1
|
Zhao T, Cui X, Zhang X, Zhao M, Rastegar-Kashkooli Y, Wang J, Li Q, Jiang C, Li N, Xing F, Han X, Zhang J, Xing N, Wang J, Wang J. Hippocampal sclerosis: A review on current research status and its mechanisms. Ageing Res Rev 2025; 108:102716. [PMID: 40058463 DOI: 10.1016/j.arr.2025.102716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 02/27/2025] [Accepted: 03/02/2025] [Indexed: 03/27/2025]
Abstract
Hippocampal sclerosis (HS) is a pathological condition characterized by significant loss of hippocampal neurons and gliosis. This condition represents the most common neuropathological change observed in patients with temporal lobe epilepsy (TLE) and is also found in aging individuals. TLE related to HS is the most prevalent type of drug-resistant epilepsy in adults, and its underlying mechanisms are not yet fully understood. Therefore, developing improved methods for predicting and treating drug-resistant patients with TLE-HS is crucial. Patients with TLE-HS often experience cognitive impairment and psychological comorbidities, significantly affecting their quality of life. Consequently, a thorough review of the current research status of TLE-HS is essential, focusing on its prediction, diagnosis, treatment, and underlying mechanisms. The hippocampus plays a pivotal role in memory and cognition. HS of aging (HS-Aging), a condition linked to dementia in the ultra-elderly, is marked by severe CA1 (cornu ammonis) neuronal loss and frequent transactive response DNA-binding protein of 43 kDa (TDP-43) proteinopathy, often misdiagnosed as Alzheimer's disease (AD). Nonetheless, clinical characteristics and patterns of hippocampal atrophy can help differentiate between the two disorders. This review aims to provide a comprehensive overview of the pathological features of HS, the relevant mechanisms underlying TLE-HS and HS-Aging, current imaging diagnostic techniques, including machine learning, and available treatment modalities. It also explores the prognosis and comorbidities related to these conditions. Future research directions include establishing animal models to clarify the poorly understood mechanisms underlying HS, particularly those related to emotional processing. Investigating post-HS behavioral and cognitive changes in these models will lay the foundation for further advancements in this field. This review is a cornerstone for future investigations and suggests additional research endeavors.
Collapse
Affiliation(s)
- Ting Zhao
- Department of Neurology, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, Henan 450003, China.
| | - Xiaoxiao Cui
- Department of Neurology, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, Henan 450003, China
| | - Xinru Zhang
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Mengke Zhao
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Yousef Rastegar-Kashkooli
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China; School of International Education, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Junyang Wang
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Qiang Li
- Department of Neurology, Shanghai Gongli Hospital of Pudong New Area, Shanghai 200135, China
| | - Chao Jiang
- Department of Neurology, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, Henan 450003, China
| | - Nan Li
- Department of Neurology, The 2nd Affiliated Hospital of Zhengzhou University, Zhengzhou 450014, China
| | - Fei Xing
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, China
| | - Xiong Han
- Department of Neurology, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, Henan 450003, China
| | - Jiewen Zhang
- Department of Neurology, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, Henan 450003, China
| | - Na Xing
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, China.
| | - Junmin Wang
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China.
| | - Jian Wang
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China.
| |
Collapse
|
2
|
Wang R, Li J, Li X, Guo Y, Chen P, Peng T. Exercise-induced modulation of miRNAs and gut microbiome: a holistic approach to neuroprotection in Alzheimer's disease. Rev Neurosci 2025:revneuro-2025-0013. [PMID: 40366727 DOI: 10.1515/revneuro-2025-0013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Accepted: 03/28/2025] [Indexed: 05/15/2025]
Abstract
Alzheimer's disease (AD), a progressive neurodegenerative disorder, is marked by cognitive decline, neuroinflammation, and neuronal loss. MicroRNAs (miRNAs) have emerged as critical regulators of gene expression, influencing key pathways involved in neuroinflammation and neurodegeneration in AD. This review delves into the multifaceted role of exercise in modulating miRNA expression and its interplay with the gut microbiome, proposing a comprehensive framework for neuroprotection in AD. By synthesizing current research, we elucidate how exercise-induced changes in miRNA profiles can mitigate inflammatory responses, promote neurogenesis, and reduce amyloid-beta and tau pathologies. Additionally, we explore the gut-brain axis, highlighting how exercise-driven alterations in gut microbiota composition can further influence miRNA expression, thereby enhancing cognitive function and reducing neuroinflammatory markers. This holistic approach underscores the potential of targeting exercise-regulated miRNAs and gut microbiome interactions as a novel, noninvasive therapeutic strategy to decelerate AD progression and improve quality of life for patients. This approach aims to decelerate disease progression and improve patient outcomes, offering a promising avenue for enhancing the effectiveness of AD management.
Collapse
Affiliation(s)
- Rui Wang
- College of Physical Education, Guizhou Normal University, GuiYang 550025, China
| | - Juan Li
- Hanyang University Erica, AnSan 15588, Korea
| | - Xiaochen Li
- School of Physical Education, Huaibei Normal University, HuaiBei 235000, China
| | - Yan Guo
- Sichuan University Jinjiang College, ChengDu 610000, China
| | - Pei Chen
- School of Physical Education, Huaibei Normal University, HuaiBei 235000, China
| | - Tian Peng
- Department of Physical Education, 12377 Zhejiang University of Science and Technology , HangZhou 310023, China
| |
Collapse
|
3
|
Strauss A, Swann P, Kigar SL, Christou R, Savinykh Yarkoni N, Turner L, Murley AG, Chouliaras L, Shapiro N, Ashton NJ, Savulich G, Bevan-Jones WR, Surendranthan A, Blennow K, Zetterberg H, O'Brien JT, Rowe JB, Malpetti M. Peripheral innate immunophenotype in neurodegenerative disease: blood-based profiles and links to survival. Mol Psychiatry 2025; 30:1985-1994. [PMID: 39472664 PMCID: PMC12015116 DOI: 10.1038/s41380-024-02809-w] [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: 01/16/2024] [Revised: 10/18/2024] [Accepted: 10/22/2024] [Indexed: 04/24/2025]
Abstract
The innate immune system plays an integral role in the progression of many neurodegenerative diseases. In addition to central innate immune cells (e.g., microglia), peripheral innate immune cells (e.g., blood monocytes, natural killer cells, and dendritic cells) may also differ in these conditions. However, the characterization of peripheral innate immune cell types across different neurodegenerative diseases remains incomplete. This study aimed to characterize peripheral innate immune profiles using flow cytometry for immunophenotyping of peripheral blood mononuclear cells in n = 148 people with Alzheimer's disease (AD), frontotemporal dementia (FTD), corticobasal syndrome (CBS), progressive supranuclear palsy (PSP), Lewy body dementia (LBD) as compared to n = 37 healthy controls. To compare groups, we used multivariate dissimilarity analysis and principal component analysis across 19 innate immune cell types. We identified pro-inflammatory profiles that significantly differ between patients with all-cause dementia and healthy controls, with some significant differences between patient groups. Regression analysis confirmed that time to death following the blood test correlated with the individuals' immune profile weighting, positively to TREM2+ and non-classical monocytes and negatively to classical monocytes. Taken together, these results describe transdiagnostic peripheral immune profiles and highlight the link between prognosis and the monocyte cellular subdivision and function (as measured by surface protein expression). The results suggest that blood-derived innate immune profiles can inform sub-populations of cells relevant for specific neurodegenerative diseases that are significantly linked to accelerated disease progression and worse survival outcomes across diagnoses. Blood-based innate immune profiles may contribute to enhanced precision medicine approaches in dementia, helping to identify and monitor therapeutic targets and stratify patients for candidate immunotherapies.
Collapse
Affiliation(s)
- Alexandra Strauss
- University of Cambridge Department of Clinical Neurosciences and Cambridge University Hospitals NHS Trust, Cambridge, UK
| | - Peter Swann
- Department of Psychiatry, University of Cambridge, Cambridge, UK
| | - Stacey L Kigar
- Department of Psychiatry, University of Cambridge, Cambridge, UK
- Department of Medicine, University Cambridge, Cambridge, UK
| | - Rafailia Christou
- University of Cambridge Department of Clinical Neurosciences and Cambridge University Hospitals NHS Trust, Cambridge, UK
| | | | - Lorinda Turner
- Department of Psychiatry, University of Cambridge, Cambridge, UK
- Department of Medicine, University Cambridge, Cambridge, UK
| | - Alexander G Murley
- University of Cambridge Department of Clinical Neurosciences and Cambridge University Hospitals NHS Trust, Cambridge, UK
| | | | - Noah Shapiro
- University of Cambridge Department of Clinical Neurosciences and Cambridge University Hospitals NHS Trust, Cambridge, UK
| | - Nicholas J Ashton
- Department of Psychiatry and Neurochemistry, University of Gothenburg, Gothenburg, Sweden
- Banner Alzheimer's Institute and University of Arizona, Phoenix, AZ, USA
- Banner Sun Health Research Institute, Sun City, AZ, USA
| | - George Savulich
- Department of Psychiatry, University of Cambridge, Cambridge, UK
| | | | | | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, University of Gothenburg, Gothenburg, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, University of Gothenburg, Gothenburg, 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
| | - John T O'Brien
- Department of Psychiatry, University of Cambridge, Cambridge, UK
| | - James B Rowe
- University of Cambridge Department of Clinical Neurosciences and Cambridge University Hospitals NHS Trust, Cambridge, UK
- Medical Research Council Cognition and Brain Sciences Unit, Cambridge, UK
| | - Maura Malpetti
- University of Cambridge Department of Clinical Neurosciences and Cambridge University Hospitals NHS Trust, Cambridge, UK.
