (G.T.)
| | - Athanasia Athanasaki
- 2nd Department of Neurology, “Attikon” General University Hospital, School of Medicine, National and Kapodistrian University of Athens, 12462 Athens, Greece; (I.T.); (A.A.); (M.-I.S.); (C.M.); (C.Z.); (S.G.P.); (A.B.); (S.G.); (G.T.)
| | - Fotini Boufidou
- Neurochemistry and Βiological Markers Unit, 1st Department of Neurology, “Eginition” Hospital, School of Medicine, National and Kapodistrian University of Athens, 11528 Athens, Greece; (F.B.); (V.C.C.); (E.K.)
| | - Vasilios C. Constantinides
- Neurochemistry and Βiological Markers Unit, 1st Department of Neurology, “Eginition” Hospital, School of Medicine, National and Kapodistrian University of Athens, 11528 Athens, Greece; (F.B.); (V.C.C.); (E.K.)
| | - Maria-Ioanna Stefanou
- 2nd Department of Neurology, “Attikon” General University Hospital, School of Medicine, National and Kapodistrian University of Athens, 12462 Athens, Greece; (I.T.); (A.A.); (M.-I.S.); (C.M.); (C.Z.); (S.G.P.); (A.B.); (S.G.); (G.T.)
| | - Christos Moschovos
- 2nd Department of Neurology, “Attikon” General University Hospital, School of Medicine, National and Kapodistrian University of Athens, 12462 Athens, Greece; (I.T.); (A.A.); (M.-I.S.); (C.M.); (C.Z.); (S.G.P.); (A.B.); (S.G.); (G.T.)
| | - Christina Zompola
- 2nd Department of Neurology, “Attikon” General University Hospital, School of Medicine, National and Kapodistrian University of Athens, 12462 Athens, Greece; (I.T.); (A.A.); (M.-I.S.); (C.M.); (C.Z.); (S.G.P.); (A.B.); (S.G.); (G.T.)
| | - Sotirios G. Paraskevas
- 2nd Department of Neurology, “Attikon” General University Hospital, School of Medicine, National and Kapodistrian University of Athens, 12462 Athens, Greece; (I.T.); (A.A.); (M.-I.S.); (C.M.); (C.Z.); (S.G.P.); (A.B.); (S.G.); (G.T.)
| | - Anastasios Bonakis
- 2nd Department of Neurology, “Attikon” General University Hospital, School of Medicine, National and Kapodistrian University of Athens, 12462 Athens, Greece; (I.T.); (A.A.); (M.-I.S.); (C.M.); (C.Z.); (S.G.P.); (A.B.); (S.G.); (G.T.)
| | - Sotirios Giannopoulos
- 2nd Department of Neurology, “Attikon” General University Hospital, School of Medicine, National and Kapodistrian University of Athens, 12462 Athens, Greece; (I.T.); (A.A.); (M.-I.S.); (C.M.); (C.Z.); (S.G.P.); (A.B.); (S.G.); (G.T.)
| | - Georgios Tsivgoulis
- 2nd Department of Neurology, “Attikon” General University Hospital, School of Medicine, National and Kapodistrian University of Athens, 12462 Athens, Greece; (I.T.); (A.A.); (M.-I.S.); (C.M.); (C.Z.); (S.G.P.); (A.B.); (S.G.); (G.T.)
- Department of Neurology, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Elisabeth Kapaki
- Neurochemistry and Βiological Markers Unit, 1st Department of Neurology, “Eginition” Hospital, School of Medicine, National and Kapodistrian University of Athens, 11528 Athens, Greece; (F.B.); (V.C.C.); (E.K.)
| | - George P. Paraskevas
- 2nd Department of Neurology, “Attikon” General University Hospital, School of Medicine, National and Kapodistrian University of Athens, 12462 Athens, Greece; (I.T.); (A.A.); (M.-I.S.); (C.M.); (C.Z.); (S.G.P.); (A.B.); (S.G.); (G.T.)
- Neurochemistry and Βiological Markers Unit, 1st Department of Neurology, “Eginition” Hospital, School of Medicine, National and Kapodistrian University of Athens, 11528 Athens, Greece; (F.B.); (V.C.C.); (E.K.)
| |
Collapse
|
5
|
Bergström S, Mravinacová S, Lindberg O, Zettergren A, Westman E, Wahlund LO, Blennow K, Zetterberg H, Kern S, Skoog I, Månberg A. CSF levels of brain-derived proteins correlate with brain ventricular volume in cognitively healthy 70-year-olds. Clin Proteomics 2024; 21:65. [PMID: 39668376 PMCID: PMC11636040 DOI: 10.1186/s12014-024-09517-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 12/02/2024] [Indexed: 12/14/2024] Open
Abstract
BACKGROUND The effect of varying brain ventricular volume on the cerebrospinal fluid (CSF) proteome has been discussed as possible confounding factors in comparative protein level analyses. However, the relationship between CSF volume and protein levels remains largely unexplored. Moreover, the few existing studies provide conflicting findings, indicating the need for further research. METHODS Here, we explored the association between levels of 88 pre-selected CSF proteins and ventricular volume derived from magnetic resonance imaging (MRI) measurements in 157 cognitively healthy 70-year-olds from the H70 Gothenburg Birth Cohort Studies, including individuals with and without pathological levels of Alzheimer's disease (AD) CSF markers (n = 123 and 34, respectively). Both left and right lateral, the inferior horn as well as the third and the fourth ventricular volumes were measured. Different antibody-based methods were employed for the protein measurements, with most being analyzed using a multiplex bead-based microarray technology. Furthermore, the associations between the protein levels and cortical thickness, fractional anisotropy, and mean diffusivity were assessed. RESULTS CSF levels of many brain-derived proteins correlated with ventricular volumes in A-T- individuals, with lower levels in individuals with larger ventricles. The strongest negative correlations with total ventricular volume were observed for neurocan (NCAN) and neurosecretory protein VGF (rho = -0.34 for both). Significant negative correlations were observed also for amyloid beta (Ab) 38, Ab40, total tau (t-tau), and phosphorylated tau (p-tau), with correlation ranging between - 0.34 and - 0.28, while no association was observed between ventricular volumes and Ab42 or neurofilament light chain (NfL). Proteins with negative correlations to ventricular volumes further demonstrated negative correlations to mean diffusivity and positive correlation to fractional anisotropy. However, only weak or no correlations were observed between the CSF protein levels and cortical thickness. A + T + individuals demonstrated higher CSF protein levels compared to A-T- individuals with the most significant differences observed for neurogranin (NRGN) and synuclein beta (SNCB). CONCLUSIONS Our findings suggest that the levels of many brain-derived proteins in CSF may be subjected to dilution effects depending on the size of the brain ventricles in healthy individuals without AD pathology. This phenomenon could potentially contribute to the inter-individual variations observed in CSF proteomic studies.
Collapse
Affiliation(s)
- Sofia Bergström
- Division of Affinity Proteomics, Department of Protein Science, KTH Royal Institute of Technology, SciLifeLab, Stockholm, Sweden
| | - Sára Mravinacová
- Division of Affinity Proteomics, Department of Protein Science, KTH Royal Institute of Technology, SciLifeLab, Stockholm, Sweden
| | - Olof Lindberg
- Division of Clinical Geriatrics, Department of Neurobiology, Center for Alzheimer Research, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Anna Zettergren
- Neuropsychiatric Epidemiology Unit, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, Centre for Ageing and Health (AGECAP), University of Gothenburg, Mölndal, Sweden
| | - Eric Westman
- Division of Clinical Geriatrics, Department of Neurobiology, Center for Alzheimer Research, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Lars-Olof Wahlund
- Division of Clinical Geriatrics, Department of Neurobiology, Center for Alzheimer Research, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Kaj Blennow
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Henrik Zetterberg
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- UK Dementia Research Institute at UCL, London, UK
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Silke Kern
- Neuropsychiatric Epidemiology Unit, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, Centre for Ageing and Health (AGECAP), University of Gothenburg, Mölndal, Sweden
- Region Västra Götaland, Sahlgrenska University Hospital, Neuropsychiatry, Mölndal, Sweden
| | - Ingmar Skoog
- Neuropsychiatric Epidemiology Unit, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, Centre for Ageing and Health (AGECAP), University of Gothenburg, Mölndal, Sweden
- Region Västra Götaland, Sahlgrenska University Hospital, Neuropsychiatry, Mölndal, Sweden
| | - Anna Månberg
- Division of Affinity Proteomics, Department of Protein Science, KTH Royal Institute of Technology, SciLifeLab, Stockholm, Sweden.
| |
Collapse
|
6
|
Le Borgne J, Gomez L, Heikkinen S, Amin N, Ahmad S, Choi SH, Bis J, Grenier-Boley B, Rodriguez OG, Kleineidam L, Young J, Tripathi KP, Wang L, Varma A, Campos-Martin R, van der Lee S, Damotte V, de Rojas I, Palmal S, Lipton R, Reiman E, McKee A, De Jager P, Bush W, Small S, Levey A, Saykin A, Foroud T, Albert M, Hyman B, Petersen R, Younkin S, Sano M, Wisniewski T, Vassar R, Schneider J, Henderson V, Roberson E, DeCarli C, LaFerla F, Brewer J, Swerdlow R, Van Eldik L, Hamilton-Nelson K, Paulson H, Naj A, Lopez O, Chui H, Crane P, Grabowski T, Kukull W, Asthana S, Craft S, Strittmatter S, Cruchaga C, Leverenz J, Goate A, Kamboh MI, George-Hyslop PS, Valladares O, Kuzma A, Cantwell L, Riemenschneider M, Morris J, Slifer S, Dalmasso C, Castillo A, Küçükali F, Peters O, Schneider A, Dichgans M, Rujescu D, Scherbaum N, Deckert J, Riedel-Heller S, Hausner L, Molina-Porcel L, Düzel E, Grimmer T, Wiltfang J, Heilmann-Heimbach S, Moebus S, Tegos T, Scarmeas N, Dols-Icardo O, Moreno F, Pérez-Tur J, Bullido MJ, Pastor P, Sánchez-Valle R, Álvarez V, Boada M, García-González P, Puerta R, Mir P, Real LM, Piñol-Ripoll G, García-Alberca JM, Royo JL, Rodriguez-Rodriguez E, Soininen H, de Mendonça A, Mehrabian S, Traykov L, Hort J, Vyhnalek M, Thomassen JQ, Pijnenburg YAL, Holstege H, van Swieten J, Ramakers I, Verhey F, Scheltens P, Graff C, Papenberg G, Giedraitis V, Boland A, Deleuze JF, Nicolas G, Dufouil C, Pasquier F, Hanon O, Debette S, Grünblatt E, Popp J, Ghidoni R, Galimberti D, Arosio B, Mecocci P, Solfrizzi V, Parnetti L, Squassina A, Tremolizzo L, Borroni B, Nacmias B, Spallazzi M, Seripa D, Rainero I, Daniele A, Bossù P, Masullo C, Rossi G, Jessen F, Fernandez V, Kehoe PG, Frikke-Schmidt R, Tsolaki M, Sánchez-Juan P, Sleegers K, Ingelsson M, Haines J, Farrer L, Mayeux R, Wang LS, Sims R, DeStefano A, Schellenberg GD, Seshadri S, Amouyel P, Williams J, van der Flier W, Ramirez A, Pericak-Vance M, Andreassen OA, Van Duijn C, Hiltunen M, Ruiz A, Dupuis J, Martin E, Lambert JC, Kunkle B, Bellenguez C. X-chromosome-wide association study for Alzheimer's disease. Mol Psychiatry 2024:10.1038/s41380-024-02838-5. [PMID: 39633006 DOI: 10.1038/s41380-024-02838-5] [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: 06/18/2024] [Revised: 11/05/2024] [Accepted: 11/07/2024] [Indexed: 12/07/2024]
Abstract
Due to methodological reasons, the X-chromosome has not been featured in the major genome-wide association studies on Alzheimer's Disease (AD). To address this and better characterize the genetic landscape of AD, we performed an in-depth X-Chromosome-Wide Association Study (XWAS) in 115,841 AD cases or AD proxy cases, including 52,214 clinically-diagnosed AD cases, and 613,671 controls. We considered three approaches to account for the different X-chromosome inactivation (XCI) states in females, i.e. random XCI, skewed XCI, and escape XCI. We did not detect any genome-wide significant signals (P ≤ 5 × 10-8) but identified seven X-chromosome-wide significant loci (P ≤ 1.6 × 10-6). The index variants were common for the Xp22.32, FRMPD4, DMD and Xq25 loci, and rare for the WNK3, PJA1, and DACH2 loci. Overall, this well-powered XWAS found no genetic risk factors for AD on the non-pseudoautosomal region of the X-chromosome, but it identified suggestive signals warranting further investigations.
Collapse
Affiliation(s)
- Julie Le Borgne
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, LabEx DISTALZ - U1167-RID-AGE Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, Lille, France
| | - Lissette Gomez
- The John P. Hussman Institute for Human Genomics, University of Miami, Miami, FL, USA
| | - Sami Heikkinen
- Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland
| | - Najaf Amin
- Nuffield Department of Population Health Oxford University, Oxford, UK
| | - Shahzad Ahmad
- Department of Epidemiology, Erasmus MC, Rotterdam, The Netherlands
| | - Seung Hoan Choi
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Joshua Bis
- Department of Medicine, Cardiovascular Health Research Unit, University of Washington, Seattle, WA, USA
| | - Benjamin Grenier-Boley
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, LabEx DISTALZ - U1167-RID-AGE Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, Lille, France
| | - Omar Garcia Rodriguez
- The John P. Hussman Institute for Human Genomics, University of Miami, Miami, FL, USA
| | - Luca Kleineidam
- Department of Old Age Psychiatry and Cognitive Disorders, University Hospital Bonn, University of Bonn, Bonn, Germany
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Juan Young
- The John P. Hussman Institute for Human Genomics, University of Miami, Miami, FL, USA
| | - Kumar Parijat Tripathi
- Division of Neurogenetics and Molecular Psychiatry, Department of Psychiatry and Psychotherapy, University of Cologne, Medical Faculty, Cologne, Germany
| | - Lily Wang
- Department of Public Health Sciences, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Achintya Varma
- The John P. Hussman Institute for Human Genomics, University of Miami, Miami, FL, USA
| | - Rafael Campos-Martin
- Division of Neurogenetics and Molecular Psychiatry, Department of Psychiatry and Psychotherapy, University of Cologne, Medical Faculty, Cologne, Germany
| | - Sven van der Lee
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije University, Amsterdam, The Netherlands
| | - Vincent Damotte
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, LabEx DISTALZ - U1167-RID-AGE Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, Lille, France
| | - Itziar de Rojas
- Research Center and Memory Clinic, ACE Alzheimer Center Barcelona, Universitat Internacional de Catalunya, Barcelona, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
| | - Sagnik Palmal
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, LabEx DISTALZ - U1167-RID-AGE Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, Lille, France
| | - Richard Lipton
- Department of Neurology, Albert Einstein College of Medicine, New York, NY, USA
| | - Eric Reiman
- Neurogenomics Division, Translational Genomics Research Institute, Phoenix, AZ, USA
- Arizona Alzheimer's Consortium, Phoenix, AZ, USA
- Banner Alzheimer's Institute, Phoenix, AZ, USA
- Department of Psychiatry, University of Arizona, Phoenix, AZ, USA
| | - Ann McKee
- Department of Neurology, Boston University, Boston, MA, USA
- Department of Pathology, Boston University, Boston, MA, USA
| | - Philip De Jager
- Program in Translational Neuro-Psychiatric Genomics, Institute for the Neurosciences, Department of Neurology & Psychiatry, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute, Cambridge, MA, USA
| | - William Bush
- Department of Epidemiology and Biostatistics, Case Western Reserve University, Cleveland, OH, USA
| | - Scott Small
- Taub Institute on Alzheimer's Disease and the Aging Brain, Department of Neurology, Columbia University, New York, NY, USA
- Department of Neurology, Columbia University, New York, NY, USA
| | - Allan Levey
- Department of Neurology, Emory University, Atlanta, GA, USA
| | - Andrew Saykin
- Department of Radiology, Indiana University, Indianapolis, IN, USA
- Department of Medical and Molecular Genetics, Indiana University, Indianapolis, IN, USA
| | - Tatiana Foroud
- Department of Medical and Molecular Genetics, Indiana University, Indianapolis, IN, USA
| | - Marilyn Albert
- Department of Neurology, Johns Hopkins University, Baltimore, MD, USA
| | - Bradley Hyman
- Department of Neurology, Massachusetts General Hospital/Harvard Medical School, Boston, MA, USA
| | | | - Steven Younkin
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Mary Sano
- Department of Psychiatry, Mount Sinai School of Medicine, New York, NY, USA
| | - Thomas Wisniewski
- Center for Cognitive Neurology and Departments of Neurology, New York University, School of Medicine, New York, NY, USA
- Department of Psychiatry, New York University, New York, NY, USA
| | - Robert Vassar
- Cognitive Neurology and Alzheimer's Disease Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Julie Schneider
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
- Department of Pathology (Neuropathology), Rush University Medical Center, Chicago, IL, USA
| | - Victor Henderson
- Department of Epidemiology and Population Health, Stanford University, Stanford, CA, USA
- Department of Neurology & Neurological Sciences, Stanford University, Stanford, CA, USA
| | - Erik Roberson
- Department of Neurology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Charles DeCarli
- Department of Neurology, University of California Davis, Sacramento, CA, USA
| | - Frank LaFerla
- Department of Neurobiology and Behavior, University of California Irvine, Irvine, CA, USA
| | - James Brewer
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| | - Russell Swerdlow
- University of Kansas Alzheimer's Disease Center, University of Kansas Medical Center, Kansas City, KS, USA
| | - Linda Van Eldik
- Sanders-Brown Center on Aging and University of Kentucky Alzheimer's Disease Research Center, Department of Neuroscience, University of Kentucky, Lexington, KY, USA
| | - Kara Hamilton-Nelson
- The John P. Hussman Institute for Human Genomics, University of Miami, Miami, FL, USA
| | - Henry Paulson
- Michigan Alzheimer's Disease Center, Department of Neurology, University of Michigan, Ann Arbor, MI, USA
| | - Adam Naj
- Penn Neurodegeneration Genomics Center, Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Oscar Lopez
- University of Pittsburgh Alzheimer's Disease Research Center, Pittsburgh, PA, USA
| | - Helena Chui
- Department of Neurology, University of Southern California, Los Angeles, CA, USA
| | - Paul Crane
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - Thomas Grabowski
- Department of Neurology, University of Washington, Seattle, WA, USA
- Department of Radiology, University of Washington, Seattle, WA, USA
| | - Walter Kukull
- Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - Sanjay Asthana
- Geriatric Research, Education and Clinical Center (GRECC), University of Wisconsin, Madison, WI, USA
- Department of Medicine, University of Wisconsin, Madison, WI, USA
- Wisconsin Alzheimer's Disease Research Center, Madison, WI, USA
| | - Suzanne Craft
- Gerontology and Geriatric Medicine Center on Diabetes, Obesity, and Metabolism, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Stephen Strittmatter
- Program in Cellular Neuroscience, Neurodegeneration & Repair, Yale University, New Haven, CT, USA
| | - Carlos Cruchaga
- Department of Psychiatry and Hope Center Program on Protein Aggregation and Neurodegeneration, Washington University School of Medicine, St. Louis, MO, USA
| | - James Leverenz
- Cleveland Clinic Lou Ruvo Center for Brain Health, Cleveland Clinic, Cleveland, OH, USA
| | - Alison Goate
- Department of Neuroscience, Mount Sinai School of Medicine, New York, NY, USA
| | - M Ilyas Kamboh
- University of Pittsburgh Alzheimer's Disease Research Center, Pittsburgh, PA, USA
- Department of Human Genetics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Peter St George-Hyslop
- Department of Medicine (Neurology), Tanz Centre for Research in Neurodegenerative Disease, Temerty Faculty of Medicine, University of Toronto, and University Health Network, Toronto, ON, USA
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Department of Neurology, Columbia University Irving Medical Center, 630 West 168th Street, New York, NY, 10032, USA
| | - Otto Valladares
- Penn Neurodegeneration Genomics Center, Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Amanda Kuzma
- Penn Neurodegeneration Genomics Center, Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Laura Cantwell
- Penn Neurodegeneration Genomics Center, Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | | | - John Morris
- Department of Neurology, Washington University, St. Louis, MO, USA
- Department of Pathology and Immunology, Washington University, St. Louis, MO, USA
| | - Susan Slifer
- The John P. Hussman Institute for Human Genomics, University of Miami, Miami, FL, USA
| | - Carolina Dalmasso
- Division of Neurogenetics and Molecular Psychiatry, Department of Psychiatry and Psychotherapy, University of Cologne, Medical Faculty, Cologne, Germany
- Estudios en Neurociencias y Sistemas Complejos (ENyS) CONICET-HEC-UNAJ, Buenos Aires, Argentina
| | - Atahualpa Castillo
- Division of Psychological Medicine and Clinical Neuroscience, School of Medicine, Cardiff University, Wales, UK
| | - Fahri Küçükali
- Complex Genetics of Alzheimer's Disease Group, VIB Center for Molecular Neurology, VIB, Antwerp, Belgium
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Oliver Peters
- German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Psychiatry and Psychotherapy, Hindenburgdamm 30, 12203, Berlin, Germany
| | - Anja Schneider
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Department for Neurodegenerative Diseases and Geriatric Psychiatry, University Hospital Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Martin Dichgans
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Dan Rujescu
- Martin-Luther-University Halle-Wittenberg, University Clinic and Outpatient Clinic for Psychiatry, Psychotherapy and Psychosomatics, Halle (Saale), Germany
| | - Norbert Scherbaum
- Department of Psychiatry and Psychotherapy, LVR-Klinikum Essen, University of Duisburg-Essen, Germany, Medical Faculty, Duisburg, Germany
| | - Jürgen Deckert
- Department of Psychiatry, Psychosomatics and Psychotherapy, Center of Mental Health, University Hospital of Würzburg, Würzburg, Germany
| | - Steffi Riedel-Heller
- Institute of Social Medicine, Occupational Health and Public Health, University of Leipzig, 04103, Leipzig, Germany
| | - Lucrezia Hausner
- Department of Geriatric Psychiatry, Central Institute for Mental Health Mannheim, Faculty Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Laura Molina-Porcel
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic of Barcelona, Fundació Recerca Clinic Barcelona- Institut d'Investigacions Biomèdiques August Pi i Sunyer (FRCB-IDIBAPS), and University of Barcelona, Barcelona, Spain
- Neurological Tissue Bank-Biobank, Hospital Clinic-FRCB-IDIBAPS, Barcelona, Spain
| | - Emrah Düzel
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
- Institute of Cognitive Neurology and Dementia Research (IKND), Otto-von-Guericke University, Magdeburg, Germany
| | - Timo Grimmer
- Center for Cognitive Disorders, Department of Psychiatry and Psychotherapy, Technical University of Munich, School of Medicine and Health, Klinikum rechts der Isar, Munich, Germany
| | - Jens Wiltfang
- Department of Psychiatry and Psychotherapy, University Medical Center Goettingen, Goettingen, Germany
- German Center for Neurodegenerative Diseases (DZNE), Goettingen, Germany
- Medical Science Department, iBiMED, Aveiro, Portugal
| | - Stefanie Heilmann-Heimbach
- Institute of Human Genetics, University of Bonn, School of Medicine & University Hospital Bonn, Bonn, Germany
| | - Susanne Moebus
- Institute for Urban Public Health, University Hospital of University Duisburg-Essen, Essen, Germany
| | - Thomas Tegos
- 1st Department of Neurology, Medical school, Aristotle University of Thessaloniki, Thessaloniki, Makedonia, Greece
| | - Nikolaos Scarmeas
- Taub Institute for Research in Alzheimer's Disease and the Aging Brain, The Gertrude H. Sergievsky Center, Depatment of Neurology, Columbia University, New York, NY, USA
- 1st Department of Neurology, Aiginition Hospital, National and Kapodistrian University of Athens, Medical School, Athens, Greece
| | - Oriol Dols-Icardo
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
- Sant Pau Memory Unit, Institut de Recerca Sant Pau (IR Sant Pau), Department of Neurology, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Fermin Moreno
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
- Department of Neurology, Hospital Universitario Donostia, San Sebastian, Spain
- Neurosciences Area, Instituto Biodonostia, San Sebastian, Spain
| | - Jordi Pérez-Tur
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
- Unitat de Genètica Molecular, Institut de Biomedicina de València-CSIC, Valencia, Spain
- Unidad Mixta de Neurologia Genètica, Instituto de Investigación Sanitaria La Fe, Valencia, Spain
| | - María J Bullido
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
- Centro de Biología Molecular Severo Ochoa (UAM-CSIC), Madrid, Spain
- Instituto de Investigacion Sanitaria 'Hospital la Paz' (IdIPaz), Madrid, Spain
- Universidad Autónoma de Madrid, Madrid, Spain
| | - Pau Pastor
- Fundació Docència i Recerca MútuaTerrassa, Terrassa, Barcelona, Spain
- Memory Disorders Unit, Department of Neurology, Hospital Universitari Mutua de Terrassa, Terrassa, Barcelona, Spain
| | - Raquel Sánchez-Valle
- Alzheimer's Disease and Other Cognitive Disorders Unit, Service of Neurology, Hospital Clínic of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer, University of Barcelona, Barcelona, Spain
| | - Victoria Álvarez
- Laboratorio de Genética, Hospital Universitario Central de Asturias, Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
| | - Mercè Boada
- Research Center and Memory Clinic, ACE Alzheimer Center Barcelona, Universitat Internacional de Catalunya, Barcelona, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
| | - Pablo García-González
- Research Center and Memory Clinic, ACE Alzheimer Center Barcelona, Universitat Internacional de Catalunya, Barcelona, Spain
| | - Raquel Puerta
- Research Center and Memory Clinic, ACE Alzheimer Center Barcelona, Universitat Internacional de Catalunya, Barcelona, Spain
| | - Pablo Mir
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
- Unidad de Trastornos del Movimiento, Servicio de Neurología y Neurofisiología, Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
| | - Luis M Real
- Unidad Clínica de Enfermedades Infecciosas y Microbiología, Hospital Universitario de Valme, Sevilla, Spain
- Depatamento de Especialidades Quirúrgicas, Bioquímica e Inmunología, Facultad de Medicina, Universidad de Málaga, Málaga, Spain
| | - Gerard Piñol-Ripoll
- Unitat Trastorns Cognitius, Hospital Universitari Santa Maria de Lleida, Lleida, Spain
- Institut de Recerca Biomedica de Lleida (IRBLLeida), Lleida, Spain
| | - Jose María García-Alberca
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
- Alzheimer Research Center & Memory Clinic, Andalusian Institute for Neuroscience, Málaga, Spain
| | - Jose Luís Royo
- Depatamento de Especialidades Quirúrgicas, Bioquímica e Inmunología, Facultad de Medicina, Universidad de Málaga, Málaga, Spain
| | - Eloy Rodriguez-Rodriguez
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
- Neurology Service, Marqués de Valdecilla University Hospital (University of Cantabria and IDIVAL), Santander, Spain
| | - Hilkka Soininen
- Institute of Clinical Medicine-Neurology, University of Eastern Finland, Kuopio, Finland
| | | | - Shima Mehrabian
- Clinic of Neurology, UH "Alexandrovska", Medical University-Sofia, Sofia, Bulgaria
| | - Latchezar Traykov
- Clinic of Neurology, UH "Alexandrovska", Medical University-Sofia, Sofia, Bulgaria
| | - Jakub Hort
- Memory Clinic, Department of Neurology, Charles University, Second Faculty of Medicine and Motol University Hospital, Praha, Czech Republic
| | - Martin Vyhnalek
- Memory Clinic, Department of Neurology, Charles University, Second Faculty of Medicine and Motol University Hospital, Praha, Czech Republic
| | - Jesper Qvist Thomassen
- Department of Clinical Biochemistry, Copenhagen University Hospital-Rigshospitalet, Copenhagen, Denmark
| | - Yolande A L Pijnenburg
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Henne Holstege
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
- Department of Clinical Genetics, VU University Medical Centre, Amsterdam, The Netherlands
| | | | - Inez Ramakers
- Maastricht University, Department of Psychiatry & Neuropsychologie, Alzheimer Center Limburg, Maastricht, The Netherlands
| | - Frans Verhey
- Maastricht University, Department of Psychiatry & Neuropsychologie, Alzheimer Center Limburg, Maastricht, The Netherlands
| | - Philip Scheltens
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Caroline Graff
- Unit for Hereditary Dementias, Theme Aging, Karolinska University Hospital-Solna, 171 64, Stockholm, Sweden
| | - Goran Papenberg
- Aging Research Center, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet and Stockholm University, Stockholm, Sweden
| | - Vilmantas Giedraitis
- Department of Public Health and Caring Sciences/Geriatrics, Uppsala University, Uppsala, Sweden
| | - Anne Boland
- Université Paris-Saclay, CEA, Centre National de Recherche en Génomique Humaine, 91057, Evry, France
| | - Jean-François Deleuze
- Université Paris-Saclay, CEA, Centre National de Recherche en Génomique Humaine, 91057, Evry, France
| | - Gael Nicolas
- Univ Rouen Normandie, Normandie Univ, Inserm U1245 and CHU Rouen, Department of Genetics and CNRMAJ, F-76000, Rouen, France
| | - Carole Dufouil
- Inserm, Bordeaux Population Health Research Center, UMR 1219, Univ. Bordeaux, ISPED, CIC 1401-EC, Univ. Bordeaux, Bordeaux, France
- CHU de Bordeaux, Pole Santé Publique, Bordeaux, France
| | - Florence Pasquier
- Univ. Lille, Inserm 1171, CHU Clinical and Research Memory Research Centre (CMRR) of Distalz, Lille, France
| | - Olivier Hanon
- Université de Paris, EA 4468, APHP, Hôpital Broca, Paris, France
| | - Stéphanie Debette
- University Bordeaux, Inserm, Bordeaux Population Health Research Center, Bordeaux, France
- Department of Neurology, Bordeaux University Hospital, Bordeaux, France
| | - Edna Grünblatt
- Department of Child and Adolescent Psychiatry and Psychotherapy, University Hospital of Psychiatry Zurich, University of Zurich, Zurich, Switzerland
- Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
- Zurich Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland
| | - Julius Popp
- Old Age Psychiatry, Department of Psychiatry, Lausanne University Hospital, Lausanne, Switzerland
- Department of Geriatric Psychiatry, University Hospital of Psychiatry Zürich, Zürich, Switzerland
- Institute for Regenerative Medicine, University of Zürich, Zurich, Switzerland
| | - Roberta Ghidoni
- Molecular Markers Laboratory, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, 25125, Italy
| | - Daniela Galimberti
- Neurodegenerative Diseases Unit, Fondazione IRCCS Ca' Granda, Ospedale Policlinico, Milan, Italy
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, Milan, Italy
| | - Beatrice Arosio
- Department of Clinical Sciences and Community Health, University of Milan, 20122, Milan, Italy
- Geriatric Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, 20122, Milan, Italy
| | - Patrizia Mecocci
- Institute of Gerontology and Geriatrics, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Vincenzo Solfrizzi
- Interdisciplinary Department of Medicine, Geriatric Medicine and Memory Unit, University of Bari "A. Moro", Bari, Italy
| | - Lucilla Parnetti
- Centre for Memory Disturbances, Lab of Clinical Neurochemistry, Section of Neurology, University of Perugia, Perugia, Italy
| | - Alessio Squassina
- Department of Biomedical Sciences, Section of Neuroscience and Clinical Pharmacology, University of Cagliari, Cagliari, Italy
| | - Lucio Tremolizzo
- Neurology Unit, "San Gerardo" Hospital, Monza and University of Milano-Bicocca, Milan, Italy
| | - Barbara Borroni
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
- Cognitive and Behavioural Neurology, Department of Continuity of Care and Frailty, ASST Spedali Civili Brescia, Brescia, Italy
| | - Benedetta Nacmias
- Department of Neuroscience, Psychology, Drug Research and Child Health University of Florence, Florence, Italy
- IRCCS Fondazione Don Carlo Gnocchi, Florence, Italy
| | - Marco Spallazzi
- Department of Medicine and Surgery, Unit of Neurology, University-Hospital of Parma, Parma, Italy
| | - Davide Seripa
- Department of Hematology and Stem Cell Transplant, Vito Fazzi Hospital, Lecce, Italy
| | - Innocenzo Rainero
- Department of Neuroscience "Rita Levi Montalcini", University of Torino, Torino, Italy
| | - Antonio Daniele
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy
- Neurology Unit, IRCCS Fondazione Policlinico Universitario A. Gemelli, Rome, Italy
| | - Paola Bossù
- Laboratory of Experimental Neuropsychobiology, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Carlo Masullo
- Institute of Neurology, Catholic University of the Sacred Heart, Rome, Italy
| | - Giacomina Rossi
- Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Frank Jessen
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Department of Psychiatry and Psychotherapy, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Cluster of Excellence Cellular Stress Responses in Aging-associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Victoria Fernandez
- Research Center and Memory Clinic, ACE Alzheimer Center Barcelona, Universitat Internacional de Catalunya, Barcelona, Spain
| | - Patrick Gavin Kehoe
- Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Ruth Frikke-Schmidt
- Department of Clinical Biochemistry, Copenhagen University Hospital-Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Magda Tsolaki
- 1st Department of Neurology, Medical school, Aristotle University of Thessaloniki, Thessaloniki, Makedonia, Greece
- Laboratory of Genetics, Immunology and Human Pathology, Faculty of Science of Tunis, University of Tunis El Manar, 2092, Tunis, Tunisia
| | - Pascual Sánchez-Juan
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
- Alzheimer's Centre Reina Sofia-CIEN Foundation-ISCIII, Madrid, Spain
| | - Kristel Sleegers
- Complex Genetics of Alzheimer's Disease Group, VIB Center for Molecular Neurology, VIB, Antwerp, Belgium
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Martin Ingelsson
- Department of Public Health and Caring Sciences/Geriatrics, Uppsala University, Uppsala, Sweden
- Krembil Brain Institute, University Health Network, Toronto, ON, Canada
- Tanz Centre for Research in Neurodegenerative Diseases, Departments of Medicine and Laboratory Medicine & Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Jonathan Haines
- Department of Population and Quantitative Health Sciences and Cleveland Institute for Computational Biology, Case Western Reserve University, Cleveland, OH, USA
| | - Lindsay Farrer
- Department of Neurology, Boston University, Boston, MA, USA
- Department of Biostatistics, Boston University, Boston, MA, USA
- Department of Epidemiology, Boston University, Boston, MA, USA
- Department of Medicine (Biomedical Genetics), Boston University, Boston, MA, USA
- Department of Ophthalmology, Boston University, Boston, MA, USA
| | - Richard Mayeux
- Taub Institute on Alzheimer's Disease and the Aging Brain, Department of Neurology, Columbia University, New York, NY, USA
- Gertrude H. Sergievsky Center, Columbia University, New York, NY, USA
| | - Li-San Wang
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Rebecca Sims
- Division of Psychological Medicine and Clinical Neuroscience, School of Medicine, Cardiff University, Wales, UK
| | - Anita DeStefano
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Gerard D Schellenberg
- Penn Neurodegeneration Genomics Center, Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Sudha Seshadri
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, San Antonio, TX, USA
- Boston University and the NHLBI's Framingham Heart Study, Boston, MA, USA
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA
| | - Philippe Amouyel
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, LabEx DISTALZ - U1167-RID-AGE Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, Lille, France
| | - Julie Williams
- Division of Psychological Medicine and Clinical Neuroscience, School of Medicine, Cardiff University, Wales, UK
- UK Dementia Research Institute, Cardiff University, Cardiff, UK
| | - Wiesje van der Flier
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Alfredo Ramirez
- Department of Old Age Psychiatry and Cognitive Disorders, University Hospital Bonn, University of Bonn, Bonn, Germany
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Division of Neurogenetics and Molecular Psychiatry, Department of Psychiatry and Psychotherapy, University of Cologne, Medical Faculty, Cologne, Germany
- Department of Psychiatry & Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, San Antonio, TX, USA
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Disease (CECAD), University of Cologne, Cologne, Germany
| | - Margaret Pericak-Vance
- The John P. Hussman Institute for Human Genomics, University of Miami, Miami, FL, USA
- Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami, Miami, FL, USA
| | - Ole A Andreassen
- NORMENT Centre, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Cornelia Van Duijn
- Nuffield Department of Population Health Oxford University, Oxford, UK
- Department of Epidemiology, Erasmus MC, Rotterdam, The Netherlands
| | - Mikko Hiltunen
- Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland
| | - Agustín Ruiz
- Research Center and Memory Clinic, ACE Alzheimer Center Barcelona, Universitat Internacional de Catalunya, Barcelona, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
| | - Josée Dupuis
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
- Department of Epidemiology, Biostatistics and Occupational Health, McGill University, Montreal, QC, Canada
| | - Eden Martin
- The John P. Hussman Institute for Human Genomics, University of Miami, Miami, FL, USA
- Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami, Miami, FL, USA
- University of Miami Miller School of Medicine, Miami, FL, USA
| | - Jean-Charles Lambert
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, LabEx DISTALZ - U1167-RID-AGE Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, Lille, France
| | - Brian Kunkle
- The John P. Hussman Institute for Human Genomics, University of Miami, Miami, FL, USA
| | - Céline Bellenguez
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, LabEx DISTALZ - U1167-RID-AGE Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, Lille, France.
| |
Collapse
|
7
|
Western D, Timsina J, Wang L, Wang C, Yang C, Phillips B, Wang Y, Liu M, Ali M, Beric A, Gorijala P, Kohlfeld P, Budde J, Levey AI, Morris JC, Perrin RJ, Ruiz A, Marquié M, Boada M, de Rojas I, Rutledge J, Oh H, Wilson EN, Le Guen Y, Reus LM, Tijms B, Visser PJ, van der Lee SJ, Pijnenburg YAL, Teunissen CE, Del Campo Milan M, Alvarez I, Aguilar M, Greicius MD, Pastor P, Pulford DJ, Ibanez L, Wyss-Coray T, Sung YJ, Cruchaga C. Proteogenomic analysis of human cerebrospinal fluid identifies neurologically relevant regulation and implicates causal proteins for Alzheimer's disease. Nat Genet 2024; 56:2672-2684. [PMID: 39528825 DOI: 10.1038/s41588-024-01972-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 10/02/2024] [Indexed: 11/16/2024]
Abstract
The integration of quantitative trait loci (QTLs) with disease genome-wide association studies (GWASs) has proven successful in prioritizing candidate genes at disease-associated loci. QTL mapping has been focused on multi-tissue expression QTLs or plasma protein QTLs (pQTLs). We generated a cerebrospinal fluid (CSF) pQTL atlas by measuring 6,361 proteins in 3,506 samples. We identified 3,885 associations for 1,883 proteins, including 2,885 new pQTLs, demonstrating unique genetic regulation in CSF. We identified CSF-enriched pleiotropic regions on chromosome (chr)3q28 near OSTN and chr19q13.32 near APOE that were enriched for neuron specificity and neurological development. We integrated our associations with Alzheimer's disease (AD) through proteome-wide association study (PWAS), colocalization and Mendelian randomization and identified 38 putative causal proteins, 15 of which have drugs available. Finally, we developed a proteomics-based AD prediction model that outperforms genetics-based models. These findings will be instrumental to further understand the biology and identify causal and druggable proteins for brain and neurological traits.