- UK Dementia Research Institute at University of Cambridge, Cambridge, UK.
| |
Collapse
|
4
|
Al Massadi O, Labarchède M, de Pins B, Longueville S, Giralt A, Irinopoulou T, Savariradjane M, Subashi E, Ginés S, Caboche J, Mariani LL, Betuing S, Girault JA. PYK2 in the dorsal striatum of Huntington's disease R6/2 mouse model. Neurobiol Dis 2025; 207:106840. [PMID: 39971200 DOI: 10.1016/j.nbd.2025.106840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2025] [Accepted: 02/11/2025] [Indexed: 02/21/2025] Open
Abstract
Huntington's disease (HD) is a devastating disease due to autosomal dominant mutation in the HTT gene. Its pathophysiology involves multiple molecular alterations including transcriptional defects. We previously showed that in HD patients and mouse model, the protein levels of the non-receptor tyrosine kinase PYK2 were decreased in the hippocampus and that viral expression of PYK2 improved the hippocampal phenotype. Here, we investigated the possible contribution of PYK2 in the striatum, a brain region particularly altered in HD. PYK2 mRNA levels were decreased in the striatum and hippocampus of R6/2 mice, a severe HD model. Striatal PYK2 protein levels were also decreased in R6/2 mice and human patients. PYK2 knockout by itself did not result in motor symptoms observed in HD mouse models. We examined whether PYK2 deficiency participated in the R6/2 mice phenotype by expressing PYK2 in their dorsal striatum using AAV vectors. With an AAV1/Camk2a promoter, we did not observe significant improvement of body weight, clasping, motor activity and coordination (rotarod) alterations observed in R6/2 mice. With an AAV9/SYN1 promoter we found a slightly higher body weight and a trend to better rotarod performance. Both viruses similarly transduced striatal projection neurons and somatostatin-positive interneurons but only AAV9/SYN1 led to PYK2 expression in cholinergic and parvalbumin-positive interneurons. Expression of PYK2 in cholinergic interneurons may contribute to the slight effects observed. We conclude that PYK2 mRNA and protein levels are decreased in the striatum as in hippocampus of HD patients and mouse models. However, in contrast to hippocampus, striatal viral expression of PYK2 has only a minor effect on the R6/2 model striatal phenotype.
Collapse
Affiliation(s)
- Omar Al Massadi
- Inserm UMR-S 1270, Paris 75005, France; Sorbonne Université, Faculty of Sciences and Engineering, Paris 75005, France; Institut du Fer à Moulin, 17 rue du Fer à Moulin, Paris 75005, France.
| | - Mélody Labarchède
- Inserm UMR-S 1270, Paris 75005, France; Sorbonne Université, Faculty of Sciences and Engineering, Paris 75005, France; Institut du Fer à Moulin, 17 rue du Fer à Moulin, Paris 75005, France; Institut du Cerveau, Paris Brain Institute, ICM, Inserm, CNRS, Sorbonne Université, Paris, France
| | - Benoit de Pins
- Inserm UMR-S 1270, Paris 75005, France; Sorbonne Université, Faculty of Sciences and Engineering, Paris 75005, France; Institut du Fer à Moulin, 17 rue du Fer à Moulin, Paris 75005, France
| | - Sophie Longueville
- Inserm UMR-S 1270, Paris 75005, France; Sorbonne Université, Faculty of Sciences and Engineering, Paris 75005, France; Institut du Fer à Moulin, 17 rue du Fer à Moulin, Paris 75005, France
| | - Albert Giralt
- Inserm UMR-S 1270, Paris 75005, France; Sorbonne Université, Faculty of Sciences and Engineering, Paris 75005, France; Institut du Fer à Moulin, 17 rue du Fer à Moulin, Paris 75005, France; Departament de Biomedicina, Facultat de Medicina i Ciències de la Salut, Institut de Neurociències, Universitat de Barcelona, 08036 Barcelona, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, Madrid, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain; Production and Validation Center of Advanced Therapies (Creatio), University of Barcelona, Barcelona, Spain
| | - Theano Irinopoulou
- Inserm UMR-S 1270, Paris 75005, France; Sorbonne Université, Faculty of Sciences and Engineering, Paris 75005, France; Institut du Fer à Moulin, 17 rue du Fer à Moulin, Paris 75005, France
| | - Mythili Savariradjane
- Inserm UMR-S 1270, Paris 75005, France; Sorbonne Université, Faculty of Sciences and Engineering, Paris 75005, France; Institut du Fer à Moulin, 17 rue du Fer à Moulin, Paris 75005, France
| | - Enejda Subashi
- Sorbonne Université, Faculty of Sciences and Engineering, Paris 75005, France; Neuroscience Paris Seine, Institut de Biologie Paris-Seine, Paris, France; CNRS UMR8246, Paris, France; INSERM U1130, Paris, France
| | - Silvia Ginés
- Departament de Biomedicina, Facultat de Medicina i Ciències de la Salut, Institut de Neurociències, Universitat de Barcelona, 08036 Barcelona, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, Madrid, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain; Production and Validation Center of Advanced Therapies (Creatio), University of Barcelona, Barcelona, Spain
| | - Jocelyne Caboche
- Sorbonne Université, Faculty of Sciences and Engineering, Paris 75005, France; Neuroscience Paris Seine, Institut de Biologie Paris-Seine, Paris, France; CNRS UMR8246, Paris, France; INSERM U1130, Paris, France
| | - Louise-Laure Mariani
- Institut du Cerveau, Paris Brain Institute, ICM, Inserm, CNRS, Sorbonne Université, Paris, France
| | - Sandrine Betuing
- Sorbonne Université, Faculty of Sciences and Engineering, Paris 75005, France; Neuroscience Paris Seine, Institut de Biologie Paris-Seine, Paris, France; CNRS UMR8246, Paris, France; INSERM U1130, Paris, France
| | - Jean-Antoine Girault
- Inserm UMR-S 1270, Paris 75005, France; Sorbonne Université, Faculty of Sciences and Engineering, Paris 75005, France; Institut du Fer à Moulin, 17 rue du Fer à Moulin, Paris 75005, France; Institut du Cerveau, Paris Brain Institute, ICM, Inserm, CNRS, Sorbonne Université, Paris, France.
| |
Collapse
|
5
|
Rodriguez-Rodriguez P, Wang W, Tsagkogianni C, Feng I, Morello-Megias A, Jain K, Alanko V, Kahvecioglu HA, Mohammadi E, Li X, Flajolet M, Sandebring-Matton A, Maioli S, Vidal N, Milosevic A, Roussarie JP. Cell-type specific profiling of human entorhinal cortex at the onset of Alzheimer's disease neuropathology. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.12.31.630881. [PMID: 39803521 PMCID: PMC11722323 DOI: 10.1101/2024.12.31.630881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/29/2025]
Abstract
Neurons located in layer II of the entorhinal cortex (ECII) are the primary site of pathological tau accumulation and neurodegeneration at preclinical stages of Alzheimer's disease (AD). Exploring the alterations that underlie the early degeneration of these cells is essential to develop therapies that curb the disease before symptom onset. Here we performed cell-type specific profiling of human EC at the onset of AD neuropathology. We identify an early response to amyloid pathology by microglia and oligodendrocytes. Importantly, we provide the first insight into neuronal alterations that coincide with incipient tau pathology: the signaling pathway for Reelin, recently shown to be a major AD resilience gene is dysregulated in ECII neurons, while the secreted synaptic organizer molecules NPTX2 and CBLN4, emerging AD biomarkers, are downregulated in surrounding neurons. By uncovering the complex multicellular landscape of EC at these early AD stages, this study paves the way for detailed characterization of the mechanisms governing NFT formation and opens long-needed novel therapeutic avenues.
Collapse
Affiliation(s)
| | - Wei Wang
- Bioinformatics Resource Center, The Rockefeller University. New York, NY, USA
| | - Christina Tsagkogianni
- Department of Neurobiology Care Sciences and Society, Karolinska Institute. Stockholm, Sweden
| | - Irena Feng
- Boston University Chobanian & Avedisian School of Medicine. Boston, MA, USA
| | - Ana Morello-Megias
- Boston University Chobanian & Avedisian School of Medicine. Boston, MA, USA
| | - Kaahini Jain
- Boston University Chobanian & Avedisian School of Medicine. Boston, MA, USA
| | - Vilma Alanko
- Department of Neurobiology Care Sciences and Society, Karolinska Institute. Stockholm, Sweden
| | | | - Elyas Mohammadi
- Department of Neurobiology Care Sciences and Society, Karolinska Institute. Stockholm, Sweden
| | - Xiaofei Li
- Department of Neurobiology Care Sciences and Society, Karolinska Institute. Stockholm, Sweden
| | | | - Anna Sandebring-Matton
- Department of Neurobiology Care Sciences and Society, Karolinska Institute. Stockholm, Sweden
| | - Silvia Maioli
- Department of Neurobiology Care Sciences and Society, Karolinska Institute. Stockholm, Sweden
| | - Noemi Vidal
- Pathology department. Biobank HUB-ICO-IDIBELL, University Hospital of Bellvitge. Barcelona, Spain
| | - Ana Milosevic
- Laboratory of Developmental Genetics, The Rockefeller University. New York, NY, USA
| | | |
Collapse
|
6
|
Han S, Hwang J, Park T, Pyun J, Lee J, Park JS, Bice PJ, Liu S, Yun S, Jeong J, Risacher SL, Saykin AJ, Byun MS, Yi D, Sung J, Lee DY, Kim S, Nho K, Park YH. Transcriptome analysis of early- and late-onset Alzheimer's disease in Korean cohorts. Alzheimers Dement 2025; 21:e14563. [PMID: 39935412 PMCID: PMC11815242 DOI: 10.1002/alz.14563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 12/24/2024] [Accepted: 12/28/2024] [Indexed: 02/13/2025]
Abstract
INTRODUCTION The molecular mechanisms underlying early-onset Alzheimer's disease (EOAD) and late-onset Alzheimer's disease (LOAD) remain incompletely understood, particularly in Asian populations. METHODS RNA-sequencing was carried out on blood samples from 248 participants in the Seoul National University Bundang Hospital cohort to perform differential gene expression (DGE) and weighted gene co-expression network analysis. Findings were replicated in an independent Korean cohort (N = 275). RESULTS DGE analysis identified 18 and 88 dysregulated genes in EOAD and LOAD, respectively. Network analysis identified a LOAD-associated module showing a significant enrichment in pathways related to mitophagy, 5' adenosine monophosphate-activated protein kinase signaling, and ubiquitin-mediated proteolysis. In the replication cohort, downregulation of SMOX and PLVAP in LOAD was replicated, and the LOAD-associated module was highly preserved. In addition, SMOX and PLVAP were associated with brain amyloid beta deposition. DISCUSSION Our findings suggest distinct molecular signatures for EOAD and LOAD in a Korean population, providing deeper understanding of their diagnostic potential and molecular mechanisms. HIGHLIGHTS Analysis identified 18 and 88 dysregulated genes in early-onset Alzheimer's disease (EOAD) and late-onset Alzheimer's disease (LOAD), respectively. Expression levels of SMOX and PLVAP were downregulated in LOAD. Expression levels of SMOX and PLVAP were associated with brain amyloid beta deposition. Pathways including mitophagy and 5' adenosine monophosphate-activated protein kinase signaling were enriched in a LOAD module. A LOAD module was highly preserved across two independent cohorts.