Collapse
Affiliation(s)
- Daniel Western
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, USA
- Division of Biology and Biomedical Sciences, Washington University School of Medicine, St. Louis, MO, USA
| | - Jigyasha Timsina
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Lihua Wang
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Ciyang Wang
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, USA
- Division of Biology and Biomedical Sciences, Washington University School of Medicine, St. Louis, MO, USA
| | - Chengran Yang
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Bridget Phillips
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, USA
- Division of Biology and Biomedical Sciences, Washington University School of Medicine, St. Louis, MO, USA
| | - Yueyao Wang
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Menghan Liu
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Muhammad Ali
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Aleksandra Beric
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Priyanka Gorijala
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Pat Kohlfeld
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, USA
| | - John Budde
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Allan I Levey
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA
| | - John C Morris
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Richard J Perrin
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
- Hope Center for Neurological Disorders, Washington University, St. Louis, MO, USA
| | - Agustin Ruiz
- ACE Alzheimer Center Barcelona, Universitat Internacional de Catalunya, Barcelona, Spain
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, University of Texas Health Science Center, San Antonio, TX, USA
| | - Marta Marquié
- ACE Alzheimer Center Barcelona, Universitat Internacional de Catalunya, Barcelona, Spain
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Mercè Boada
- ACE Alzheimer Center Barcelona, Universitat Internacional de Catalunya, Barcelona, Spain
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Itziar de Rojas
- ACE Alzheimer Center Barcelona, Universitat Internacional de Catalunya, Barcelona, Spain
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Jarod Rutledge
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
| | - Hamilton Oh
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
| | - Edward N Wilson
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
| | - Yann Le Guen
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
- Quantitative Sciences Unit, Department of Medicine, Stanford University, Stanford, CA, USA
| | - Lianne M Reus
- 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
| | - Betty 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
| | - 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
| | - 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
- Genomics of Neurodegenerative Diseases and Aging, Department of Human Genetics, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
| | - 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
| | - Charlotte E Teunissen
- Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Amsterdam University Medical Centers, Location VUmc, Amsterdam, the Netherlands
| | - Marta Del Campo Milan
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad San Pablo CEU, CEU Universities, Madrid, Spain
- Barcelonaβeta Brain Research Center, Pasqual Maragall Foundation, Barcelona, Spain
| | - Ignacio Alvarez
- Memory Disorders Unit, Department of Neurology, University Hospital Mutua Terrassa, Terrassa, Spain
| | - Miquel Aguilar
- Memory Disorders Unit, Department of Neurology, University Hospital Mutua Terrassa, Terrassa, Spain
| | - Michael D Greicius
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
| | - Pau Pastor
- Memory Disorders Unit, Department of Neurology, University Hospital Mutua Terrassa, Terrassa, Spain
- Unit of Neurodegenerative Diseases, Department of Neurology, University Hospital Germans Trias i Pujol and the Germans Trias i Pujol Research Institute (IGTP) Badalona, Barcelona, Spain
| | | | - Laura Ibanez
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, USA
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Tony Wyss-Coray
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
| | - Yun Ju Sung
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, USA
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO, USA
| | - Carlos Cruchaga
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA.
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, USA.
- Hope Center for Neurological Disorders, Washington University, St. Louis, MO, USA.
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
8
|
Moore A, Ritchie MD. Is the Relationship Between Cardiovascular Disease and Alzheimer's Disease Genetic? A Scoping Review. Genes (Basel) 2024; 15:1509. [PMID: 39766777 PMCID: PMC11675426 DOI: 10.3390/genes15121509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 11/20/2024] [Accepted: 11/22/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND/OBJECTIVES Cardiovascular disease (CVD) and Alzheimer's disease (AD) are two diseases highly prevalent in the aging population and often co-occur. The exact relationship between the two diseases is uncertain, though epidemiological studies have demonstrated that CVDs appear to increase the risk of AD and vice versa. This scoping review aims to examine the current identified overlapping genetics between CVDs and AD at the individual gene level and at the shared pathway level. METHODS Following PRISMA-ScR guidelines for a scoping review, we searched the PubMed and Scopus databases from 1990 to October 2024 for articles that involved (1) CVDs, (2) AD, and (3) used statistical methods to parse genetic relationships. RESULTS Our search yielded 2918 articles, of which 274 articles passed screening and were organized into two main sections: (1) evidence of shared genetic risk; and (2) shared mechanisms. The genes APOE, PSEN1, and PSEN2 reportedly have wide effects across the AD and CVD spectrum, affecting both cardiac and brain tissues. Mechanistically, changes in three main pathways (lipid metabolism, blood pressure regulation, and the breakdown of the blood-brain barrier (BBB)) contribute to subclinical and etiological changes that promote both AD and CVD progression. However, genetic studies continue to be limited by the availability of longitudinal data and lack of cohorts that are representative of diverse populations. CONCLUSIONS Highly penetrant familial genes simultaneously increase the risk of CVDs and AD. However, in most cases, sets of dysregulated genes within larger-scale mechanisms, like changes in lipid metabolism, blood pressure regulation, and BBB breakdown, increase the risk of both AD and CVDs and contribute to disease progression.
Collapse
Affiliation(s)
- Anni Moore
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA;
| | - Marylyn D. Ritchie
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA;
- Division of Informatics, Department of Biostatistics, Epidemiology & Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Penn Institute for Biomedical Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
9
|
Escarcega RD, Vijay Kumar MJ, Kyriakopoulos VE, Ortiz GJ, Gusdon AM, Fan H, Peesh P, Conesa MPB, Colpo GD, Ahnstedt HW, Couture L, Kim SH, Hinojosa M, Farrell CM, Marrelli SP, Urayama A, Ganesh BP, Schulz PE, McCullough LD, Tsvetkov AS. Serum metabolome profiling in patients with mild cognitive impairment reveals sex differences in lipid metabolism. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.11.623108. [PMID: 39605322 PMCID: PMC11601308 DOI: 10.1101/2024.11.11.623108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Alzheimer's disease (AD) affects more women than men. Although women live longer than men, it is not longevity alone, but other factors, including metabolic changes, that contribute to the higher risk of AD in women. Metabolic pathways have been implicated in AD progression, but studies to date examined targeted pathways, leaving many metabolites unmeasured. Sex is often a neglected biological variable, and most metabolomic studies were not designed to investigate sex differences in metabolomic profiles. Here, we performed untargeted metabolomic profiling of sera from male and female patients with mild cognitive impairment (MCI), a common precursor to AD, and matched controls. We discovered significant metabolic changes in individuals with MCI, and found several pathways that were strongly associated with sex. Peptide energy metabolism demonstrated sexual dimorphism. Lipid pathways exhibited the strongest differences between female and male MCI patients, including specific phosphatidylcholine lipids, lysophospholipids, long-chain fatty acids, and monoacylglycerols. 1-palmitoleoyl glycerol and 1-arachidonoyl glycerol were higher in female MCI subjects than in male MCI subjects with no differences between control males and females. Conversely, specific dicarboxylic fatty acids were lower in female MCI subjects than male MCI subjects. In cultured astrocytes, 1-arachidonoyl glycerol promoted phosphorylation of the transcriptional regulator sphingosine kinase 2, which was inhibited by the transient receptor potential vanilloid 1 receptor antagonists, as well as chromatin remodelling. Overall, we identified novel sex-specific metabolites in MCI patients that could serve as biomarkers of MCI in both sexes, help further define AD etiology, and reveal new potential prevention strategies for AD.
Collapse
Affiliation(s)
- Rocio Diaz Escarcega
- Department of Neurology, the University of Texas McGovern Medical School at Houston, TX, USA
| | - M. J. Vijay Kumar
- Department of Neurology, the University of Texas McGovern Medical School at Houston, TX, USA
| | | | - Guadalupe J. Ortiz
- Department of Neurology, the University of Texas McGovern Medical School at Houston, TX, USA
| | - Aaron M. Gusdon
- Department of Neurosurgery, the University of Texas McGovern Medical School at Houston, TX, USA
| | - Huihui Fan
- Department of Neurology, the University of Texas McGovern Medical School at Houston, TX, USA
| | - Pedram Peesh
- Department of Neurology, the University of Texas McGovern Medical School at Houston, TX, USA
| | - Maria P. Blasco Conesa
- Department of Neurology, the University of Texas McGovern Medical School at Houston, TX, USA
| | - Gabriela Delevati Colpo
- Department of Neurology, the University of Texas McGovern Medical School at Houston, TX, USA
| | - Hilda W. Ahnstedt
- Department of Neurology, the University of Texas McGovern Medical School at Houston, TX, USA
| | - Lucy Couture
- Department of Neurology, the University of Texas McGovern Medical School at Houston, TX, USA
| | - Stella H. Kim
- Department of Neurology, the University of Texas McGovern Medical School at Houston, TX, USA
- The University of Texas Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Miriam Hinojosa
- Department of Neurology, the University of Texas McGovern Medical School at Houston, TX, USA
| | - Christine M. Farrell
- Department of Neurology, the University of Texas McGovern Medical School at Houston, TX, USA
| | - Sean P. Marrelli
- Department of Neurology, the University of Texas McGovern Medical School at Houston, TX, USA
| | - Akihiko Urayama
- Department of Neurology, the University of Texas McGovern Medical School at Houston, TX, USA
| | - Bhanu P. Ganesh
- Department of Neurology, the University of Texas McGovern Medical School at Houston, TX, USA
| | - Paul E. Schulz
- Department of Neurology, the University of Texas McGovern Medical School at Houston, TX, USA
| | - Louise D. McCullough
- Department of Neurology, the University of Texas McGovern Medical School at Houston, TX, USA
- The University of Texas Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Andrey S. Tsvetkov
- Department of Neurology, the University of Texas McGovern Medical School at Houston, TX, USA
- The University of Texas Graduate School of Biomedical Sciences, Houston, TX, USA
- UTHealth Consortium on Aging, the University of Texas McGovern Medical School, Houston, TX, USA
| |
Collapse
|
10
|
Wang X, Yang H, Zhan D, Sun H, Huang Q, Zhang Y, Lin Y, Wei G, Hua F, Liu L, Chen S. Novel targets for the treatment and prevention of Alzheimer's disease in the European population, inspiration from amyloid beta and tau protein. Heliyon 2024; 10:e39013. [PMID: 39492919 PMCID: PMC11531621 DOI: 10.1016/j.heliyon.2024.e39013] [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: 07/03/2024] [Revised: 09/30/2024] [Accepted: 10/04/2024] [Indexed: 11/05/2024] Open
Abstract
Alzheimer's disease (AD) is a gradual neurodegenerative ailment that lacks any disease-modifying intervention. Our objective was to pinpoint pharmacological targets with a focus on amyloid beta (Aβ) and tau to treat and prevent AD in the European population. A proteome-wide Mendelian randomization (MR) analysis was carried out to estimate the associations between proteins and cerebrospinal fluid (CSF) Aβ-42 and phosphorylated Tau (p-Tau). We utilized colocalization and MR analysis to investigate whether the identified proteins were associated with the risk of AD. Additionally, we expanded our investigation to include non-AD phenotypes by conducting a phenome-wide MR analysis of 1646 disease traits based on the FinnGen and UK Biobank databases to explore potential side effects. We identified 11 proteins that were genetically associated with both CSF Aβ-42 and p-Tau levels. The genetically predicted levels of three proteins, GAL3ST2, POLR1C, and BIN1, were found to be associated with an increased risk of AD with high colocalization. In the phenome-wide MR analysis, two out of the three biomarkers were associated with at least one disease, except for GAL3ST2, which was not associated with any disease under the threshold of FDR <0.1. POLR1C was found to be associated with the most disease traits, and all disease associations with genetically inhibited BIN1 were protective. The proteome-wide MR investigation revealed 11 proteins that were associated with the level of CSF Aβ-42 and p-Tau. Among them, GAL3ST2, POLR1C, and BIN1 were identified as potential therapeutic targets for AD and warrant further investigation.
Collapse
Affiliation(s)
- Xifeng Wang
- Department of Anesthesiology, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, 330006, 17# Yong Wai Zheng Street, Nanchang City, Jiangxi Province, PR China
- Department of Neuroscience, Tat Chee Avenue City University of Hong Kong, 999077, Hong Kong City, PR China
- Key Laboratory of Anesthesiology of Jiangxi Province, 330006, 1# Minde Road, Nanchang, Jiangxi Province, PR China
| | - Huayu Yang
- Clinical Medical College, Nanchang Medical College, 330052, 689# Huiren Big Road, Nanchang City, Jiangxi Province, PR China
- Key Laboratory of Anesthesiology of Jiangxi Province, 330006, 1# Minde Road, Nanchang, Jiangxi Province, PR China
| | - Dengcheng Zhan
- Department of Neuroscience, Tat Chee Avenue City University of Hong Kong, 999077, Hong Kong City, PR China
| | - Haiying Sun
- Department of Anesthesiology, Jiujiang Women and Children's Healthcare Hospital, 332001, 61# Gansang South Road, Jiujiang City, Jiangxi Province, PR China
| | - Qiang Huang
- Department of Anesthesiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, 1 Minde Road, Nanchang, 330006, Jiangxi Province, PR China
- Key Laboratory of Anesthesiology of Jiangxi Province, 330006, 1# Minde Road, Nanchang, Jiangxi Province, PR China
| | - Yiping Zhang
- Department of Anesthesiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, 1 Minde Road, Nanchang, 330006, Jiangxi Province, PR China
- Key Laboratory of Anesthesiology of Jiangxi Province, 330006, 1# Minde Road, Nanchang, Jiangxi Province, PR China
| | - Yue Lin
- Department of Anesthesiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, 1 Minde Road, Nanchang, 330006, Jiangxi Province, PR China
- Key Laboratory of Anesthesiology of Jiangxi Province, 330006, 1# Minde Road, Nanchang, Jiangxi Province, PR China
| | - Gen Wei
- Department of Anesthesiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, 1 Minde Road, Nanchang, 330006, Jiangxi Province, PR China
- Key Laboratory of Anesthesiology of Jiangxi Province, 330006, 1# Minde Road, Nanchang, Jiangxi Province, PR China
| | - Fuzhou Hua
- Department of Anesthesiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, 1 Minde Road, Nanchang, 330006, Jiangxi Province, PR China
- Key Laboratory of Anesthesiology of Jiangxi Province, 330006, 1# Minde Road, Nanchang, Jiangxi Province, PR China
| | - Li Liu
- Department of Anesthesiology, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, 330006, 17# Yong Wai Zheng Street, Nanchang City, Jiangxi Province, PR China
- Key Laboratory of Anesthesiology of Jiangxi Province, 330006, 1# Minde Road, Nanchang, Jiangxi Province, PR China
| | - Shibiao Chen
- Department of Anesthesiology, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, 330006, 17# Yong Wai Zheng Street, Nanchang City, Jiangxi Province, PR China
- Key Laboratory of Anesthesiology of Jiangxi Province, 330006, 1# Minde Road, Nanchang, Jiangxi Province, PR China
| |
Collapse
|
11
|
Masurkar AV, Marsh K, Morgan B, Leitner D, Wisniewski T. Factors Affecting Resilience and Prevention of Alzheimer's Disease and Related Dementias. Ann Neurol 2024; 96:633-649. [PMID: 39152774 PMCID: PMC11534551 DOI: 10.1002/ana.27055] [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: 02/24/2024] [Revised: 07/27/2024] [Accepted: 07/31/2024] [Indexed: 08/19/2024]
Abstract
Alzheimer's disease (AD) is a devastating, age-associated neurodegenerative disorder and the most common cause of dementia. The clinical continuum of AD spans from preclinical disease to subjective cognitive decline, mild cognitive impairment, and dementia stages (mild, moderate, and severe). Neuropathologically, AD is defined by the accumulation of amyloid β (Aβ) into extracellular plaques in the brain parenchyma and in the cerebral vasculature, and by abnormally phosphorylated tau that accumulates intraneuronally forming neurofibrillary tangles (NFTs). Development of treatment approaches that prevent or even reduce the cognitive decline because of AD has been slow compared to other major causes of death. Recently, the United States Food and Drug Administration gave full approval to 2 different Aβ-targeting monoclonal antibodies. However, this breakthrough disease modifying approach only applies to a limited subset of patients in the AD continuum and there are stringent eligibility criteria. Furthermore, these approaches do not prevent progression of disease, because other AD-related pathologies, such as NFTs, are not directly targeted. A non-mutually exclusive alternative is to address lifestyle interventions that can help reduce the risk of AD and AD-related dementias (ADRD). It is estimated that addressing such modifiable risk factors could potentially delay up to 40% of AD/ADRD cases. In this review, we discuss some of the many modifiable risk factors that may be associated with prevention of AD/ADRD and/or increasing brain resilience, as well as other factors that may interact with these modifiable risk factors to influence AD/ADRD progression. [Color figure can be viewed at www.annalsofneurology.org] ANN NEUROL 2024;96:633-649.
Collapse
Affiliation(s)
- Arjun V. Masurkar
- Department of Neurology, New York University Grossman School of Medicine, 560 First Avenue, New York, NY 10016
- Center for Cognitive Neurology, New York University Grossman School of Medicine, 560 First Avenue, New York, NY 10016
- Department of Neuroscience and Physiology, New York University Grossman School of Medicine, 560 First Avenue, New York, NY 10016
| | - Karyn Marsh
- Department of Neurology, New York University Grossman School of Medicine, 560 First Avenue, New York, NY 10016
- Center for Cognitive Neurology, New York University Grossman School of Medicine, 560 First Avenue, New York, NY 10016
| | - Brianna Morgan
- Department of Medicine, New York University Grossman School of Medicine, 560 First Avenue, New York, NY 10016
| | - Dominique Leitner
- Department of Neurology, New York University Grossman School of Medicine, 560 First Avenue, New York, NY 10016
- Center for Cognitive Neurology, New York University Grossman School of Medicine, 560 First Avenue, New York, NY 10016
| | - Thomas Wisniewski
- Department of Neurology, New York University Grossman School of Medicine, 560 First Avenue, New York, NY 10016
- Center for Cognitive Neurology, New York University Grossman School of Medicine, 560 First Avenue, New York, NY 10016
- Department of Pathology, New York University Grossman School of Medicine, 560 First Avenue, New York, NY 10016
- Department of Psychiatry, New York University Grossman School of Medicine, 560 First Avenue, New York, NY 10016
| |
Collapse
|
12
|
Hu Y, Cao C, Li M, He H, Luo L, Guo Y. Association between idiopathic normal pressure hydrocephalus and Alzheimer's disease: a bidirectional Mendelian randomization study. Sci Rep 2024; 14:22744. [PMID: 39349954 PMCID: PMC11443089 DOI: 10.1038/s41598-024-72559-w] [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: 05/29/2024] [Accepted: 09/09/2024] [Indexed: 10/04/2024] Open
Abstract
Observational studies have suggested a bidirectional relationship between idiopathic normal pressure hydrocephalus (iNPH) and Alzheimer's disease (AD). However, the causal association between these two neurodegenerative disorders remains unclear. This study aimed to explore the causal relationship between iNPH and AD using a two-sample bidirectional Mendelian randomization (MR) method. Large-scale genome-wide association studies of iNPH (Ncase = 767, Ncontrol = 375,610) and AD (Ncase/proxy = 111,326, Ncontrol = 677,663) in European individuals were used to screen genetic instruments for MR analysis. Inverse variance-weighted (IVW) method was used as the main analysis, other MR methods and a series of sensitivity analyses were performed to ensure the reliability. In the forward MR analysis, genetic predisposition to iNPH had no effects on the risk of AD development. Likewise, in the reverse MR analysis, AD did not demonstrate a significant causal effect on iNPH. Sensitivity analyses bolstered the reliability of the MR results. Our MR study indicated no genetic evidence supporting a suggestive association between AD and iNPH in either direction, and provided evidence on the dichotomy between true iNPH and neurodegenerative NPH.
Collapse
Affiliation(s)
- Yuanjun Hu
- Department of Neurosurgery, The Third Affiliated Hospital of Sun Yat-Sen University, 600 Tianhe Road, Guangzhou, 510630, China
| | - Chengan Cao
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Manting Li
- Department of Neurosurgery, The Third Affiliated Hospital of Sun Yat-Sen University, 600 Tianhe Road, Guangzhou, 510630, China
| | - Haiyong He
- Department of Neurosurgery, The Third Affiliated Hospital of Sun Yat-Sen University, 600 Tianhe Road, Guangzhou, 510630, China
| | - Lun Luo
- Department of Neurosurgery, The Third Affiliated Hospital of Sun Yat-Sen University, 600 Tianhe Road, Guangzhou, 510630, China.
| | - Ying Guo
- Department of Neurosurgery, The Third Affiliated Hospital of Sun Yat-Sen University, 600 Tianhe Road, Guangzhou, 510630, China.
| |
Collapse
|
13
|
Nho K, Risacher SL, Apostolova LG, Bice PJ, Brosch JR, Deardorff R, Faber K, Farlow MR, Foroud T, Gao S, Rosewood T, Kim JP, Nudelman K, Yu M, Aisen P, Sperling R, Hooli B, Shcherbinin S, Svaldi D, Jack CR, Jagust WJ, Landau S, Vasanthakumar A, Waring JF, Doré V, Laws SM, Masters CL, Porter T, Rowe CC, Villemagne VL, Dumitrescu L, Hohman TJ, Libby JB, Mormino E, Buckley RF, Johnson K, Yang HS, Petersen RC, Ramanan VK, Ertekin-Taner N, Vemuri P, Cohen AD, Fan KH, Kamboh MI, Lopez OL, Bennett DA, Ali M, Benzinger T, Cruchaga C, Hobbs D, De Jager PL, Fujita M, Jadhav V, Lamb BT, Tsai AP, Castanho I, Mill J, Weiner MW, Saykin AJ. CYP1B1-RMDN2 Alzheimer's disease endophenotype locus identified for cerebral tau PET. Nat Commun 2024; 15:8251. [PMID: 39304655 PMCID: PMC11415491 DOI: 10.1038/s41467-024-52298-2] [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: 11/14/2023] [Accepted: 09/01/2024] [Indexed: 09/22/2024] Open
Abstract
Determining the genetic architecture of Alzheimer's disease pathologies can enhance mechanistic understanding and inform precision medicine strategies. Here, we perform a genome-wide association study of cortical tau quantified by positron emission tomography in 3046 participants from 12 independent studies. The CYP1B1-RMDN2 locus is associated with tau deposition. The most significant signal is at rs2113389, explaining 4.3% of the variation in cortical tau, while APOE4 rs429358 accounts for 3.6%. rs2113389 is associated with higher tau and faster cognitive decline. Additive effects, but no interactions, are observed between rs2113389 and diagnosis, APOE4, and amyloid beta positivity. CYP1B1 expression is upregulated in AD. rs2113389 is associated with higher CYP1B1 expression and methylation levels. Mouse model studies provide additional functional evidence for a relationship between CYP1B1 and tau deposition but not amyloid beta. These results provide insight into the genetic basis of cerebral tau deposition and support novel pathways for therapeutic development in AD.
Collapse
Affiliation(s)
- Kwangsik Nho
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, USA
- Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, USA
- Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, USA
- Department of BioHealth Informatics, Indiana University, Indianapolis, USA
| | - Shannon L Risacher
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, USA
- Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, USA
| | - Liana G Apostolova
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, USA
- Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, USA
- Department of Neurology, Indiana University School of Medicine, Indianapolis, USA
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, USA
| | - Paula J Bice
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, USA
- Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, USA
| | - Jared R Brosch
- Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, USA
- Department of Neurology, Indiana University School of Medicine, Indianapolis, USA
| | - Rachael Deardorff
- Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, USA
- Department of Neurology, Indiana University School of Medicine, Indianapolis, USA
| | - Kelley Faber
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, USA
- National Centralized Repository for Alzheimer's Disease and Related Dementias, Indiana University School of Medicine, Indianapolis, USA
| | - Martin R Farlow
- Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, USA
- Department of Neurology, Indiana University School of Medicine, Indianapolis, USA
| | - Tatiana Foroud
- Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, USA
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, USA
- National Centralized Repository for Alzheimer's Disease and Related Dementias, Indiana University School of Medicine, Indianapolis, USA
| | - Sujuan Gao
- Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, USA
- Department of Biostatistics, Indiana University School of Medicine, Indianapolis, USA
| | - Thea Rosewood
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, USA
- Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, USA
- Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, USA
| | - Jun Pyo Kim
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, USA
- Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, USA
- Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, USA
| | - Kelly Nudelman
- Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, USA
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, USA
- National Centralized Repository for Alzheimer's Disease and Related Dementias, Indiana University School of Medicine, Indianapolis, USA
| | - Meichen Yu
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, USA
- Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, USA
| | - Paul Aisen
- Department of Neurology, Keck School of Medicine, University of Southern California, San Diego, USA
| | - Reisa Sperling
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, USA
| | | | | | | | | | - William J Jagust
- UC Berkeley Helen Wills Neuroscience Institute, University of California - Berkeley, Berkeley, USA
| | - Susan Landau
- UC Berkeley Helen Wills Neuroscience Institute, University of California - Berkeley, Berkeley, USA
| | | | | | - Vincent Doré
- CSIRO Health and Biosecurity, Melbourne, Australia
- Department of Molecular Imaging & Therapy, Austin Health, Heidelberg, Australia
| | - Simon M Laws
- Centre for Precision Health, School of Medical and Health Sciences, Edith Cowan University, Joondalup, Australia
| | - Colin L Masters
- Florey Institute of Neuroscience and Mental Health and The University of Melbourne, Parkville, Australia
| | - Tenielle Porter
- Centre for Precision Health, School of Medical and Health Sciences, Edith Cowan University, Joondalup, Australia
| | - Christopher C Rowe
- Department of Molecular Imaging & Therapy, Austin Health, Heidelberg, Australia
- Florey Institute of Neuroscience and Mental Health and The University of Melbourne, Parkville, Australia
| | - Victor L Villemagne
- Department of Molecular Imaging & Therapy, Austin Health, Heidelberg, Australia
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, USA
| | - Logan Dumitrescu
- Vanderbilt Memory & Alzheimer's Center, Vanderbilt University Medical Center, Nashville, USA
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, USA
| | - Timothy J Hohman
- Vanderbilt Memory & Alzheimer's Center, Vanderbilt University Medical Center, Nashville, USA
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, USA
| | - Julia B Libby
- Vanderbilt Memory & Alzheimer's Center, Vanderbilt University Medical Center, Nashville, USA
| | - Elizabeth Mormino
- Department of Neurology & Neurological Sciences, Stanford University, Stanford, USA
| | - Rachel F Buckley
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, USA
| | - Keith Johnson
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, USA
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, USA
| | - Hyun-Sik Yang
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, USA
- Center for Alzheimer's Research and Treatment, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, USA
| | | | | | - Nilüfer Ertekin-Taner
- Department of Neurology, Mayo Clinic, Jacksonville, USA
- Department of Neuroscience, Mayo Clinic, Jacksonville, USA
| | | | - Ann D Cohen
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, USA
| | - Kang-Hsien Fan
- Department of Human Genetics, University of Pittsburgh, Pittsburgh, USA
| | - M Ilyas Kamboh
- Department of Human Genetics, University of Pittsburgh, Pittsburgh, USA
| | - Oscar L Lopez
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, USA
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, USA
| | - David A Bennett
- Department of Neurological Sciences, Rush Medical College, Rush University, Chicago, USA
| | - Muhammad Ali
- Department of Psychiatry, Washington University, St. Louis, USA
| | - Tammie Benzinger
- Department of Radiology, Washington University School of Medicine, St. Louis, USA
| | - Carlos Cruchaga
- Department of Psychiatry, Washington University, St. Louis, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, USA
| | - Diana Hobbs
- Department of Radiology, Washington University School of Medicine, St. Louis, USA
| | - Philip L De Jager
- Center for Translational and Computational Neuroimmunology, Department of Neurology and Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, USA
| | - Masashi Fujita
- Center for Translational and Computational Neuroimmunology, Department of Neurology and Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, USA
| | - Vaishnavi Jadhav
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, USA
- Stark Neuroscience Research Institute, Indiana University School of Medicine, Indianapolis, USA
| | - Bruce T Lamb
- Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, USA
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, USA
- Stark Neuroscience Research Institute, Indiana University School of Medicine, Indianapolis, USA
| | - Andy P Tsai
- Department of Neurology & Neurological Sciences, Stanford University, Stanford, USA
- Stark Neuroscience Research Institute, Indiana University School of Medicine, Indianapolis, USA
- Wu Tsai Neurosciences Institute, Stanford University School of Medicine, Stanford, USA
| | - Isabel Castanho
- Department for Clinical and Biomedical Sciences, University of Exeter Medical School, University of Exeter, Exeter, UK
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, USA
| | - Jonathan Mill
- Department for Clinical and Biomedical Sciences, University of Exeter Medical School, University of Exeter, Exeter, UK
| | - Michael W Weiner
- Departments of Radiology, Medicine, and Psychiatry, University of California-San Francisco, San Francisco, USA
- Department of Veterans Affairs Medical Center, San Francisco, USA
| | - Andrew J Saykin
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, USA.
- Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, USA.
- Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, USA.
- Department of Neurology, Indiana University School of Medicine, Indianapolis, USA.
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, USA.
| |
Collapse
|
14
|
Räsänen J, Heikkinen S, Mäklin K, Lipponen A, Kuulasmaa T, Mehtonen J, Korhonen VE, Junkkari A, Grenier-Boley B, Bellenguez C, Oinas M, Avellan C, Frantzén J, Kotkansalo A, Rinne J, Ronkainen A, Kauppinen M, von Und Zu Fraunberg M, Lönnrot K, Satopää J, Perola M, Koivisto AM, Julkunen V, Portaankorva AM, Mannermaa A, Soininen H, Helisalmi S, Jääskeläinen JE, Lambert JC, Eide PK, Palotie A, Kurki MI, Hiltunen M, Leinonen V. Risk Variants Associated With Normal Pressure Hydrocephalus: Genome-Wide Association Study in the FinnGen Cohort. Neurology 2024; 103:e209694. [PMID: 39141892 PMCID: PMC11361830 DOI: 10.1212/wnl.0000000000209694] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Accepted: 05/24/2024] [Indexed: 08/16/2024] Open
Abstract
BACKGROUND AND OBJECTIVES Large-scale genome-wide studies of chronic hydrocephalus have been lacking. We conducted a genome-wide association study (GWAS) in normal pressure hydrocephalus (NPH). METHODS We used a case-control study design implementing FinnGen data containing 473,691 Finns with genotypes and nationwide health records. Patients with NPH were selected based on ICD-10 G91.2 diagnosis. To select patients with idiopathic NPH (iNPH) for sensitivity analysis, we excluded patients with a potentially known etiology of the condition using an algorithm on their disease history. The controls were the remaining non-hydrocephalic participants. For a replication analysis, the NPH cohort from UK Biobank (UKBB) was used. RESULTS We included 1,522 patients with NPH (mean age 72.2 years, 53% women) and 451,091 controls (mean age 60.5 years, 44% women). In the GWAS comparing patients with NPH with the controls, we identified 6 gene regions significantly (p < 5.0e-8) associated with NPH that replicated in a meta-analysis with UKBB (NPH n = 173). The top loci near the following genes were rs7962263, SLCO1A2 (odds ratio [OR] 0.71, 95% CI 0.65-0.78, p = 1.0e-14); rs798495, AMZ1/GNA12 (OR 1.29, 95% CI 1.20-1.39, p = 2.9e-12); rs10828247, MLLT10 (OR 0.77, 95% CI 0.71-0.83, p = 1.5e-11); rs561699566 and rs371919113, CDCA2 (OR 0.76, 95% CI 0.70-0.82, p = 1.5e-11); rs56023709, C16orf95 (OR 1.24, 95% CI 1.16-1.33, p = 3.0e-9); and rs62434144, PLEKHG1 (OR 1.23, 95% CI 1.14-1.32, p = 1.4e-8). In the sensitivity analysis comparing only patients with iNPH (n = 1,055) with the controls (n = 451,091), 4 top loci near the following genes remained significant: rs7962263, SLCO1A2 (OR 0.70, 95% CI 0.63-0.78, p = 2.1e-11); rs10828247, MLLT10 (OR 0.74, 95% CI 0.62-0.82, p = 4.6e-10); rs798511, AMZ1/GNA12 (OR 1.28, 95% CI 1.17-1.39, p = 1.7e-8); and rs56023709, C16orf95 (OR 1.28, 95% CI 1.17-1.39, p = 1.7e-8). DISCUSSION We identified 6 loci significantly associated with NPH in the thus far largest GWAS in chronic hydrocephalus. The genes near the top loci have previously been associated with blood-brain barrier and blood-CSF barrier function and with increased lateral brain ventricle volume. The effect sizes and allele frequencies remained similar in NPH and iNPH cohorts, indicating the identified loci are risk determinants for iNPH and likely not explained by associations with other etiologies. However, the exact role of these loci is still unknown, warranting further studies.