Collapse
Affiliation(s)
- Sang‐Won Han
- Department of NeurologySoonchunhyang University Seoul HospitalSeoulRepublic of Korea
- Department of NeurologyChuncheon Sacred Heart Hospital, Hallym University College of MedicineChuncheon‐siRepublic of Korea
| | - Jiyun Hwang
- Genome and Health Big Data Laboratory, Graduate School of Public HealthSeoul National UniversitySeoulRepublic of Korea
| | - Tamina Park
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana Alzheimer's Disease Research CenterIndiana University School of MedicineIndianapolisIndianaUSA
| | - Jung‐Min Pyun
- Department of NeurologySoonchunhyang University Seoul HospitalSeoulRepublic of Korea
| | - Joo‐Yeon Lee
- Genome and Health Big Data Laboratory, Graduate School of Public HealthSeoul National UniversitySeoulRepublic of Korea
- Institute of Health and EnvironmentsSeoul National UniversitySeoulRepublic of Korea
| | - Jeong Su Park
- Department of Laboratory MedicineSeoul National University Bundang Hospital, Seoul National University College of MedicineSeongnam‐siRepublic of Korea
| | - Paula J. Bice
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana Alzheimer's Disease Research CenterIndiana University School of MedicineIndianapolisIndianaUSA
| | - Shiwei Liu
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana Alzheimer's Disease Research CenterIndiana University School of MedicineIndianapolisIndianaUSA
| | - Sunmin Yun
- Precision Medicine CenterSeoul National University Bundang HospitalSeongnam‐siRepublic of Korea
| | - Jibin Jeong
- Precision Medicine CenterSeoul National University Bundang HospitalSeongnam‐siRepublic of Korea
| | - Shannon L. Risacher
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana Alzheimer's Disease Research CenterIndiana University School of MedicineIndianapolisIndianaUSA
| | - Andrew J. Saykin
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana Alzheimer's Disease Research CenterIndiana University School of MedicineIndianapolisIndianaUSA
| | - Min Soo Byun
- Department of PsychiatrySeoul National University College of MedicineSeoulRepublic of Korea
| | - Dahyun Yi
- Institute of Human Behavioral Medicine, Medical Research CenterSeoul National UniversitySeoulRepublic of Korea
| | - Joohon Sung
- Genome and Health Big Data Laboratory, Graduate School of Public HealthSeoul National UniversitySeoulRepublic of Korea
- Institute of Health and EnvironmentsSeoul National UniversitySeoulRepublic of Korea
| | - Dong Young Lee
- Department of PsychiatrySeoul National University College of MedicineSeoulRepublic of Korea
| | - SangYun Kim
- Department of NeurologySeoul National University Bundang Hospital and Seoul National University College of MedicineSeongnam‐siRepublic of Korea
| | - Kwangsik Nho
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana Alzheimer's Disease Research CenterIndiana University School of MedicineIndianapolisIndianaUSA
| | - Young Ho Park
- Department of NeurologySeoul National University Bundang Hospital and Seoul National University College of MedicineSeongnam‐siRepublic of Korea
| |
Collapse
|
7
|
Ayyanar MP, Vijayan M. A review on gut microbiota and miRNA crosstalk: implications for Alzheimer's disease. GeroScience 2025; 47:339-385. [PMID: 39562408 PMCID: PMC11872870 DOI: 10.1007/s11357-024-01432-5] [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: 09/23/2024] [Accepted: 11/07/2024] [Indexed: 11/21/2024] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder characterized by cognitive decline and progressive neuronal damage. Recent research has highlighted the significant roles of the gut microbiota and microRNAs (miRNAs) in the pathogenesis of AD. This review explores the intricate interaction between gut microbiota and miRNAs, emphasizing their combined impact on Alzheimer's progression. First, we discuss the bidirectional communication within the gut-brain axis and how gut dysbiosis contributes to neuroinflammation and neurodegeneration in AD. Changes in gut microbiota composition in Alzheimer's patients have been linked to inflammation, which exacerbates disease progression. Next, we delve into the biology of miRNAs, focusing on their roles in gene regulation, neurodevelopment, and neurodegeneration. Dysregulated miRNAs are implicated in AD pathogenesis, influencing key processes like inflammation, tau pathology, and amyloid deposition. We then examine how the gut microbiota modulates miRNA expression, particularly in the brain, potentially altering neuroinflammatory responses and synaptic plasticity. The interplay between gut microbiota and miRNAs also affects blood-brain barrier integrity, further contributing to Alzheimer's pathology. Lastly, we explore therapeutic strategies targeting this gut microbiota-miRNA axis, including probiotics, prebiotics, and dietary interventions, aiming to modulate miRNA expression and improve AD outcomes. While promising, challenges remain in fully elucidating these interactions and translating them into effective therapies. This review highlights the importance of understanding the gut microbiota-miRNA relationship in AD, offering potential pathways for novel therapeutic approaches aimed at mitigating the disease's progression.
Collapse
Affiliation(s)
- Maruthu Pandian Ayyanar
- Department of Biology, The Gandhigram Rural Institute (Deemed to be University), Gandhigram, 624302, Tamil Nadu, India
| | - Murali Vijayan
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, 79430, USA.
| |
Collapse
|
8
|
Jin Y, Topaloudi A, Shekhar S, Chen G, Scott AN, Colon BD, Drineas P, Rochet C, Paschou P. Neuropathology-based approach reveals novel Alzheimer's Disease genes and highlights female-specific pathways and causal links to disrupted lipid metabolism: insights into a vicious cycle. Acta Neuropathol Commun 2025; 13:1. [PMID: 39755674 DOI: 10.1186/s40478-024-01909-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Accepted: 12/05/2024] [Indexed: 01/06/2025] Open
Abstract
Dementia refers to an umbrella phenotype of many different underlying pathologies with Alzheimer's disease (AD) being the most common type. Neuropathological examination remains the gold standard for accurate AD diagnosis, however, most that we know about AD genetics is based on Genome-Wide Association Studies (GWAS) of clinically defined AD. Such studies have identified multiple AD susceptibility variants with a significant portion of the heritability unexplained and highlighting the phenotypic and genetic heterogeneity of the clinically defined entity. Furthermore, despite women's increased susceptibility to dementia, there is a lack of sex-specific genetic studies and understanding of sex-specific background for the disorder. Here, we aim to tackle the heterogeneity of AD by specifically concentrating on neuropathological features and pursuing sex-specific analysis. We bring together 14 different genomic and neuropathology datasets (6960 individuals) and we integrate our GWAS findings with transcriptomic and phenotypic data aiming to also identify biomarkers for AD progression. We uncover novel genetic associations to AD neuropathology, including BIN1 and OPCML. Our sex-specific analysis points to a role for BIN1 specifically in women as well as novel AD loci including QRFPR and SGCZ. Post-GWAS analyses illuminate the functional and biological mechanisms underlying AD and reveal sex-specific differences. Finally, through PheWAS and Mendelian Randomization analysis, we identify causal links with AD neuropathology pointing to disrupted lipid metabolism, as well as impaired peripheral immune response and liver dysfunction as part of a vicious cycle that fuels neurodegeneration.
Collapse
Affiliation(s)
- Yin Jin
- Department of Biological Sciences, Purdue University, 915 Mitch Daniels Blvd, West Lafayette, IN, USA
| | - Apostolia Topaloudi
- Department of Biological Sciences, Purdue University, 915 Mitch Daniels Blvd, West Lafayette, IN, USA
| | - Sudhanshu Shekhar
- Department of Biological Sciences, Purdue University, 915 Mitch Daniels Blvd, West Lafayette, IN, USA
| | - Guangxin Chen
- Department of Biological Sciences, Purdue University, 915 Mitch Daniels Blvd, West Lafayette, IN, USA
| | - Alicia Nicole Scott
- Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, USA
| | - Bryce David Colon
- Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, USA
| | - Petros Drineas
- Computer Science, Purdue University, West Lafayette, IN, USA
| | - Chris Rochet
- Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, USA
| | - Peristera Paschou
- Department of Biological Sciences, Purdue University, 915 Mitch Daniels Blvd, West Lafayette, IN, USA.
| |
Collapse
|
9
|
Borràs C, Canyelles M, Santos D, Rotllan N, Núñez E, Vázquez J, Maspoch D, Cano-Sarabia M, Carmona-Iragui M, Sirisi S, Lleó A, Fortea J, Alcolea D, Blanco-Vaca F, Escolà-Gil JC, Tondo M. Impaired Cerebrospinal Fluid Lipoprotein-Mediated Cholesterol Delivery to Neurons in Alzheimer's Disease. RESEARCH SQUARE 2024:rs.3.rs-5682870. [PMID: 39764088 PMCID: PMC11703344 DOI: 10.21203/rs.3.rs-5682870/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/15/2025]
Abstract
In the central nervous system, apolipoprotein (APO) E-containing high-density lipoprotein (HDL)-like particles mediate the transport of glial-derived cholesterol to neurons, which is essential for neuronal membrane remodeling and maintenance of the myelin sheath. Despite this, the role of HDL-like cholesterol trafficking on Alzheimer's disease (AD) pathogenesis remains poorly understood. We aimed to examine cholesterol transport via HDL-like particles in cerebrospinal fluid (CSF) of AD patients compared to control individuals. Additionally, we explored the ability of reconstituted HDL containing different APOE isoforms to regulate cholesterol transport. We evaluated the capacity of CSF HDL-like particles to facilitate radiolabeled unesterified cholesterol efflux from A172 human glioblastoma astrocytes and to deliver cholesterol to SH-SY5Y human neuronal cells. The HDL-like proteome in the AD and control groups was analyzed by liquid chromatography-mass spectrometry (LC-MS/MS). Reconstituted HDL nanoparticles were prepared by combining phospholipids and cholesterol with human APOE3 or APOE4, followed by radiolabeling with unesterified cholesterol. Our results showed that cholesterol efflux from astrocytes to CSF were similar between AD patients and controls, both under baseline conditions and after activation of ATP-binding cassette transporters A1 and G1. However, CSF HDL-like particle-mediated neuronal cholesterol uptake was significantly reduced in the AD group. LC-MS/MS analysis identified 775 proteins associated with HDL-like particles in both groups, with no major alterations in proteins linked to cholesterol metabolism. However, 27 proteins involved in non-cholesterol-related processes were differentially expressed. Notably, synthetic reconstituted HDL particles containing APOE4 exhibited reduced capacity to deliver cholesterol to neurons compared to those with APOE3. These findings indicate that CSF HDL-like particles from patients with AD demonstrate impaired cholesterol delivery to neurons. Our study highlights APOE4 as a critical contributor to abnormal neuronal cholesterol uptake in AD pathophysiology.
Collapse
Affiliation(s)
| | | | - David Santos
- Centro de Investigación Biomédica en Red Diabetes y Enfermedades Metabólicas Asociadas
| | | | - Estefanía Núñez
- Centro Nacional de Investigaciones Cardiovasculares Carlos III
| | - Jesús Vázquez
- Centro Nacional de Investigaciones Cardiovasculares Carlos III
| | - Daniel Maspoch
- Catalan Institute of Nanoscience and Nanotechnology (ICN2)
| | | | | | | | | | | | | | | | | | | |
Collapse
|
10
|
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
|
11
|
Godrich D, Pasteris J, Martin ER, Rundek T, Schellenberg G, Foroud T, Vance JM, Pericak-Vance MA, Cuccaro ML, Scott WK, Kukull W, Montine TJ, Beecham GW. Cerebral amyloid angiopathy impacts neurofibrillary tangle burden and cognition. Brain Commun 2024; 6:fcae369. [PMID: 39584156 PMCID: PMC11581998 DOI: 10.1093/braincomms/fcae369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 07/01/2024] [Accepted: 11/20/2024] [Indexed: 11/26/2024] Open
Abstract
Cerebral amyloid angiopathy commonly co-occurs with amyloid β plaques and neurofibrillary degeneration and is proposed to contribute to cognitive impairment. However, the interplay among these pathologic changes of Alzheimer disease is not well understood. Here we replicate and extend findings of a recent study that suggested the association of cerebral amyloid angiopathy and cognitive impairment is mediated by neurofibrillary degeneration. We employed similar approaches but in a larger, clinical-based (as opposed to community-based) set of 4915 autopsied National Alzheimer's Coordinating Center participants (60% with dementia). Neuropathologic lesions were measured ordinally; longitudinal change in cognition was used to measure cognitive impairment. Statistical analyses included ordinal logistic regression, mediation analyses and extension of models to include presence of APOE e4. We show a statistical interaction between cerebral amyloid angiopathy and neuritic plaques that impacts the burden of neurofibrillary tangles. Mediation analyses show that cerebral amyloid angiopathy is associated with cognitive impairment, but only by modifying the impact of neurofibrillary tangles on cognition. We expanded the mediation analysis to include APOE e4 and show similar results. Findings indicate that cerebral amyloid angiopathy plays an important role in the burden and impact of neurofibrillary degeneration contributing to cognitive impairment.
Collapse
Affiliation(s)
- Dana Godrich
- Dr. John T Macdonald Foundation Department of Human Genetics, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Jeremy Pasteris
- Dr. John T Macdonald Foundation Department of Human Genetics, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
- John P. Hussman Institute for Human Genomics, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Eden R Martin
- Dr. John T Macdonald Foundation Department of Human Genetics, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
- John P. Hussman Institute for Human Genomics, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Tatjana Rundek
- Department of Neurology and Evelyn F. McKnight Brain Institute, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Gerard Schellenberg
- Penn Neurodegeneration Genomics Center, Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 190104, USA
| | - Tatiana Foroud
- Department of Medical and Molecular Genetics, Indiana University, Indianapolis, IN 46202, USA
| | - Jeffery M Vance
- Dr. John T Macdonald Foundation Department of Human Genetics, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
- John P. Hussman Institute for Human Genomics, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Margaret A Pericak-Vance
- Dr. John T Macdonald Foundation Department of Human Genetics, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
- John P. Hussman Institute for Human Genomics, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Michael L Cuccaro
- Dr. John T Macdonald Foundation Department of Human Genetics, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
- John P. Hussman Institute for Human Genomics, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - William K Scott
- Dr. John T Macdonald Foundation Department of Human Genetics, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
- John P. Hussman Institute for Human Genomics, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Walter Kukull
- Department of Epidemiology, University of Washington, Seattle, WA 351619, USA
| | - Thomas J Montine
- Department of Pathology, Stanford University, Stanford, CA 94305, USA
| | - Gary W Beecham
- Dr. John T Macdonald Foundation Department of Human Genetics, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
- John P. Hussman Institute for Human Genomics, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| |
Collapse
|
12
|
Gunasekaran TI, Reyes‐Dumeyer D, Faber KM, Goate A, Boeve B, Cruchaga C, Pericak‐Vance M, Haines JL, Rosenberg R, Tsuang D, Mejia DR, Medrano M, Lantigua RA, Sweet RA, Bennett DA, Wilson RS, Alba C, Dalgard C, Foroud T, Vardarajan BN, Mayeux R. Missense and loss-of-function variants at GWAS loci in familial Alzheimer's disease. Alzheimers Dement 2024; 20:7580-7594. [PMID: 39233587 PMCID: PMC11567820 DOI: 10.1002/alz.14221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 07/10/2024] [Accepted: 08/06/2024] [Indexed: 09/06/2024]
Abstract
BACKGROUND Few rare variants have been identified in genetic loci from genome-wide association studies (GWAS) of Alzheimer's disease (AD), limiting understanding of mechanisms, risk assessment, and genetic counseling. METHODS Using genome sequencing data from 197 families in the National Institute on Aging Alzheimer's Disease Family Based Study and 214 Caribbean Hispanic families, we searched for rare coding variants within known GWAS loci from the largest published study. RESULTS Eighty-six rare missense or loss-of-function (LoF) variants completely segregated in 17.5% of families, but in 91 (22.1%) families Apolipoprotein E (APOE)-𝜀4 was the only variant segregating. However, in 60.3% of families, APOE 𝜀4, missense, and LoF variants were not found within the GWAS loci. DISCUSSION Although APOE 𝜀4and several rare variants were found to segregate in both family datasets, many families had no variant accounting for their disease. This suggests that familial AD may be the result of unidentified rare variants. HIGHLIGHTS Rare coding variants from GWAS loci segregate in familial Alzheimer's disease. Missense or loss of function variants were found segregating in nearly 7% of families. APOE-𝜀4 was the only segregating variant in 29.7% in familial Alzheimer's disease. In Hispanic and non-Hispanic families, different variants were found in segregating genes. No coding variants were found segregating in many Hispanic and non-Hispanic families.
Collapse
Affiliation(s)
- Tamil Iniyan Gunasekaran
- Department of NeurologyTaub Institute for Research on Alzheimer's Disease and the Aging Brain and the Gertrude H. Sergievsky CenterColumbia UniversityNew YorkNew YorkUSA
| | - Dolly Reyes‐Dumeyer
- Department of NeurologyTaub Institute for Research on Alzheimer's Disease and the Aging Brain and the Gertrude H. Sergievsky CenterColumbia UniversityNew YorkNew YorkUSA
| | - Kelley M. Faber
- Department of Medical and Molecular GeneticsNational Centralized Repository for Alzheimer's Disease and Related Dementias (NCRAD), 410 W. 10th St., HS 4000. Indiana University School of MedicineIndianapolisIndianaUSA
| | - Alison Goate
- Department of Genetics & Genomic SciencesRonald M. Loeb Center for Alzheimer's diseaseIcahn School of Medicine at Mount SinaiIcahn Bldg., One Gustave L. Levy PlaceNew YorkNew YorkUSA
| | - Brad Boeve
- Department of Neurology, Mayo ClinicRochesterMinnesotaUSA
| | - Carlos Cruchaga
- Department of PsychiatryWashington University in St. Louis, Rand Johnson Building, 600 S Euclid Ave., Wohl Hospital BuildingSt. LouisMissouriUSA
| | - Margaret Pericak‐Vance
- John P Hussman Institute for Human GenomicsDr. John T Macdonald Foundation Department of Human GeneticsUniversity of Miami Miller School of MedicineMiamiFloridaUSA
| | - Jonathan L. Haines
- Department of Population & Quantitative Health Sciences and Cleveland Institute for Computational Biology. Case Western Reserve UniversityClevelandOhioUSA
| | - Roger Rosenberg
- Department of NeurologyUniversity of Texas Southwestern Medical CenterDallasTexasUSA
| | - Debby Tsuang
- Department of Psychiatry and Behavioral SciencesUniversity of Washington, GRECC VA Puget Sound, 1660 South Columbian WaySeattleWashingtonUSA
| | - Diones Rivera Mejia
- Los Centros de Diagnóstico y Medicina Avanzada y de Conferencias Médicas y TelemedicinaCEDIMAT, Arturo LogroñoPlaza de la Salud, Dr. Juan Manuel Taveras Rodríguez, C. Pepillo Salcedo esqSanto DomingoDominican Republic
- Universidad Pedro Henríquez Urena, Av. John F. Kennedy Km. 7‐1/2 Santo Domingo 1423Santo DomingoDominican Republic
| | - Martin Medrano
- Pontíficia Universidad Católica Madre y Maestra (PUCMM), Autopista Duarte Km 1 1/2Santiago de los CaballerosDominican Republic
| | - Rafael A. Lantigua
- Department of NeurologyTaub Institute for Research on Alzheimer's Disease and the Aging Brain and the Gertrude H. Sergievsky CenterColumbia UniversityNew YorkNew YorkUSA
- Department of MedicineVagelos College of Physicians and SurgeonsColumbia University, and the New York Presbyterian HospitalNew YorkNew YorkUSA
| | - Robert A. Sweet
- Departments of Psychiatry and NeurologyUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - David A. Bennett
- Rush Alzheimer's Disease CenterRush University Medical Center, 1750, West Harrison StChicagoIllinoisUSA
| | - Robert S. Wilson
- Rush Alzheimer's Disease CenterRush University Medical Center, 1750, West Harrison StChicagoIllinoisUSA
| | - Camille Alba
- Department of AnatomyPhysiology and GeneticsUniformed Services University of the Health SciencesBethesdaMarylandUSA
| | - Clifton Dalgard
- Department of AnatomyPhysiology and GeneticsUniformed Services University of the Health SciencesBethesdaMarylandUSA
| | - Tatiana Foroud
- Department of Medical and Molecular GeneticsNational Centralized Repository for Alzheimer's Disease and Related Dementias (NCRAD), 410 W. 10th St., HS 4000. Indiana University School of MedicineIndianapolisIndianaUSA
| | - Badri N. Vardarajan
- Department of NeurologyTaub Institute for Research on Alzheimer's Disease and the Aging Brain and the Gertrude H. Sergievsky CenterColumbia UniversityNew YorkNew YorkUSA
| | - Richard Mayeux
- Department of NeurologyTaub Institute for Research on Alzheimer's Disease and the Aging Brain and the Gertrude H. Sergievsky CenterColumbia UniversityNew YorkNew YorkUSA
| |
Collapse
|
13
|
Shade LMP, Katsumata Y, Abner EL, Aung KZ, Claas SA, Qiao Q, Heberle BA, Brandon JA, Page ML, Hohman TJ, Mukherjee S, Mayeux RP, Farrer LA, Schellenberg GD, Haines JL, Kukull WA, Nho K, Saykin AJ, Bennett DA, Schneider JA, Ebbert MTW, Nelson PT, Fardo DW. GWAS of multiple neuropathology endophenotypes identifies new risk loci and provides insights into the genetic risk of dementia. Nat Genet 2024; 56:2407-2421. [PMID: 39379761 PMCID: PMC11549054 DOI: 10.1038/s41588-024-01939-9] [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: 05/05/2023] [Accepted: 08/30/2024] [Indexed: 10/10/2024]
Abstract
Genome-wide association studies (GWAS) have identified >80 Alzheimer's disease and related dementias (ADRD)-associated genetic loci. However, the clinical outcomes used in most previous studies belie the complex nature of underlying neuropathologies. Here we performed GWAS on 11 ADRD-related neuropathology endophenotypes with participants drawn from the following three sources: the National Alzheimer's Coordinating Center, the Religious Orders Study and Rush Memory and Aging Project, and the Adult Changes in Thought study (n = 7,804 total autopsied participants). We identified eight independent significantly associated loci, of which four were new (COL4A1, PIK3R5, LZTS1 and APOC2). Separately testing known ADRD loci, 19 loci were significantly associated with at least one neuropathology after false-discovery rate adjustment. Genetic colocalization analyses identified pleiotropic effects and quantitative trait loci. Methylation in the cerebral cortex at two sites near APOC2 was associated with cerebral amyloid angiopathy. Studies that include neuropathology endophenotypes are an important step in understanding the mechanisms underlying genetic ADRD risk.