Collapse
Affiliation(s)
- Joel Räsänen
- From the Department of Neurosurgery (J. Räsänen, K.M., V.E.K., M.O., J.E.J., V.L.), Kuopio University Hospital and Institute of Clinical Medicine-Neurosurgery, and Institute of Biomedicine (S. Heikkinen, K.M., A.L., T.K., M.H.), University of Eastern Finland, Kuopio; Institute for Molecular Medicine Finland (FIMM) (J.M., A.P.), Helsinki Institute of Life Science (HiLIFE), University of Helsinki; Department of Neurology (A.J.), Clinical Neurosciences, Helsinki University Hospital and University of Helsinki, Finland; Univ. Lille (B.G.-B., C.B., J.-C.L.), Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, France; Department of Neurosurgery (M.O., K.L., J.S.), University of Helsinki and Helsinki University Hospital; Clinical Neurosciences (C.A., J.F., A.K., J. Rinne), Department of Neurosurgery, University of Turku and Turku University Hospital; Department of Neurosurgery (A.R.), Tampere University Hospital; Unit of Clinical Neuroscience (M.K., M.v.u.z.F.), Neurosurgery, University of Oulu and Medical Research Center, Oulu University Hospital; Finnish Institute for Health and Welfare (THL) (M.P.); University of Helsinki (M.P.); Department of Neurosciences (A.M.K., A.M.P.), University of Helsinki; Department of Geriatrics (A.M.K.), Helsinki University Hospital; NeuroCenter (A.M.K.), Kuopio University Hospital; Institute of Clinical Medicine-Neurology (V.J., H.S.), University of Eastern Finland; School of Medicine (A.M.), Institute of Clinical Medicine, Pathology and Forensic Medicine, and Translational Cancer Research Area, University of Eastern Finland; Department of Clinical Pathology (A.M.), Kuopio University Hospital; Unit of Clinical Medicine (S. Helisalmi), University of Eastern Finland, Kuopio, Finland; Department of Neurosurgery (P.K.E.), Oslo University Hospital-Rikshospitalet; Institute of Clinical Medicine (P.K.E.), Faculty of Medicine, and KG Jebsen Centre for Brain Fluid Research (P.K.E.), University of Oslo, Norway; Analytical and Translational Genetics Unit (A.P., M.I.K.), Department of Medicine, Massachusetts General Hospital, Boston; Program in Medical and Population Genetics (A.P., M.I.K.), and Stanley Center for Psychiatric Research (A.P., M.I.K.), Broad Institute for Harvard and MIT, Cambridge, MA
| | - Sami Heikkinen
- From the Department of Neurosurgery (J. Räsänen, K.M., V.E.K., M.O., J.E.J., V.L.), Kuopio University Hospital and Institute of Clinical Medicine-Neurosurgery, and Institute of Biomedicine (S. Heikkinen, K.M., A.L., T.K., M.H.), University of Eastern Finland, Kuopio; Institute for Molecular Medicine Finland (FIMM) (J.M., A.P.), Helsinki Institute of Life Science (HiLIFE), University of Helsinki; Department of Neurology (A.J.), Clinical Neurosciences, Helsinki University Hospital and University of Helsinki, Finland; Univ. Lille (B.G.-B., C.B., J.-C.L.), Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, France; Department of Neurosurgery (M.O., K.L., J.S.), University of Helsinki and Helsinki University Hospital; Clinical Neurosciences (C.A., J.F., A.K., J. Rinne), Department of Neurosurgery, University of Turku and Turku University Hospital; Department of Neurosurgery (A.R.), Tampere University Hospital; Unit of Clinical Neuroscience (M.K., M.v.u.z.F.), Neurosurgery, University of Oulu and Medical Research Center, Oulu University Hospital; Finnish Institute for Health and Welfare (THL) (M.P.); University of Helsinki (M.P.); Department of Neurosciences (A.M.K., A.M.P.), University of Helsinki; Department of Geriatrics (A.M.K.), Helsinki University Hospital; NeuroCenter (A.M.K.), Kuopio University Hospital; Institute of Clinical Medicine-Neurology (V.J., H.S.), University of Eastern Finland; School of Medicine (A.M.), Institute of Clinical Medicine, Pathology and Forensic Medicine, and Translational Cancer Research Area, University of Eastern Finland; Department of Clinical Pathology (A.M.), Kuopio University Hospital; Unit of Clinical Medicine (S. Helisalmi), University of Eastern Finland, Kuopio, Finland; Department of Neurosurgery (P.K.E.), Oslo University Hospital-Rikshospitalet; Institute of Clinical Medicine (P.K.E.), Faculty of Medicine, and KG Jebsen Centre for Brain Fluid Research (P.K.E.), University of Oslo, Norway; Analytical and Translational Genetics Unit (A.P., M.I.K.), Department of Medicine, Massachusetts General Hospital, Boston; Program in Medical and Population Genetics (A.P., M.I.K.), and Stanley Center for Psychiatric Research (A.P., M.I.K.), Broad Institute for Harvard and MIT, Cambridge, MA
| | - Kiira Mäklin
- From the Department of Neurosurgery (J. Räsänen, K.M., V.E.K., M.O., J.E.J., V.L.), Kuopio University Hospital and Institute of Clinical Medicine-Neurosurgery, and Institute of Biomedicine (S. Heikkinen, K.M., A.L., T.K., M.H.), University of Eastern Finland, Kuopio; Institute for Molecular Medicine Finland (FIMM) (J.M., A.P.), Helsinki Institute of Life Science (HiLIFE), University of Helsinki; Department of Neurology (A.J.), Clinical Neurosciences, Helsinki University Hospital and University of Helsinki, Finland; Univ. Lille (B.G.-B., C.B., J.-C.L.), Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, France; Department of Neurosurgery (M.O., K.L., J.S.), University of Helsinki and Helsinki University Hospital; Clinical Neurosciences (C.A., J.F., A.K., J. Rinne), Department of Neurosurgery, University of Turku and Turku University Hospital; Department of Neurosurgery (A.R.), Tampere University Hospital; Unit of Clinical Neuroscience (M.K., M.v.u.z.F.), Neurosurgery, University of Oulu and Medical Research Center, Oulu University Hospital; Finnish Institute for Health and Welfare (THL) (M.P.); University of Helsinki (M.P.); Department of Neurosciences (A.M.K., A.M.P.), University of Helsinki; Department of Geriatrics (A.M.K.), Helsinki University Hospital; NeuroCenter (A.M.K.), Kuopio University Hospital; Institute of Clinical Medicine-Neurology (V.J., H.S.), University of Eastern Finland; School of Medicine (A.M.), Institute of Clinical Medicine, Pathology and Forensic Medicine, and Translational Cancer Research Area, University of Eastern Finland; Department of Clinical Pathology (A.M.), Kuopio University Hospital; Unit of Clinical Medicine (S. Helisalmi), University of Eastern Finland, Kuopio, Finland; Department of Neurosurgery (P.K.E.), Oslo University Hospital-Rikshospitalet; Institute of Clinical Medicine (P.K.E.), Faculty of Medicine, and KG Jebsen Centre for Brain Fluid Research (P.K.E.), University of Oslo, Norway; Analytical and Translational Genetics Unit (A.P., M.I.K.), Department of Medicine, Massachusetts General Hospital, Boston; Program in Medical and Population Genetics (A.P., M.I.K.), and Stanley Center for Psychiatric Research (A.P., M.I.K.), Broad Institute for Harvard and MIT, Cambridge, MA
| | - Anssi Lipponen
- From the Department of Neurosurgery (J. Räsänen, K.M., V.E.K., M.O., J.E.J., V.L.), Kuopio University Hospital and Institute of Clinical Medicine-Neurosurgery, and Institute of Biomedicine (S. Heikkinen, K.M., A.L., T.K., M.H.), University of Eastern Finland, Kuopio; Institute for Molecular Medicine Finland (FIMM) (J.M., A.P.), Helsinki Institute of Life Science (HiLIFE), University of Helsinki; Department of Neurology (A.J.), Clinical Neurosciences, Helsinki University Hospital and University of Helsinki, Finland; Univ. Lille (B.G.-B., C.B., J.-C.L.), Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, France; Department of Neurosurgery (M.O., K.L., J.S.), University of Helsinki and Helsinki University Hospital; Clinical Neurosciences (C.A., J.F., A.K., J. Rinne), Department of Neurosurgery, University of Turku and Turku University Hospital; Department of Neurosurgery (A.R.), Tampere University Hospital; Unit of Clinical Neuroscience (M.K., M.v.u.z.F.), Neurosurgery, University of Oulu and Medical Research Center, Oulu University Hospital; Finnish Institute for Health and Welfare (THL) (M.P.); University of Helsinki (M.P.); Department of Neurosciences (A.M.K., A.M.P.), University of Helsinki; Department of Geriatrics (A.M.K.), Helsinki University Hospital; NeuroCenter (A.M.K.), Kuopio University Hospital; Institute of Clinical Medicine-Neurology (V.J., H.S.), University of Eastern Finland; School of Medicine (A.M.), Institute of Clinical Medicine, Pathology and Forensic Medicine, and Translational Cancer Research Area, University of Eastern Finland; Department of Clinical Pathology (A.M.), Kuopio University Hospital; Unit of Clinical Medicine (S. Helisalmi), University of Eastern Finland, Kuopio, Finland; Department of Neurosurgery (P.K.E.), Oslo University Hospital-Rikshospitalet; Institute of Clinical Medicine (P.K.E.), Faculty of Medicine, and KG Jebsen Centre for Brain Fluid Research (P.K.E.), University of Oslo, Norway; Analytical and Translational Genetics Unit (A.P., M.I.K.), Department of Medicine, Massachusetts General Hospital, Boston; Program in Medical and Population Genetics (A.P., M.I.K.), and Stanley Center for Psychiatric Research (A.P., M.I.K.), Broad Institute for Harvard and MIT, Cambridge, MA
| | - Teemu Kuulasmaa
- From the Department of Neurosurgery (J. Räsänen, K.M., V.E.K., M.O., J.E.J., V.L.), Kuopio University Hospital and Institute of Clinical Medicine-Neurosurgery, and Institute of Biomedicine (S. Heikkinen, K.M., A.L., T.K., M.H.), University of Eastern Finland, Kuopio; Institute for Molecular Medicine Finland (FIMM) (J.M., A.P.), Helsinki Institute of Life Science (HiLIFE), University of Helsinki; Department of Neurology (A.J.), Clinical Neurosciences, Helsinki University Hospital and University of Helsinki, Finland; Univ. Lille (B.G.-B., C.B., J.-C.L.), Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, France; Department of Neurosurgery (M.O., K.L., J.S.), University of Helsinki and Helsinki University Hospital; Clinical Neurosciences (C.A., J.F., A.K., J. Rinne), Department of Neurosurgery, University of Turku and Turku University Hospital; Department of Neurosurgery (A.R.), Tampere University Hospital; Unit of Clinical Neuroscience (M.K., M.v.u.z.F.), Neurosurgery, University of Oulu and Medical Research Center, Oulu University Hospital; Finnish Institute for Health and Welfare (THL) (M.P.); University of Helsinki (M.P.); Department of Neurosciences (A.M.K., A.M.P.), University of Helsinki; Department of Geriatrics (A.M.K.), Helsinki University Hospital; NeuroCenter (A.M.K.), Kuopio University Hospital; Institute of Clinical Medicine-Neurology (V.J., H.S.), University of Eastern Finland; School of Medicine (A.M.), Institute of Clinical Medicine, Pathology and Forensic Medicine, and Translational Cancer Research Area, University of Eastern Finland; Department of Clinical Pathology (A.M.), Kuopio University Hospital; Unit of Clinical Medicine (S. Helisalmi), University of Eastern Finland, Kuopio, Finland; Department of Neurosurgery (P.K.E.), Oslo University Hospital-Rikshospitalet; Institute of Clinical Medicine (P.K.E.), Faculty of Medicine, and KG Jebsen Centre for Brain Fluid Research (P.K.E.), University of Oslo, Norway; Analytical and Translational Genetics Unit (A.P., M.I.K.), Department of Medicine, Massachusetts General Hospital, Boston; Program in Medical and Population Genetics (A.P., M.I.K.), and Stanley Center for Psychiatric Research (A.P., M.I.K.), Broad Institute for Harvard and MIT, Cambridge, MA
| | - Juha Mehtonen
- From the Department of Neurosurgery (J. Räsänen, K.M., V.E.K., M.O., J.E.J., V.L.), Kuopio University Hospital and Institute of Clinical Medicine-Neurosurgery, and Institute of Biomedicine (S. Heikkinen, K.M., A.L., T.K., M.H.), University of Eastern Finland, Kuopio; Institute for Molecular Medicine Finland (FIMM) (J.M., A.P.), Helsinki Institute of Life Science (HiLIFE), University of Helsinki; Department of Neurology (A.J.), Clinical Neurosciences, Helsinki University Hospital and University of Helsinki, Finland; Univ. Lille (B.G.-B., C.B., J.-C.L.), Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, France; Department of Neurosurgery (M.O., K.L., J.S.), University of Helsinki and Helsinki University Hospital; Clinical Neurosciences (C.A., J.F., A.K., J. Rinne), Department of Neurosurgery, University of Turku and Turku University Hospital; Department of Neurosurgery (A.R.), Tampere University Hospital; Unit of Clinical Neuroscience (M.K., M.v.u.z.F.), Neurosurgery, University of Oulu and Medical Research Center, Oulu University Hospital; Finnish Institute for Health and Welfare (THL) (M.P.); University of Helsinki (M.P.); Department of Neurosciences (A.M.K., A.M.P.), University of Helsinki; Department of Geriatrics (A.M.K.), Helsinki University Hospital; NeuroCenter (A.M.K.), Kuopio University Hospital; Institute of Clinical Medicine-Neurology (V.J., H.S.), University of Eastern Finland; School of Medicine (A.M.), Institute of Clinical Medicine, Pathology and Forensic Medicine, and Translational Cancer Research Area, University of Eastern Finland; Department of Clinical Pathology (A.M.), Kuopio University Hospital; Unit of Clinical Medicine (S. Helisalmi), University of Eastern Finland, Kuopio, Finland; Department of Neurosurgery (P.K.E.), Oslo University Hospital-Rikshospitalet; Institute of Clinical Medicine (P.K.E.), Faculty of Medicine, and KG Jebsen Centre for Brain Fluid Research (P.K.E.), University of Oslo, Norway; Analytical and Translational Genetics Unit (A.P., M.I.K.), Department of Medicine, Massachusetts General Hospital, Boston; Program in Medical and Population Genetics (A.P., M.I.K.), and Stanley Center for Psychiatric Research (A.P., M.I.K.), Broad Institute for Harvard and MIT, Cambridge, MA
| | - Ville E Korhonen
- From the Department of Neurosurgery (J. Räsänen, K.M., V.E.K., M.O., J.E.J., V.L.), Kuopio University Hospital and Institute of Clinical Medicine-Neurosurgery, and Institute of Biomedicine (S. Heikkinen, K.M., A.L., T.K., M.H.), University of Eastern Finland, Kuopio; Institute for Molecular Medicine Finland (FIMM) (J.M., A.P.), Helsinki Institute of Life Science (HiLIFE), University of Helsinki; Department of Neurology (A.J.), Clinical Neurosciences, Helsinki University Hospital and University of Helsinki, Finland; Univ. Lille (B.G.-B., C.B., J.-C.L.), Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, France; Department of Neurosurgery (M.O., K.L., J.S.), University of Helsinki and Helsinki University Hospital; Clinical Neurosciences (C.A., J.F., A.K., J. Rinne), Department of Neurosurgery, University of Turku and Turku University Hospital; Department of Neurosurgery (A.R.), Tampere University Hospital; Unit of Clinical Neuroscience (M.K., M.v.u.z.F.), Neurosurgery, University of Oulu and Medical Research Center, Oulu University Hospital; Finnish Institute for Health and Welfare (THL) (M.P.); University of Helsinki (M.P.); Department of Neurosciences (A.M.K., A.M.P.), University of Helsinki; Department of Geriatrics (A.M.K.), Helsinki University Hospital; NeuroCenter (A.M.K.), Kuopio University Hospital; Institute of Clinical Medicine-Neurology (V.J., H.S.), University of Eastern Finland; School of Medicine (A.M.), Institute of Clinical Medicine, Pathology and Forensic Medicine, and Translational Cancer Research Area, University of Eastern Finland; Department of Clinical Pathology (A.M.), Kuopio University Hospital; Unit of Clinical Medicine (S. Helisalmi), University of Eastern Finland, Kuopio, Finland; Department of Neurosurgery (P.K.E.), Oslo University Hospital-Rikshospitalet; Institute of Clinical Medicine (P.K.E.), Faculty of Medicine, and KG Jebsen Centre for Brain Fluid Research (P.K.E.), University of Oslo, Norway; Analytical and Translational Genetics Unit (A.P., M.I.K.), Department of Medicine, Massachusetts General Hospital, Boston; Program in Medical and Population Genetics (A.P., M.I.K.), and Stanley Center for Psychiatric Research (A.P., M.I.K.), Broad Institute for Harvard and MIT, Cambridge, MA
| | - Antti Junkkari
- From the Department of Neurosurgery (J. Räsänen, K.M., V.E.K., M.O., J.E.J., V.L.), Kuopio University Hospital and Institute of Clinical Medicine-Neurosurgery, and Institute of Biomedicine (S. Heikkinen, K.M., A.L., T.K., M.H.), University of Eastern Finland, Kuopio; Institute for Molecular Medicine Finland (FIMM) (J.M., A.P.), Helsinki Institute of Life Science (HiLIFE), University of Helsinki; Department of Neurology (A.J.), Clinical Neurosciences, Helsinki University Hospital and University of Helsinki, Finland; Univ. Lille (B.G.-B., C.B., J.-C.L.), Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, France; Department of Neurosurgery (M.O., K.L., J.S.), University of Helsinki and Helsinki University Hospital; Clinical Neurosciences (C.A., J.F., A.K., J. Rinne), Department of Neurosurgery, University of Turku and Turku University Hospital; Department of Neurosurgery (A.R.), Tampere University Hospital; Unit of Clinical Neuroscience (M.K., M.v.u.z.F.), Neurosurgery, University of Oulu and Medical Research Center, Oulu University Hospital; Finnish Institute for Health and Welfare (THL) (M.P.); University of Helsinki (M.P.); Department of Neurosciences (A.M.K., A.M.P.), University of Helsinki; Department of Geriatrics (A.M.K.), Helsinki University Hospital; NeuroCenter (A.M.K.), Kuopio University Hospital; Institute of Clinical Medicine-Neurology (V.J., H.S.), University of Eastern Finland; School of Medicine (A.M.), Institute of Clinical Medicine, Pathology and Forensic Medicine, and Translational Cancer Research Area, University of Eastern Finland; Department of Clinical Pathology (A.M.), Kuopio University Hospital; Unit of Clinical Medicine (S. Helisalmi), University of Eastern Finland, Kuopio, Finland; Department of Neurosurgery (P.K.E.), Oslo University Hospital-Rikshospitalet; Institute of Clinical Medicine (P.K.E.), Faculty of Medicine, and KG Jebsen Centre for Brain Fluid Research (P.K.E.), University of Oslo, Norway; Analytical and Translational Genetics Unit (A.P., M.I.K.), Department of Medicine, Massachusetts General Hospital, Boston; Program in Medical and Population Genetics (A.P., M.I.K.), and Stanley Center for Psychiatric Research (A.P., M.I.K.), Broad Institute for Harvard and MIT, Cambridge, MA
| | - Benjamin Grenier-Boley
- From the Department of Neurosurgery (J. Räsänen, K.M., V.E.K., M.O., J.E.J., V.L.), Kuopio University Hospital and Institute of Clinical Medicine-Neurosurgery, and Institute of Biomedicine (S. Heikkinen, K.M., A.L., T.K., M.H.), University of Eastern Finland, Kuopio; Institute for Molecular Medicine Finland (FIMM) (J.M., A.P.), Helsinki Institute of Life Science (HiLIFE), University of Helsinki; Department of Neurology (A.J.), Clinical Neurosciences, Helsinki University Hospital and University of Helsinki, Finland; Univ. Lille (B.G.-B., C.B., J.-C.L.), Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, France; Department of Neurosurgery (M.O., K.L., J.S.), University of Helsinki and Helsinki University Hospital; Clinical Neurosciences (C.A., J.F., A.K., J. Rinne), Department of Neurosurgery, University of Turku and Turku University Hospital; Department of Neurosurgery (A.R.), Tampere University Hospital; Unit of Clinical Neuroscience (M.K., M.v.u.z.F.), Neurosurgery, University of Oulu and Medical Research Center, Oulu University Hospital; Finnish Institute for Health and Welfare (THL) (M.P.); University of Helsinki (M.P.); Department of Neurosciences (A.M.K., A.M.P.), University of Helsinki; Department of Geriatrics (A.M.K.), Helsinki University Hospital; NeuroCenter (A.M.K.), Kuopio University Hospital; Institute of Clinical Medicine-Neurology (V.J., H.S.), University of Eastern Finland; School of Medicine (A.M.), Institute of Clinical Medicine, Pathology and Forensic Medicine, and Translational Cancer Research Area, University of Eastern Finland; Department of Clinical Pathology (A.M.), Kuopio University Hospital; Unit of Clinical Medicine (S. Helisalmi), University of Eastern Finland, Kuopio, Finland; Department of Neurosurgery (P.K.E.), Oslo University Hospital-Rikshospitalet; Institute of Clinical Medicine (P.K.E.), Faculty of Medicine, and KG Jebsen Centre for Brain Fluid Research (P.K.E.), University of Oslo, Norway; Analytical and Translational Genetics Unit (A.P., M.I.K.), Department of Medicine, Massachusetts General Hospital, Boston; Program in Medical and Population Genetics (A.P., M.I.K.), and Stanley Center for Psychiatric Research (A.P., M.I.K.), Broad Institute for Harvard and MIT, Cambridge, MA
| | - Celine Bellenguez
- From the Department of Neurosurgery (J. Räsänen, K.M., V.E.K., M.O., J.E.J., V.L.), Kuopio University Hospital and Institute of Clinical Medicine-Neurosurgery, and Institute of Biomedicine (S. Heikkinen, K.M., A.L., T.K., M.H.), University of Eastern Finland, Kuopio; Institute for Molecular Medicine Finland (FIMM) (J.M., A.P.), Helsinki Institute of Life Science (HiLIFE), University of Helsinki; Department of Neurology (A.J.), Clinical Neurosciences, Helsinki University Hospital and University of Helsinki, Finland; Univ. Lille (B.G.-B., C.B., J.-C.L.), Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, France; Department of Neurosurgery (M.O., K.L., J.S.), University of Helsinki and Helsinki University Hospital; Clinical Neurosciences (C.A., J.F., A.K., J. Rinne), Department of Neurosurgery, University of Turku and Turku University Hospital; Department of Neurosurgery (A.R.), Tampere University Hospital; Unit of Clinical Neuroscience (M.K., M.v.u.z.F.), Neurosurgery, University of Oulu and Medical Research Center, Oulu University Hospital; Finnish Institute for Health and Welfare (THL) (M.P.); University of Helsinki (M.P.); Department of Neurosciences (A.M.K., A.M.P.), University of Helsinki; Department of Geriatrics (A.M.K.), Helsinki University Hospital; NeuroCenter (A.M.K.), Kuopio University Hospital; Institute of Clinical Medicine-Neurology (V.J., H.S.), University of Eastern Finland; School of Medicine (A.M.), Institute of Clinical Medicine, Pathology and Forensic Medicine, and Translational Cancer Research Area, University of Eastern Finland; Department of Clinical Pathology (A.M.), Kuopio University Hospital; Unit of Clinical Medicine (S. Helisalmi), University of Eastern Finland, Kuopio, Finland; Department of Neurosurgery (P.K.E.), Oslo University Hospital-Rikshospitalet; Institute of Clinical Medicine (P.K.E.), Faculty of Medicine, and KG Jebsen Centre for Brain Fluid Research (P.K.E.), University of Oslo, Norway; Analytical and Translational Genetics Unit (A.P., M.I.K.), Department of Medicine, Massachusetts General Hospital, Boston; Program in Medical and Population Genetics (A.P., M.I.K.), and Stanley Center for Psychiatric Research (A.P., M.I.K.), Broad Institute for Harvard and MIT, Cambridge, MA
| | - Minna Oinas
- From the Department of Neurosurgery (J. Räsänen, K.M., V.E.K., M.O., J.E.J., V.L.), Kuopio University Hospital and Institute of Clinical Medicine-Neurosurgery, and Institute of Biomedicine (S. Heikkinen, K.M., A.L., T.K., M.H.), University of Eastern Finland, Kuopio; Institute for Molecular Medicine Finland (FIMM) (J.M., A.P.), Helsinki Institute of Life Science (HiLIFE), University of Helsinki; Department of Neurology (A.J.), Clinical Neurosciences, Helsinki University Hospital and University of Helsinki, Finland; Univ. Lille (B.G.-B., C.B., J.-C.L.), Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, France; Department of Neurosurgery (M.O., K.L., J.S.), University of Helsinki and Helsinki University Hospital; Clinical Neurosciences (C.A., J.F., A.K., J. Rinne), Department of Neurosurgery, University of Turku and Turku University Hospital; Department of Neurosurgery (A.R.), Tampere University Hospital; Unit of Clinical Neuroscience (M.K., M.v.u.z.F.), Neurosurgery, University of Oulu and Medical Research Center, Oulu University Hospital; Finnish Institute for Health and Welfare (THL) (M.P.); University of Helsinki (M.P.); Department of Neurosciences (A.M.K., A.M.P.), University of Helsinki; Department of Geriatrics (A.M.K.), Helsinki University Hospital; NeuroCenter (A.M.K.), Kuopio University Hospital; Institute of Clinical Medicine-Neurology (V.J., H.S.), University of Eastern Finland; School of Medicine (A.M.), Institute of Clinical Medicine, Pathology and Forensic Medicine, and Translational Cancer Research Area, University of Eastern Finland; Department of Clinical Pathology (A.M.), Kuopio University Hospital; Unit of Clinical Medicine (S. Helisalmi), University of Eastern Finland, Kuopio, Finland; Department of Neurosurgery (P.K.E.), Oslo University Hospital-Rikshospitalet; Institute of Clinical Medicine (P.K.E.), Faculty of Medicine, and KG Jebsen Centre for Brain Fluid Research (P.K.E.), University of Oslo, Norway; Analytical and Translational Genetics Unit (A.P., M.I.K.), Department of Medicine, Massachusetts General Hospital, Boston; Program in Medical and Population Genetics (A.P., M.I.K.), and Stanley Center for Psychiatric Research (A.P., M.I.K.), Broad Institute for Harvard and MIT, Cambridge, MA
| | - Cecilia Avellan
- From the Department of Neurosurgery (J. Räsänen, K.M., V.E.K., M.O., J.E.J., V.L.), Kuopio University Hospital and Institute of Clinical Medicine-Neurosurgery, and Institute of Biomedicine (S. Heikkinen, K.M., A.L., T.K., M.H.), University of Eastern Finland, Kuopio; Institute for Molecular Medicine Finland (FIMM) (J.M., A.P.), Helsinki Institute of Life Science (HiLIFE), University of Helsinki; Department of Neurology (A.J.), Clinical Neurosciences, Helsinki University Hospital and University of Helsinki, Finland; Univ. Lille (B.G.-B., C.B., J.-C.L.), Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, France; Department of Neurosurgery (M.O., K.L., J.S.), University of Helsinki and Helsinki University Hospital; Clinical Neurosciences (C.A., J.F., A.K., J. Rinne), Department of Neurosurgery, University of Turku and Turku University Hospital; Department of Neurosurgery (A.R.), Tampere University Hospital; Unit of Clinical Neuroscience (M.K., M.v.u.z.F.), Neurosurgery, University of Oulu and Medical Research Center, Oulu University Hospital; Finnish Institute for Health and Welfare (THL) (M.P.); University of Helsinki (M.P.); Department of Neurosciences (A.M.K., A.M.P.), University of Helsinki; Department of Geriatrics (A.M.K.), Helsinki University Hospital; NeuroCenter (A.M.K.), Kuopio University Hospital; Institute of Clinical Medicine-Neurology (V.J., H.S.), University of Eastern Finland; School of Medicine (A.M.), Institute of Clinical Medicine, Pathology and Forensic Medicine, and Translational Cancer Research Area, University of Eastern Finland; Department of Clinical Pathology (A.M.), Kuopio University Hospital; Unit of Clinical Medicine (S. Helisalmi), University of Eastern Finland, Kuopio, Finland; Department of Neurosurgery (P.K.E.), Oslo University Hospital-Rikshospitalet; Institute of Clinical Medicine (P.K.E.), Faculty of Medicine, and KG Jebsen Centre for Brain Fluid Research (P.K.E.), University of Oslo, Norway; Analytical and Translational Genetics Unit (A.P., M.I.K.), Department of Medicine, Massachusetts General Hospital, Boston; Program in Medical and Population Genetics (A.P., M.I.K.), and Stanley Center for Psychiatric Research (A.P., M.I.K.), Broad Institute for Harvard and MIT, Cambridge, MA
| | - Janek Frantzén
- From the Department of Neurosurgery (J. Räsänen, K.M., V.E.K., M.O., J.E.J., V.L.), Kuopio University Hospital and Institute of Clinical Medicine-Neurosurgery, and Institute of Biomedicine (S. Heikkinen, K.M., A.L., T.K., M.H.), University of Eastern Finland, Kuopio; Institute for Molecular Medicine Finland (FIMM) (J.M., A.P.), Helsinki Institute of Life Science (HiLIFE), University of Helsinki; Department of Neurology (A.J.), Clinical Neurosciences, Helsinki University Hospital and University of Helsinki, Finland; Univ. Lille (B.G.-B., C.B., J.-C.L.), Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, France; Department of Neurosurgery (M.O., K.L., J.S.), University of Helsinki and Helsinki University Hospital; Clinical Neurosciences (C.A., J.F., A.K., J. Rinne), Department of Neurosurgery, University of Turku and Turku University Hospital; Department of Neurosurgery (A.R.), Tampere University Hospital; Unit of Clinical Neuroscience (M.K., M.v.u.z.F.), Neurosurgery, University of Oulu and Medical Research Center, Oulu University Hospital; Finnish Institute for Health and Welfare (THL) (M.P.); University of Helsinki (M.P.); Department of Neurosciences (A.M.K., A.M.P.), University of Helsinki; Department of Geriatrics (A.M.K.), Helsinki University Hospital; NeuroCenter (A.M.K.), Kuopio University Hospital; Institute of Clinical Medicine-Neurology (V.J., H.S.), University of Eastern Finland; School of Medicine (A.M.), Institute of Clinical Medicine, Pathology and Forensic Medicine, and Translational Cancer Research Area, University of Eastern Finland; Department of Clinical Pathology (A.M.), Kuopio University Hospital; Unit of Clinical Medicine (S. Helisalmi), University of Eastern Finland, Kuopio, Finland; Department of Neurosurgery (P.K.E.), Oslo University Hospital-Rikshospitalet; Institute of Clinical Medicine (P.K.E.), Faculty of Medicine, and KG Jebsen Centre for Brain Fluid Research (P.K.E.), University of Oslo, Norway; Analytical and Translational Genetics Unit (A.P., M.I.K.), Department of Medicine, Massachusetts General Hospital, Boston; Program in Medical and Population Genetics (A.P., M.I.K.), and Stanley Center for Psychiatric Research (A.P., M.I.K.), Broad Institute for Harvard and MIT, Cambridge, MA
| | - Anna Kotkansalo
- From the Department of Neurosurgery (J. Räsänen, K.M., V.E.K., M.O., J.E.J., V.L.), Kuopio University Hospital and Institute of Clinical Medicine-Neurosurgery, and Institute of Biomedicine (S. Heikkinen, K.M., A.L., T.K., M.H.), University of Eastern Finland, Kuopio; Institute for Molecular Medicine Finland (FIMM) (J.M., A.P.), Helsinki Institute of Life Science (HiLIFE), University of Helsinki; Department of Neurology (A.J.), Clinical Neurosciences, Helsinki University Hospital and University of Helsinki, Finland; Univ. Lille (B.G.-B., C.B., J.-C.L.), Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, France; Department of Neurosurgery (M.O., K.L., J.S.), University of Helsinki and Helsinki University Hospital; Clinical Neurosciences (C.A., J.F., A.K., J. Rinne), Department of Neurosurgery, University of Turku and Turku University Hospital; Department of Neurosurgery (A.R.), Tampere University Hospital; Unit of Clinical Neuroscience (M.K., M.v.u.z.F.), Neurosurgery, University of Oulu and Medical Research Center, Oulu University Hospital; Finnish Institute for Health and Welfare (THL) (M.P.); University of Helsinki (M.P.); Department of Neurosciences (A.M.K., A.M.P.), University of Helsinki; Department of Geriatrics (A.M.K.), Helsinki University Hospital; NeuroCenter (A.M.K.), Kuopio University Hospital; Institute of Clinical Medicine-Neurology (V.J., H.S.), University of Eastern Finland; School of Medicine (A.M.), Institute of Clinical Medicine, Pathology and Forensic Medicine, and Translational Cancer Research Area, University of Eastern Finland; Department of Clinical Pathology (A.M.), Kuopio University Hospital; Unit of Clinical Medicine (S. Helisalmi), University of Eastern Finland, Kuopio, Finland; Department of Neurosurgery (P.K.E.), Oslo University Hospital-Rikshospitalet; Institute of Clinical Medicine (P.K.E.), Faculty of Medicine, and KG Jebsen Centre for Brain Fluid Research (P.K.E.), University of Oslo, Norway; Analytical and Translational Genetics Unit (A.P., M.I.K.), Department of Medicine, Massachusetts General Hospital, Boston; Program in Medical and Population Genetics (A.P., M.I.K.), and Stanley Center for Psychiatric Research (A.P., M.I.K.), Broad Institute for Harvard and MIT, Cambridge, MA
| | - Jaakko Rinne
- From the Department of Neurosurgery (J. Räsänen, K.M., V.E.K., M.O., J.E.J., V.L.), Kuopio University Hospital and Institute of Clinical Medicine-Neurosurgery, and Institute of Biomedicine (S. Heikkinen, K.M., A.L., T.K., M.H.), University of Eastern Finland, Kuopio; Institute for Molecular Medicine Finland (FIMM) (J.M., A.P.), Helsinki Institute of Life Science (HiLIFE), University of Helsinki; Department of Neurology (A.J.), Clinical Neurosciences, Helsinki University Hospital and University of Helsinki, Finland; Univ. Lille (B.G.-B., C.B., J.-C.L.), Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, France; Department of Neurosurgery (M.O., K.L., J.S.), University of Helsinki and Helsinki University Hospital; Clinical Neurosciences (C.A., J.F., A.K., J. Rinne), Department of Neurosurgery, University of Turku and Turku University Hospital; Department of Neurosurgery (A.R.), Tampere University Hospital; Unit of Clinical Neuroscience (M.K., M.v.u.z.F.), Neurosurgery, University of Oulu and Medical Research Center, Oulu University Hospital; Finnish Institute for Health and Welfare (THL) (M.P.); University of Helsinki (M.P.); Department of Neurosciences (A.M.K., A.M.P.), University of Helsinki; Department of Geriatrics (A.M.K.), Helsinki University Hospital; NeuroCenter (A.M.K.), Kuopio University Hospital; Institute of Clinical Medicine-Neurology (V.J., H.S.), University of Eastern Finland; School of Medicine (A.M.), Institute of Clinical Medicine, Pathology and Forensic Medicine, and Translational Cancer Research Area, University of Eastern Finland; Department of Clinical Pathology (A.M.), Kuopio University Hospital; Unit of Clinical Medicine (S. Helisalmi), University of Eastern Finland, Kuopio, Finland; Department of Neurosurgery (P.K.E.), Oslo University Hospital-Rikshospitalet; Institute of Clinical Medicine (P.K.E.), Faculty of Medicine, and KG Jebsen Centre for Brain Fluid Research (P.K.E.), University of Oslo, Norway; Analytical and Translational Genetics Unit (A.P., M.I.K.), Department of Medicine, Massachusetts General Hospital, Boston; Program in Medical and Population Genetics (A.P., M.I.K.), and Stanley Center for Psychiatric Research (A.P., M.I.K.), Broad Institute for Harvard and MIT, Cambridge, MA