Collapse
Affiliation(s)
- Lincoln M P Shade
- Department of Biostatistics, College of Public Health, University of Kentucky, Lexington, KY, USA
| | - Yuriko Katsumata
- Department of Biostatistics, College of Public Health, University of Kentucky, Lexington, KY, USA
- Sanders-Brown Center on Aging and Alzheimer's Disease Research Center, University of Kentucky, Lexington, KY, USA
| | - Erin L Abner
- Sanders-Brown Center on Aging and Alzheimer's Disease Research Center, University of Kentucky, Lexington, KY, USA
- Department of Epidemiology and Environmental Health, College of Public Health, University of Kentucky, Lexington, KY, USA
| | - Khine Zin Aung
- Department of Biostatistics, College of Public Health, University of Kentucky, Lexington, KY, USA
- Sanders-Brown Center on Aging and Alzheimer's Disease Research Center, University of Kentucky, Lexington, KY, USA
| | - Steven A Claas
- Department of Biostatistics, College of Public Health, University of Kentucky, Lexington, KY, USA
| | - Qi Qiao
- Department of Biostatistics, College of Public Health, University of Kentucky, Lexington, KY, USA
- Sanders-Brown Center on Aging and Alzheimer's Disease Research Center, University of Kentucky, Lexington, KY, USA
| | - Bernardo Aguzzoli Heberle
- Sanders-Brown Center on Aging and Alzheimer's Disease Research Center, University of Kentucky, Lexington, KY, USA
- Department of Neuroscience, University of Kentucky College of Medicine, Lexington, KY, USA
| | - J Anthony Brandon
- Sanders-Brown Center on Aging and Alzheimer's Disease Research Center, University of Kentucky, Lexington, KY, USA
- Department of Neuroscience, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Madeline L Page
- Sanders-Brown Center on Aging and Alzheimer's Disease Research Center, University of Kentucky, Lexington, KY, USA
- Department of Neuroscience, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Timothy J Hohman
- Vanderbilt Memory and Alzheimer's Center, Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
| | | | - Richard P Mayeux
- Department of Neurology, Columbia University, New York City, NY, USA
| | - Lindsay A Farrer
- Department of Medicine (Biomedical Genetics), Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
- Department of Neurology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
- Department of Ophthalmology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
- Department of Biostatistics, School of Public Health, Boston University, Boston, MA, USA
- Department of Epidemiology, School of Public Health, Boston University, Boston, MA, USA
| | - Gerard D Schellenberg
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Penn Neurodegeneration Genomics Center, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Jonathan L Haines
- Cleveland Institute for Computational Biology, Case Western Reserve University, Cleveland, OH, USA
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, OH, USA
| | - Walter A Kukull
- National Alzheimer's Coordinating Center, Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - Kwangsik Nho
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA
- Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, USA
- Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Andrew J Saykin
- Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, USA
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA
| | - David A Bennett
- Department of Neurological Sciences, Rush Medical College, Chicago, IL, USA
- Rush Alzheimer's Disease Center, Rush Medical College, Chicago, IL, USA
| | - Julie A Schneider
- Department of Neurological Sciences, Rush Medical College, Chicago, IL, USA
- Rush Alzheimer's Disease Center, Rush Medical College, Chicago, IL, USA
- Department of Pathology, Rush Medical College, Chicago, IL, USA
| | - Mark T W Ebbert
- Sanders-Brown Center on Aging and Alzheimer's Disease Research Center, University of Kentucky, Lexington, KY, USA
- Department of Neuroscience, University of Kentucky College of Medicine, Lexington, KY, USA
- Division of Biomedical Informatics, Department of Internal Medicine, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Peter T Nelson
- Sanders-Brown Center on Aging and Alzheimer's Disease Research Center, University of Kentucky, Lexington, KY, USA
- Department of Pathology and Laboratory Medicine, University of Kentucky College of Medicine, Lexington, KY, USA
| | - David W Fardo
- Department of Biostatistics, College of Public Health, University of Kentucky, Lexington, KY, USA.
- Sanders-Brown Center on Aging and Alzheimer's Disease Research Center, University of Kentucky, Lexington, KY, USA.
| |
Collapse
|
14
|
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, Alzheimer’s Disease Neuroimaging Initiative.. 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
|
15
|
Lee JW, Mizuno K, Watanabe H, Lee IH, Tsumita T, Hida K, Yawaka Y, Kitagawa Y, Hasebe A, Iimura T, Kong SW. Enhanced phagocytosis associated with multinucleated microglia via Pyk2 inhibition in an acute β-amyloid infusion model. J Neuroinflammation 2024; 21:196. [PMID: 39107821 PMCID: PMC11301859 DOI: 10.1186/s12974-024-03192-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 07/31/2024] [Indexed: 08/10/2024] Open
Abstract
Multinucleated microglia have been observed in contexts associated with infection, inflammation, and aging. Though commonly linked to pathological conditions, the larger cell size of multinucleated microglia might enhance their phagocytic functions, potentially aiding in the clearance of brain debris and suggesting a reassessment of their pathological significance. To assess the phagocytic capacity of multinucleated microglia and its implications for brain debris clearance, we induced their formation by inhibiting Pyk2 activity using the pharmacological inhibitor PF-431396, which triggers cytokinesis regression. Multinucleated microglia demonstrate enhanced phagocytic function, as evidenced by their increased capacity to engulf β-amyloid (Aβ) oligomers. Concurrently, the phosphorylation of Pyk2, induced by Aβ peptide, was diminished upon treatment with a Pyk2 inhibitor (Pyk2-Inh, PF-431396). Furthermore, the increased expression of Lamp1, a lysosomal marker, with Pyk2-inh treatment, suggests an enhancement in proteolytic activity. In vivo, we generated an acute Alzheimer's disease (AD) model by infusing Aβ into the brains of Iba-1 EGFP transgenic (Tg) mice. The administration of the Pyk2-Inh led to an increased migration of microglia toward amyloid deposits in the brains of Iba-1 EGFP Tg mice, accompanied by morphological activation, suggesting a heightened affinity for Aβ. In human microglia, lipopolysaccharide (LPS)-induced inflammatory responses showed that inhibition of Pyk2 signaling significantly reduced the transcription and protein expression of pro-inflammatory markers. These results suggest that Pyk2 inhibition can modulate microglial functions, potentially reducing neuroinflammation and aiding in the clearance of neurodegenerative disease markers. This highlights Pyk2 as a promising target for therapeutic intervention in neurodegenerative diseases.
Collapse
Affiliation(s)
- Ji-Won Lee
- Microbiology, Department of Oral Pathobiological Science, Faculty and Graduate School of Dental Medicine, Hokkaido University, Kita13 Nishi7, Kita-Ku, Sapporo, 060-8586, Japan.
| | - Kaito Mizuno
- Microbiology, Department of Oral Pathobiological Science, Faculty and Graduate School of Dental Medicine, Hokkaido University, Kita13 Nishi7, Kita-Ku, Sapporo, 060-8586, Japan
- Dentistry for Children and Disabled Persons, Department of Oral Functional Science, Faculty of Dental Medicine, Hokkaido University, Kita13 Nishi7, Kita-Ku, Sapporo, 060-8586, Japan
| | - Haruhisa Watanabe
- Department of Pharmacology, Faculty and Graduate School of Dental Medicine, Hokkaido University, Kita13 Nishi7, Kita-Ku, Sapporo, 060-8586, Japan
- Oral Diagnosis and Medicine, Department of Oral Pathobiological Science, Faculty of Dental Medicine, Hokkaido University, Kita13 Nishi7, Kita-Ku, Sapporo, 060-8586, Japan
| | - In-Hee Lee
- Computational Health and Informatics Program, Boston Children's Hospital, Boston, MA, 02215, USA
| | - Takuya Tsumita
- Department of Vascular Biology and Molecular Pathology, Faculty and Graduate School of Dental Medicine, Hokkaido University, Kita13 Nishi7, Kita-Ku, Sapporo, 060-8586, Japan
| | - Kyoko Hida
- Department of Vascular Biology and Molecular Pathology, Faculty and Graduate School of Dental Medicine, Hokkaido University, Kita13 Nishi7, Kita-Ku, Sapporo, 060-8586, Japan
| | - Yasutaka Yawaka
- Dentistry for Children and Disabled Persons, Department of Oral Functional Science, Faculty of Dental Medicine, Hokkaido University, Kita13 Nishi7, Kita-Ku, Sapporo, 060-8586, Japan
| | - Yoshimasa Kitagawa
- Oral Diagnosis and Medicine, Department of Oral Pathobiological Science, Faculty of Dental Medicine, Hokkaido University, Kita13 Nishi7, Kita-Ku, Sapporo, 060-8586, Japan
| | - Akira Hasebe
- Microbiology, Department of Oral Pathobiological Science, Faculty and Graduate School of Dental Medicine, Hokkaido University, Kita13 Nishi7, Kita-Ku, Sapporo, 060-8586, Japan
| | - Tadahiro Iimura
- Department of Pharmacology, Faculty and Graduate School of Dental Medicine, Hokkaido University, Kita13 Nishi7, Kita-Ku, Sapporo, 060-8586, Japan
| | - Sek Won Kong
- Computational Health and Informatics Program, Boston Children's Hospital, Boston, MA, 02215, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, 02115, USA
| |
Collapse
|
16
|
Gunasekaran TI, Reyes-Dumeyer D, Faber KM, Goate A, Boeve B, Cruchaga C, Pericak-Vance M, Haines JL, Rosenberg R, Tsuang D, Mejia DR, Medrano M, Lantigua RA, Sweet RA, Bennett DA, Wilson RS, Alba C, Dalgard C, Foroud T, Vardarajan BN, Mayeux R. Missense and Loss of Function Variants at GWAS Loci in Familial Alzheimer's Disease. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2023.12.18.23300145. [PMID: 38196599 PMCID: PMC10775337 DOI: 10.1101/2023.12.18.23300145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2024]
Abstract
BACKGROUND Few rare variants have been identified in genetic loci from genome wide association studies of Alzheimer's disease (AD), limiting understanding of mechanisms and risk assessment, and genetic counseling. METHODS Using genome sequencing data from 197 families in The NIA Alzheimer's Disease Family Based Study, and 214 Caribbean Hispanic families, we searched for rare coding variants within known GWAS loci from the largest published study. RESULTS Eighty-six rare missense or loss of function (LoF) variants completely segregated in 17.5% of families, but in 91 (22.1%) of families APOE-e4 was the only variant segregating. However, in 60.3% of families neither APOE-e4 nor missense or LoF variants were found within the GWAS loci. DISCUSSION Although APOE-ε4 and several rare variants were found to segregate in both family datasets, many families had no variant accounting for their disease. This suggests that familial AD may be the result of unidentified rare variants.
Collapse
|
17
|
Rodriguez-Rodriguez P, Arroyo-Garcia LE, Tsagkogianni C, Li L, Wang W, Végvári Á, Salas-Allende I, Plautz Z, Cedazo-Minguez A, Sinha SC, Troyanskaya O, Flajolet M, Yao V, Roussarie JP. A cell autonomous regulator of neuronal excitability modulates tau in Alzheimer's disease vulnerable neurons. Brain 2024; 147:2384-2399. [PMID: 38462574 PMCID: PMC11224620 DOI: 10.1093/brain/awae051] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 01/12/2024] [Accepted: 01/19/2024] [Indexed: 03/12/2024] Open
Abstract
Neurons from layer II of the entorhinal cortex (ECII) are the first to accumulate tau protein aggregates and degenerate during prodromal Alzheimer's disease. Gaining insight into the molecular mechanisms underlying this vulnerability will help reveal genes and pathways at play during incipient stages of the disease. Here, we use a data-driven functional genomics approach to model ECII neurons in silico and identify the proto-oncogene DEK as a regulator of tau pathology. We show that epigenetic changes caused by Dek silencing alter activity-induced transcription, with major effects on neuronal excitability. This is accompanied by the gradual accumulation of tau in the somatodendritic compartment of mouse ECII neurons in vivo, reactivity of surrounding microglia, and microglia-mediated neuron loss. These features are all characteristic of early Alzheimer's disease. The existence of a cell-autonomous mechanism linking Alzheimer's disease pathogenic mechanisms in the precise neuron type where the disease starts provides unique evidence that synaptic homeostasis dysregulation is of central importance in the onset of tau pathology in Alzheimer's disease.
Collapse
Affiliation(s)
| | | | - Christina Tsagkogianni
- Department of Neurobiology Care Sciences and Society, Karolinska Institutet, 17 164, Solna, Sweden
| | - Lechuan Li
- Department of Computer Science, Rice University, Houston, TX 77004, USA
| | - Wei Wang
- Bioinformatics Resource Center, The Rockefeller University, New York, NY 10065, USA
| | - Ákos Végvári
- Division of Chemistry I, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 17 164, Solna, Sweden
| | - Isabella Salas-Allende
- Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, New York, NY 10065, USA
| | - Zakary Plautz
- Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, New York, NY 10065, USA
| | - Angel Cedazo-Minguez
- Department of Neurobiology Care Sciences and Society, Karolinska Institutet, 17 164, Solna, Sweden
| | - Subhash C Sinha
- Helen and Robert Appel Alzheimer’s Disease Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10065, USA
| | - Olga Troyanskaya
- Department of Computer Science, Princeton University, Princeton, NJ 08540, USA
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA
- Center for Computational Biology, Flatiron Institute, Simons Foundation, New York, NY 10010, USA
| | - Marc Flajolet
- Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, New York, NY 10065, USA
| | - Vicky Yao
- Division of Chemistry I, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 17 164, Solna, Sweden
| | - Jean-Pierre Roussarie
- Department of Anatomy & Neurobiology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA 02118, USA
| |
Collapse
|
18
|
Tomé SO, Gawor K, Thal DR. LATE-NC in Alzheimer's disease: Molecular aspects and synergies. Brain Pathol 2024; 34:e13213. [PMID: 37793659 PMCID: PMC11189776 DOI: 10.1111/bpa.13213] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 09/01/2023] [Indexed: 10/06/2023] Open
Abstract
Alzheimer's disease (AD) is classically characterized by senile plaques and neurofibrillary tangles (NFTs). However, multiple copathologies can be observed in the AD brain and contribute to the development of cognitive decline. Limbic-predominant age-related TDP-43 encephalopathy neuropathological changes (LATE-NC) accumulates in the majority of AD cases and leads to more severe cognitive decline compared with AD pathology alone. In this review, we focus on the synergistic relationship between LATE-NC and tau in AD, highlighting the aggravating role of TDP-43 aggregates on tau pathogenesis and its impact on the clinical picture and therapeutic strategies. Additionally, we discuss to what extent the molecular patterns of LATE-NC in AD differ from frontotemporal lobar degeneration with TDP-43 pathology (FTLD-TDP) neuropathological changes. Thus, we highlight the importance of tau and TDP-43 synergies for subtyping AD patients, which may respond differently to therapeutic interventions depending on the presence of comorbid LATE-NC.
Collapse
Grants
- 10810 Alzheimer Forschung Initiative (Germany)
- 13803 Alzheimer Forschung Initiative (Germany)
- 22-AAIIA-963171 Alzheimer's Association (USA)
- A2022019F BrightFocus Foundation (USA)
- TH-624-4-1 Deutsche Forschungsgemeinschaft (DFG, Germany)
- 4-2 Deutsche Forschungsgemeinschaft (DFG, Germany)
- 6-1 Deutsche Forschungsgemeinschaft (DFG, Germany)
- G065721N Fonds Wetenschappelijk Onderzoek (FWO, Belgium)
- G0F8516N Fonds Wetenschappelijk Onderzoek (FWO, Belgium)
- 2020/017 Stichting Alzheimer Onderzoek (SAO/FRA, Belgium)
- C3/20/057 Onderzoeksraad, KU Leuven (Belgium)
- PDMT2/21/069 Onderzoeksraad, KU Leuven (Belgium)
- IWT 135043 Vlaamse Impulsfinanciering voor Netwerken voor Dementie-onderzoek (Belgium)
- Alzheimer Forschung Initiative (Germany)
- Alzheimer's Association (USA)
- BrightFocus Foundation (USA)
- Deutsche Forschungsgemeinschaft (DFG, Germany)
- Fonds Wetenschappelijk Onderzoek (FWO, Belgium)
- Onderzoeksraad, KU Leuven (Belgium)
Collapse
Affiliation(s)
- Sandra O. Tomé
- Laboratory for Neuropathology, Department of Imaging and Pathology and Leuven Brain InstituteKU LeuvenLeuvenBelgium
| | - Klara Gawor
- Laboratory for Neuropathology, Department of Imaging and Pathology and Leuven Brain InstituteKU LeuvenLeuvenBelgium
| | - Dietmar Rudolf Thal
- Laboratory for Neuropathology, Department of Imaging and Pathology and Leuven Brain InstituteKU LeuvenLeuvenBelgium
- Department of PathologyUniversity Hospitals of LeuvenLeuvenBelgium
| |
Collapse
|
19
|
Huang YH, Chen YC, Ho WM, Lee RG, Chung RH, Liu YL, Chang PY, Chang SC, Wang CW, Chung WH, Tsai SJ, Kuo PH, Lee YS, Hsiao CC. Classifying Alzheimer's disease and normal subjects using machine learning techniques and genetic-environmental features. J Formos Med Assoc 2024; 123:701-709. [PMID: 38044212 DOI: 10.1016/j.jfma.2023.10.021] [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: 06/01/2023] [Revised: 08/24/2023] [Accepted: 10/23/2023] [Indexed: 12/05/2023] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is complicated by multiple environmental and polygenetic factors. The accuracy of artificial neural networks (ANNs) incorporating the common factors for identifying AD has not been evaluated. METHODS A total of 184 probable AD patients and 3773 healthy individuals aged 65 and over were enrolled. AD-related genes (51 SNPs) and 8 environmental factors were selected as features for multilayer ANN modeling. Random Forest (RF) and Support Vector Machine with RBF kernel (SVM) were also employed for comparison. Model results were verified using traditional statistics. RESULTS The ANN achieved high accuracy (0.98), sensitivity (0.95), and specificity (0.96) in the intrinsic test for AD classification. Excluding age and genetic data still yielded favorable results (accuracy: 0.97, sensitivity: 0.94, specificity: 0.96). The assigned weights to ANN features highlighted the importance of mental evaluation, years of education, and specific genetic variations (CASS4 rs7274581, PICALM rs3851179, and TOMM40 rs2075650) for AD classification. Receiver operating characteristic analysis revealed AUC values of 0.99 (intrinsic test), 0.60 (TWB-GWA), and 0.72 (CG-WGS), with slightly lower AUC values (0.96, 0.80, 0.52) when excluding age in ANN. The performance of the ANN model in AD classification was comparable to RF, SVM (linear kernel), and SVM (RBF kernel). CONCLUSION The ANN model demonstrated good sensitivity, specificity, and accuracy in AD classification. The top-weighted SNPs for AD prediction were CASS4 rs7274581, PICALM rs3851179, and TOMM40 rs2075650. The ANN model performed similarly to RF and SVM, indicating its capability to handle the complexity of AD as a disease entity.