| | - Antti Ronkainen
- From the Department of Neurosurgery (J. Räsänen, K.M., V.E.K., M.O., J.E.J., V.L.), Kuopio University Hospital and Institute of Clinical Medicine-Neurosurgery, and Institute of Biomedicine (S. Heikkinen, K.M., A.L., T.K., M.H.), University of Eastern Finland, Kuopio; Institute for Molecular Medicine Finland (FIMM) (J.M., A.P.), Helsinki Institute of Life Science (HiLIFE), University of Helsinki; Department of Neurology (A.J.), Clinical Neurosciences, Helsinki University Hospital and University of Helsinki, Finland; Univ. Lille (B.G.-B., C.B., J.-C.L.), Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, France; Department of Neurosurgery (M.O., K.L., J.S.), University of Helsinki and Helsinki University Hospital; Clinical Neurosciences (C.A., J.F., A.K., J. Rinne), Department of Neurosurgery, University of Turku and Turku University Hospital; Department of Neurosurgery (A.R.), Tampere University Hospital; Unit of Clinical Neuroscience (M.K., M.v.u.z.F.), Neurosurgery, University of Oulu and Medical Research Center, Oulu University Hospital; Finnish Institute for Health and Welfare (THL) (M.P.); University of Helsinki (M.P.); Department of Neurosciences (A.M.K., A.M.P.), University of Helsinki; Department of Geriatrics (A.M.K.), Helsinki University Hospital; NeuroCenter (A.M.K.), Kuopio University Hospital; Institute of Clinical Medicine-Neurology (V.J., H.S.), University of Eastern Finland; School of Medicine (A.M.), Institute of Clinical Medicine, Pathology and Forensic Medicine, and Translational Cancer Research Area, University of Eastern Finland; Department of Clinical Pathology (A.M.), Kuopio University Hospital; Unit of Clinical Medicine (S. Helisalmi), University of Eastern Finland, Kuopio, Finland; Department of Neurosurgery (P.K.E.), Oslo University Hospital-Rikshospitalet; Institute of Clinical Medicine (P.K.E.), Faculty of Medicine, and KG Jebsen Centre for Brain Fluid Research (P.K.E.), University of Oslo, Norway; Analytical and Translational Genetics Unit (A.P., M.I.K.), Department of Medicine, Massachusetts General Hospital, Boston; Program in Medical and Population Genetics (A.P., M.I.K.), and Stanley Center for Psychiatric Research (A.P., M.I.K.), Broad Institute for Harvard and MIT, Cambridge, MA
| | - Mikko Kauppinen
- From the Department of Neurosurgery (J. Räsänen, K.M., V.E.K., M.O., J.E.J., V.L.), Kuopio University Hospital and Institute of Clinical Medicine-Neurosurgery, and Institute of Biomedicine (S. Heikkinen, K.M., A.L., T.K., M.H.), University of Eastern Finland, Kuopio; Institute for Molecular Medicine Finland (FIMM) (J.M., A.P.), Helsinki Institute of Life Science (HiLIFE), University of Helsinki; Department of Neurology (A.J.), Clinical Neurosciences, Helsinki University Hospital and University of Helsinki, Finland; Univ. Lille (B.G.-B., C.B., J.-C.L.), Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, France; Department of Neurosurgery (M.O., K.L., J.S.), University of Helsinki and Helsinki University Hospital; Clinical Neurosciences (C.A., J.F., A.K., J. Rinne), Department of Neurosurgery, University of Turku and Turku University Hospital; Department of Neurosurgery (A.R.), Tampere University Hospital; Unit of Clinical Neuroscience (M.K., M.v.u.z.F.), Neurosurgery, University of Oulu and Medical Research Center, Oulu University Hospital; Finnish Institute for Health and Welfare (THL) (M.P.); University of Helsinki (M.P.); Department of Neurosciences (A.M.K., A.M.P.), University of Helsinki; Department of Geriatrics (A.M.K.), Helsinki University Hospital; NeuroCenter (A.M.K.), Kuopio University Hospital; Institute of Clinical Medicine-Neurology (V.J., H.S.), University of Eastern Finland; School of Medicine (A.M.), Institute of Clinical Medicine, Pathology and Forensic Medicine, and Translational Cancer Research Area, University of Eastern Finland; Department of Clinical Pathology (A.M.), Kuopio University Hospital; Unit of Clinical Medicine (S. Helisalmi), University of Eastern Finland, Kuopio, Finland; Department of Neurosurgery (P.K.E.), Oslo University Hospital-Rikshospitalet; Institute of Clinical Medicine (P.K.E.), Faculty of Medicine, and KG Jebsen Centre for Brain Fluid Research (P.K.E.), University of Oslo, Norway; Analytical and Translational Genetics Unit (A.P., M.I.K.), Department of Medicine, Massachusetts General Hospital, Boston; Program in Medical and Population Genetics (A.P., M.I.K.), and Stanley Center for Psychiatric Research (A.P., M.I.K.), Broad Institute for Harvard and MIT, Cambridge, MA
| | - Mikael von Und Zu Fraunberg
- From the Department of Neurosurgery (J. Räsänen, K.M., V.E.K., M.O., J.E.J., V.L.), Kuopio University Hospital and Institute of Clinical Medicine-Neurosurgery, and Institute of Biomedicine (S. Heikkinen, K.M., A.L., T.K., M.H.), University of Eastern Finland, Kuopio; Institute for Molecular Medicine Finland (FIMM) (J.M., A.P.), Helsinki Institute of Life Science (HiLIFE), University of Helsinki; Department of Neurology (A.J.), Clinical Neurosciences, Helsinki University Hospital and University of Helsinki, Finland; Univ. Lille (B.G.-B., C.B., J.-C.L.), Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, France; Department of Neurosurgery (M.O., K.L., J.S.), University of Helsinki and Helsinki University Hospital; Clinical Neurosciences (C.A., J.F., A.K., J. Rinne), Department of Neurosurgery, University of Turku and Turku University Hospital; Department of Neurosurgery (A.R.), Tampere University Hospital; Unit of Clinical Neuroscience (M.K., M.v.u.z.F.), Neurosurgery, University of Oulu and Medical Research Center, Oulu University Hospital; Finnish Institute for Health and Welfare (THL) (M.P.); University of Helsinki (M.P.); Department of Neurosciences (A.M.K., A.M.P.), University of Helsinki; Department of Geriatrics (A.M.K.), Helsinki University Hospital; NeuroCenter (A.M.K.), Kuopio University Hospital; Institute of Clinical Medicine-Neurology (V.J., H.S.), University of Eastern Finland; School of Medicine (A.M.), Institute of Clinical Medicine, Pathology and Forensic Medicine, and Translational Cancer Research Area, University of Eastern Finland; Department of Clinical Pathology (A.M.), Kuopio University Hospital; Unit of Clinical Medicine (S. Helisalmi), University of Eastern Finland, Kuopio, Finland; Department of Neurosurgery (P.K.E.), Oslo University Hospital-Rikshospitalet; Institute of Clinical Medicine (P.K.E.), Faculty of Medicine, and KG Jebsen Centre for Brain Fluid Research (P.K.E.), University of Oslo, Norway; Analytical and Translational Genetics Unit (A.P., M.I.K.), Department of Medicine, Massachusetts General Hospital, Boston; Program in Medical and Population Genetics (A.P., M.I.K.), and Stanley Center for Psychiatric Research (A.P., M.I.K.), Broad Institute for Harvard and MIT, Cambridge, MA
| | - Kimmo Lönnrot
- From the Department of Neurosurgery (J. Räsänen, K.M., V.E.K., M.O., J.E.J., V.L.), Kuopio University Hospital and Institute of Clinical Medicine-Neurosurgery, and Institute of Biomedicine (S. Heikkinen, K.M., A.L., T.K., M.H.), University of Eastern Finland, Kuopio; Institute for Molecular Medicine Finland (FIMM) (J.M., A.P.), Helsinki Institute of Life Science (HiLIFE), University of Helsinki; Department of Neurology (A.J.), Clinical Neurosciences, Helsinki University Hospital and University of Helsinki, Finland; Univ. Lille (B.G.-B., C.B., J.-C.L.), Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, France; Department of Neurosurgery (M.O., K.L., J.S.), University of Helsinki and Helsinki University Hospital; Clinical Neurosciences (C.A., J.F., A.K., J. Rinne), Department of Neurosurgery, University of Turku and Turku University Hospital; Department of Neurosurgery (A.R.), Tampere University Hospital; Unit of Clinical Neuroscience (M.K., M.v.u.z.F.), Neurosurgery, University of Oulu and Medical Research Center, Oulu University Hospital; Finnish Institute for Health and Welfare (THL) (M.P.); University of Helsinki (M.P.); Department of Neurosciences (A.M.K., A.M.P.), University of Helsinki; Department of Geriatrics (A.M.K.), Helsinki University Hospital; NeuroCenter (A.M.K.), Kuopio University Hospital; Institute of Clinical Medicine-Neurology (V.J., H.S.), University of Eastern Finland; School of Medicine (A.M.), Institute of Clinical Medicine, Pathology and Forensic Medicine, and Translational Cancer Research Area, University of Eastern Finland; Department of Clinical Pathology (A.M.), Kuopio University Hospital; Unit of Clinical Medicine (S. Helisalmi), University of Eastern Finland, Kuopio, Finland; Department of Neurosurgery (P.K.E.), Oslo University Hospital-Rikshospitalet; Institute of Clinical Medicine (P.K.E.), Faculty of Medicine, and KG Jebsen Centre for Brain Fluid Research (P.K.E.), University of Oslo, Norway; Analytical and Translational Genetics Unit (A.P., M.I.K.), Department of Medicine, Massachusetts General Hospital, Boston; Program in Medical and Population Genetics (A.P., M.I.K.), and Stanley Center for Psychiatric Research (A.P., M.I.K.), Broad Institute for Harvard and MIT, Cambridge, MA
| | - Jarno Satopää
- From the Department of Neurosurgery (J. Räsänen, K.M., V.E.K., M.O., J.E.J., V.L.), Kuopio University Hospital and Institute of Clinical Medicine-Neurosurgery, and Institute of Biomedicine (S. Heikkinen, K.M., A.L., T.K., M.H.), University of Eastern Finland, Kuopio; Institute for Molecular Medicine Finland (FIMM) (J.M., A.P.), Helsinki Institute of Life Science (HiLIFE), University of Helsinki; Department of Neurology (A.J.), Clinical Neurosciences, Helsinki University Hospital and University of Helsinki, Finland; Univ. Lille (B.G.-B., C.B., J.-C.L.), Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, France; Department of Neurosurgery (M.O., K.L., J.S.), University of Helsinki and Helsinki University Hospital; Clinical Neurosciences (C.A., J.F., A.K., J. Rinne), Department of Neurosurgery, University of Turku and Turku University Hospital; Department of Neurosurgery (A.R.), Tampere University Hospital; Unit of Clinical Neuroscience (M.K., M.v.u.z.F.), Neurosurgery, University of Oulu and Medical Research Center, Oulu University Hospital; Finnish Institute for Health and Welfare (THL) (M.P.); University of Helsinki (M.P.); Department of Neurosciences (A.M.K., A.M.P.), University of Helsinki; Department of Geriatrics (A.M.K.), Helsinki University Hospital; NeuroCenter (A.M.K.), Kuopio University Hospital; Institute of Clinical Medicine-Neurology (V.J., H.S.), University of Eastern Finland; School of Medicine (A.M.), Institute of Clinical Medicine, Pathology and Forensic Medicine, and Translational Cancer Research Area, University of Eastern Finland; Department of Clinical Pathology (A.M.), Kuopio University Hospital; Unit of Clinical Medicine (S. Helisalmi), University of Eastern Finland, Kuopio, Finland; Department of Neurosurgery (P.K.E.), Oslo University Hospital-Rikshospitalet; Institute of Clinical Medicine (P.K.E.), Faculty of Medicine, and KG Jebsen Centre for Brain Fluid Research (P.K.E.), University of Oslo, Norway; Analytical and Translational Genetics Unit (A.P., M.I.K.), Department of Medicine, Massachusetts General Hospital, Boston; Program in Medical and Population Genetics (A.P., M.I.K.), and Stanley Center for Psychiatric Research (A.P., M.I.K.), Broad Institute for Harvard and MIT, Cambridge, MA
| | - Markus Perola
- From the Department of Neurosurgery (J. Räsänen, K.M., V.E.K., M.O., J.E.J., V.L.), Kuopio University Hospital and Institute of Clinical Medicine-Neurosurgery, and Institute of Biomedicine (S. Heikkinen, K.M., A.L., T.K., M.H.), University of Eastern Finland, Kuopio; Institute for Molecular Medicine Finland (FIMM) (J.M., A.P.), Helsinki Institute of Life Science (HiLIFE), University of Helsinki; Department of Neurology (A.J.), Clinical Neurosciences, Helsinki University Hospital and University of Helsinki, Finland; Univ. Lille (B.G.-B., C.B., J.-C.L.), Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, France; Department of Neurosurgery (M.O., K.L., J.S.), University of Helsinki and Helsinki University Hospital; Clinical Neurosciences (C.A., J.F., A.K., J. Rinne), Department of Neurosurgery, University of Turku and Turku University Hospital; Department of Neurosurgery (A.R.), Tampere University Hospital; Unit of Clinical Neuroscience (M.K., M.v.u.z.F.), Neurosurgery, University of Oulu and Medical Research Center, Oulu University Hospital; Finnish Institute for Health and Welfare (THL) (M.P.); University of Helsinki (M.P.); Department of Neurosciences (A.M.K., A.M.P.), University of Helsinki; Department of Geriatrics (A.M.K.), Helsinki University Hospital; NeuroCenter (A.M.K.), Kuopio University Hospital; Institute of Clinical Medicine-Neurology (V.J., H.S.), University of Eastern Finland; School of Medicine (A.M.), Institute of Clinical Medicine, Pathology and Forensic Medicine, and Translational Cancer Research Area, University of Eastern Finland; Department of Clinical Pathology (A.M.), Kuopio University Hospital; Unit of Clinical Medicine (S. Helisalmi), University of Eastern Finland, Kuopio, Finland; Department of Neurosurgery (P.K.E.), Oslo University Hospital-Rikshospitalet; Institute of Clinical Medicine (P.K.E.), Faculty of Medicine, and KG Jebsen Centre for Brain Fluid Research (P.K.E.), University of Oslo, Norway; Analytical and Translational Genetics Unit (A.P., M.I.K.), Department of Medicine, Massachusetts General Hospital, Boston; Program in Medical and Population Genetics (A.P., M.I.K.), and Stanley Center for Psychiatric Research (A.P., M.I.K.), Broad Institute for Harvard and MIT, Cambridge, MA
| | - Anne M Koivisto
- From the Department of Neurosurgery (J. Räsänen, K.M., V.E.K., M.O., J.E.J., V.L.), Kuopio University Hospital and Institute of Clinical Medicine-Neurosurgery, and Institute of Biomedicine (S. Heikkinen, K.M., A.L., T.K., M.H.), University of Eastern Finland, Kuopio; Institute for Molecular Medicine Finland (FIMM) (J.M., A.P.), Helsinki Institute of Life Science (HiLIFE), University of Helsinki; Department of Neurology (A.J.), Clinical Neurosciences, Helsinki University Hospital and University of Helsinki, Finland; Univ. Lille (B.G.-B., C.B., J.-C.L.), Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, France; Department of Neurosurgery (M.O., K.L., J.S.), University of Helsinki and Helsinki University Hospital; Clinical Neurosciences (C.A., J.F., A.K., J. Rinne), Department of Neurosurgery, University of Turku and Turku University Hospital; Department of Neurosurgery (A.R.), Tampere University Hospital; Unit of Clinical Neuroscience (M.K., M.v.u.z.F.), Neurosurgery, University of Oulu and Medical Research Center, Oulu University Hospital; Finnish Institute for Health and Welfare (THL) (M.P.); University of Helsinki (M.P.); Department of Neurosciences (A.M.K., A.M.P.), University of Helsinki; Department of Geriatrics (A.M.K.), Helsinki University Hospital; NeuroCenter (A.M.K.), Kuopio University Hospital; Institute of Clinical Medicine-Neurology (V.J., H.S.), University of Eastern Finland; School of Medicine (A.M.), Institute of Clinical Medicine, Pathology and Forensic Medicine, and Translational Cancer Research Area, University of Eastern Finland; Department of Clinical Pathology (A.M.), Kuopio University Hospital; Unit of Clinical Medicine (S. Helisalmi), University of Eastern Finland, Kuopio, Finland; Department of Neurosurgery (P.K.E.), Oslo University Hospital-Rikshospitalet; Institute of Clinical Medicine (P.K.E.), Faculty of Medicine, and KG Jebsen Centre for Brain Fluid Research (P.K.E.), University of Oslo, Norway; Analytical and Translational Genetics Unit (A.P., M.I.K.), Department of Medicine, Massachusetts General Hospital, Boston; Program in Medical and Population Genetics (A.P., M.I.K.), and Stanley Center for Psychiatric Research (A.P., M.I.K.), Broad Institute for Harvard and MIT, Cambridge, MA
| | - Valtteri Julkunen
- From the Department of Neurosurgery (J. Räsänen, K.M., V.E.K., M.O., J.E.J., V.L.), Kuopio University Hospital and Institute of Clinical Medicine-Neurosurgery, and Institute of Biomedicine (S. Heikkinen, K.M., A.L., T.K., M.H.), University of Eastern Finland, Kuopio; Institute for Molecular Medicine Finland (FIMM) (J.M., A.P.), Helsinki Institute of Life Science (HiLIFE), University of Helsinki; Department of Neurology (A.J.), Clinical Neurosciences, Helsinki University Hospital and University of Helsinki, Finland; Univ. Lille (B.G.-B., C.B., J.-C.L.), Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, France; Department of Neurosurgery (M.O., K.L., J.S.), University of Helsinki and Helsinki University Hospital; Clinical Neurosciences (C.A., J.F., A.K., J. Rinne), Department of Neurosurgery, University of Turku and Turku University Hospital; Department of Neurosurgery (A.R.), Tampere University Hospital; Unit of Clinical Neuroscience (M.K., M.v.u.z.F.), Neurosurgery, University of Oulu and Medical Research Center, Oulu University Hospital; Finnish Institute for Health and Welfare (THL) (M.P.); University of Helsinki (M.P.); Department of Neurosciences (A.M.K., A.M.P.), University of Helsinki; Department of Geriatrics (A.M.K.), Helsinki University Hospital; NeuroCenter (A.M.K.), Kuopio University Hospital; Institute of Clinical Medicine-Neurology (V.J., H.S.), University of Eastern Finland; School of Medicine (A.M.), Institute of Clinical Medicine, Pathology and Forensic Medicine, and Translational Cancer Research Area, University of Eastern Finland; Department of Clinical Pathology (A.M.), Kuopio University Hospital; Unit of Clinical Medicine (S. Helisalmi), University of Eastern Finland, Kuopio, Finland; Department of Neurosurgery (P.K.E.), Oslo University Hospital-Rikshospitalet; Institute of Clinical Medicine (P.K.E.), Faculty of Medicine, and KG Jebsen Centre for Brain Fluid Research (P.K.E.), University of Oslo, Norway; Analytical and Translational Genetics Unit (A.P., M.I.K.), Department of Medicine, Massachusetts General Hospital, Boston; Program in Medical and Population Genetics (A.P., M.I.K.), and Stanley Center for Psychiatric Research (A.P., M.I.K.), Broad Institute for Harvard and MIT, Cambridge, MA
| | - Anne M Portaankorva
- From the Department of Neurosurgery (J. Räsänen, K.M., V.E.K., M.O., J.E.J., V.L.), Kuopio University Hospital and Institute of Clinical Medicine-Neurosurgery, and Institute of Biomedicine (S. Heikkinen, K.M., A.L., T.K., M.H.), University of Eastern Finland, Kuopio; Institute for Molecular Medicine Finland (FIMM) (J.M., A.P.), Helsinki Institute of Life Science (HiLIFE), University of Helsinki; Department of Neurology (A.J.), Clinical Neurosciences, Helsinki University Hospital and University of Helsinki, Finland; Univ. Lille (B.G.-B., C.B., J.-C.L.), Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, France; Department of Neurosurgery (M.O., K.L., J.S.), University of Helsinki and Helsinki University Hospital; Clinical Neurosciences (C.A., J.F., A.K., J. Rinne), Department of Neurosurgery, University of Turku and Turku University Hospital; Department of Neurosurgery (A.R.), Tampere University Hospital; Unit of Clinical Neuroscience (M.K., M.v.u.z.F.), Neurosurgery, University of Oulu and Medical Research Center, Oulu University Hospital; Finnish Institute for Health and Welfare (THL) (M.P.); University of Helsinki (M.P.); Department of Neurosciences (A.M.K., A.M.P.), University of Helsinki; Department of Geriatrics (A.M.K.), Helsinki University Hospital; NeuroCenter (A.M.K.), Kuopio University Hospital; Institute of Clinical Medicine-Neurology (V.J., H.S.), University of Eastern Finland; School of Medicine (A.M.), Institute of Clinical Medicine, Pathology and Forensic Medicine, and Translational Cancer Research Area, University of Eastern Finland; Department of Clinical Pathology (A.M.), Kuopio University Hospital; Unit of Clinical Medicine (S. Helisalmi), University of Eastern Finland, Kuopio, Finland; Department of Neurosurgery (P.K.E.), Oslo University Hospital-Rikshospitalet; Institute of Clinical Medicine (P.K.E.), Faculty of Medicine, and KG Jebsen Centre for Brain Fluid Research (P.K.E.), University of Oslo, Norway; Analytical and Translational Genetics Unit (A.P., M.I.K.), Department of Medicine, Massachusetts General Hospital, Boston; Program in Medical and Population Genetics (A.P., M.I.K.), and Stanley Center for Psychiatric Research (A.P., M.I.K.), Broad Institute for Harvard and MIT, Cambridge, MA
| | - Arto Mannermaa
- From the Department of Neurosurgery (J. Räsänen, K.M., V.E.K., M.O., J.E.J., V.L.), Kuopio University Hospital and Institute of Clinical Medicine-Neurosurgery, and Institute of Biomedicine (S. Heikkinen, K.M., A.L., T.K., M.H.), University of Eastern Finland, Kuopio; Institute for Molecular Medicine Finland (FIMM) (J.M., A.P.), Helsinki Institute of Life Science (HiLIFE), University of Helsinki; Department of Neurology (A.J.), Clinical Neurosciences, Helsinki University Hospital and University of Helsinki, Finland; Univ. Lille (B.G.-B., C.B., J.-C.L.), Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, France; Department of Neurosurgery (M.O., K.L., J.S.), University of Helsinki and Helsinki University Hospital; Clinical Neurosciences (C.A., J.F., A.K., J. Rinne), Department of Neurosurgery, University of Turku and Turku University Hospital; Department of Neurosurgery (A.R.), Tampere University Hospital; Unit of Clinical Neuroscience (M.K., M.v.u.z.F.), Neurosurgery, University of Oulu and Medical Research Center, Oulu University Hospital; Finnish Institute for Health and Welfare (THL) (M.P.); University of Helsinki (M.P.); Department of Neurosciences (A.M.K., A.M.P.), University of Helsinki; Department of Geriatrics (A.M.K.), Helsinki University Hospital; NeuroCenter (A.M.K.), Kuopio University Hospital; Institute of Clinical Medicine-Neurology (V.J., H.S.), University of Eastern Finland; School of Medicine (A.M.), Institute of Clinical Medicine, Pathology and Forensic Medicine, and Translational Cancer Research Area, University of Eastern Finland; Department of Clinical Pathology (A.M.), Kuopio University Hospital; Unit of Clinical Medicine (S. Helisalmi), University of Eastern Finland, Kuopio, Finland; Department of Neurosurgery (P.K.E.), Oslo University Hospital-Rikshospitalet; Institute of Clinical Medicine (P.K.E.), Faculty of Medicine, and KG Jebsen Centre for Brain Fluid Research (P.K.E.), University of Oslo, Norway; Analytical and Translational Genetics Unit (A.P., M.I.K.), Department of Medicine, Massachusetts General Hospital, Boston; Program in Medical and Population Genetics (A.P., M.I.K.), and Stanley Center for Psychiatric Research (A.P., M.I.K.), Broad Institute for Harvard and MIT, Cambridge, MA
| | - Hilkka Soininen
- From the Department of Neurosurgery (J. Räsänen, K.M., V.E.K., M.O., J.E.J., V.L.), Kuopio University Hospital and Institute of Clinical Medicine-Neurosurgery, and Institute of Biomedicine (S. Heikkinen, K.M., A.L., T.K., M.H.), University of Eastern Finland, Kuopio; Institute for Molecular Medicine Finland (FIMM) (J.M., A.P.), Helsinki Institute of Life Science (HiLIFE), University of Helsinki; Department of Neurology (A.J.), Clinical Neurosciences, Helsinki University Hospital and University of Helsinki, Finland; Univ. Lille (B.G.-B., C.B., J.-C.L.), Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, France; Department of Neurosurgery (M.O., K.L., J.S.), University of Helsinki and Helsinki University Hospital; Clinical Neurosciences (C.A., J.F., A.K., J. Rinne), Department of Neurosurgery, University of Turku and Turku University Hospital; Department of Neurosurgery (A.R.), Tampere University Hospital; Unit of Clinical Neuroscience (M.K., M.v.u.z.F.), Neurosurgery, University of Oulu and Medical Research Center, Oulu University Hospital; Finnish Institute for Health and Welfare (THL) (M.P.); University of Helsinki (M.P.); Department of Neurosciences (A.M.K., A.M.P.), University of Helsinki; Department of Geriatrics (A.M.K.), Helsinki University Hospital; NeuroCenter (A.M.K.), Kuopio University Hospital; Institute of Clinical Medicine-Neurology (V.J., H.S.), University of Eastern Finland; School of Medicine (A.M.), Institute of Clinical Medicine, Pathology and Forensic Medicine, and Translational Cancer Research Area, University of Eastern Finland; Department of Clinical Pathology (A.M.), Kuopio University Hospital; Unit of Clinical Medicine (S. Helisalmi), University of Eastern Finland, Kuopio, Finland; Department of Neurosurgery (P.K.E.), Oslo University Hospital-Rikshospitalet; Institute of Clinical Medicine (P.K.E.), Faculty of Medicine, and KG Jebsen Centre for Brain Fluid Research (P.K.E.), University of Oslo, Norway; Analytical and Translational Genetics Unit (A.P., M.I.K.), Department of Medicine, Massachusetts General Hospital, Boston; Program in Medical and Population Genetics (A.P., M.I.K.), and Stanley Center for Psychiatric Research (A.P., M.I.K.), Broad Institute for Harvard and MIT, Cambridge, MA
| | - Seppo Helisalmi
- From the Department of Neurosurgery (J. Räsänen, K.M., V.E.K., M.O., J.E.J., V.L.), Kuopio University Hospital and Institute of Clinical Medicine-Neurosurgery, and Institute of Biomedicine (S. Heikkinen, K.M., A.L., T.K., M.H.), University of Eastern Finland, Kuopio; Institute for Molecular Medicine Finland (FIMM) (J.M., A.P.), Helsinki Institute of Life Science (HiLIFE), University of Helsinki; Department of Neurology (A.J.), Clinical Neurosciences, Helsinki University Hospital and University of Helsinki, Finland; Univ. Lille (B.G.-B., C.B., J.-C.L.), Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, France; Department of Neurosurgery (M.O., K.L., J.S.), University of Helsinki and Helsinki University Hospital; Clinical Neurosciences (C.A., J.F., A.K., J. Rinne), Department of Neurosurgery, University of Turku and Turku University Hospital; Department of Neurosurgery (A.R.), Tampere University Hospital; Unit of Clinical Neuroscience (M.K., M.v.u.z.F.), Neurosurgery, University of Oulu and Medical Research Center, Oulu University Hospital; Finnish Institute for Health and Welfare (THL) (M.P.); University of Helsinki (M.P.); Department of Neurosciences (A.M.K., A.M.P.), University of Helsinki; Department of Geriatrics (A.M.K.), Helsinki University Hospital; NeuroCenter (A.M.K.), Kuopio University Hospital; Institute of Clinical Medicine-Neurology (V.J., H.S.), University of Eastern Finland; School of Medicine (A.M.), Institute of Clinical Medicine, Pathology and Forensic Medicine, and Translational Cancer Research Area, University of Eastern Finland; Department of Clinical Pathology (A.M.), Kuopio University Hospital; Unit of Clinical Medicine (S. Helisalmi), University of Eastern Finland, Kuopio, Finland; Department of Neurosurgery (P.K.E.), Oslo University Hospital-Rikshospitalet; Institute of Clinical Medicine (P.K.E.), Faculty of Medicine, and KG Jebsen Centre for Brain Fluid Research (P.K.E.), University of Oslo, Norway; Analytical and Translational Genetics Unit (A.P., M.I.K.), Department of Medicine, Massachusetts General Hospital, Boston; Program in Medical and Population Genetics (A.P., M.I.K.), and Stanley Center for Psychiatric Research (A.P., M.I.K.), Broad Institute for Harvard and MIT, Cambridge, MA
| | - Juha E Jääskeläinen
- From the Department of Neurosurgery (J. Räsänen, K.M., V.E.K., M.O., J.E.J., V.L.), Kuopio University Hospital and Institute of Clinical Medicine-Neurosurgery, and Institute of Biomedicine (S. Heikkinen, K.M., A.L., T.K., M.H.), University of Eastern Finland, Kuopio; Institute for Molecular Medicine Finland (FIMM) (J.M., A.P.), Helsinki Institute of Life Science (HiLIFE), University of Helsinki; Department of Neurology (A.J.), Clinical Neurosciences, Helsinki University Hospital and University of Helsinki, Finland; Univ. Lille (B.G.-B., C.B., J.-C.L.), Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, France; Department of Neurosurgery (M.O., K.L., J.S.), University of Helsinki and Helsinki University Hospital; Clinical Neurosciences (C.A., J.F., A.K., J. Rinne), Department of Neurosurgery, University of Turku and Turku University Hospital; Department of Neurosurgery (A.R.), Tampere University Hospital; Unit of Clinical Neuroscience (M.K., M.v.u.z.F.), Neurosurgery, University of Oulu and Medical Research Center, Oulu University Hospital; Finnish Institute for Health and Welfare (THL) (M.P.); University of Helsinki (M.P.); Department of Neurosciences (A.M.K., A.M.P.), University of Helsinki; Department of Geriatrics (A.M.K.), Helsinki University Hospital; NeuroCenter (A.M.K.), Kuopio University Hospital; Institute of Clinical Medicine-Neurology (V.J., H.S.), University of Eastern Finland; School of Medicine (A.M.), Institute of Clinical Medicine, Pathology and Forensic Medicine, and Translational Cancer Research Area, University of Eastern Finland; Department of Clinical Pathology (A.M.), Kuopio University Hospital; Unit of Clinical Medicine (S. Helisalmi), University of Eastern Finland, Kuopio, Finland; Department of Neurosurgery (P.K.E.), Oslo University Hospital-Rikshospitalet; Institute of Clinical Medicine (P.K.E.), Faculty of Medicine, and KG Jebsen Centre for Brain Fluid Research (P.K.E.), University of Oslo, Norway; Analytical and Translational Genetics Unit (A.P., M.I.K.), Department of Medicine, Massachusetts General Hospital, Boston; Program in Medical and Population Genetics (A.P., M.I.K.), and Stanley Center for Psychiatric Research (A.P., M.I.K.), Broad Institute for Harvard and MIT, Cambridge, MA
| | - Jean-Charles Lambert
- From the Department of Neurosurgery (J. Räsänen, K.M., V.E.K., M.O., J.E.J., V.L.), Kuopio University Hospital and Institute of Clinical Medicine-Neurosurgery, and Institute of Biomedicine (S. Heikkinen, K.M., A.L., T.K., M.H.), University of Eastern Finland, Kuopio; Institute for Molecular Medicine Finland (FIMM) (J.M., A.P.), Helsinki Institute of Life Science (HiLIFE), University of Helsinki; Department of Neurology (A.J.), Clinical Neurosciences, Helsinki University Hospital and University of Helsinki, Finland; Univ. Lille (B.G.-B., C.B., J.-C.L.), Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, France; Department of Neurosurgery (M.O., K.L., J.S.), University of Helsinki and Helsinki University Hospital; Clinical Neurosciences (C.A., J.F., A.K., J. Rinne), Department of Neurosurgery, University of Turku and Turku University Hospital; Department of Neurosurgery (A.R.), Tampere University Hospital; Unit of Clinical Neuroscience (M.K., M.v.u.z.F.), Neurosurgery, University of Oulu and Medical Research Center, Oulu University Hospital; Finnish Institute for Health and Welfare (THL) (M.P.); University of Helsinki (M.P.); Department of Neurosciences (A.M.K., A.M.P.), University of Helsinki; Department of Geriatrics (A.M.K.), Helsinki University Hospital; NeuroCenter (A.M.K.), Kuopio University Hospital; Institute of Clinical Medicine-Neurology (V.J., H.S.), University of Eastern Finland; School of Medicine (A.M.), Institute of Clinical Medicine, Pathology and Forensic Medicine, and Translational Cancer Research Area, University of Eastern Finland; Department of Clinical Pathology (A.M.), Kuopio University Hospital; Unit of Clinical Medicine (S. Helisalmi), University of Eastern Finland, Kuopio, Finland; Department of Neurosurgery (P.K.E.), Oslo University Hospital-Rikshospitalet; Institute of Clinical Medicine (P.K.E.), Faculty of Medicine, and KG Jebsen Centre for Brain Fluid Research (P.K.E.), University of Oslo, Norway; Analytical and Translational Genetics Unit (A.P., M.I.K.), Department of Medicine, Massachusetts General Hospital, Boston; Program in Medical and Population Genetics (A.P., M.I.K.), and Stanley Center for Psychiatric Research (A.P., M.I.K.), Broad Institute for Harvard and MIT, Cambridge, MA
| | - Per K Eide