Collapse
Affiliation(s)
- Yu-Hua Huang
- Department of Neurology, Chang Gung Memorial Hospital Linkou Medical Center and College of Medicine, Chang-Gung University, Taoyuan, Taiwan
| | - Yi-Chun Chen
- Department of Neurology, Chang Gung Memorial Hospital Linkou Medical Center and College of Medicine, Chang-Gung University, Taoyuan, Taiwan
| | - Wei-Min Ho
- Department of Neurology, Chang Gung Memorial Hospital Linkou Medical Center and College of Medicine, Chang-Gung University, Taoyuan, Taiwan
| | - Ren-Guey Lee
- Department of Electronics Engineering, National Taipei University of Technology, Taipei, Taiwan
| | - Ren-Hua Chung
- Division of Biostatistics and Bioinformatics, Institute of Population Health Sciences, National Health Research Institutes, Zhunan, Taiwan
| | - Yu-Li Liu
- Center for Neuropsychiatric Research, National Health Research Institutes, Miaoli, Taiwan
| | - Pi-Yueh Chang
- Department of Laboratory Medicine, Chang Gung Memorial Hospital Linkou Medical Center, Taoyuan, Taiwan; Department of Medical Biotechnology and Laboratory Science, Chang Gung University, Taoyuan, Taiwan
| | - Shih-Cheng Chang
- Department of Laboratory Medicine, Chang Gung Memorial Hospital Linkou Medical Center, Taoyuan, Taiwan; Department of Medical Biotechnology and Laboratory Science, Chang Gung University, Taoyuan, Taiwan
| | - Chaung-Wei Wang
- Department of Dermatology, Drug Hypersensitivity Clinical and Research Center, Chang Gung Memorial Hospital, Linkou, Taipei and Keelung, Taiwan; Cancer Vaccine and Immune Cell Therapy Core Laboratory, Chang Gung Memorial Hospital, Linkou, Taiwan; Whole-Genome Research Core Laboratory of Human Diseases, Chang Gung Memorial Hospital, Keelung, Taiwan
| | - Wen-Hung Chung
- Department of Dermatology, Drug Hypersensitivity Clinical and Research Center, Chang Gung Memorial Hospital, Linkou, Taipei and Keelung, Taiwan; Cancer Vaccine and Immune Cell Therapy Core Laboratory, Chang Gung Memorial Hospital, Linkou, Taiwan; Whole-Genome Research Core Laboratory of Human Diseases, Chang Gung Memorial Hospital, Keelung, Taiwan
| | - Shih-Jen Tsai
- Department of Psychiatry, Taipei Veterans General Hospital, Taipei, Taiwan; Division of Psychiatry, National Yang-Ming University, Taipei, Taiwan
| | - Po-Hsiu Kuo
- Department of Public Health, Institute of Epidemiology and Preventive Medicine, National Taiwan University, Taipei, Taiwan
| | - Yun-Shien Lee
- Department of Biotechnology, Ming Chuan University, Taoyuan, Taiwan; Genomic Medicine Research Core Laboratory, Chang Gung Memorial Hospital, Taoyuan, Taiwan.
| | - Chun-Chieh Hsiao
- Department of Electrical Engineering, National Taiwan University, Taipei, Taiwan; Department of Computer Information and Network Engineering, Lunghwa University of Science and Technology, Taoyuan, Taiwan.
| |
Collapse
|
20
|
Yan D, Hu B, Darst BF, Mukherjee S, Kunkle BW, Deming Y, Dumitrescu L, Wang Y, Naj A, Kuzma A, Zhao Y, Kang H, Johnson SC, Carlos C, Hohman TJ, Crane PK, Engelman CD, Alzheimer’s Disease Genetics Consortium (ADGC), Lu Q. Biobank-wide association scan identifies risk factors for late-onset Alzheimer's disease and endophenotypes. eLife 2024; 12:RP91360. [PMID: 38787369 PMCID: PMC11126309 DOI: 10.7554/elife.91360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/25/2024] Open
Abstract
Rich data from large biobanks, coupled with increasingly accessible association statistics from genome-wide association studies (GWAS), provide great opportunities to dissect the complex relationships among human traits and diseases. We introduce BADGERS, a powerful method to perform polygenic score-based biobank-wide association scans. Compared to traditional approaches, BADGERS uses GWAS summary statistics as input and does not require multiple traits to be measured in the same cohort. We applied BADGERS to two independent datasets for late-onset Alzheimer's disease (AD; n=61,212). Among 1738 traits in the UK biobank, we identified 48 significant associations for AD. Family history, high cholesterol, and numerous traits related to intelligence and education showed strong and independent associations with AD. Furthermore, we identified 41 significant associations for a variety of AD endophenotypes. While family history and high cholesterol were strongly associated with AD subgroups and pathologies, only intelligence and education-related traits predicted pre-clinical cognitive phenotypes. These results provide novel insights into the distinct biological processes underlying various risk factors for AD.
Collapse
Affiliation(s)
- Donghui Yan
- University of Wisconsin-MadisonMadisonUnited States
| | - Bowen Hu
- Department of Statistics, University of Wisconsin-MadisonMadisonUnited States
| | - Burcu F Darst
- Department of Population Health Sciences, University of Wisconsin-MadisonMadisonUnited States
| | - Shubhabrata Mukherjee
- Division of General Internal Medicine, Department of Medicine, University of WashingtonSeattleUnited States
| | - Brian W Kunkle
- University of Miami Miller School of MedicineMiamiUnited States
| | - Yuetiva Deming
- Department of Population Health Sciences, University of Wisconsin-MadisonMadisonUnited States
| | - Logan Dumitrescu
- Vanderbilt Memory and Alzheimer’s Center, Vanderbilt University Medical Center, Vanderbilt University School of MedicineNashvilleUnited States
| | - Yunling Wang
- University of Wisconsin-MadisonMadisonUnited States
| | - Adam Naj
- School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - Amanda Kuzma
- School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - Yi Zhao
- School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - Hyunseung Kang
- Department of Statistics, University of Wisconsin-MadisonMadisonUnited States
| | - Sterling C Johnson
- Wisconsin Alzheimer’s Institute, University of Wisconsin School of Medicine and Public HealthMadisonUnited States
- Geriatric Research Education and Clinical Center, Wm. S. Middleton Memorial VA HospitalMadisonUnited States
- Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public HealthMadisonUnited States
| | - Cruchaga Carlos
- Department of Psychiatry, Washington University in St. LouisSt. LouisUnited States
| | - Timothy J Hohman
- Vanderbilt Memory and Alzheimer’s Center, Vanderbilt University Medical Center, Vanderbilt University School of MedicineNashvilleUnited States
| | - Paul K Crane
- Division of General Internal Medicine, Department of Medicine, University of WashingtonSeattleUnited States
| | - Corinne D Engelman
- Department of Population Health Sciences, University of Wisconsin-MadisonMadisonUnited States
- Wisconsin Alzheimer’s Institute, University of Wisconsin School of Medicine and Public HealthMadisonUnited States
- Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public HealthMadisonUnited States
| | | | - Qiongshi Lu
- Department of Statistics, University of Wisconsin-MadisonMadisonUnited States
- Department of Biostatistics and Medical Informatics, University of Wisconsin-MadisonMadisonUnited States
| |
Collapse
|
21
|
Nelson PT, Fardo DW, Wu X, Aung KZ, Cykowski MD, Katsumata Y. Limbic-predominant age-related TDP-43 encephalopathy (LATE-NC): Co-pathologies and genetic risk factors provide clues about pathogenesis. J Neuropathol Exp Neurol 2024; 83:396-415. [PMID: 38613823 PMCID: PMC11110076 DOI: 10.1093/jnen/nlae032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/15/2024] Open
Abstract
Limbic-predominant age-related TDP-43 encephalopathy neuropathologic change (LATE-NC) is detectable at autopsy in more than one-third of people beyond age 85 years and is robustly associated with dementia independent of other pathologies. Although LATE-NC has a large impact on public health, there remain uncertainties about the underlying biologic mechanisms. Here, we review the literature from human studies that may shed light on pathogenetic mechanisms. It is increasingly clear that certain combinations of pathologic changes tend to coexist in aging brains. Although "pure" LATE-NC is not rare, LATE-NC often coexists in the same brains with Alzheimer disease neuropathologic change, brain arteriolosclerosis, hippocampal sclerosis of aging, and/or age-related tau astrogliopathy (ARTAG). The patterns of pathologic comorbidities provide circumstantial evidence of mechanistic interactions ("synergies") between the pathologies, and also suggest common upstream influences. As to primary mediators of vulnerability to neuropathologic changes, genetics may play key roles. Genes associated with LATE-NC include TMEM106B, GRN, APOE, SORL1, ABCC9, and others. Although the anatomic distribution of TDP-43 pathology defines the condition, important cofactors for LATE-NC may include Tau pathology, endolysosomal pathways, and blood-brain barrier dysfunction. A review of the human phenomenology offers insights into disease-driving mechanisms, and may provide clues for diagnostic and therapeutic targets.