- From the Department of Neurosurgery (J. Räsänen, K.M., V.E.K., M.O., J.E.J., V.L.), Kuopio University Hospital and Institute of Clinical Medicine-Neurosurgery, and Institute of Biomedicine (S. Heikkinen, K.M., A.L., T.K., M.H.), University of Eastern Finland, Kuopio; Institute for Molecular Medicine Finland (FIMM) (J.M., A.P.), Helsinki Institute of Life Science (HiLIFE), University of Helsinki; Department of Neurology (A.J.), Clinical Neurosciences, Helsinki University Hospital and University of Helsinki, Finland; Univ. Lille (B.G.-B., C.B., J.-C.L.), Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, France; Department of Neurosurgery (M.O., K.L., J.S.), University of Helsinki and Helsinki University Hospital; Clinical Neurosciences (C.A., J.F., A.K., J. Rinne), Department of Neurosurgery, University of Turku and Turku University Hospital; Department of Neurosurgery (A.R.), Tampere University Hospital; Unit of Clinical Neuroscience (M.K., M.v.u.z.F.), Neurosurgery, University of Oulu and Medical Research Center, Oulu University Hospital; Finnish Institute for Health and Welfare (THL) (M.P.); University of Helsinki (M.P.); Department of Neurosciences (A.M.K., A.M.P.), University of Helsinki; Department of Geriatrics (A.M.K.), Helsinki University Hospital; NeuroCenter (A.M.K.), Kuopio University Hospital; Institute of Clinical Medicine-Neurology (V.J., H.S.), University of Eastern Finland; School of Medicine (A.M.), Institute of Clinical Medicine, Pathology and Forensic Medicine, and Translational Cancer Research Area, University of Eastern Finland; Department of Clinical Pathology (A.M.), Kuopio University Hospital; Unit of Clinical Medicine (S. Helisalmi), University of Eastern Finland, Kuopio, Finland; Department of Neurosurgery (P.K.E.), Oslo University Hospital-Rikshospitalet; Institute of Clinical Medicine (P.K.E.), Faculty of Medicine, and KG Jebsen Centre for Brain Fluid Research (P.K.E.), University of Oslo, Norway; Analytical and Translational Genetics Unit (A.P., M.I.K.), Department of Medicine, Massachusetts General Hospital, Boston; Program in Medical and Population Genetics (A.P., M.I.K.), and Stanley Center for Psychiatric Research (A.P., M.I.K.), Broad Institute for Harvard and MIT, Cambridge, MA
| | - Aarno Palotie
- From the Department of Neurosurgery (J. Räsänen, K.M., V.E.K., M.O., J.E.J., V.L.), Kuopio University Hospital and Institute of Clinical Medicine-Neurosurgery, and Institute of Biomedicine (S. Heikkinen, K.M., A.L., T.K., M.H.), University of Eastern Finland, Kuopio; Institute for Molecular Medicine Finland (FIMM) (J.M., A.P.), Helsinki Institute of Life Science (HiLIFE), University of Helsinki; Department of Neurology (A.J.), Clinical Neurosciences, Helsinki University Hospital and University of Helsinki, Finland; Univ. Lille (B.G.-B., C.B., J.-C.L.), Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, France; Department of Neurosurgery (M.O., K.L., J.S.), University of Helsinki and Helsinki University Hospital; Clinical Neurosciences (C.A., J.F., A.K., J. Rinne), Department of Neurosurgery, University of Turku and Turku University Hospital; Department of Neurosurgery (A.R.), Tampere University Hospital; Unit of Clinical Neuroscience (M.K., M.v.u.z.F.), Neurosurgery, University of Oulu and Medical Research Center, Oulu University Hospital; Finnish Institute for Health and Welfare (THL) (M.P.); University of Helsinki (M.P.); Department of Neurosciences (A.M.K., A.M.P.), University of Helsinki; Department of Geriatrics (A.M.K.), Helsinki University Hospital; NeuroCenter (A.M.K.), Kuopio University Hospital; Institute of Clinical Medicine-Neurology (V.J., H.S.), University of Eastern Finland; School of Medicine (A.M.), Institute of Clinical Medicine, Pathology and Forensic Medicine, and Translational Cancer Research Area, University of Eastern Finland; Department of Clinical Pathology (A.M.), Kuopio University Hospital; Unit of Clinical Medicine (S. Helisalmi), University of Eastern Finland, Kuopio, Finland; Department of Neurosurgery (P.K.E.), Oslo University Hospital-Rikshospitalet; Institute of Clinical Medicine (P.K.E.), Faculty of Medicine, and KG Jebsen Centre for Brain Fluid Research (P.K.E.), University of Oslo, Norway; Analytical and Translational Genetics Unit (A.P., M.I.K.), Department of Medicine, Massachusetts General Hospital, Boston; Program in Medical and Population Genetics (A.P., M.I.K.), and Stanley Center for Psychiatric Research (A.P., M.I.K.), Broad Institute for Harvard and MIT, Cambridge, MA
| | - Mitja I Kurki
- From the Department of Neurosurgery (J. Räsänen, K.M., V.E.K., M.O., J.E.J., V.L.), Kuopio University Hospital and Institute of Clinical Medicine-Neurosurgery, and Institute of Biomedicine (S. Heikkinen, K.M., A.L., T.K., M.H.), University of Eastern Finland, Kuopio; Institute for Molecular Medicine Finland (FIMM) (J.M., A.P.), Helsinki Institute of Life Science (HiLIFE), University of Helsinki; Department of Neurology (A.J.), Clinical Neurosciences, Helsinki University Hospital and University of Helsinki, Finland; Univ. Lille (B.G.-B., C.B., J.-C.L.), Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, France; Department of Neurosurgery (M.O., K.L., J.S.), University of Helsinki and Helsinki University Hospital; Clinical Neurosciences (C.A., J.F., A.K., J. Rinne), Department of Neurosurgery, University of Turku and Turku University Hospital; Department of Neurosurgery (A.R.), Tampere University Hospital; Unit of Clinical Neuroscience (M.K., M.v.u.z.F.), Neurosurgery, University of Oulu and Medical Research Center, Oulu University Hospital; Finnish Institute for Health and Welfare (THL) (M.P.); University of Helsinki (M.P.); Department of Neurosciences (A.M.K., A.M.P.), University of Helsinki; Department of Geriatrics (A.M.K.), Helsinki University Hospital; NeuroCenter (A.M.K.), Kuopio University Hospital; Institute of Clinical Medicine-Neurology (V.J., H.S.), University of Eastern Finland; School of Medicine (A.M.), Institute of Clinical Medicine, Pathology and Forensic Medicine, and Translational Cancer Research Area, University of Eastern Finland; Department of Clinical Pathology (A.M.), Kuopio University Hospital; Unit of Clinical Medicine (S. Helisalmi), University of Eastern Finland, Kuopio, Finland; Department of Neurosurgery (P.K.E.), Oslo University Hospital-Rikshospitalet; Institute of Clinical Medicine (P.K.E.), Faculty of Medicine, and KG Jebsen Centre for Brain Fluid Research (P.K.E.), University of Oslo, Norway; Analytical and Translational Genetics Unit (A.P., M.I.K.), Department of Medicine, Massachusetts General Hospital, Boston; Program in Medical and Population Genetics (A.P., M.I.K.), and Stanley Center for Psychiatric Research (A.P., M.I.K.), Broad Institute for Harvard and MIT, Cambridge, MA
| | - Mikko Hiltunen
- From the Department of Neurosurgery (J. Räsänen, K.M., V.E.K., M.O., J.E.J., V.L.), Kuopio University Hospital and Institute of Clinical Medicine-Neurosurgery, and Institute of Biomedicine (S. Heikkinen, K.M., A.L., T.K., M.H.), University of Eastern Finland, Kuopio; Institute for Molecular Medicine Finland (FIMM) (J.M., A.P.), Helsinki Institute of Life Science (HiLIFE), University of Helsinki; Department of Neurology (A.J.), Clinical Neurosciences, Helsinki University Hospital and University of Helsinki, Finland; Univ. Lille (B.G.-B., C.B., J.-C.L.), Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, France; Department of Neurosurgery (M.O., K.L., J.S.), University of Helsinki and Helsinki University Hospital; Clinical Neurosciences (C.A., J.F., A.K., J. Rinne), Department of Neurosurgery, University of Turku and Turku University Hospital; Department of Neurosurgery (A.R.), Tampere University Hospital; Unit of Clinical Neuroscience (M.K., M.v.u.z.F.), Neurosurgery, University of Oulu and Medical Research Center, Oulu University Hospital; Finnish Institute for Health and Welfare (THL) (M.P.); University of Helsinki (M.P.); Department of Neurosciences (A.M.K., A.M.P.), University of Helsinki; Department of Geriatrics (A.M.K.), Helsinki University Hospital; NeuroCenter (A.M.K.), Kuopio University Hospital; Institute of Clinical Medicine-Neurology (V.J., H.S.), University of Eastern Finland; School of Medicine (A.M.), Institute of Clinical Medicine, Pathology and Forensic Medicine, and Translational Cancer Research Area, University of Eastern Finland; Department of Clinical Pathology (A.M.), Kuopio University Hospital; Unit of Clinical Medicine (S. Helisalmi), University of Eastern Finland, Kuopio, Finland; Department of Neurosurgery (P.K.E.), Oslo University Hospital-Rikshospitalet; Institute of Clinical Medicine (P.K.E.), Faculty of Medicine, and KG Jebsen Centre for Brain Fluid Research (P.K.E.), University of Oslo, Norway; Analytical and Translational Genetics Unit (A.P., M.I.K.), Department of Medicine, Massachusetts General Hospital, Boston; Program in Medical and Population Genetics (A.P., M.I.K.), and Stanley Center for Psychiatric Research (A.P., M.I.K.), Broad Institute for Harvard and MIT, Cambridge, MA
| | - Ville Leinonen
- From the Department of Neurosurgery (J. Räsänen, K.M., V.E.K., M.O., J.E.J., V.L.), Kuopio University Hospital and Institute of Clinical Medicine-Neurosurgery, and Institute of Biomedicine (S. Heikkinen, K.M., A.L., T.K., M.H.), University of Eastern Finland, Kuopio; Institute for Molecular Medicine Finland (FIMM) (J.M., A.P.), Helsinki Institute of Life Science (HiLIFE), University of Helsinki; Department of Neurology (A.J.), Clinical Neurosciences, Helsinki University Hospital and University of Helsinki, Finland; Univ. Lille (B.G.-B., C.B., J.-C.L.), Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, France; Department of Neurosurgery (M.O., K.L., J.S.), University of Helsinki and Helsinki University Hospital; Clinical Neurosciences (C.A., J.F., A.K., J. Rinne), Department of Neurosurgery, University of Turku and Turku University Hospital; Department of Neurosurgery (A.R.), Tampere University Hospital; Unit of Clinical Neuroscience (M.K., M.v.u.z.F.), Neurosurgery, University of Oulu and Medical Research Center, Oulu University Hospital; Finnish Institute for Health and Welfare (THL) (M.P.); University of Helsinki (M.P.); Department of Neurosciences (A.M.K., A.M.P.), University of Helsinki; Department of Geriatrics (A.M.K.), Helsinki University Hospital; NeuroCenter (A.M.K.), Kuopio University Hospital; Institute of Clinical Medicine-Neurology (V.J., H.S.), University of Eastern Finland; School of Medicine (A.M.), Institute of Clinical Medicine, Pathology and Forensic Medicine, and Translational Cancer Research Area, University of Eastern Finland; Department of Clinical Pathology (A.M.), Kuopio University Hospital; Unit of Clinical Medicine (S. Helisalmi), University of Eastern Finland, Kuopio, Finland; Department of Neurosurgery (P.K.E.), Oslo University Hospital-Rikshospitalet; Institute of Clinical Medicine (P.K.E.), Faculty of Medicine, and KG Jebsen Centre for Brain Fluid Research (P.K.E.), University of Oslo, Norway; Analytical and Translational Genetics Unit (A.P., M.I.K.), Department of Medicine, Massachusetts General Hospital, Boston; Program in Medical and Population Genetics (A.P., M.I.K.), and Stanley Center for Psychiatric Research (A.P., M.I.K.), Broad Institute for Harvard and MIT, Cambridge, MA
| |
Collapse
|
15
|
Puerta R, de Rojas I, García-González P, Olivé C, Sotolongo-Grau O, García-Sánchez A, García-Gutiérrez F, Montrreal L, Pablo Tartari J, Sanabria Á, Pytel V, Lage C, Quintela I, Aguilera N, Rodriguez-Rodriguez E, Alarcón-Martín E, Orellana A, Pastor P, Pérez-Tur J, Piñol-Ripoll G, de Munian AL, García-Alberca JM, Royo JL, Bullido MJ, Álvarez V, Real LM, Anchuelo AC, Gómez-Garre D, Larrad MTM, Franco-Macías E, Mir P, Medina M, Sánchez-Valle R, Dols-Icardo O, Sáez ME, Carracedo Á, Tárraga L, Alegret M, Valero S, Marquié M, Boada M, Juan PS, Cavazos JE, Cabrera A, Cano A. Connecting genomic and proteomic signatures of amyloid burden in the brain. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.09.06.24313124. [PMID: 39281766 PMCID: PMC11398581 DOI: 10.1101/2024.09.06.24313124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/18/2024]
Abstract
Background Alzheimer's disease (AD) has a high heritable component characteristic of complex diseases, yet many of the genetic risk factors remain unknown. We combined genome-wide association studies (GWAS) on amyloid endophenotypes measured in cerebrospinal fluid (CSF) and positron emission tomography (PET) as surrogates of amyloid pathology, which may be helpful to understand the underlying biology of the disease. Methods We performed a meta-analysis of GWAS of CSF Aβ42 and PET measures combining six independent cohorts (n=2,076). Due to the opposite effect direction of Aβ phenotypes in CSF and PET measures, only genetic signals in the opposite direction were considered for analysis (n=376,599). Polygenic risk scores (PRS) were calculated and evaluated for AD status and amyloid endophenotypes. We then searched the CSF proteome signature of brain amyloidosis using SOMAscan proteomic data (Ace cohort, n=1,008) and connected it with GWAS results of loci modulating amyloidosis. Finally, we compared our results with a large meta-analysis using publicly available datasets in CSF (n=13,409) and PET (n=13,116). This combined approach enabled the identification of overlapping genes and proteins associated with amyloid burden and the assessment of their biological significance using enrichment analyses. Results After filtering the meta-GWAS, we observed genome-wide significance in the rs429358-APOE locus and nine suggestive hits were annotated. We replicated the APOE loci using the large CSF-PET meta-GWAS and identified multiple AD-associated genes as well as the novel GADL1 locus. Additionally, we found a significant association between the AD PRS and amyloid levels, whereas no significant association was found between any Aβ PRS with AD risk. CSF SOMAscan analysis identified 1,387 FDR-significant proteins associated with CSF Aβ42 levels. The overlap among GWAS loci and proteins associated with amyloid burden was very poor (n=35). The enrichment analysis of overlapping hits strongly suggested several signalling pathways connecting amyloidosis with the anchored component of the plasma membrane, synapse physiology and mental disorders that were replicated in the large CSF-PET meta-analysis. Conclusions The strategy of combining CSF and PET amyloid endophenotypes GWAS with CSF proteome analyses might be effective for identifying signals associated with the AD pathological process and elucidate causative molecular mechanisms behind the amyloid mobilization in AD.
Collapse
Affiliation(s)
- Raquel Puerta
- Ace Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Spain
- Universitat de Barcelona (UB)
| | - Itziar de Rojas
- Ace Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
| | - Pablo García-González
- Ace Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
| | - Clàudia Olivé
- Ace Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Spain
| | | | | | | | - Laura Montrreal
- Ace Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Spain
| | - Juan Pablo Tartari
- Ace Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Spain
| | - Ángela Sanabria
- Ace Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
| | - Vanesa Pytel
- Ace Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
| | - Carmen Lage
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
- Neurology Service, Marqués de Valdecilla University Hospital (University of Cantabria and IDIVAL), Santander, Spain
| | - Inés Quintela
- Grupo de Medicina Xenómica, Centro Nacional de Genotipado (CEGEN-PRB3-ISCIII). Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Nuria Aguilera
- Ace Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Spain
| | - Eloy Rodriguez-Rodriguez
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
- Neurology Service, Marqués de Valdecilla University Hospital (University of Cantabria and IDIVAL), Santander, Spain
| | | | - Adelina Orellana
- Ace Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
| | - Pau Pastor
- Unit of Neurodegenerative diseases, Department of Neurology, University Hospital Germans Trias i Pujol, Badalona, Barcelona, Spain
- The Germans Trias i Pujol Research Institute (IGTP), Badalona, Barcelona, Spain
| | - Jordi Pérez-Tur
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
- Unitat de Genètica Molecular, Institut de Biomedicina de València-CSIC, Valencia, Spain
- Unidad Mixta de Neurologia Genètica, Instituto de Investigación Sanitaria La Fe, Valencia, Spain
| | - Gerard Piñol-Ripoll
- Unitat Trastorns Cognitius, Hospital Universitari Santa Maria de Lleida, Lleida, Spain
- Institut de Recerca Biomedica de Lleida (IRBLLeida), Lleida, Spain
| | - Adolfo López de Munian
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
- Department of Neurology. Hospital Universitario Donostia. San Sebastian, Spain
- Department of Neurosciences. Faculty of Medicine and Nursery. University of the Basque Country, San Sebastián, Spain
- Neurosciences Area. Instituto Biodonostia. San Sebastian, Spain
| | - Jose María García-Alberca
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
- Alzheimer Research Center & Memory Clinic, Andalusian Institute for Neuroscience, Málaga, Spain
| | - Jose Luís Royo
- Departamento de Especialidades Quirúrgicas, Bioquímica e Inmunología. School of Medicine. University of Malaga. Málaga, Spain
| | - María Jesús Bullido
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
- Centro de Biología Molecular Severo Ochoa (UAM-CSIC)
- Instituto de Investigacion Sanitaria ‘Hospital la Paz’ (IdIPaz), Madrid, Spain
- Universidad Autónoma de Madrid
| | - Victoria Álvarez
- Laboratorio de Genética. Hospital Universitario Central de Asturias, Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA)
| | - Luis Miguel Real
- Departamento de Especialidades Quirúrgicas, Bioquímica e Inmunología. School of Medicine. University of Malaga. Málaga, Spain
- Unidad Clínica de Enfermedades Infecciosas y Microbiología.Hospital Universitario de Valme, Sevilla, Spain
| | - Arturo Corbatón Anchuelo
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Hospital Clínico San Carlos
| | - Dulcenombre Gómez-Garre
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Hospital Clínico San Carlos
- Laboratorio de Riesgo Cardiovascular y Microbiota, Hospital Clínico San Carlos; Departamento de Fisiología, Facultad de Medicina, Universidad Complutense de Madrid (UCM)
- Biomedical Research Networking Center in Cardiovascular Diseases (CIBERCV), Madrid, Spain
| | - María Teresa Martínez Larrad
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Hospital Clínico San Carlos
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM)
| | - Emilio Franco-Macías
- Dementia Unit, Department of Neurology, Hospital Universitario Virgen del Rocío, Instituto de Biomedicina de Sevilla (IBiS), Sevilla, Spain
| | - Pablo Mir
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
- Unidad de Trastornos del Movimiento, Servicio de Neurología y Neurofisiología. Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
| | - Miguel Medina
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
- CIEN Foundation/Queen Sofia Foundation Alzheimer Center
| | - Raquel Sánchez-Valle
- Alzheimer’s disease and other cognitive disorders unit. Service of Neurology. Hospital Clínic of Barcelona. Institut d’Investigacions Biomèdiques August Pi i Sunyer, University of Barcelona, Barcelona, Spain
| | - Oriol Dols-Icardo
- Department of Neurology, Sant Pau Memory Unit, Sant Pau Biomedical Research Institute, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
| | | | - Ángel Carracedo
- Grupo de Medicina Xenómica, Centro Nacional de Genotipado (CEGEN-PRB3-ISCIII). Universidade de Santiago de Compostela, Santiago de Compostela, Spain
- Fundación Pública Galega de Medicina Xenómica – CIBERER-IDIS, Santiago de Compostela, Spain
| | - Lluís Tárraga
- Ace Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
| | - Montse Alegret
- Ace Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
| | - Sergi Valero
- Ace Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
| | - Marta Marquié
- Ace Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
| | - Mercè Boada
- Ace Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
| | - Pascual Sánchez Juan
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
- Neurology Service, Marqués de Valdecilla University Hospital (University of Cantabria and IDIVAL), Santander, Spain
| | - Jose Enrique Cavazos
- South Texas Medical Science Training Program, University of Texas Health San Antonio, San Antonio
- Glenn Biggs Institute for Alzheimer’s & Neurodegenerative Diseases, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Dr, San Antonio, TX 78229 USA
| | - Alfredo Cabrera
- Neuroscience Therapeutic Area, Janssen Research & Development, Turnhoutseweg 30, 2340 Beerse, Belgium
| | - Amanda Cano
- Ace Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
| | - Alzheimer’s Disease Neuroimaging Initiative.
- Ace Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
- Glenn Biggs Institute for Alzheimer’s & Neurodegenerative Diseases, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Dr, San Antonio, TX 78229 USA
| |
Collapse
|
16
|
Hu T, Liu Q, Dai Q, Parrish RL, Buchman AS, Tasaki S, Seyfried NT, Wang Y, Bennett DA, De Jager PL, Epstein MP, Yang J. Proteome-wide association studies using summary pQTL data of three tissues identified 30 risk genes of Alzheimer's disease dementia. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.03.28.24305044. [PMID: 38585769 PMCID: PMC10996749 DOI: 10.1101/2024.03.28.24305044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
Background Proteome-wide association study (PWAS) integrating proteomic data with genome-wide association study (GWAS) summary data is a powerful tool for studying Alzheimer's disease (AD) dementia. Existing PWAS analyses of AD often rely on the availability of individual-level proteomic and genetic data of a reference panel. Leveraging summary protein quantitative trait loci (pQTL) reference data of multiple AD-relevant tissues is expected to improve PWAS findings of AD dementia. Methods We conducted PWAS of AD dementia by integrating publicly available summary pQTL data of brain, cerebrospinal fluid (CSF), and plasma tissues, with the latest GWAS summary data of AD dementia. For each target protein per tissue, we employed our recently published OTTERS tool to obtain omnibus PWAS p-value, to test whether the genetically regulated protein abundance in the corresponding tissue is associated with AD dementia. Protein-protein interactions and enriched pathways of identified significant PWAS risk genes were analyzed by STRING. The potential causal effects of these PWAS risk genes were assessed by probabilistic Mendelian randomization analyses. Results We identified 30 unique significant PWAS risk genes for AD dementia, including 11 for brain, 9 for CSF, and 16 for plasma tissues. Four of these were shared by at least two tissues, and gene MAPK3 was found in all three tissues. We found that 11 of these PWAS risk genes were associated with AD or AD pathological hall marks as shown in GWAS Catalog; 18 of these were detected by transcriptome-wide association studies (TWAS); and 25 of these, including 8 out of 9 novel genes, were interconnected within a protein-protein interaction network involving the well-known AD risk gene APOE. Especially, these PWAS risk genes were enriched in immune response, glial cell proliferation, and high-density lipoprotein particle clearance pathways. Mediated causal effects were validated for 13 PWAS risk genes (43.3%). Conclusions Our findings provide novel insights into the genetic mechanisms of AD dementia in brain, CSF, and plasma tissues, and targets for developing therapeutic interventions. We also demonstrated the effectiveness of integrating summary pQTL and GWAS data for mapping risk genes of complex human diseases.
Collapse
Affiliation(s)
- Tingyang Hu
- Center for Computational and Quantitative Genetics, Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Division of Biostatistics and Bioinformatics, Department of Public Health Sciences, Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA
| | - Qiang Liu
- Center for Computational and Quantitative Genetics, Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Department of Biostatistics and Bioinformatics, Emory University School of Public Health, Atlanta, GA, 30322, USA
| | - Qile Dai
- Center for Computational and Quantitative Genetics, Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Department of Biostatistics and Bioinformatics, Emory University School of Public Health, Atlanta, GA, 30322, USA
| | - Randy L. Parrish
- Center for Computational and Quantitative Genetics, Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Department of Biostatistics and Bioinformatics, Emory University School of Public Health, Atlanta, GA, 30322, USA
| | - Aron S. Buchman
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL, 60612, USA
| | - Shinya Tasaki
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL, 60612, USA
| | - Nicholas T. Seyfried
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Yanling Wang
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL, 60612, USA
| | - David A. Bennett
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL, 60612, USA
| | - Philip L. De Jager
- Center for Translational and Computational Neuroimmunology, Department of Neurology and Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY10032, USA
| | - Michael P. Epstein
- Center for Computational and Quantitative Genetics, Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Jingjing Yang
- Center for Computational and Quantitative Genetics, Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, 30322, USA
| |
Collapse
|
17
|
Saha O, Melo de Farias AR, Pelletier A, Siedlecki-Wullich D, Landeira BS, Gadaut J, Carrier A, Vreulx AC, Guyot K, Shen Y, Bonnefond A, Amouyel P, Tcw J, Kilinc D, Queiroz CM, Delahaye F, Lambert JC, Costa MR. The Alzheimer's disease risk gene BIN1 regulates activity-dependent gene expression in human-induced glutamatergic neurons. Mol Psychiatry 2024; 29:2634-2646. [PMID: 38514804 PMCID: PMC11420064 DOI: 10.1038/s41380-024-02502-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Revised: 02/22/2024] [Accepted: 02/27/2024] [Indexed: 03/23/2024]
Abstract
Bridging Integrator 1 (BIN1) is the second most important Alzheimer's disease (AD) risk gene, but its physiological roles in neurons and its contribution to brain pathology remain largely elusive. In this work, we show that BIN1 plays a critical role in the regulation of calcium homeostasis, electrical activity, and gene expression of glutamatergic neurons. Using single-cell RNA-sequencing on cerebral organoids generated from isogenic BIN1 wild type (WT), heterozygous (HET) and homozygous knockout (KO) human-induced pluripotent stem cells (hiPSCs), we show that BIN1 is mainly expressed by oligodendrocytes and glutamatergic neurons, like in the human brain. Both BIN1 HET and KO cerebral organoids show specific transcriptional alterations, mainly associated with ion transport and synapses in glutamatergic neurons. We then demonstrate that BIN1 cell-autonomously regulates gene expression in glutamatergic neurons by using a novel protocol to generate pure culture of hiPSC-derived induced neurons (hiNs). Using this system, we also show that BIN1 plays a key role in the regulation of neuronal calcium transients and electrical activity via its interaction with the L-type voltage-gated calcium channel Cav1.2. BIN1 KO hiNs show reduced activity-dependent internalization and higher Cav1.2 expression compared to WT hiNs. Pharmacological blocking of this channel with clinically relevant doses of nifedipine, a calcium channel blocker, partly rescues electrical and gene expression alterations in BIN1 KO glutamatergic neurons. Further, we show that transcriptional alterations in BIN1 KO hiNs that affect biological processes related to calcium homeostasis are also present in glutamatergic neurons of the human brain at late stages of AD pathology. Together, these findings suggest that BIN1-dependent alterations in neuronal properties could contribute to AD pathophysiology and that treatment with low doses of clinically approved calcium blockers should be considered as an option to slow disease-onset and progression.
Collapse
Affiliation(s)
- Orthis Saha
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, DISTALZ, 1 rue du Professeur Calmette, 59019, Lille, France
| | - Ana Raquel Melo de Farias
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, DISTALZ, 1 rue du Professeur Calmette, 59019, Lille, France
- Brain Institute, Federal University of Rio Grande do Norte, Av. Senador Salgado Filho, 3000, Campus Universitário, Lagoa, Nova, 59078-970, Natal, Brazil
| | - Alexandre Pelletier
- Univ. Lille, Inserm, CNRS, CHU Lille, Institut Pasteur de Lille, U1283-UMR 8199 EGID, Pôle Recherche, 1 Place de Verdun, 59045, Lille, Cedex, France
- Department of Pharmacology, Physiology & Biophysics, Boston University, Chobanian & Avedisian School of Medicine, Boston, MA, 02118, USA
| | - Dolores Siedlecki-Wullich
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, DISTALZ, 1 rue du Professeur Calmette, 59019, Lille, France
| | - Bruna Soares Landeira
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, DISTALZ, 1 rue du Professeur Calmette, 59019, Lille, France
| | - Johanna Gadaut
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, DISTALZ, 1 rue du Professeur Calmette, 59019, Lille, France
| | - Arnaud Carrier
- Univ. Lille, Inserm, CNRS, CHU Lille, Institut Pasteur de Lille, U1283-UMR 8199 EGID, Pôle Recherche, 1 Place de Verdun, 59045, Lille, Cedex, France
| | - Anaïs-Camille Vreulx
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, DISTALZ, 1 rue du Professeur Calmette, 59019, Lille, France
| | - Karine Guyot
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, DISTALZ, 1 rue du Professeur Calmette, 59019, Lille, France
| | - Yun Shen
- Department of Pharmacology, Physiology & Biophysics, Boston University, Chobanian & Avedisian School of Medicine, Boston, MA, 02118, USA
| | - Amelie Bonnefond
- Univ. Lille, Inserm, CNRS, CHU Lille, Institut Pasteur de Lille, U1283-UMR 8199 EGID, Pôle Recherche, 1 Place de Verdun, 59045, Lille, Cedex, France
| | - Philippe Amouyel
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, DISTALZ, 1 rue du Professeur Calmette, 59019, Lille, France
| | - Julia Tcw
- Department of Pharmacology, Physiology & Biophysics, Boston University, Chobanian & Avedisian School of Medicine, Boston, MA, 02118, USA
- Bioinformatics Program, Faculty of Computing & Data Sciences, Boston University, Boston, MA, 02115, USA
| | - Devrim Kilinc
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, DISTALZ, 1 rue du Professeur Calmette, 59019, Lille, France
| | - Claudio Marcos Queiroz
- Brain Institute, Federal University of Rio Grande do Norte, Av. Senador Salgado Filho, 3000, Campus Universitário, Lagoa, Nova, 59078-970, Natal, Brazil
| | - Fabien Delahaye
- Univ. Lille, Inserm, CNRS, CHU Lille, Institut Pasteur de Lille, U1283-UMR 8199 EGID, Pôle Recherche, 1 Place de Verdun, 59045, Lille, Cedex, France
| | - Jean-Charles Lambert
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, DISTALZ, 1 rue du Professeur Calmette, 59019, Lille, France
| | - Marcos R Costa
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, DISTALZ, 1 rue du Professeur Calmette, 59019, Lille, France.
- Brain Institute, Federal University of Rio Grande do Norte, Av. Senador Salgado Filho, 3000, Campus Universitário, Lagoa, Nova, 59078-970, Natal, Brazil.
| |
Collapse
|
18
|
Liu XT, Chen X, Zhao N, Geng F, Zhu MM, Ren QG. Synergism of ApoE4 and systemic infectious burden is mediated by the APOE-NLRP3 axis in Alzheimer's disease. Psychiatry Clin Neurosci 2024; 78:517-526. [PMID: 39011734 DOI: 10.1111/pcn.13704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 04/12/2024] [Accepted: 06/03/2024] [Indexed: 07/17/2024]
Abstract
BACKGROUND Systemic infections are associated with the development of AD, especially in individuals carrying the APOE4 genotype. However, the detailed mechanism through which APOE4 affects microglia inflammatory response remains unclear. METHODS We obtained human snRNA-seq data from the Synapse AD Knowledge Portal and assessed the DEGs between APOE3 and APOE4 isoforms in microglia. To verify the interaction between ApoE and infectious products, we used ApoE to stimulate in vitro and in vivo models in the presence or absence of LPS (or ATP). The NLRP3 gene knockout experiment was performed to demonstrate whether the APOE-NLRP3 axis was indispensable for microglia to regulate inflammation and mitochondrial autophagy. Results were evaluated by biochemical analyses and fluorescence imaging. RESULTS Compared with APOE3, up-regulated genes in APOE4 gene carriers were involved in pro-inflammatory responses. ApoE4-stimulation significantly increased the levels of NLRP3 inflammasomes and ROS in microglia. Moreover, compared with ApoE4 alone, the co-incubation of ApoE4 with LPS (or ATP) markedly promoted pyroptosis. Both NF-κB activation and mitochondrial autophagy dysfunction were contributed by the increased level of NLRP3 inflammasomes induced by ApoE4. Furthermore, the pathological impairment induced by ApoE4 could be reversed by NLRP3 KO. CONCLUSIONS Our study highlights the importance of NLRP3 inflammasomes in linking ApoE4 with microglia innate immune function. These findings not only provide a molecular basis for APOE4-mediated neuroinflammatory but also reveal the potential reason for the increased risk of AD in APOE4 gene carriers after contracting infectious diseases.
Collapse
Affiliation(s)
- Xue-Ting Liu
- School of Medicine, Southeast University, Nanjing, China
| | - Xiu Chen
- School of Medicine, Southeast University, Nanjing, China
| | - Na Zhao
- School of Medicine, Southeast University, Nanjing, China
| | - Fan Geng
- School of Medicine, Southeast University, Nanjing, China
| | - Meng-Meng Zhu
- School of Medicine, Southeast University, Nanjing, China
| | - Qing-Guo Ren
- Department of Neurology, Affiliated ZhongDa Hospital, Southeast University, Nanjing, China
| |
Collapse
|
19
|
Ge Y, Chen S, Wu B, Zhang Y, Wang J, He X, Liu W, Chen Y, Ou Y, Shen X, Huang Y, Gan Y, Yang L, Ma L, Ma Y, Chen K, Chen S, Cui M, Tan L, Dong Q, Zhao Q, Wang Y, Jia J, Yu J. Genome-wide meta-analysis identifies ancestry-specific loci for Alzheimer's disease. Alzheimers Dement 2024; 20:6243-6256. [PMID: 39023044 PMCID: PMC11497642 DOI: 10.1002/alz.14121] [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: 12/13/2023] [Revised: 06/03/2024] [Accepted: 06/10/2024] [Indexed: 07/20/2024]
Abstract
INTRODUCTION Alzheimer's disease (AD) is a devastating neurological disease with complex genetic etiology. Yet most known loci have only identified from the late-onset type AD in populations of European ancestry. METHODS We performed a two-stage genome-wide association study (GWAS) of AD totaling 6878 Chinese and 63,926 European individuals. RESULTS In addition to the apolipoprotein E (APOE) locus, our GWAS of two independent Chinese samples uncovered three novel AD susceptibility loci (KIAA2013, SLC52A3, and TCN2) and a novel ancestry-specific variant within EGFR (rs1815157). More replicated variants were observed in the Chinese (31%) than in the European samples (15%). In combining genome-wide associations and functional annotations, EGFR and TCN2 were prioritized as two of the most biologically significant genes. Phenome-wide Mendelian randomization suggests that high mean corpuscular hemoglobin concentration might protect against AD. DISCUSSION The current study reveals novel AD susceptibility loci, emphasizes the importance of diverse populations in AD genetic research, and advances our understanding of disease etiology. HIGHLIGHTS Loci KIAA2013, SLC52A3, and TCN2 were associated with Alzheimer's disease (AD) in Chinese populations. rs1815157 within the EGFR locus was associated with AD in Chinese populations. The genetic architecture of AD varied between Chinese and European populations. EGFR and TCN2 were prioritized as two of the most biologically significant genes. High mean corpuscular hemoglobin concentrations might have protective effects against AD.
Collapse
Affiliation(s)
- Yi‐Jun Ge
- Department of Neurology and Institute of NeurologyHuashan HospitalState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceShanghai Medical CollegeNational Center for Neurological DisordersFudan UniversityShanghaiChina
| | - Shi‐Dong Chen
- Department of Neurology and Institute of NeurologyHuashan HospitalState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceShanghai Medical CollegeNational Center for Neurological DisordersFudan UniversityShanghaiChina
| | - Bang‐Sheng Wu
- Department of Neurology and Institute of NeurologyHuashan HospitalState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceShanghai Medical CollegeNational Center for Neurological DisordersFudan UniversityShanghaiChina
| | - Ya‐Ru Zhang
- Department of Neurology and Institute of NeurologyHuashan HospitalState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceShanghai Medical CollegeNational Center for Neurological DisordersFudan UniversityShanghaiChina
| | - Jun Wang
- Department of Neurology and Centre for Clinical NeuroscienceDaping HospitalThird Military Medical UniversityChongqingChina
| | - Xiao‐Yu He
- Department of Neurology and Institute of NeurologyHuashan HospitalState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceShanghai Medical CollegeNational Center for Neurological DisordersFudan UniversityShanghaiChina
| | - Wei‐Shi Liu
- Department of Neurology and Institute of NeurologyHuashan HospitalState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceShanghai Medical CollegeNational Center for Neurological DisordersFudan UniversityShanghaiChina
| | - Yi‐Lin Chen
- Department of Neurology and Institute of NeurologyHuashan HospitalState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceShanghai Medical CollegeNational Center for Neurological DisordersFudan UniversityShanghaiChina
| | - Ya‐Nan Ou
- Department of NeurologyQingdao Municipal HospitalQingdao UniversityQingdaoChina
| | - Xue‐Ning Shen
- Department of Neurology and Institute of NeurologyHuashan HospitalState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceShanghai Medical CollegeNational Center for Neurological DisordersFudan UniversityShanghaiChina
| | - Yu‐Yuan Huang
- Department of Neurology and Institute of NeurologyHuashan HospitalState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceShanghai Medical CollegeNational Center for Neurological DisordersFudan UniversityShanghaiChina
| | - Yi‐Han Gan
- Department of Neurology and Institute of NeurologyHuashan HospitalState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceShanghai Medical CollegeNational Center for Neurological DisordersFudan UniversityShanghaiChina
| | - Liu Yang
- Department of Neurology and Institute of NeurologyHuashan HospitalState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceShanghai Medical CollegeNational Center for Neurological DisordersFudan UniversityShanghaiChina
| | - Ling‐Zhi Ma
- Department of NeurologyQingdao Municipal HospitalQingdao UniversityQingdaoChina
| | - Ya‐Hui Ma
- Department of NeurologyThe Affiliated Hospital of Qingdao UniversityQingdaoChina
| | - Ke‐Liang Chen
- Department of Neurology and Institute of NeurologyHuashan HospitalState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceShanghai Medical CollegeNational Center for Neurological DisordersFudan UniversityShanghaiChina
| | - Shu‐Fen Chen
- Department of Neurology and Institute of NeurologyHuashan HospitalState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceShanghai Medical CollegeNational Center for Neurological DisordersFudan UniversityShanghaiChina
| | - Mei Cui
- Department of Neurology and Institute of NeurologyHuashan HospitalState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceShanghai Medical CollegeNational Center for Neurological DisordersFudan UniversityShanghaiChina
| | - Lan Tan
- Department of NeurologyQingdao Municipal HospitalQingdao UniversityQingdaoChina
| | - Qiang Dong
- Department of Neurology and Institute of NeurologyHuashan HospitalState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceShanghai Medical CollegeNational Center for Neurological DisordersFudan UniversityShanghaiChina
| | - Qian‐Hua Zhao
- Department of Neurology and Institute of NeurologyHuashan HospitalState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceShanghai Medical CollegeNational Center for Neurological DisordersFudan UniversityShanghaiChina
| | - Yan‐Jiang Wang
- Department of Neurology and Centre for Clinical NeuroscienceDaping HospitalThird Military Medical UniversityChongqingChina
| | - Jian‐Ping Jia
- Innovation Center for Neurological Disorders and Department of NeurologyNational Clinical Research Center for Geriatric DiseasesXuanwu HospitalCapital Medical UniversityBeijingChina
| | - Jin‐Tai Yu
- Department of Neurology and Institute of NeurologyHuashan HospitalState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceShanghai Medical CollegeNational Center for Neurological DisordersFudan UniversityShanghaiChina
| |
Collapse
|
20
|
Zhang L, Cai R, Wang C, Liu J, Kuang Z, Wang H. Prediction of Multiple Degenerative Diseases Based on DNA Methylation in a Co-Physiology Mechanisms Perspective. Int J Mol Sci 2024; 25:9514. [PMID: 39273460 PMCID: PMC11395594 DOI: 10.3390/ijms25179514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 08/26/2024] [Accepted: 08/27/2024] [Indexed: 09/15/2024] Open
Abstract
Degenerative diseases oftentimes occur within the continuous process of aging, and the corresponding clinical manifestations may be neurodegeneration, neoplastic diseases, or various human complex diseases. DNA methylation provides the opportunity to explore aging and degenerative diseases as epigenetic traits. It has already been applied to age prediction and disease diagnosis. It has been shown that various degenerative diseases share co-physiology mechanisms with each other, clues of which may be gained from studying the aging process. Here, we endeavor to predict the risk of degenerative diseases in an aging-relevant comorbid mechanism perspective. Firstly, an epigenetic clock method was implemented based on a multi-scale convolutional neural network, and a Shapley feature attribution analysis was applied to discover the aging-related CpG sites. Then, these sites were further screened to a smaller subset composed of 196 sites by using biomics analysis according to their biological functions and mechanisms. Finally, we constructed a multilayer perceptron (MLP)-based degenerative disease risk prediction model, Mlp-DDR, which was well trained and tested to accurately classify nine degenerative diseases. Recent studies also suggest that DNA methylation plays a significant role in conditions like osteoporosis and osteoarthritis, broadening the potential applications of our model. This approach significantly advances the ability to understand degenerative diseases and represents a substantial shift from traditional diagnostic methods. Despite the promising results, limitations regarding model complexity and dataset diversity suggest directions for future research, including the development of tissue-specific epigenetic clocks and the inclusion of a wider range of diseases.
Collapse
Affiliation(s)
- Li Zhang
- College of Computer Science and Engineering, Changchun University of Technology, Changchun 130051, China
| | - Ruirui Cai
- School of Information Science and Technology, Institute of Computational Biology, Northeast Normal University, Changchun 130117, China
| | - Chencai Wang
- College of Computer Science and Engineering, Changchun University of Technology, Changchun 130051, China
| | - Jialong Liu
- College of Computer Science and Engineering, Changchun University of Technology, Changchun 130051, China
| | - Zhejun Kuang
- School of Cyber Security, School of Computer Science and Technology, Changchun University, Changchun 130022, China
| | - Han Wang
- School of Information Science and Technology, Institute of Computational Biology, Northeast Normal University, Changchun 130117, China
| |
Collapse
|
21
|
Chen Q, Aguirre L, Liang G, Zhao H, Dong T, Borrego F, de Rojas I, Hu Q, Reyes C, Su LY, Zhang B, Lechleiter JD, Göring HHH, De Jager PL, Kleinman JE, Hyde TM, Li PP, Ruiz A, Weinberger DR, Seshadri S, Ma L. Identification of a specific APOE transcript and functional elements associated with Alzheimer's disease. Mol Neurodegener 2024; 19:63. [PMID: 39210471 PMCID: PMC11361112 DOI: 10.1186/s13024-024-00751-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 08/16/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND The APOE gene is the strongest genetic risk factor for late-onset Alzheimer's Disease (LOAD). However, the gene regulatory mechanisms at this locus remain incompletely characterized. METHODS To identify novel AD-linked functional elements within the APOE locus, we integrated SNP variants with multi-omics data from human postmortem brains including 2,179 RNA-seq samples from 3 brain regions and two ancestries (European and African), 667 DNA methylation samples, and ChIP-seq samples. Additionally, we plotted the expression trajectory of APOE transcripts in human brains during development. RESULTS We identified an AD-linked APOE transcript (jxn1.2.2) particularly observed in the dorsolateral prefrontal cortex (DLPFC). The APOE jxn1.2.2 transcript is associated with brain neuropathological features, cognitive impairment, and the presence of the APOE4 allele in DLPFC. We prioritized two independent functional SNPs (rs157580 and rs439401) significantly associated with jxn1.2.2 transcript abundance and DNA methylation levels. These SNPs are located within active chromatin regions and affect brain-related transcription factor-binding affinities. The two SNPs shared effects on the jxn1.2.2 transcript between European and African ethnic groups. CONCLUSION The novel APOE functional elements provide potential therapeutic targets with mechanistic insight into the disease etiology.
Collapse
Affiliation(s)
- Qiang Chen
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
- Glenn Biggs Institute for Alzheimer's & Neurodegenerative Diseases, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Dr, San Antonio, TX, 78229, USA
| | - Luis Aguirre
- Glenn Biggs Institute for Alzheimer's & Neurodegenerative Diseases, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Dr, San Antonio, TX, 78229, USA
| | - Guoming Liang
- College of Animal Science, Shanxi Agricultural University, Taigu, Shanxi, China
| | - Huanhuan Zhao
- Bioinformatics Program, University of Texas at El Paso, El Paso, TX, USA
| | - Tao Dong
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Felix Borrego
- Glenn Biggs Institute for Alzheimer's & Neurodegenerative Diseases, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Dr, San Antonio, TX, 78229, USA
| | - Itziar de Rojas
- Research Center and Memory Clinic, Ace Alzheimer Center Barcelona - Universitat Internacional de Catalunya, Barcelona, Spain
- Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
| | - Qichan Hu
- Glenn Biggs Institute for Alzheimer's & Neurodegenerative Diseases, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Dr, San Antonio, TX, 78229, USA
| | - Christopher Reyes
- Glenn Biggs Institute for Alzheimer's & Neurodegenerative Diseases, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Dr, San Antonio, TX, 78229, USA
| | - Ling-Yan Su
- College of Food Science and Technology, Yunnan Agricultural University, Kunming, Yunnan, China
| | - Bao Zhang
- College of Forensic Medicine, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
| | - James D Lechleiter
- Department of Cell Systems and Anatomy, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Harald H H Göring
- South Texas Diabetes and Obesity Institute and Division of Human Genetics, University of Texas Rio Grande Valley School of Medicine, San Antonio, TX, USA
| | - Philip L De Jager
- Center for Translational and Computational Neuroimmunology, Department of Neurology and Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Medical Center, New York, NY, USA
| | - Joel E Kleinman
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Thomas M Hyde
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Pan P Li
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Agustín Ruiz
- Glenn Biggs Institute for Alzheimer's & Neurodegenerative Diseases, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Dr, San Antonio, TX, 78229, USA
- Research Center and Memory Clinic, Ace Alzheimer Center Barcelona - Universitat Internacional de Catalunya, Barcelona, Spain
- Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
| | - Daniel R Weinberger
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Departments of Neurology, Neuroscience, and Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sudha Seshadri
- Glenn Biggs Institute for Alzheimer's & Neurodegenerative Diseases, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Dr, San Antonio, TX, 78229, USA.
- Department of Neurology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA.
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA.
| | - Liang Ma
- Glenn Biggs Institute for Alzheimer's & Neurodegenerative Diseases, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Dr, San Antonio, TX, 78229, USA.
- Department of Pharmacology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA.
| |
Collapse
|
22
|
Zhou X, Wang S, Yu D, Niu T. Investigating CR1 as an indicated Gene for mild cognitive impairment in type 2 diabetes mellitus. Diabetol Metab Syndr 2024; 16:206. [PMID: 39182129 PMCID: PMC11344402 DOI: 10.1186/s13098-024-01449-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Accepted: 08/19/2024] [Indexed: 08/27/2024] Open
Abstract
OBJECTIVE Type 2 diabetes mellitus (T2DM) has beenis known as an important risk factor for cognitive impairment. Meanwhile, the liver plays a central role in the development of T2DM and insulin resistance. The present study attempted to identify and validate marker genes for mild cognitive impairment (MCI) in patients with T2DM. METHODS In this study, insulin resistance-related differentially expressed genes were identified from the liver tissues of individuals with T2DM and those with normal glucose tolerance using the Gene Expression Omnibus database and MCI-associated genes were identified using the GeneCards database. Next, enrichment analysis was performed with overlapping T2DM and MCI genes, followed by the identification of specific genes using the LASSO logistic regression and SVM-RFE algorithms. An important experiment involved the implementation of clinical and in vitro validation using real-time quantitative polymerase chain reaction (RT-qPCR). Finally, multiple linear regression, binary logistic regression, and receiver operating characteristic curve analyses were performed to investigate the relationship between the key gene and cognitive function in these patients. RESULT The present study identified 40 overlapping genes between MCI and T2DM, with subsequent enrichment analysis revealing their significant association with the roles of neuronal and glial projections. The marker gene complement receptor 1(CR1) was identified for both diseases using two regression algorithms. Based on RT-qPCR validation in 65 T2DM patients with MCI (MCI group) and 65 T2DM patients without MCI (NC group), a significant upregulation of CR1 mRNA in peripheral blood mononuclear cells was observed in the MCI group (P < 0.001). Furthermore, the CR1 gene level was significantly negatively associated with MoCA and MMSE scores, which reflect the overall cognitive function, and positively correlated with TMTB scores, which indicate the executive function. Finally, elevated CR1 mRNA levels were identified as an independent risk factor for MCI (OR = 1.481, P < 0.001). CONCLUSION These findings suggest that CR1 is an important predictor of MCI in patients with T2DM. Thus, CR1 has potential clinical significance, which may offer new ideas and directions for the management and treatment of T2DM. The identification and clinical validation of dysregulated marker genes in both T2DM and MCI can offer valuable insights into the intrinsic association between these two conditions. The current study insights may inspire the development of novel strategies for addressing the complicated issues related to cognitive impairment associated with diabetes.
Collapse
Affiliation(s)
- Xueling Zhou
- School of Medicine, Southeast University, Nanjing, China
- Department of Endocrinology, Affiliated Zhongda Hospital of Southeast University, Nanjing, China
| | - Shaohua Wang
- School of Medicine, Southeast University, Nanjing, China.
- Department of Endocrinology, Affiliated Zhongda Hospital of Southeast University, Nanjing, China.
| | - Dandan Yu
- School of Medicine, Southeast University, Nanjing, China
- Department of Endocrinology, Affiliated Zhongda Hospital of Southeast University, Nanjing, China
| | - Tong Niu
- School of Medicine, Southeast University, Nanjing, China
- Department of Endocrinology, Affiliated Zhongda Hospital of Southeast University, Nanjing, China
| |
Collapse
|
23
|
Altmann A, Aksman LM, Oxtoby NP, Young AL, Alexander DC, Barkhof F, Shoai M, Hardy J, Schott JM. Towards cascading genetic risk in Alzheimer's disease. Brain 2024; 147:2680-2690. [PMID: 38820112 PMCID: PMC11292901 DOI: 10.1093/brain/awae176] [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: 12/21/2023] [Revised: 04/22/2024] [Accepted: 05/06/2024] [Indexed: 06/02/2024] Open
Abstract
Alzheimer's disease typically progresses in stages, which have been defined by the presence of disease-specific biomarkers: amyloid (A), tau (T) and neurodegeneration (N). This progression of biomarkers has been condensed into the ATN framework, in which each of the biomarkers can be either positive (+) or negative (-). Over the past decades, genome-wide association studies have implicated ∼90 different loci involved with the development of late-onset Alzheimer's disease. Here, we investigate whether genetic risk for Alzheimer's disease contributes equally to the progression in different disease stages or whether it exhibits a stage-dependent effect. Amyloid (A) and tau (T) status was defined using a combination of available PET and CSF biomarkers in the Alzheimer's Disease Neuroimaging Initiative cohort. In 312 participants with biomarker-confirmed A-T- status, we used Cox proportional hazards models to estimate the contribution of APOE and polygenic risk scores (beyond APOE) to convert to A+T- status (65 conversions). Furthermore, we repeated the analysis in 290 participants with A+T- status and investigated the genetic contribution to conversion to A+T+ (45 conversions). Both survival analyses were adjusted for age, sex and years of education. For progression from A-T- to A+T-, APOE-e4 burden showed a significant effect [hazard ratio (HR) = 2.88; 95% confidence interval (CI): 1.70-4.89; P < 0.001], whereas polygenic risk did not (HR = 1.09; 95% CI: 0.84-1.42; P = 0.53). Conversely, for the transition from A+T- to A+T+, the contribution of APOE-e4 burden was reduced (HR = 1.62; 95% CI: 1.05-2.51; P = 0.031), whereas the polygenic risk showed an increased contribution (HR = 1.73; 95% CI: 1.27-2.36; P < 0.001). The marginal APOE effect was driven by e4 homozygotes (HR = 2.58; 95% CI: 1.05-6.35; P = 0.039) as opposed to e4 heterozygotes (HR = 1.74; 95% CI: 0.87-3.49; P = 0.12). The genetic risk for late-onset Alzheimer's disease unfolds in a disease stage-dependent fashion. A better understanding of the interplay between disease stage and genetic risk can lead to a more mechanistic understanding of the transition between ATN stages and a better understanding of the molecular processes leading to Alzheimer's disease, in addition to opening therapeutic windows for targeted interventions.
Collapse
Affiliation(s)
- Andre Altmann
- UCL Centre for Medical Image Computing, Department of Medical Physics and Biomedical Engineering, University College London, London, WC1E 6BT, UK
| | - Leon M Aksman
- Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Neil P Oxtoby
- UCL Centre for Medical Image Computing, Department of Computer Science, University College London, London, WC1E 6BT, UK
| | - Alexandra L Young
- UCL Centre for Medical Image Computing, Department of Computer Science, University College London, London, WC1E 6BT, UK
| | - Daniel C Alexander
- UCL Centre for Medical Image Computing, Department of Computer Science, University College London, London, WC1E 6BT, UK
| | - Frederik Barkhof
- UCL Centre for Medical Image Computing, Department of Medical Physics and Biomedical Engineering, University College London, London, WC1E 6BT, UK
- UCL Queen Square Institute of Neurology, University College London, London, WC1N 3BG, UK
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Center, Amsterdam, 1081 HV, The Netherlands
| | - Maryam Shoai
- UCL Queen Square Institute of Neurology, University College London, London, WC1N 3BG, UK
- UK Dementia Research Institute, University College London, London, WC1E 6BT, UK
| | - John Hardy
- UCL Queen Square Institute of Neurology, University College London, London, WC1N 3BG, UK
- UK Dementia Research Institute, University College London, London, WC1E 6BT, UK
| | - Jonathan M Schott
- UK Dementia Research Institute, University College London, London, WC1E 6BT, UK
- Dementia Research Centre, UCL Queen Square Institute of Neurology, University College London, London, WC1N 3AR, UK
| |
Collapse
|
24
|
Ge YJ, Fu Y, Gong W, Cheng W, Yu JT. Genetic architecture of brain morphology and overlap with neuropsychiatric traits. Trends Genet 2024; 40:706-717. [PMID: 38702264 DOI: 10.1016/j.tig.2024.04.005] [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: 02/12/2024] [Revised: 04/05/2024] [Accepted: 04/12/2024] [Indexed: 05/06/2024]
Abstract
Uncovering the genetic architectures of brain morphology offers valuable insights into brain development and disease. Genetic association studies of brain morphological phenotypes have discovered thousands of loci. However, interpretation of these loci presents a significant challenge. One potential solution is exploring the genetic overlap between brain morphology and disorders, which can improve our understanding of their complex relationships, ultimately aiding in clinical applications. In this review, we examine current evidence on the genetic associations between brain morphology and neuropsychiatric traits. We discuss the impact of these associations on the diagnosis, prediction, and treatment of neuropsychiatric diseases, along with suggestions for future research directions.
Collapse
Affiliation(s)
- Yi-Jun Ge
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yan Fu
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, 266071, China
| | - Weikang Gong
- School of Data Science, Fudan University, Shanghai, China; Centre for Functional MRI of the Brain (FMRIB), Nuffield Department of Clinical Neurosciences, Wellcome Centre for Integrative Neuroimaging, University of Oxford, Oxford, OX3 9DU, UK
| | - Wei Cheng
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China; Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China; Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Fudan University, Ministry of Education, Shanghai, China.
| | - Jin-Tai Yu
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China.
| |
Collapse
|
25
|
Fernandez MV, Liu M, Beric A, Johnson M, Cetin A, Patel M, Budde J, Kohlfeld P, Bergmann K, Lowery J, Flynn A, Brock W, Sanchez Montejo B, Gentsch J, Sykora N, Norton J, Gentsch J, Valdez O, Gorijala P, Sanford J, Sun Y, Wang C, Western D, Timsina J, Mangetti Goncalves T, Do AN, Sung YJ, Zhao G, Morris JC, Moulder K, Holtzman DM, Bateman RJ, Karch C, Hassenstab J, Xiong C, Schindler SE, Balls-Berry JJ, Benzinger TLS, Perrin RJ, Denny A, Snider BJ, Stark SL, Ibanez L, Cruchaga C. Genetic and multi-omic resources for Alzheimer disease and related dementia from the Knight Alzheimer Disease Research Center. Sci Data 2024; 11:768. [PMID: 38997326 PMCID: PMC11245521 DOI: 10.1038/s41597-024-03485-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 06/06/2024] [Indexed: 07/14/2024] Open
Abstract
The Knight-Alzheimer Disease Research Center (Knight-ADRC) at Washington University in St. Louis has pioneered and led worldwide seminal studies that have expanded our clinical, social, pathological, and molecular understanding of Alzheimer Disease. Over more than 40 years, research volunteers have been recruited to participate in cognitive, neuropsychologic, imaging, fluid biomarkers, genomic and multi-omic studies. Tissue and longitudinal data collected to foster, facilitate, and support research on dementia and aging. The Genetics and high throughput -omics core (GHTO) have collected of more than 26,000 biological samples from 6,625 Knight-ADRC participants. Samples available include longitudinal DNA, RNA, non-fasted plasma, cerebrospinal fluid pellets, and peripheral blood mononuclear cells. The GHTO has performed deep molecular profiling (genomic, transcriptomic, epigenomic, proteomic, and metabolomic) from large number of brain (n = 2,117), CSF (n = 2,012) and blood/plasma (n = 8,265) samples with the goal of identifying novel risk and protective variants, identify novel molecular biomarkers and causal and druggable targets. Overall, the resources available at GHTO support the increase of our understanding of Alzheimer Disease.
Collapse
Affiliation(s)
- Maria Victoria Fernandez
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Research Center and Memory Clinic, ACE Alzheimer Center, Barcelona, Spain
| | - Menghan Liu
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Aleksandra Beric
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Matt Johnson
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Arda Cetin
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Maulik Patel
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - John Budde
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Pat Kohlfeld
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Kristy Bergmann
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Joseph Lowery
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Allison Flynn
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - William Brock
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Brenda Sanchez Montejo
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Jen Gentsch
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Nicholas Sykora
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Joanne Norton
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Jen Gentsch
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Olga Valdez
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Priyanka Gorijala
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Jessie Sanford
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Yichen Sun
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Ciyang Wang
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Dan Western
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Jigyasha Timsina
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | | | - Anh N Do
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Yun Ju Sung
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Guoyan Zhao
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Pathology and Immunology Department, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - John C Morris
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Krista Moulder
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - David M Holtzman
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, USA
| | - Randall J Bateman
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, USA
- Dominantly Inherited Alzheimer Disease Network (DIAN), St. Louis, USA
| | - Celeste Karch
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, USA
- Dominantly Inherited Alzheimer Disease Network (DIAN), St. Louis, USA
| | - Jason Hassenstab
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Chengjie Xiong
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
- Dominantly Inherited Alzheimer Disease Network (DIAN), St. Louis, USA
| | - Suzanne E Schindler
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Joyce Joy Balls-Berry
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Tammie L S Benzinger
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
- Dominantly Inherited Alzheimer Disease Network (DIAN), St. Louis, USA
- Radiology Department, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Richard J Perrin
- Pathology and Immunology Department, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
- Dominantly Inherited Alzheimer Disease Network (DIAN), St. Louis, USA
| | - Andrea Denny
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - B Joy Snider
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, USA
| | - Susan L Stark
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
- Occupational Therapy, Neurology and Social Work, St. Louis, USA
| | - Laura Ibanez
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA.
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, 63110, USA.
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, 63110, USA.
- Dominantly Inherited Alzheimer Disease Network (DIAN), St. Louis, USA.
| | - Carlos Cruchaga
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA.
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, 63110, USA.
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA.
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, 63110, USA.
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA.
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, USA.
- Dominantly Inherited Alzheimer Disease Network (DIAN), St. Louis, USA.
| |
Collapse
|
26
|
Ando K, Küçükali F, Doeraene E, Nagaraj S, Antonelli EM, Thazin Htut M, Yilmaz Z, Kosa AC, Lopez-Guitierrez L, Quintanilla-Sánchez C, Aydin E, Ramos AR, Mansour S, Turbant S, Schurmans S, Sleegers K, Erneux C, Brion JP, Leroy K. Alteration of gene expression and protein solubility of the PI 5-phosphatase SHIP2 are correlated with Alzheimer's disease pathology progression. Acta Neuropathol 2024; 147:94. [PMID: 38833073 PMCID: PMC11150309 DOI: 10.1007/s00401-024-02745-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 05/21/2024] [Accepted: 05/21/2024] [Indexed: 06/06/2024]
Abstract
A recent large genome-wide association study has identified EGFR (encoding the epidermal growth factor EGFR) as a new genetic risk factor for late-onset AD. SHIP2, encoded by INPPL1, is taking part in the signalling and interactome of several growth factor receptors, such as the EGFR. While INPPL1 has been identified as one of the most significant genes whose RNA expression correlates with cognitive decline, the potential alteration of SHIP2 expression and localization during the progression of AD remains largely unknown. Here we report that gene expression of both EGFR and INPPL1 was upregulated in AD brains. SHIP2 immunoreactivity was predominantly detected in plaque-associated astrocytes and dystrophic neurites and its increase was correlated with amyloid load in the brain of human AD and of 5xFAD transgenic mouse model of AD. While mRNA of INPPL1 was increased in AD, SHIP2 protein undergoes a significant solubility change being depleted from the soluble fraction of AD brain homogenates and co-enriched with EGFR in the insoluble fraction. Using FRET-based flow cytometry biosensor assay for tau-tau interaction, overexpression of SHIP2 significantly increased the FRET signal while siRNA-mediated downexpression of SHIP2 significantly decreased FRET signal. Genetic association analyses suggest that some variants in INPPL1 locus are associated with the level of CSF pTau. Our data support the hypothesis that SHIP2 is an intermediate key player of EGFR and AD pathology linking amyloid and tau pathologies in human AD.
Collapse
Affiliation(s)
- Kunie Ando
- Alzheimer and Other Tauopathies Research Group, ULB Neuroscience Institute (UNI), ULB Center for Diabetes Research (UCDR), Faculty of Medicine, Université Libre de Bruxelles, 808 Route de Lennik, Bldg GE, 1070, Brussels, Belgium.
| | - Fahri Küçükali
- Complex Genetics of Alzheimer's Disease Group, VIB Center for Molecular Neurology, Antwerp, Belgium
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Emilie Doeraene
- Alzheimer and Other Tauopathies Research Group, ULB Neuroscience Institute (UNI), ULB Center for Diabetes Research (UCDR), Faculty of Medicine, Université Libre de Bruxelles, 808 Route de Lennik, Bldg GE, 1070, Brussels, Belgium
| | - Siranjeevi Nagaraj
- Alzheimer and Other Tauopathies Research Group, ULB Neuroscience Institute (UNI), ULB Center for Diabetes Research (UCDR), Faculty of Medicine, Université Libre de Bruxelles, 808 Route de Lennik, Bldg GE, 1070, Brussels, Belgium
| | - Eugenia Maria Antonelli
- Alzheimer and Other Tauopathies Research Group, ULB Neuroscience Institute (UNI), ULB Center for Diabetes Research (UCDR), Faculty of Medicine, Université Libre de Bruxelles, 808 Route de Lennik, Bldg GE, 1070, Brussels, Belgium
| | - May Thazin Htut
- Alzheimer and Other Tauopathies Research Group, ULB Neuroscience Institute (UNI), ULB Center for Diabetes Research (UCDR), Faculty of Medicine, Université Libre de Bruxelles, 808 Route de Lennik, Bldg GE, 1070, Brussels, Belgium
| | - Zehra Yilmaz
- Laboratory of Histology, Neuroanatomy and Neuropathology, Faculty of Medicine, Université Libre de Bruxelles, ULB Neuroscience Institute (UNI), 808 Route de Lennik, 1070, Brussels, Belgium
| | - Andreea-Claudia Kosa
- Alzheimer and Other Tauopathies Research Group, ULB Neuroscience Institute (UNI), ULB Center for Diabetes Research (UCDR), Faculty of Medicine, Université Libre de Bruxelles, 808 Route de Lennik, Bldg GE, 1070, Brussels, Belgium
| | - Lidia Lopez-Guitierrez
- Alzheimer and Other Tauopathies Research Group, ULB Neuroscience Institute (UNI), ULB Center for Diabetes Research (UCDR), Faculty of Medicine, Université Libre de Bruxelles, 808 Route de Lennik, Bldg GE, 1070, Brussels, Belgium
| | - Carolina Quintanilla-Sánchez
- Alzheimer and Other Tauopathies Research Group, ULB Neuroscience Institute (UNI), ULB Center for Diabetes Research (UCDR), Faculty of Medicine, Université Libre de Bruxelles, 808 Route de Lennik, Bldg GE, 1070, Brussels, Belgium
| | - Emmanuel Aydin
- Alzheimer and Other Tauopathies Research Group, ULB Neuroscience Institute (UNI), ULB Center for Diabetes Research (UCDR), Faculty of Medicine, Université Libre de Bruxelles, 808 Route de Lennik, Bldg GE, 1070, Brussels, Belgium
| | - Ana Raquel Ramos
- Institute of Interdisciplinary Research in Molecular Human Biology (IRIBHM), Université Libre de Bruxelles, 808 Route de Lennik, 1070, Brussels, Belgium
| | - Salwa Mansour
- Laboratory of Histology, Neuroanatomy and Neuropathology, Faculty of Medicine, Université Libre de Bruxelles, ULB Neuroscience Institute (UNI), 808 Route de Lennik, 1070, Brussels, Belgium
| | - Sabrina Turbant
- Biobanque Neuro-CEB, Hôpital de la Pitié-Salpétrière, Paris, France
- Plateforme de Ressources Biologiques (PRB), Hôpital de La Pitié-Salpêtrière, AP-HP, Paris, France
| | - Stéphane Schurmans
- Laboratory of Functional Genetics, GIGA Research Centre, University of Liège, Liège, Belgium
| | - Kristel Sleegers
- Complex Genetics of Alzheimer's Disease Group, VIB Center for Molecular Neurology, Antwerp, Belgium
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Christophe Erneux
- Institute of Interdisciplinary Research in Molecular Human Biology (IRIBHM), Université Libre de Bruxelles, 808 Route de Lennik, 1070, Brussels, Belgium
| | - Jean-Pierre Brion
- Alzheimer and Other Tauopathies Research Group, ULB Neuroscience Institute (UNI), ULB Center for Diabetes Research (UCDR), Faculty of Medicine, Université Libre de Bruxelles, 808 Route de Lennik, Bldg GE, 1070, Brussels, Belgium
| | - Karelle Leroy
- Alzheimer and Other Tauopathies Research Group, ULB Neuroscience Institute (UNI), ULB Center for Diabetes Research (UCDR), Faculty of Medicine, Université Libre de Bruxelles, 808 Route de Lennik, Bldg GE, 1070, Brussels, Belgium.
| |
Collapse
|
27
|
Santorelli FM, McLoughlin HS, Wolter JM, Galatolo D, Synofzik M, Mengel D, Opal P. Standards of Fluid Biomarker Collection and Pre-analytical Processes in Humans and Mice: Recommendations by the Ataxia Global Initiative Working Group on Biomarkers. CEREBELLUM (LONDON, ENGLAND) 2024; 23:881-886. [PMID: 37243885 DOI: 10.1007/s12311-023-01561-1] [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] [Accepted: 04/25/2023] [Indexed: 05/29/2023]
Abstract
The Ataxia Global Initiative (AGI) aims to serve as a platform to facilitate clinical trial readiness for the hereditary ataxias. Clinical trials for these diseases have been hampered by the lack of objective measures to study disease onset, progression, and treatment efficacy. While these issues are not unique to the genetic ataxias, the relative rarity of these diseases makes the need for such measures even more pressing to achieve statistical power in clinical trials. In this report, we have described the efforts of the AGI fluid biomarker working group (WG) in developing uniform protocols for biomarker sampling and storage, both for human and preclinical studies in mice. By reducing collection variability, we anticipate reduced noise in downstream biomarker analysis that will improve statistical power and minimize the necessary sample size. The emphasis has been on defining and standardizing the sampling and pre-analytical work-up of minimal set of biological samples, specifically blood plasma and serum, keeping in mind the need for harmonization of collection and storage that can be achieved with relatively limited cost and resources. An optional package is detailed for those centers that have the resources and commitment for additional biofluids/sample processing and storage. Finally, we have delineated similar standardized protocols for mice that will be important for preclinical studies in the field.
Collapse
Affiliation(s)
- Filippo M Santorelli
- Molecular Medicine and Neurogenetics, IRCCS Fondazione Stella Maris, Pisa, Italy.
| | | | - Justin M Wolter
- UNC Neuroscience Center, UNC School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, 27599, USA
| | - Daniele Galatolo
- Molecular Medicine and Neurogenetics, IRCCS Fondazione Stella Maris, Pisa, Italy
| | - Matthis Synofzik
- Division Translational Genomics of Neurodegenerative Diseases, Hertie Institute for Clinical Brain Research and Center of Neurology, University of Tübingen, Tübingen, Germany
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
| | - David Mengel
- Division Translational Genomics of Neurodegenerative Diseases, Hertie Institute for Clinical Brain Research and Center of Neurology, University of Tübingen, Tübingen, Germany.
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany.
| | - Puneet Opal
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
| |
Collapse
|
28
|
Yaghoobi A, Malekpour SA. Unraveling the genetic architecture of blood unfolded p-53 among non-demented elderlies: novel candidate genes for early Alzheimer's disease. BMC Genomics 2024; 25:440. [PMID: 38702606 PMCID: PMC11067101 DOI: 10.1186/s12864-024-10363-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 04/29/2024] [Indexed: 05/06/2024] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is a heritable neurodegenerative disease whose long asymptomatic phase makes the early diagnosis of it pivotal. Blood U-p53 has recently emerged as a superior predictive biomarker for AD in the early stages. We hypothesized that genetic variants associated with blood U-p53 could reveal novel loci and pathways involved in the early stages of AD. RESULTS We performed a blood U-p53 Genome-wide association study (GWAS) on 484 healthy and mild cognitively impaired subjects from the ADNI cohort using 612,843 Single nucleotide polymorphisms (SNPs). We performed a pathway analysis and prioritized candidate genes using an AD single-cell gene program. We fine-mapped the intergenic SNPs by leveraging a cell-type-specific enhancer-to-gene linking strategy using a brain single-cell multimodal dataset. We validated the candidate genes in an independent brain single-cell RNA-seq and the ADNI blood transcriptome datasets. The rs279686 between AASS and FEZF1 genes was the most significant SNP (p-value = 4.82 × 10-7). Suggestive pathways were related to the immune and nervous systems. Twenty-three candidate genes were prioritized at 27 suggestive loci. Fine-mapping of 5 intergenic loci yielded nine cell-specific candidate genes. Finally, 15 genes were validated in the independent single-cell RNA-seq dataset, and five were validated in the ADNI blood transcriptome dataset. CONCLUSIONS We underlined the importance of performing a GWAS on an early-stage biomarker of AD and leveraging functional omics datasets for pinpointing causal genes in AD. Our study prioritized nine genes (SORCS1, KIF5C, TMEFF2, TMEM63C, HLA-E, ATAT1, TUBB, ARID1B, and RUNX1) strongly implicated in the early stages of AD.
Collapse
Affiliation(s)
- Arash Yaghoobi
- School of Biological Sciences, Institute for Research in Fundamental Sciences (IPM), Tehran, 19395-5746, Iran
| | - Seyed Amir Malekpour
- School of Biological Sciences, Institute for Research in Fundamental Sciences (IPM), Tehran, 19395-5746, Iran.
| |
Collapse
|
29
|
Karlsson L, Vogel J, Arvidsson I, Åström K, Janelidze S, Blennow K, Palmqvist S, Stomrud E, Mattsson-Carlgren N, Hansson O. Cerebrospinal fluid reference proteins increase accuracy and interpretability of biomarkers for brain diseases. Nat Commun 2024; 15:3676. [PMID: 38693142 PMCID: PMC11063138 DOI: 10.1038/s41467-024-47971-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 04/17/2024] [Indexed: 05/03/2024] Open
Abstract
Cerebrospinal fluid (CSF) biomarkers reflect brain pathophysiology and are used extensively in translational research as well as in clinical practice for diagnosis of neurological diseases, e.g., Alzheimer's disease (AD). However, CSF biomarker concentrations may be influenced by non-disease related inter-individual variability. Here we use a data-driven approach to demonstrate the existence of inter-individual variability in mean standardized CSF protein levels. We show that these non-disease related differences cause many commonly reported CSF biomarkers to be highly correlated, thereby producing misleading results if not accounted for. To adjust for this inter-individual variability, we identified and evaluated high-performing reference proteins which improved the diagnostic accuracy of key CSF AD biomarkers. Our reference protein method attenuates the risk for false positive findings, and improves the sensitivity and specificity of CSF biomarkers, with broad implications for both research and clinical practice.
Collapse
Affiliation(s)
- Linda Karlsson
- Department of Clinical Sciences in Malmö, Clinical Memory Research Unit, Lund University, Lund, Sweden.
| | - Jacob Vogel
- Department of Clinical Sciences in Malmö, Clinical Memory Research Unit, Lund University, Lund, Sweden
- Department of Clinical Sciences, Clinical Memory Research Unit, SciLifeLab, Lund University, Lund, Sweden
| | - Ida Arvidsson
- Centre for Mathematical Sciences, Lund University, Lund, Sweden
| | - Kalle Åström
- Centre for Mathematical Sciences, Lund University, Lund, Sweden
| | - Shorena Janelidze
- Department of Clinical Sciences in Malmö, Clinical Memory Research Unit, Lund University, Lund, Sweden
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Sebastian Palmqvist
- Department of Clinical Sciences in Malmö, Clinical Memory Research Unit, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Erik Stomrud
- Department of Clinical Sciences in Malmö, Clinical Memory Research Unit, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Niklas Mattsson-Carlgren
- Department of Clinical Sciences in Malmö, Clinical Memory Research Unit, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Oskar Hansson
- Department of Clinical Sciences in Malmö, Clinical Memory Research Unit, Lund University, Lund, Sweden.
- Memory Clinic, Skåne University Hospital, Malmö, Sweden.
| |
Collapse
|
30
|
Bousiges O, Cretin B, Muller C, Botzung A, Sanna L, Anthony P, Philippi N, Demuynck C, Blanc F. Involvement of ApoE4 in dementia with Lewy bodies in the prodromal and demented stages: evaluation of the Strasbourg cohort. GeroScience 2024; 46:1527-1542. [PMID: 37653269 PMCID: PMC10828291 DOI: 10.1007/s11357-023-00883-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Accepted: 07/19/2023] [Indexed: 09/02/2023] Open
Abstract
ApoE4 as a risk factor for dementia with Lewy bodies (DLB) is still an issue. We sought to determine the involvement of ApoE4 according to different clinical parameters in our cohort of patients from Strasbourg, France. ApoE genotyping was performed on the AlphaLewyMA cohort. In this cohort, 197 patients were genotyped: 105 DLB patients, 37 Alzheimer's disease (AD) patients, 29 patients with AD/DLB comorbidity, and 26 control subjects (CS). The groups of patients were also classified according to the stage of evolution of the disease: prodromal or demented. We analyzed other parameters in relation to ApoE4 status, such as years of education (YOE) and Alzheimer CSF biomarkers. We observed a higher proportion of ApoE4 carriers in the AD (51.4%) and AD/DLB (72.4%) groups compared to the DLB (25.7%) and CS (11.5%) groups (p < 0.0001). We found a correlation between age at disease onset and YOE in the AD group (p = 0.039) but not in the DLB group (p = 0.056). Interestingly, in the DLB group, the subgroup of patients with high YOE (≥ 11) had significantly more patients with ApoE4 than the subgroup with low YOE (< 11). AD biomarkers did not seem to be impacted by the presence of ApoE4, except for Aβ42: DLB ApoE4-positive demented patients showed a more marked Aβ42 decrease. ApoE4 does not appear to be a risk factor for "pure" DLB patients. These results suggest a strong link between ApoE4 and amyloidopathy and consequently with AD. Trial registration: AlphaLewyMa, Identifier: NCT01876459, date of registration: June 12, 2013.
Collapse
Affiliation(s)
- Olivier Bousiges
- ICube Laboratory UMR 7357 and FMTS (Fédération de Médecine Translationnelle de Strasbourg), IMIS Team and IMAGeS Team, University of Strasbourg and CNRS, Strasbourg, France.
- Laboratory of Biochemistry and Molecular Biology, University Hospital of Strasbourg, Strasbourg, France.
| | - Benjamin Cretin
- ICube Laboratory UMR 7357 and FMTS (Fédération de Médecine Translationnelle de Strasbourg), IMIS Team and IMAGeS Team, University of Strasbourg and CNRS, Strasbourg, France
- CM2R (Research and Resources Memory Center), Geriatrics Department, Geriatric Day Hospital, Neurogeriatric Service, University Hospital of Strasbourg, Strasbourg, France
| | - Candice Muller
- CM2R (Research and Resources Memory Center), Geriatrics Department, Geriatric Day Hospital, Neurogeriatric Service, University Hospital of Strasbourg, Strasbourg, France
| | - Anne Botzung
- ICube Laboratory UMR 7357 and FMTS (Fédération de Médecine Translationnelle de Strasbourg), IMIS Team and IMAGeS Team, University of Strasbourg and CNRS, Strasbourg, France
- CM2R (Research and Resources Memory Center), Geriatrics Department, Geriatric Day Hospital, Neurogeriatric Service, University Hospital of Strasbourg, Strasbourg, France
| | - Lea Sanna
- CM2R (Research and Resources Memory Center), Geriatrics Department, Geriatric Day Hospital, Neurogeriatric Service, University Hospital of Strasbourg, Strasbourg, France
| | - Pierre Anthony
- CM2R, Neuropsychology Unit, Head and Neck Department, Neurology Department, University of Strasbourg, Strasbourg, France
- CM2R, Geriatrics Department and Neurology Department, Colmar General Hospital, Colmar, France
| | - Nathalie Philippi
- ICube Laboratory UMR 7357 and FMTS (Fédération de Médecine Translationnelle de Strasbourg), IMIS Team and IMAGeS Team, University of Strasbourg and CNRS, Strasbourg, France
- CM2R, Neuropsychology Unit, Head and Neck Department, Neurology Department, University of Strasbourg, Strasbourg, France
| | - Catherine Demuynck
- CM2R (Research and Resources Memory Center), Geriatrics Department, Geriatric Day Hospital, Neurogeriatric Service, University Hospital of Strasbourg, Strasbourg, France
| | - Frédéric Blanc
- ICube Laboratory UMR 7357 and FMTS (Fédération de Médecine Translationnelle de Strasbourg), IMIS Team and IMAGeS Team, University of Strasbourg and CNRS, Strasbourg, France
- CM2R (Research and Resources Memory Center), Geriatrics Department, Geriatric Day Hospital, Neurogeriatric Service, University Hospital of Strasbourg, Strasbourg, France
| |
Collapse
|
31
|
Beveridge J, Kaniecki E, Naidu A, Silverglate BD, Grossberg G. How promising are the latest monoclonal antibodies targeting amyloid-β for the treatment of early Alzheimer's disease? Expert Opin Emerg Drugs 2024; 29:35-43. [PMID: 38193477 DOI: 10.1080/14728214.2024.2304059] [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: 10/02/2023] [Accepted: 01/08/2024] [Indexed: 01/10/2024]
Abstract
INTRODUCTION Monoclonal antibodies targeting amyloid-β are the first disease-modifying treatments for Alzheimer disease to have received FDA-approval. There are three different drugs approved or pending FDA-approval: aducanumab, lecanemab, and donanemab. These three drugs are each in different stages of regulatory approval by the FDA. AREAS COVERED We discuss the development of these drugs, the data regarding their clinical efficacy, their dosing regimens, and side effects. In addition, we examine pragmatic issues with their potential implementation as common treatments to slow the rate of decline in Alzheimer disease, and what unanswered questions remain regarding this new class of drugs. EXPERT OPINION We conclude that these new monoclonal antibodies that target amyloid-β represent a genuine advance in the treatment of Alzheimer disease. However, questions remain regarding their clinical significance. Additionally, it is presently unclear which patients would most benefit from these expensive drugs given the risk of side effects and the logistical difficulties concerning administration and the determination of eligibility.
Collapse
Affiliation(s)
- Jordan Beveridge
- Saint Louis University School of Medicine, Department of Psychiatry and Behavioral Neuroscience, Division of Geriatric Psychiatry, St. Louis, MO, USA
| | - Eileen Kaniecki
- Saint Louis University School of Medicine, Department of Psychiatry and Behavioral Neuroscience, Division of Geriatric Psychiatry, St. Louis, MO, USA
| | - Aniketh Naidu
- Saint Louis University School of Medicine, Department of Psychiatry and Behavioral Neuroscience, Division of Geriatric Psychiatry, St. Louis, MO, USA
| | - Bret David Silverglate
- Saint Louis University School of Medicine, Department of Psychiatry and Behavioral Neuroscience, Division of Geriatric Psychiatry, St. Louis, MO, USA
| | - George Grossberg
- Saint Louis University School of Medicine, Department of Psychiatry and Behavioral Neuroscience, Division of Geriatric Psychiatry, St. Louis, MO, USA
| |
Collapse
|
32
|
Wang L, Nykänen NP, Western D, Gorijala P, Timsina J, Li F, Wang Z, Ali M, Yang C, Liu M, Brock W, Marquié M, Boada M, Alvarez I, Aguilar M, Pastor P, Ruiz A, Puerta R, Orellana A, Rutledge J, Oh H, Greicius MD, Le Guen Y, Perrin RJ, Wyss-Coray T, Jefferson A, Hohman TJ, Graff-Radford N, Mori H, Goate A, Levin J, Sung YJ, Cruchaga C. Proteo-genomics of soluble TREM2 in cerebrospinal fluid provides novel insights and identifies novel modulators for Alzheimer's disease. Mol Neurodegener 2024; 19:1. [PMID: 38172904 PMCID: PMC10763080 DOI: 10.1186/s13024-023-00687-4] [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: 06/08/2023] [Accepted: 11/28/2023] [Indexed: 01/05/2024] Open
Abstract
Triggering receptor expressed on myeloid cells 2 (TREM2) plays a critical role in microglial activation, survival, and apoptosis, as well as in Alzheimer's disease (AD) pathogenesis. We previously reported the MS4A locus as a key modulator for soluble TREM2 (sTREM2) in cerebrospinal fluid (CSF). To identify additional novel genetic modifiers of sTREM2, we performed the largest genome-wide association study (GWAS) and identified four loci for CSF sTREM2 in 3,350 individuals of European ancestry. Through multi-ethnic fine mapping, we identified two independent missense variants (p.M178V in MS4A4A and p.A112T in MS4A6A) that drive the association in MS4A locus and showed an epistatic effect for sTREM2 levels and AD risk. The novel TREM2 locus on chr 6 contains two rare missense variants (rs75932628 p.R47H, P=7.16×10-19; rs142232675 p.D87N, P=2.71×10-10) associated with sTREM2 and AD risk. The third novel locus in the TGFBR2 and RBMS3 gene region (rs73823326, P=3.86×10-9) included a regulatory variant with a microglia-specific chromatin loop for the promoter of TGFBR2. Using cell-based assays we demonstrate that overexpression and knock-down of TGFBR2, but not RBMS3, leads to significant changes of sTREM2. The last novel locus is located on the APOE region (rs11666329, P=2.52×10-8), but we demonstrated that this signal was independent of APOE genotype. This signal colocalized with cis-eQTL of NECTIN2 in the brain cortex and cis-pQTL of NECTIN2 in CSF. Overexpression of NECTIN2 led to an increase of sTREM2 supporting the genetic findings. To our knowledge, this is the largest study to date aimed at identifying genetic modifiers of CSF sTREM2. This study provided novel insights into the MS4A and TREM2 loci, two well-known AD risk genes, and identified TGFBR2 and NECTIN2 as additional modulators involved in TREM2 biology.
Collapse
Affiliation(s)
- Lihua Wang
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Niko-Petteri Nykänen
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Daniel Western
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Priyanka Gorijala
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Jigyasha Timsina
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Fuhai Li
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, USA
| | - Zhaohua Wang
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Muhammad Ali
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Chengran Yang
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Menghan Liu
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, USA
| | - William Brock
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Marta Marquié
- Networking Research Center on Neurodegenerative Disease (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
- Research Center and Memory Clinic, ACE Alzheimer Center Barcelona, Universitat Internacional de Catalunya, Barcelona, Spain
| | - Mercè Boada
- Networking Research Center on Neurodegenerative Disease (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
- Research Center and Memory Clinic, ACE Alzheimer Center Barcelona, Universitat Internacional de Catalunya, Barcelona, Spain
| | - Ignacio Alvarez
- Memory Disorders Unit, Department of Neurology, University Hospital Mutua Terrassa, Terrassa, Spain
| | - Miquel Aguilar
- Memory Disorders Unit, Department of Neurology, University Hospital Mutua Terrassa, Terrassa, Spain
| | - Pau Pastor
- Unit of Neurodegenerative diseases, Department of Neurology, University Hospital Germans Trias i Pujol and The Germans Trias i Pujol Research Institute (IGTP) Badalona, Barcelona, Spain
| | - Agustín Ruiz
- Networking Research Center on Neurodegenerative Disease (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
- Research Center and Memory Clinic, ACE Alzheimer Center Barcelona, Universitat Internacional de Catalunya, Barcelona, Spain
| | - Raquel Puerta
- Networking Research Center on Neurodegenerative Disease (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
- Research Center and Memory Clinic, ACE Alzheimer Center Barcelona, Universitat Internacional de Catalunya, Barcelona, Spain
| | - Adelina Orellana
- Networking Research Center on Neurodegenerative Disease (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
- Research Center and Memory Clinic, ACE Alzheimer Center Barcelona, Universitat Internacional de Catalunya, Barcelona, Spain
| | - Jarod Rutledge
- Wu-Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
| | - Hamilton Oh
- Wu-Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
| | | | - Yann Le Guen
- Wu-Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
| | - Richard J Perrin
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Tony Wyss-Coray
- Wu-Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
| | - Angela Jefferson
- Vanderbilt Memory & Alzheimer's Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Timothy J Hohman
- Vanderbilt Memory & Alzheimer's Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | | | | | - Alison Goate
- Department of Genetics & Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Johannes Levin
- Department of Neurology, University Hospital of Munich, Ludwig-Maximilians-Universität (LMU) Munich, Munich, Germany
| | - Yun Ju Sung
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, USA
- Division of Biostatistics, Washington University School of Medicine, BJC Institute of Health, 425 S. Euclid Ave, Box 8134, St. Louis, MO, 63110, USA
| | - Carlos Cruchaga
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA.
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, USA.
- Hope Center for Neurologic Diseases, Washington University, St. Louis, MO, USA.
| |
Collapse
|
33
|
Timsina J, Ali M, Do A, Wang L, Western D, Sung YJ, Cruchaga C. Harmonization of CSF and imaging biomarkers in Alzheimer's disease: Need and practical applications for genetics studies and preclinical classification. Neurobiol Dis 2024; 190:106373. [PMID: 38072165 DOI: 10.1016/j.nbd.2023.106373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 10/06/2023] [Accepted: 12/05/2023] [Indexed: 12/19/2023] Open
Abstract
In Alzheimer's disease (AD) research, cerebrospinal fluid (CSF) Amyloid beta (Aβ), Tau and pTau are the most accepted and well validated biomarkers. Several methods and platforms exist to measure those biomarkers, leading to challenges in combining data across studies. Thus, there is a need to identify methods that harmonize and standardize these values. We used a Z-score based approach to harmonize CSF and amyloid imaging data from multiple cohorts and compared GWAS results using this approach with currently accepted methods. We also used a generalized mixture model to calculate the threshold for biomarker-positivity. Based on our findings, our normalization approach performed as well as meta-analysis and did not lead to any spurious results. In terms of dichotomization, cutoffs calculated with this approach were very similar to those reported previously. These findings show that the Z-score based harmonization approach can be applied to heterogeneous platforms and provides biomarker cut-offs consistent with the classical approaches without requiring any additional data.
Collapse
Affiliation(s)
- Jigyasha Timsina
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110, USA; NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Muhammad Ali
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110, USA; NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Anh Do
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO 63110, USA; Division of Biology and Biomedical Sciences, Washington University School of Medicine, St. Louis, MO, USA
| | - Lihua Wang
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110, USA; NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Daniel Western
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110, USA; NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Yun Ju Sung
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110, USA; NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO 63110, USA; Division of Biostatistics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Carlos Cruchaga
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110, USA; NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO 63110, USA; Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
34
|
Ge YJ, Wu BS, Zhang Y, Chen SD, Zhang YR, Kang JJ, Deng YT, Ou YN, He XY, Zhao YL, Kuo K, Ma Q, Banaschewski T, Barker GJ, Bokde ALW, Desrivières S, Flor H, Grigis A, Garavan H, Gowland P, Heinz A, Brühl R, Martinot JL, Martinot MLP, Artiges E, Nees F, Orfanos DP, Lemaitre H, Paus T, Poustka L, Hohmann S, Millenet S, Fröhner JH, Smolka MN, Vaidya N, Walter H, Whelan R, Feng JF, Tan L, Dong Q, Schumann G, Cheng W, Yu JT. Genetic architectures of cerebral ventricles and their overlap with neuropsychiatric traits. Nat Hum Behav 2024; 8:164-180. [PMID: 37857874 DOI: 10.1038/s41562-023-01722-6] [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: 04/23/2023] [Accepted: 09/12/2023] [Indexed: 10/21/2023]
Abstract
The cerebral ventricles are recognized as windows into brain development and disease, yet their genetic architectures, underlying neural mechanisms and utility in maintaining brain health remain elusive. Here we aggregated genetic and neuroimaging data from 61,974 participants (age range, 9 to 98 years) in five cohorts to elucidate the genetic basis of ventricular morphology and examined their overlap with neuropsychiatric traits. Genome-wide association analysis in a discovery sample of 31,880 individuals identified 62 unique loci and 785 candidate genes associated with ventricular morphology. We replicated over 80% of loci in a well-matched cohort of lateral ventricular volume. Gene set analysis revealed enrichment of ventricular-trait-associated genes in biological processes and disease pathogenesis during both early brain development and degeneration. We explored the age-dependent genetic associations in cohorts of different age groups to investigate the possible roles of ventricular-trait-associated loci in neurodevelopmental and neurodegenerative processes. We describe the genetic overlap between ventricular and neuropsychiatric traits through comprehensive integrative approaches under correlative and causal assumptions. We propose the volume of the inferior lateral ventricles as a heritable endophenotype to predict the risk of Alzheimer's disease, which might be a consequence of prodromal Alzheimer's disease. Our study provides an advance in understanding the genetics of the cerebral ventricles and demonstrates the potential utility of ventricular measurements in tracking brain disorders and maintaining brain health across the lifespan.
Collapse
Affiliation(s)
- Yi-Jun Ge
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Bang-Sheng Wu
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yi Zhang
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Shi-Dong Chen
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ya-Ru Zhang
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ju-Jiao Kang
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China
| | - Yue-Ting Deng
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ya-Nan Ou
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Xiao-Yu He
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yong-Li Zhao
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Kevin Kuo
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Qing Ma
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China
| | - Tobias Banaschewski
- Department of Child and Adolescent Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Gareth J Barker
- Department of Neuroimaging, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Arun L W Bokde
- Discipline of Psychiatry, School of Medicine and Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland
| | - Sylvane Desrivières
- Centre for Population Neuroscience and Precision Medicine, Institute of Psychiatry, Psychology & Neuroscience, SGDP Centre, King's College London, London, UK
| | - Herta Flor
- Institute of Cognitive and Clinical Neuroscience, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Department of Psychology, School of Social Sciences, University of Mannheim, Mannheim, Germany
| | - Antoine Grigis
- NeuroSpin, CEA, Université Paris-Saclay, Gif-sur-Yvette, France
| | - Hugh Garavan
- Departments of Psychiatry and Psychology, University of Vermont, Burlington, VT, USA
| | - Penny Gowland
- Sir Peter Mansfield Imaging Centre, School of Physics and Astronomy, University of Nottingham, Nottingham, UK
| | - Andreas Heinz
- Department of Psychiatry and Psychotherapy CCM, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
| | - Rüdiger Brühl
- Physikalisch-Technische Bundesanstalt, Braunschweig and Berlin, Germany
| | - Jean-Luc Martinot
- Institut National de la Santé et de la Recherche Médicale, INSERM U 1299 'Trajectoires développementales & psychiatrie', University Paris-Saclay, CNRS; Ecole Normale Supérieure Paris-Saclay, Centre Borelli, Gif-sur-Yvette, France
| | - Marie-Laure Paillère Martinot
- Institut National de la Santé et de la Recherche Médicale, INSERM U 1299 'Trajectoires développementales & psychiatrie', University Paris-Saclay, CNRS; Ecole Normale Supérieure Paris-Saclay, Centre Borelli, Gif-sur-Yvette, France
- AP-HP, Sorbonne University, Department of Child and Adolescent Psychiatry, Pitié-Salpêtrière Hospital, Paris, France
| | - Eric Artiges
- Institut National de la Santé et de la Recherche Médicale, INSERM U 1299 'Trajectoires développementales & psychiatrie', University Paris-Saclay, CNRS; Ecole Normale Supérieure Paris-Saclay, Centre Borelli, Gif-sur-Yvette, France
- Psychiatry Department, EPS Barthélémy Durand, Etampes, France
| | - Frauke Nees
- Department of Child and Adolescent Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Institute of Cognitive and Clinical Neuroscience, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Institute of Medical Psychology and Medical Sociology, University Medical Center Schleswig Holstein, Kiel University, Kiel, Germany
| | | | - Herve Lemaitre
- NeuroSpin, CEA, Université Paris-Saclay, Gif-sur-Yvette, France
- Institut des Maladies Neurodégénératives, UMR 5293, CNRS, CEA, Université de Bordeaux, Bordeaux, France
| | - Tomáš Paus
- Departments of Psychiatry and Neuroscience, Faculty of Medicine and Centre Hospitalier Universitaire Sainte-Justine, University of Montreal, Montreal, Quebec, Canada
- Departments of Psychiatry and Psychology, University of Toronto, Toronto, Ontario, Canada
| | - Luise Poustka
- Department of Child and Adolescent Psychiatry and Psychotherapy, University Medical Centre Göttingen, Göttingen, Germany
| | - Sarah Hohmann
- Department of Child and Adolescent Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Sabina Millenet
- Department of Child and Adolescent Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Juliane H Fröhner
- Department of Psychiatry and Neuroimaging Center, Technische Universität Dresden, Dresden, Germany
| | - Michael N Smolka
- Department of Psychiatry and Neuroimaging Center, Technische Universität Dresden, Dresden, Germany
| | - Nilakshi Vaidya
- Centre for Population Neuroscience and Stratified Medicine, Department of Psychiatry and Neuroscience, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Henrik Walter
- Department of Psychiatry and Psychotherapy CCM, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
| | - Robert Whelan
- School of Psychology and Global Brain Health Institute, Trinity College Dublin, Dublin, Ireland
| | - Jian-Feng Feng
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China
- Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence (Fudan University), Ministry of Education, Beijing, China
- Fudan ISTBI-ZJNU Algorithm Centre for Brain-Inspired Intelligence, Zhejiang Normal University, Jinhua, China
- MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
- Zhangjiang Fudan International Innovation Center, Shanghai, China
| | - Lan Tan
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Qiang Dong
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Gunter Schumann
- Centre for Population Neuroscience and Stratified Medicine, Department of Psychiatry and Neuroscience, Charité Universitätsmedizin Berlin, Berlin, Germany
- Centre for Population Neuroscience and Precision Medicine, Institute for Science and Technology of Brain-Inspired Intelligence, Fudan University, Shanghai, China
| | - Wei Cheng
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China.
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China.
- Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence (Fudan University), Ministry of Education, Beijing, China.
- Fudan ISTBI-ZJNU Algorithm Centre for Brain-Inspired Intelligence, Zhejiang Normal University, Jinhua, China.
- Shanghai Medical College and Zhongshan Hospital Immunotherapy Technology Transfer 79 Center, Shanghai, China.
| | - Jin-Tai Yu
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China.
| |
Collapse
|
35
|
Qian XH, Ding GY, Chen SY, Liu XL, Zhang M, Tang HD. Blood Cathepsins on the Risk of Alzheimer's Disease and Related Pathological Biomarkers: Results from Observational Cohort and Mendelian Randomization Study. J Prev Alzheimers Dis 2024; 11:1834-1842. [PMID: 39559895 PMCID: PMC11573867 DOI: 10.14283/jpad.2024.107] [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: 04/30/2024] [Accepted: 05/29/2024] [Indexed: 11/20/2024]
Abstract
BACKGROUND Alzheimer's disease (AD), the main type of dementia, involves in complex pathophysiological processes, including abnormal lysosomes function. Cathepsins are the predominant proteases responsible for the degradation of diverse substrates in the endo-lysosomal system. However, there was still a lack of systematic study on the causal association between cathepsins and AD. METHODS This study utilized Mendelian randomization (MR) to investigate the association between blood cathepsins and the risk of AD, as well as the level of amyloid-β (Aβ) and p-Tau in cerebrospinal fluid. Furthermore, an independent dataset was employed to corroborate the above result. Importantly, this study incorporated the Alzheimer's disease Immunization and Microbiota Initiative study Cohort to further validate the alteration of blood cathepsins expression level and examine its correlation with cognitive level and plasma AD-related pathological markers. RESULTS Using MR method, we observed that high level of cathepsin L (CTSL) was associated with a lower risk of AD in both training and validation data. In observational cohort, we found there was decreased blood CTSL expression level in Aβ+ cognitive impaired (CI) group, compared with Aβ- cognitive unimpaired (CU) group. Correlation analysis revealed that blood CTSL expression level was negatively correlated with Mini-Mental State Examination (MMSE) and Montreal Cognitive Assessment (MoCA) score, plasma Aβ42 and Aβ42/40 level in Aβ+ CI group. Mediation analysis showed that plasma Aβ42/40 level was the key mediator in the association between blood CTSL and MMSE score in Aβ+ CI participants. CONCLUSION This study revealed that blood CTSL was an important factor affecting the risk of AD, and it affected the cognitive level of AD patients through plasma Aβ42/40 level.
Collapse
Affiliation(s)
- X.-H. Qian
- Department of Geriatrics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Medical Center on Aging of Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - G.-Y. Ding
- Department of Neurology, Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - S.-Y. Chen
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiao-li Liu
- Department of Neurology, Shanghai Fengxian District Central Hospital, Shanghai University of Medicine & Health Science Affiliated Sixth People’s Hospital South Campus, Shanghai, China
| | - Miao Zhang
- Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hui-dong Tang
- Department of Geriatrics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Medical Center on Aging of Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
36
|
Duchateau L, Küҫükali F, De Roeck A, Wittens MMJ, Temmerman J, Weets I, Timmers M, Engelborghs S, Bjerke M, Sleegers K. CSF biomarker analysis of ABCA7 mutation carriers suggests altered APP processing and reduced inflammatory response. Alzheimers Res Ther 2023; 15:195. [PMID: 37946268 PMCID: PMC10634183 DOI: 10.1186/s13195-023-01338-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 10/24/2023] [Indexed: 11/12/2023]
Abstract
BACKGROUND The Alzheimer's disease (AD) risk gene ABCA7 has suggested functions in lipid metabolism and the immune system. Rare premature termination codon (PTC) mutations and an expansion of a variable number of tandem repeats (VNTR) polymorphism in the gene, both likely cause a lower ABCA7 expression and hereby increased risk for AD. However, the exact mechanism of action remains unclear. By studying CSF biomarkers reflecting different types of AD-related pathological processes, we aim to get a better insight in those processes and establish a biomarker profile of mutation carriers. METHODS The study population consisted of 229 AD patients for whom CSF was available and ABCA7 sequencing and VNTR genotyping had been performed. This included 28 PTC mutation and 16 pathogenic expansion carriers. CSF levels of Aβ1-42, Aβ1-40, P-tau181, T-tau, sAPPα, sAPPβ, YKL-40, and hFABP were determined using ELISA and Meso Scale Discovery assays. We compared differences in levels of these biomarkers and the Aβ ratio between AD patients with or without an ABCA7 PTC mutation or expansion using linear regression on INT-transformed data with APOE-status, age and sex as covariates. RESULTS Carriers of ABCA7 expansion mutations had significantly lower Aβ1-42 levels (P = 0.022) compared with non-carrier patients. The effect of the presence of ABCA7 mutations on CSF levels was especially pronounced in APOE ε4-negative carriers. In addition, VNTR expansion carriers had reduced Aβ1-40 (P = 0.023), sAPPα (P = 0.047), sAPPβ (P = 0.016), and YKL-40 (P = 0.0036) levels. CONCLUSIONS Our results are suggestive for an effect on APP processing by repeat expansions given the changes in the amyloid-related CSF biomarkers that were found in carriers. The decrease in YKL-40 levels in expansion carriers moreover suggests that these patients potentially have a reduced inflammatory response to AD damage. Moreover, our findings suggest the existence of a mechanism, independent of lowered expression, affecting neuropathology in expansion carriers.
Collapse
Affiliation(s)
- Lena Duchateau
- Complex Genetics of Alzheimer's Disease Group, VIB-UAntwerp Center for Molecular Neurology, VIB, Building V, Universiteitsplein 1, Wilrijk, Antwerp, B-2610, Belgium
- Department of Biomedical Sciences, University of Antwerp, Universiteitsplein 1, Wilrijk, Antwerp, 2610, Belgium
| | - Fahri Küҫükali
- Complex Genetics of Alzheimer's Disease Group, VIB-UAntwerp Center for Molecular Neurology, VIB, Building V, Universiteitsplein 1, Wilrijk, Antwerp, B-2610, Belgium
- Department of Biomedical Sciences, University of Antwerp, Universiteitsplein 1, Wilrijk, Antwerp, 2610, Belgium
| | - Arne De Roeck
- Complex Genetics of Alzheimer's Disease Group, VIB-UAntwerp Center for Molecular Neurology, VIB, Building V, Universiteitsplein 1, Wilrijk, Antwerp, B-2610, Belgium
- Department of Biomedical Sciences, University of Antwerp, Universiteitsplein 1, Wilrijk, Antwerp, 2610, Belgium
- Present Address: Argenx, Ghent, Belgium
| | - Mandy M J Wittens
- Department of Biomedical Sciences, University of Antwerp, Universiteitsplein 1, Wilrijk, Antwerp, 2610, Belgium
- Clinical Neurochemistry Laboratory, Department of Clinical Biology, University Hospital Brussels, Generaal Jacqueslaan 137, Elsene, Brussels, 1050, Belgium
| | - Joke Temmerman
- Department of Biomedical Sciences, University of Antwerp, Universiteitsplein 1, Wilrijk, Antwerp, 2610, Belgium
- Neuroprotection & Neuromodulation (NEUR) Research Group, Center for Neurosciences (C4N), Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, Jette, Brussels, 1090, Belgium
| | - Ilse Weets
- Clinical Neurochemistry Laboratory, Department of Clinical Biology, University Hospital Brussels, Generaal Jacqueslaan 137, Elsene, Brussels, 1050, Belgium
- Experimental Pharmacology (EFAR) Research Group, Center for Neurosciences (C4N), Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, Brussels, 1090, Belgium
| | - Maarten Timmers
- Reference Center for Biological Markers of Dementia, Department of Biomedical Sciences, University of Antwerp, Universiteitsplein 1, Wilrijk, Antwerp, 2610, Belgium
- Janssen Research and Development, a Division of Janssen Pharmaceutica NV, Turnhoutseweg 30, Beerse, 2340, Belgium
| | - Sebastiaan Engelborghs
- Neuroprotection & Neuromodulation (NEUR) Research Group, Center for Neurosciences (C4N), Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, Jette, Brussels, 1090, Belgium
- Reference Center for Biological Markers of Dementia, Department of Biomedical Sciences, University of Antwerp, Universiteitsplein 1, Wilrijk, Antwerp, 2610, Belgium
- Department of Neurology and Bru-BRAIN, University Hospital Brussels, Generaal Jacqueslaan 137, Elsene, Brussels, 1050, Belgium
| | - Maria Bjerke
- Department of Biomedical Sciences, University of Antwerp, Universiteitsplein 1, Wilrijk, Antwerp, 2610, Belgium
- Clinical Neurochemistry Laboratory, Department of Clinical Biology, University Hospital Brussels, Generaal Jacqueslaan 137, Elsene, Brussels, 1050, Belgium
- Neuroprotection & Neuromodulation (NEUR) Research Group, Center for Neurosciences (C4N), Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, Jette, Brussels, 1090, Belgium
| | - Kristel Sleegers
- Complex Genetics of Alzheimer's Disease Group, VIB-UAntwerp Center for Molecular Neurology, VIB, Building V, Universiteitsplein 1, Wilrijk, Antwerp, B-2610, Belgium.
- Department of Biomedical Sciences, University of Antwerp, Universiteitsplein 1, Wilrijk, Antwerp, 2610, Belgium.
| |
Collapse
|
37
|
Su WM, Gu XJ, Dou M, Duan QQ, Jiang Z, Yin KF, Cai WC, Cao B, Wang Y, Chen YP. Systematic druggable genome-wide Mendelian randomisation identifies therapeutic targets for Alzheimer's disease. J Neurol Neurosurg Psychiatry 2023; 94:954-961. [PMID: 37349091 PMCID: PMC10579488 DOI: 10.1136/jnnp-2023-331142] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 06/05/2023] [Indexed: 06/24/2023]
Abstract
BACKGROUND Alzheimer's disease (AD) is the leading cause of dementia. Currently, there are no effective disease-modifying treatments for AD. Mendelian randomisation (MR) has been widely used to repurpose licensed drugs and discover novel therapeutic targets. Thus, we aimed to identify novel therapeutic targets for AD and analyse their pathophysiological mechanisms and potential side effects. METHODS A two-sample MR integrating the identified druggable genes was performed to estimate the causal effects of blood and brain druggable expression quantitative trait loci (eQTLs) on AD. A repeat study was conducted using different blood and brain eQTL data sources to validate the identified genes. Using AD markers with available genome-wide association studies data, we evaluated the causal relationship between established AD markers to explore possible mechanisms. Finally, the potential side effects of the druggable genes for AD treatment were assessed using a phenome-wide MR. RESULTS Overall, 5883 unique druggable genes were aggregated; 33 unique potential druggable genes for AD were identified in at least one dataset (brain or blood), and 5 were validated in a different dataset. Among them, three prior druggable genes (epoxide hydrolase 2 (EPHX2), SERPINB1 and SIGLEC11) reached significant levels in both blood and brain tissues. EPHX2 may mediate the pathogenesis of AD by affecting the entire hippocampal volume. Further phenome-wide MR analysis revealed no potential side effects of treatments targeting EPHX2, SERPINB1 or SIGLEC11. CONCLUSIONS This study provides genetic evidence supporting the potential therapeutic benefits of targeting the three druggable genes for AD treatment, which will be useful for prioritising AD drug development.
Collapse
Affiliation(s)
- Wei-Ming Su
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Institute of Brain Science and Brain-inspired Technology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xiao-Jing Gu
- Department of Mental Health Center, West China Hospital, Sichuan University, Chengdu, China
| | - Meng Dou
- Chengdu Computer Application Institute, Chinese Academy of Sciences, Chengdu, China
| | - Qing-Qing Duan
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Institute of Brain Science and Brain-inspired Technology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zheng Jiang
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Institute of Brain Science and Brain-inspired Technology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Kang-Fu Yin
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Institute of Brain Science and Brain-inspired Technology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Wei-Chen Cai
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Institute of Brain Science and Brain-inspired Technology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Bei Cao
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Institute of Brain Science and Brain-inspired Technology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yi Wang
- Department of Pathophysiology, West China College of Basic medical sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Yong-Ping Chen
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Institute of Brain Science and Brain-inspired Technology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
38
|
Chen Q, Aguirre L, Zhao H, Borrego F, de Rojas I, Su L, Li PP, Zhang B, Kokovay E, Lechleiter JD, Göring HH, De Jager PL, Kleinman JE, Hyde TM, Ruiz A, Weinberger DR, Seshadri S, Ma L. Identification of a specific APOE transcript and functional elements associated with Alzheimer's disease. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.10.30.23297431. [PMID: 37961425 PMCID: PMC10635228 DOI: 10.1101/2023.10.30.23297431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
INTRODUCTION The APOE gene is the strongest genetic risk factor for late-onset Alzheimer's Disease (LOAD). However, the gene regulatory mechanisms at this locus have not been fully characterized. METHODS To identify novel AD-linked functional elements within the APOE locus, we integrated SNP variants with RNA-seq, DNA methylation, and ChIP-seq data from human postmortem brains. RESULTS We identified an AD-linked APOE transcript (jxn1.2.2) observed in the dorsolateral prefrontal cortex (DLPFC). The APOE jxn1.2.2 transcript is associated with brain neuropathological features in DLPFC. We prioritized an independent functional SNP, rs157580, significantly associated with jxn1.2.2 transcript abundance and DNA methylation levels. rs157580 is located within active chromatin regions and predicted to affect brain-related transcriptional factors binding affinity. rs157580 shared the effects on the jxn1.2.2 transcript between European and African ethnic groups. DISCUSSION The novel APOE functional elements provide potential therapeutic targets with mechanistic insight into the disease's etiology.
Collapse
|
39
|
Logue MW, Dasgupta S, Farrer LA. Genetics of Alzheimer's Disease in the African American Population. J Clin Med 2023; 12:5189. [PMID: 37629231 PMCID: PMC10455208 DOI: 10.3390/jcm12165189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 08/02/2023] [Accepted: 08/06/2023] [Indexed: 08/27/2023] Open
Abstract
Black/African American (AA) individuals have a higher risk of Alzheimer's disease (AD) than White non-Hispanic persons of European ancestry (EUR) for reasons that may include economic disparities, cardiovascular health, quality of education, and biases in the methods used to diagnose AD. AD is also heritable, and some of the differences in risk may be due to genetics. Many AD-associated variants have been identified by candidate gene studies, genome-wide association studies (GWAS), and genome-sequencing studies. However, most of these studies have been performed using EUR cohorts. In this paper, we review the genetics of AD and AD-related traits in AA individuals. Importantly, studies of genetic risk factors in AA cohorts can elucidate the molecular mechanisms underlying AD risk in AA and other populations. In fact, such studies are essential to enable reliable precision medicine approaches in persons with considerable African ancestry. Furthermore, genetic studies of AA cohorts allow exploration of the ways the impact of genes can vary by ancestry, culture, and economic and environmental disparities. They have yielded important gains in our knowledge of AD genetics, and increasing AA individual representation within genetic studies should remain a priority for inclusive genetic study design.
Collapse
Affiliation(s)
- Mark W. Logue
- National Center for PTSD, Behavioral Sciences Division, VA Boston Healthcare System, Boston, MA 02130, USA;
- Department of Psychiatry, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
- Department of Medicine (Biomedical Genetics), Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA;
- Department of Biostatistics, Boston University School of Public Health, Boston, MA 02118, USA
| | - Shoumita Dasgupta
- Department of Medicine (Biomedical Genetics), Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA;
- Department of Medical Sciences and Education, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
| | - Lindsay A. Farrer
- Department of Medicine (Biomedical Genetics), Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA;
- Department of Biostatistics, Boston University School of Public Health, Boston, MA 02118, USA
- Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
- Department of Ophthalmology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
- Department of Epidemiology, Boston University School of Public Health, Boston, MA 02118, USA
- Alzheimer’s Disease Research Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
| |
Collapse
|
40
|
Fang Y, Si X, Wang J, Wang Z, Chen Y, Liu Y, Yan Y, Tian J, Zhang B, Pu J. Alzheimer Disease and Epilepsy: A Mendelian Randomization Study. Neurology 2023; 101:e399-e409. [PMID: 37225432 PMCID: PMC10435057 DOI: 10.1212/wnl.0000000000207423] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 04/03/2023] [Indexed: 05/26/2023] Open
Abstract
BACKGROUND AND OBJECTIVES Observational studies suggested a bidirectional relationship between Alzheimer disease (AD) and epilepsies. However, it remains debated whether and in which direction a causal association exists. This study aims to explore the relationship between genetic predisposition to AD, CSF biomarkers of AD (β-amyloid [Aβ] 42 and phosphorylated tau [pTau]), and epilepsies with 2-sample, bidirectional Mendelian randomization (MR) method. METHODS Genetic instruments were obtained from large-scale genome-wide meta-analysis of AD (Ncase/proxy = 111,326, Ncontrol = 677,663), CSF biomarkers of AD (Aβ42 and pTau, N = 13,116), and epilepsy (Ncase = 15,212, Ncontrol = 29,677) of European ancestry. Epilepsy phenotypes included all epilepsy, generalized epilepsy, focal epilepsy, childhood absence epilepsy, juvenile absence epilepsy, juvenile myoclonic epilepsy, generalized epilepsy with tonic-clonic seizures, focal epilepsy with hippocampal sclerosis (focal HS), and lesion-negative focal epilepsy. Main analyses were performed using generalized summary data-based MR. Sensitivity analyses included inverse variance weighted, MR pleiotropy residual sum and outlier, MR-Egger, weighted mode, and weighted median. RESULTS For forward analysis, genetic predisposition to AD was associated with an increased risk of generalized epilepsy (odds ratio [OR] 1.053, 95% CI 1.002-1.105, p = 0.038) and focal HS (OR 1.013, 95% CI 1.004-1.022, p = 0.004). These associations were consistent across sensitivity analyses and replicated using a separate set of genetic instruments from another AD genome-wide association study. For reverse analysis, there was a suggestive effect of focal HS on AD (OR 3.994, 95% CI 1.172-13.613, p = 0.027). In addition, genetically predicted lower CSF Aβ42 was associated with an increased risk of generalized epilepsy (β = 0.090, 95% CI 0.022-0.158, p = 0.010). DISCUSSION This MR study supports a causal link between AD, amyloid pathology, and generalized epilepsy. This study also indicates a close association between AD and focal HS. More effort should be made to screen seizure in AD, unravel its clinical implications, and explore its role as a putative modifiable risk factor.
Collapse
Affiliation(s)
- Yi Fang
- From the Department of Neurology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaoli Si
- From the Department of Neurology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jiali Wang
- From the Department of Neurology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhiyun Wang
- From the Department of Neurology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ying Chen
- From the Department of Neurology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yi Liu
- From the Department of Neurology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yaping Yan
- From the Department of Neurology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jun Tian
- From the Department of Neurology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Baorong Zhang
- From the Department of Neurology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jiali Pu
- From the Department of Neurology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
41
|
Bradley J, Gorijala P, Schindler SE, Sung YJ, Ances B, Fernandez MV, Cruchaga C. Genetic architecture of plasma Alzheimer disease biomarkers. Hum Mol Genet 2023; 32:2532-2543. [PMID: 37208024 PMCID: PMC10360384 DOI: 10.1093/hmg/ddad087] [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: 12/23/2022] [Revised: 04/29/2023] [Accepted: 05/16/2023] [Indexed: 05/21/2023] Open
Abstract
Genome-wide association studies (GWAS) of cerebrospinal fluid (CSF) Alzheimer's Disease (AD) biomarker levels have identified novel genes implicated in disease risk, onset and progression. However, lumbar punctures have limited availability and may be perceived as invasive. Blood collection is readily available and well accepted, but it is not clear whether plasma biomarkers will be informative for genetic studies. Here we perform genetic analyses on concentrations of plasma amyloid-β peptides Aβ40 (n = 1,467) and Aβ42 (n = 1,484), Aβ42/40 (n = 1467) total tau (n = 504), tau phosphorylated (p-tau181; n = 1079) and neurofilament light (NfL; n = 2,058). GWAS and gene-based analysis was used to identify single variant and genes associated with plasma levels. Finally, polygenic risk score and summary statistics were used to investigate overlapping genetic architecture between plasma biomarkers, CSF biomarkers and AD risk. We found a total of six genome-wide significant signals. APOE was associated with plasma Aβ42, Aβ42/40, tau, p-tau181 and NfL. We proposed 10 candidate functional genes on the basis of 12 single nucleotide polymorphism-biomarker pairs and brain differential gene expression analysis. We found a significant genetic overlap between CSF and plasma biomarkers. We also demonstrate that it is possible to improve the specificity and sensitivity of these biomarkers, when genetic variants regulating protein levels are included in the model. This current study using plasma biomarker levels as quantitative traits can be critical to identification of novel genes that impact AD and more accurate interpretation of plasma biomarker levels.
Collapse
Affiliation(s)
- Joseph Bradley
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO 63110, USA
- Hope Center for Neurologic Diseases, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Priyanka Gorijala
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Suzanne E Schindler
- Hope Center for Neurologic Diseases, Washington University in St. Louis, St. Louis, MO 63110, USA
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Yun J Sung
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO 63110, USA
- Hope Center for Neurologic Diseases, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Beau Ances
- Hope Center for Neurologic Diseases, Washington University in St. Louis, St. Louis, MO 63110, USA
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Maria V Fernandez
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Carlos Cruchaga
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO 63110, USA
- Hope Center for Neurologic Diseases, Washington University in St. Louis, St. Louis, MO 63110, USA
| |
Collapse
|
42
|
Lambert JC, Ramirez A, Grenier-Boley B, Bellenguez C. Step by step: towards a better understanding of the genetic architecture of Alzheimer's disease. Mol Psychiatry 2023; 28:2716-2727. [PMID: 37131074 PMCID: PMC10615767 DOI: 10.1038/s41380-023-02076-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 04/12/2023] [Accepted: 04/14/2023] [Indexed: 05/04/2023]
Abstract
Alzheimer's disease (AD) is considered to have a large genetic component. Our knowledge of this component has progressed over the last 10 years, thanks notably to the advent of genome-wide association studies and the establishment of large consortia that make it possible to analyze hundreds of thousands of cases and controls. The characterization of dozens of chromosomal regions associated with the risk of developing AD and (in some loci) the causal genes responsible for the observed disease signal has confirmed the involvement of major pathophysiological pathways (such as amyloid precursor protein metabolism) and opened up new perspectives (such as the central role of microglia and inflammation). Furthermore, large-scale sequencing projects are starting to reveal the major impact of rare variants - even in genes like APOE - on the AD risk. This increasingly comprehensive knowledge is now being disseminated through translational research; in particular, the development of genetic risk/polygenic risk scores is helping to identify the subpopulations more at risk or less at risk of developing AD. Although it is difficult to assess the efforts still needed to comprehensively characterize the genetic component of AD, several lines of research can be improved or initiated. Ultimately, genetics (in combination with other biomarkers) might help to redefine the boundaries and relationships between various neurodegenerative diseases.
Collapse
Affiliation(s)
- Jean-Charles Lambert
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE Facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, Lille, France.
| | - Alfredo Ramirez
- Division of Neurogenetics and Molecular Psychiatry, Department of Psychiatry and Psychotherapy, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Department of Neurodegenerative diseases and Geriatric Psychiatry, University Hospital Bonn, Medical Faculty, Bonn, Germany
- Department of Psychiatry & Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, San Antonio, TX, USA
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Cluster of Excellence Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Benjamin Grenier-Boley
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE Facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, Lille, France
| | - Céline Bellenguez
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE Facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, Lille, France
| |
Collapse
|
43
|
Vanninen A, Lukkarinen H, Kokkola T, Koivisto AM, Kokki M, Musialowicz T, Hiltunen M, Zetterberg H, Leinonen V, Herukka SK, Rauramaa T. Cerebrospinal Fluid Diagnostics of Alzheimer's Disease in Patients with Idiopathic Normal Pressure Hydrocephalus. J Alzheimers Dis 2023:JAD230144. [PMID: 37334597 PMCID: PMC10357203 DOI: 10.3233/jad-230144] [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: 06/20/2023]
Abstract
BACKGROUND Alzheimer's disease (AD) is the most common cause of dementia worldwide and a frequent comorbidity in idiopathic normal pressure hydrocephalus (iNPH). The presence of AD pathology is associated with worse outcomes after a shunt procedure in iNPH. Preoperative diagnosis of AD is challenging in patients with iNPH, which involves reduced concentrations of the cerebrospinal fluid (CSF) AD biomarkers. OBJECTIVE Our aim was to estimate the effect size of iNPH as a factor in CSF levels of AD biomarkers and to test if correction could be used to improve diagnostic value. METHODS Our cohort included 222 iNPH patients with data in the Kuopio NPH registry and brain biopsy and CSF samples available. We divided the patients into groups according to AD pathology per brain biopsy. For control cohorts, we had CSF samples from cognitively healthy individuals (n = 33) and patients with diagnosed AD and no iNPH (n = 39).*-31ptResults:Levels of all investigated biomarkers differed significantly between groups, with the exception of t-Tau levels between healthy individuals and iNPH patients with AD pathology. Applying a correction factor for each biomarker (0.842*Aβ 1 - 42, 0.779*t-Tau, and 0.610*P-Tau181) for the effect of iNPH yielded a sensitivity of 2.4% and specificity of 100%. The ratio of P-Tau181 to Aβ 1 - 42 was moderately effective in aiding recognition of AD pathology in iNPH patients (sensitivity 0.79, specificity 0.76, area under the curve 0.824). CONCLUSION Correcting for iNPH as a factor failed to improve diagnostic effectiveness, but the P-Tau181/Aβ 1 - 42 ratio showed some utility in the diagnosis of AD in iNPH patients.
Collapse
Affiliation(s)
- Aleksi Vanninen
- Department of Neurosurgery, Kuopio University Hospital, Kuopio, Finland
- Neurosurgery, Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland
| | - Heikki Lukkarinen
- Department of Neurosurgery, Kuopio University Hospital, Kuopio, Finland
- Neurosurgery, Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland
| | - Tarja Kokkola
- Neurology, Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland
| | - Anne M Koivisto
- Neurology, Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland
- Department of Neurology, Kuopio University Hospital, Kuopio, Finland
- Department of Neurosciences, University of Helsinki, Helsinki, Finland
- Department of Geriatrics, Helsinki University Hospital, Helsinki, Finland
| | - Merja Kokki
- School of Medicine, University of Eastern Finland, Kuopio, Finland
- Department of Anaesthesia and Intensive Care Medicine, Kuopio University Hospital, Kuopio, Finland
| | - Tadeusz Musialowicz
- Department of Anaesthesia and Intensive Care Medicine, Kuopio University Hospital, Kuopio, Finland
| | - Mikko Hiltunen
- Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK
- UK Dementia Research Institute at UCL, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Ville Leinonen
- Department of Neurosurgery, Kuopio University Hospital, Kuopio, Finland
- Neurosurgery, Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland
| | - Sanna-Kaisa Herukka
- Neurology, Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland
- Department of Neurology, Kuopio University Hospital, Kuopio, Finland
| | - Tuomas Rauramaa
- Department of Pathology, Kuopio University Hospital, Kuopio, Finland
- Pathology, Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland
| |
Collapse
|
44
|
Cruchaga C, Western D, Timsina J, Wang L, Wang C, Yang C, Ali M, Beric A, Gorijala P, Kohlfeld P, Budde J, Levey A, Morris J, Perrin R, Ruiz A, Marquié M, Boada M, de Rojas I, Rutledge J, Oh H, Wilson E, Guen YL, Alvarez I, Aguilar M, Greicius M, Pastor P, Pulford D, Ibanez L, Wyss-Coray T, Sung YJ, Phillips B. Proteogenomic analysis of human cerebrospinal fluid identifies neurologically relevant regulation and informs causal proteins for Alzheimer's disease. RESEARCH SQUARE 2023:rs.3.rs-2814616. [PMID: 37333337 PMCID: PMC10275048 DOI: 10.21203/rs.3.rs-2814616/v1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
The integration of quantitative trait loci (QTL) with disease genome-wide association studies (GWAS) has proven successful at prioritizing candidate genes at disease-associated loci. QTL mapping has mainly been focused on multi-tissue expression QTL or plasma protein QTL (pQTL). Here we generated the largest-to-date cerebrospinal fluid (CSF) pQTL atlas by analyzing 7,028 proteins in 3,107 samples. We identified 3,373 independent study-wide associations for 1,961 proteins, including 2,448 novel pQTLs of which 1,585 are unique to CSF, demonstrating unique genetic regulation of the CSF proteome. In addition to the established chr6p22.2-21.32 HLA region, we identified pleiotropic regions on chr3q28 near OSTN and chr19q13.32 near APOE that were enriched for neuron-specificity and neurological development. We also integrated this pQTL atlas with the latest Alzheimer's disease (AD) GWAS through PWAS, colocalization and Mendelian Randomization and identified 42 putative causal proteins for AD, 15 of which have drugs available. Finally, we developed a proteomics-based risk score for AD that outperforms genetics-based polygenic risk scores. These findings will be instrumental to further understand the biology and identify causal and druggable proteins for brain and neurological traits.
Collapse
Affiliation(s)
| | - Dan Western
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
| | - Jigyasha Timsina
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
| | - Lihua Wang
- Washington University School of Medicine
| | | | | | | | | | | | - Patsy Kohlfeld
- Washington University School of Medicine, St Louis, MO, USA
| | | | | | | | | | | | | | - Mercè Boada
- Memory Clinic of Fundaciò ACE, Catalan Institute of Applied Neurosciences
| | | | | | | | | | | | - Ignacio Alvarez
- Fundació Docència i Recerca Mútua Terrassa, Terrassa, Barcelona, Spain
| | | | | | - Pau Pastor
- University Hospital Germans Trias i Pujol
| | | | | | | | | | | |
Collapse
|
45
|
Ou YN, Ge YJ, Wu BS, Zhang Y, Jiang YC, Kuo K, Yang L, Tan L, Feng JF, Cheng W, Yu JT. The genetic architecture of fornix white matter microstructure and their involvement in neuropsychiatric disorders. Transl Psychiatry 2023; 13:180. [PMID: 37236919 DOI: 10.1038/s41398-023-02475-6] [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: 10/26/2022] [Revised: 05/03/2023] [Accepted: 05/12/2023] [Indexed: 05/28/2023] Open
Abstract
The fornix is a white matter bundle located in the center of the hippocampaldiencephalic limbic circuit that controls memory and executive functions, yet its genetic architectures and involvement in brain disorders remain largely unknown. We carried out a genome-wide association analysis of 30,832 UK Biobank individuals of the six fornix diffusion magnetic resonance imaging (dMRI) traits. The post-GWAS analysis allowed us to identify causal genetic variants in phenotypes at the single nucleotide polymorphisms (SNP), locus, and gene levels, as well as genetic overlap with brain health-related traits. We further generalized our GWAS in adolescent brain cognitive development (ABCD) cohort. The GWAS identified 63 independent significant variants within 20 genomic loci associated (P < 8.33 × 10-9) with the six fornix dMRI traits. Geminin coiled-coil domain containing (GMNC) and NUAK family SNF1-like kinase 1 (NUAK1) gene were highlighted, which were found in UKB and replicated in ABCD. The heritability of the six traits ranged from 10% to 27%. Gene mapping strategies identified 213 genes, where 11 were supported by all of four methods. Gene-based analyses revealed pathways relating to cell development and differentiation, with astrocytes found to be significantly enriched. Pleiotropy analyses with eight neurological and psychiatric disorders revealed shared variants, especially with schizophrenia under the conjFDR threshold of 0.05. These findings advance our understanding of the complex genetic architectures of fornix and their relevance in neurological and psychiatric disorders.
Collapse
Affiliation(s)
- Ya-Nan Ou
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Yi-Jun Ge
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Bang-Sheng Wu
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, National Center for Neurological Disorders, Shanghai, China
| | - Yi Zhang
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, National Center for Neurological Disorders, Shanghai, China
| | - Yu-Chao Jiang
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China
- Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence (Fudan University), Ministry of Education, Shanghai, China
| | - Kevin Kuo
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, National Center for Neurological Disorders, Shanghai, China
| | - Liu Yang
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, National Center for Neurological Disorders, Shanghai, China
| | - Lan Tan
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Jian-Feng Feng
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China
- Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence (Fudan University), Ministry of Education, Shanghai, China
- Fudan ISTBI-ZJNU Algorithm Centre for Brain-Inspired Intelligence, Zhejiang Normal University, Jinhua, China
- MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
- Zhangjiang Fudan International Innovation Center, Shanghai, China
| | - Wei Cheng
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, National Center for Neurological Disorders, Shanghai, China.
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China.
- Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence (Fudan University), Ministry of Education, Shanghai, China.
- Fudan ISTBI-ZJNU Algorithm Centre for Brain-Inspired Intelligence, Zhejiang Normal University, Jinhua, China.
| | - Jin-Tai Yu
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, National Center for Neurological Disorders, Shanghai, China.
| |
Collapse
|
46
|
Timsina J, Ali M, Do A, Wang L, Sung YJ, Cruchaga C. Harmonization of CSF and imaging biomarkers for Alzheimer's disease biomarkers: need and practical applications for genetics studies and preclinical classification. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.24.542118. [PMID: 37292823 PMCID: PMC10245826 DOI: 10.1101/2023.05.24.542118] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
INTRODUCTION In Alzheimer's disease (AD) research, cerebrospinal fluid (CSF) Amyloid beta (Aβ), Tau and pTau are the most accepted and well validated biomarkers. Several methods and platforms exist to measure those biomarkers which leads to challenges in combining data across studies. Thus, there is a need to identify methods that harmonize and standardize these values. METHODS We used a Z-score based approach to harmonize CSF and amyloid imaging data from multiple cohorts and compared GWAS result using this method with currently accepted methods. We also used a generalized mixture modelling to calculate the threshold for biomarker-positivity. RESULTS Z-scores method performed as well as meta-analysis and did not lead to any spurious results. Cutoffs calculated with this approach were found to be very similar to those reported previously. DISCUSSION This approach can be applied to heterogeneous platforms and provides biomarker cut-offs consistent with the classical approaches without requiring any additional data.
Collapse
Affiliation(s)
- Jigyasha Timsina
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Muhammad Ali
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Anh Do
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO 63110, USA
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Lihua Wang
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Yun Ju Sung
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO 63110, USA
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Carlos Cruchaga
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO 63110, USA
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO 63110, USA
| |
Collapse
|
47
|
Reitz C, Pericak-Vance MA, Foroud T, Mayeux R. A global view of the genetic basis of Alzheimer disease. Nat Rev Neurol 2023; 19:261-277. [PMID: 37024647 PMCID: PMC10686263 DOI: 10.1038/s41582-023-00789-z] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/21/2023] [Indexed: 04/08/2023]
Abstract
The risk of Alzheimer disease (AD) increases with age, family history and informative genetic variants. Sadly, there is still no cure or means of prevention. As in other complex diseases, uncovering genetic causes of AD could identify underlying pathological mechanisms and lead to potential treatments. Rare, autosomal dominant forms of AD occur in middle age as a result of highly penetrant genetic mutations, but the most common form of AD occurs later in life. Large-scale, genome-wide analyses indicate that 70 or more genes or loci contribute to AD. One of the major factors limiting progress is that most genetic data have been obtained from non-Hispanic white individuals in Europe and North America, preventing the development of personalized approaches to AD in individuals of other ethnicities. Fortunately, emerging genetic data from other regions - including Africa, Asia, India and South America - are now providing information on the disease from a broader range of ethnicities. Here, we summarize the current knowledge on AD genetics in populations across the world. We predominantly focus on replicated genetic discoveries but also include studies in ethnic groups where replication might not be feasible. We attempt to identify gaps that need to be addressed to achieve a complete picture of the genetic and molecular factors that drive AD in individuals across the globe.
Collapse
Affiliation(s)
- Christiane Reitz
- The Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY, USA
- The Gertrude H. Sergievsky Center, Columbia University, New York, NY, USA
- Department of Neurology, Columbia University, New York, NY, USA
- Department of Epidemiology, Columbia University, New York, NY, USA
| | - Margaret A Pericak-Vance
- The John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, USA
- The Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Tatiana Foroud
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
- National Centralized Repository for Alzheimer's Disease and Related Dementias, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Richard Mayeux
- The Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY, USA.
- The Gertrude H. Sergievsky Center, Columbia University, New York, NY, USA.
- Department of Neurology, Columbia University, New York, NY, USA.
- Department of Epidemiology, Columbia University, New York, NY, USA.
| |
Collapse
|
48
|
van der Flier WM, de Vugt ME, Smets EMA, Blom M, Teunissen CE. Towards a future where Alzheimer's disease pathology is stopped before the onset of dementia. NATURE AGING 2023; 3:494-505. [PMID: 37202515 DOI: 10.1038/s43587-023-00404-2] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 03/21/2023] [Indexed: 05/20/2023]
Abstract
Alzheimer's disease (AD) is a major healthcare challenge with no curative treatment at present. To address this challenge, we need a paradigm shift, where we focus on pre-dementia stages of AD. In this Perspective, we outline a strategy to move towards a future with personalized medicine for AD by preparing for and investing in effective and patient-orchestrated diagnosis, prediction and prevention of the dementia stage. While focusing on AD, this Perspective also discusses studies that do not specify the cause of dementia. Future personalized prevention strategies encompass multiple components, including tailored combinations of disease-modifying interventions and lifestyle. By empowering the public and patients to be more actively engaged in the management of their health and disease and by developing improved strategies for diagnosis, prediction and prevention, we can pave the way for a future with personalized medicine, in which AD pathology is stopped to prevent or delay the onset of dementia.
Collapse
Affiliation(s)
- Wiesje M van der Flier
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, the Netherlands.
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, the Netherlands.
- Epidemiology and Data Science, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, the Netherlands.
| | - Marjolein E de Vugt
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Alzheimer Centre Limburg, Maastricht University, Maastricht, the Netherlands
| | - Ellen M A Smets
- Medical Psychology, Amsterdam UMC location AMC, Amsterdam, the Netherlands
| | - Marco Blom
- Alzheimer Nederland, Amersfoort, Utrecht, the Netherlands
| | - Charlotte E Teunissen
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, the Netherlands
- Neurochemistry Laboratory, Clinical Chemistry, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, the Netherlands
| |
Collapse
|
49
|
Keep RF, Jones HC, Hamilton MG, Drewes LR. A year in review: brain barriers and brain fluids research in 2022. Fluids Barriers CNS 2023; 20:30. [PMID: 37085841 PMCID: PMC10120509 DOI: 10.1186/s12987-023-00429-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Indexed: 04/23/2023] Open
Abstract
This aim of this editorial is to highlight progress made in brain barrier and brain fluid research in 2022. It covers studies on the blood-brain, blood-retina and blood-CSF barriers (choroid plexus and meninges), signaling within the neurovascular unit and elements of the brain fluid systems. It further discusses how brain barriers and brain fluid systems are impacted in CNS diseases, their role in disease progression and progress being made in treating such diseases.
Collapse
Affiliation(s)
- Richard F Keep
- Department of Neurosurgery, University of Michigan, R5018 BSRB 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA.
| | | | - Mark G Hamilton
- Department of Clinical Neurosciences, Division of Neurosurgery, University of Calgary, Alberta, Canada
| | - Lester R Drewes
- Department of Biomedical Sciences, University of Minnesota Medical School Duluth, Duluth, MN, 55812, USA
| |
Collapse
|
50
|
Andrews SJ, Renton AE, Fulton-Howard B, Podlesny-Drabiniok A, Marcora E, Goate AM. The complex genetic architecture of Alzheimer's disease: novel insights and future directions. EBioMedicine 2023; 90:104511. [PMID: 36907103 PMCID: PMC10024184 DOI: 10.1016/j.ebiom.2023.104511] [Citation(s) in RCA: 90] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 02/20/2023] [Indexed: 03/12/2023] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is a complex multifactorial neurodegenerative disorder and the most common form of dementia. AD is highly heritable, with heritability estimates of ∼70% from twin studies. Progressively larger genome-wide association studies (GWAS) have continued to expand our knowledge of AD/dementia genetic architecture. Until recently these efforts had identified 39 disease susceptibility loci in European ancestry populations. RECENT DEVELOPMENTS Two new AD/dementia GWAS have dramatically expanded the sample sizes and the number of disease susceptibility loci. The first increased total sample size to 1,126,563-with an effective sample size of 332,376-by predominantly including new biobank and population-based dementia datasets. The second, expands on an earlier GWAS from the International Genomics of Alzheimer's Project (IGAP) by increasing the number of clinically-defined AD cases/controls in addition to incorporating biobank dementia datasets, resulting in a total sample size to 788,989 and an effective sample size of 382,472. Collectively both GWAS identified 90 independent variants across 75 AD/dementia susceptibility loci, including 42 novel loci. Pathway analyses indicate the susceptibility loci are enriched for genes involved in amyloid plaque and neurofibrillary tangle formation, cholesterol metabolism, endocytosis/phagocytosis, and the innate immune system. Gene prioritization efforts for the novel loci identified 62 candidate causal genes. Many of the candidate genes from known and newly discovered loci play key roles in macrophages and highlight phagocytic clearance of cholesterol-rich brain tissue debris by microglia (efferocytosis) as a core pathogenetic hub and putative therapeutic target for AD. WHERE NEXT?: While GWAS in European ancestry populations have substantially enhanced our understanding of AD genetic architecture, heritability estimates from population based GWAS cohorts are markedly smaller than those from twin studies. While this missing heritability is likely due to a combination of factors, it highlights that our understanding of AD genetic architecture and genetic risk mechanisms remains incomplete. These knowledge gaps result from several underexplored areas in AD research. First, rare variants remain understudied due to methodological issues in identifying them and the cost of generating sufficiently powered whole exome/genome sequencing datasets. Second, sample sizes of non-European ancestry populations in AD GWAS remain small. Third, GWAS of AD neuroimaging and cerebrospinal fluid endophenotypes remains limited due to low compliance and high costs associated with measuring amyloid-β and tau levels and other disease-relevant biomarkers. Studies generating sequencing data, including diverse populations, and incorporating blood-based AD biomarkers are set to substantially improve our knowledge of AD genetic architecture.
Collapse
Affiliation(s)
- Shea J Andrews
- Department of Psychiatry and Behavioral Sciences, University of California San Francisco, San Francisco, CA, USA.
| | - Alan E Renton
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Brian Fulton-Howard
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Anna Podlesny-Drabiniok
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Edoardo Marcora
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Alison M Goate
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|