Collapse
Affiliation(s)
- Peter T Nelson
- Department of Pathology and Laboratory Medicine, University of Kentucky, Lexington, Kentucky, USA
- Department of Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, USA
| | - David W Fardo
- Department of Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, USA
- Department of Biostatistics, University of Kentucky, Lexington, Kentucky, USA
| | - Xian Wu
- Department of Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, USA
- Department of Biostatistics, University of Kentucky, Lexington, Kentucky, USA
| | - Khine Zin Aung
- Department of Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, USA
- Department of Biostatistics, University of Kentucky, Lexington, Kentucky, USA
| | - Matthew D Cykowski
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Houston, Texas, USA
| | - Yuriko Katsumata
- Department of Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, USA
- Department of Biostatistics, University of Kentucky, Lexington, Kentucky, USA
| |
Collapse
|
22
|
Malamon JS, Farrell JJ, Xia LC, Dombroski BA, Das RG, Way J, Kuzma AB, Valladares O, Leung YY, Scanlon AJ, Lopez IAB, Brehony J, Worley KC, Zhang NR, Wang LS, Farrer LA, Schellenberg GD, Lee WP, Vardarajan BN. A comparative study of structural variant calling in WGS from Alzheimer's disease families. Life Sci Alliance 2024; 7:e202302181. [PMID: 38418088 PMCID: PMC10902710 DOI: 10.26508/lsa.202302181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 02/07/2024] [Accepted: 02/08/2024] [Indexed: 03/01/2024] Open
Abstract
Detecting structural variants (SVs) in whole-genome sequencing poses significant challenges. We present a protocol for variant calling, merging, genotyping, sensitivity analysis, and laboratory validation for generating a high-quality SV call set in whole-genome sequencing from the Alzheimer's Disease Sequencing Project comprising 578 individuals from 111 families. Employing two complementary pipelines, Scalpel and Parliament, for SV/indel calling, we assessed sensitivity through sample replicates (N = 9) with in silico variant spike-ins. We developed a novel metric, D-score, to evaluate caller specificity for deletions. The accuracy of deletions was evaluated by Sanger sequencing. We generated a high-quality call set of 152,301 deletions of diverse sizes. Sanger sequencing validated 114 of 146 detected deletions (78.1%). Scalpel excelled in accuracy for deletions ≤100 bp, whereas Parliament was optimal for deletions >900 bp. Overall, 83.0% and 72.5% of calls by Scalpel and Parliament were validated, respectively, including all 11 deletions called by both Parliament and Scalpel between 101 and 900 bp. Our flexible protocol successfully generated a high-quality deletion call set and a truth set of Sanger sequencing-validated deletions with precise breakpoints spanning 1-17,000 bp.
Collapse
Affiliation(s)
- John S Malamon
- Department of Pathology and Laboratory Medicine, Penn Neurodegeneration Genomics Center, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - John J Farrell
- Biomedical Genetics Section, Department of Medicine, Boston University School of Medicine, Boston University, Boston, MA, USA
| | - Li Charlie Xia
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Statistics, The Wharton School, University of Pennsylvania, Philadelphia, PA, USA
| | - Beth A Dombroski
- Department of Pathology and Laboratory Medicine, Penn Neurodegeneration Genomics Center, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Rueben G Das
- Department of Pathology and Laboratory Medicine, Penn Neurodegeneration Genomics Center, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Jessica Way
- Broad Institute, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Amanda B Kuzma
- Department of Pathology and Laboratory Medicine, Penn Neurodegeneration Genomics Center, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Otto Valladares
- Department of Pathology and Laboratory Medicine, Penn Neurodegeneration Genomics Center, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Yuk Yee Leung
- Department of Pathology and Laboratory Medicine, Penn Neurodegeneration Genomics Center, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Allison J Scanlon
- Department of Pathology and Laboratory Medicine, Penn Neurodegeneration Genomics Center, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Irving Antonio Barrera Lopez
- Department of Pathology and Laboratory Medicine, Penn Neurodegeneration Genomics Center, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Jack Brehony
- Department of Pathology and Laboratory Medicine, Penn Neurodegeneration Genomics Center, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Kim C Worley
- Human Genome Sequencing Center, and Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Nancy R Zhang
- Department of Statistics, The Wharton School, University of Pennsylvania, Philadelphia, PA, USA
| | - Li-San Wang
- Department of Pathology and Laboratory Medicine, Penn Neurodegeneration Genomics Center, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Lindsay A Farrer
- Biomedical Genetics Section, Department of Medicine, Boston University School of Medicine, Boston University, Boston, MA, USA
- Departments of Neurology and Ophthalmology, Boston University School of Medicine, Boston University, Boston, MA, USA
- Departments of Epidemiology and Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Gerard D Schellenberg
- Department of Pathology and Laboratory Medicine, Penn Neurodegeneration Genomics Center, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Wan-Ping Lee
- Department of Pathology and Laboratory Medicine, Penn Neurodegeneration Genomics Center, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Badri N Vardarajan
- Gertrude H. Sergievsky Center and Taub Institute of Aging Brain, Department of Neurology, Columbia University Medical Center, New York, NY, USA
| |
Collapse
|
23
|
Pillai JA, Bena J, Tousi B, Rothenberg K, Keene CD, Leverenz JB. Lewy body pathology modifies risk factors for cerebral amyloid angiopathy when comorbid with Alzheimer's disease pathology. Alzheimers Dement 2024; 20:2564-2574. [PMID: 38353367 PMCID: PMC11032524 DOI: 10.1002/alz.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] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 11/28/2023] [Accepted: 12/16/2023] [Indexed: 02/18/2024]
Abstract
INTRODUCTION Cerebral amyloid angiopathy (CAA) often accompanies dementia-associated pathologies and is important in the context of anti-amyloid monoclonal therapies and risk of hemorrhage. METHODS We conducted a retrospective neuropathology-confirmed study of 2384 participants in the National Alzheimer Coordinating Center cohort (Alzheimer's disease [AD], n = 1175; Lewy body pathology [LBP], n = 316; and mixed AD and LBP [AD-LBP], n = 893). We used logistic regression to evaluate age, sex, education, APOE ε4, neuritic plaques, and neurofibrillary tangles (NFTs) in CAA risk. RESULTS APOE ε4 increased CAA risk in all three groups, while younger age and higher NFT stages increased risk in AD and AD-LBP. In AD-LBP, male sex and lower education were additional risk factors. The odds of APOE ε4 carrier homozygosity related to CAA was higher in LBP (25.69) and AD-LBP (9.50) than AD (3.17). DISCUSSION AD and LBPs modify risk factors for CAA and should be considered in reviewing the risk of CAA. HIGHLIGHTS Lewy body pathology modifies risk factors for cerebral amyloid angiopathy (CAA) when present along with Alzheimer's disease (AD) neuropathology. In the context of anti-amyloid monoclonal therapies and their associated risks for hemorrhage, the risk of underlying CAA in mixed dementia with Lewy body pathology needs to be considered.
Collapse
Affiliation(s)
- Jagan A. Pillai
- Lou Ruvo Center for Brain HealthCleveland ClinicClevelandOhioUSA
- Neurological InstituteCleveland ClinicClevelandOhioUSA
- Department of NeurologyCleveland ClinicClevelandOhioUSA
| | - James Bena
- Quantitative Health SciencesCleveland ClinicClevelandOhioUSA
| | - Babak Tousi
- Lou Ruvo Center for Brain HealthCleveland ClinicClevelandOhioUSA
- Neurological InstituteCleveland ClinicClevelandOhioUSA
| | - Kasia Rothenberg
- Lou Ruvo Center for Brain HealthCleveland ClinicClevelandOhioUSA
- Neurological InstituteCleveland ClinicClevelandOhioUSA
| | - C. Dirk Keene
- Department of Laboratory Medicine and PathologyUniversity of Washington School of MedicineSeattleWashingtonUSA
| | - James B. Leverenz
- Lou Ruvo Center for Brain HealthCleveland ClinicClevelandOhioUSA
- Neurological InstituteCleveland ClinicClevelandOhioUSA
- Department of NeurologyCleveland ClinicClevelandOhioUSA
| |
Collapse
|
24
|
Yazdi MK, Alavi MS, Roohbakhsh A. The role of ATP-binding cassette transporter G1 (ABCG1) in Alzheimer's disease: A review of the mechanisms. Basic Clin Pharmacol Toxicol 2024; 134:423-438. [PMID: 38275217 DOI: 10.1111/bcpt.13981] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 01/04/2024] [Accepted: 01/08/2024] [Indexed: 01/27/2024]
Abstract
The maintenance of cholesterol homeostasis is essential for central nervous system function. Consequently, factors that affect cholesterol homeostasis are linked to neurological disorders and pathologies. Among them, ATP-binding cassette transporter G1 (ABCG1) plays a significant role in atherosclerosis. However, its role in Alzheimer's disease (AD) is unclear. There is inconsistent information regarding ABCG1's role in AD. It can increase or decrease amyloid β (Aβ) levels in animals' brains. Clinical studies show that ABCG1 is involved in AD patients' impairment of cholesterol efflux capacity (CEC) in the cerebrospinal fluid (CSF). Lower Aβ levels in the CSF are correlated with ABCG1-mediated CEC dysfunction. ABCG1 modulates α-, β-, and γ-secretase activities in the plasma membrane and may affect Aβ production in the mitochondria-associated endoplasmic reticulum (ER) membrane (MAM) cell compartment. Despite contradictory findings regarding ABCG1's role in AD, this review shows that ABCG1 has a role in Aβ generation via modulation of membrane secretases. It is, however, necessary to investigate the underlying mechanism(s). ABCG1 may also contribute to AD pathology through its role in apoptosis and oxidative stress. As a result, ABCG1 plays a role in AD and is a candidate for drug development.
Collapse
Affiliation(s)
- Mohsen Karbasi Yazdi
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohaddeseh Sadat Alavi
- Pharmacological Research Center of Medicinal Plants, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Pharmacology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ali Roohbakhsh
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
25
|
Ho P, Yu WH, Tee BL, Lee W, Li C, Gu Y, Yokoyama JS, Reyes‐Dumeyer D, Choi Y, Yang H, Vardarajan BN, Tzuang M, Lieu K, Lu A, Faber KM, Potter ZD, Revta C, Kirsch M, McCallum J, Mei D, Booth B, Cantwell LB, Chen F, Chou S, Clark D, Deng M, Hong TH, Hwang L, Jiang L, Joo Y, Kang Y, Kim ES, Kim H, Kim K, Kuzma AB, Lam E, Lanata SC, Lee K, Li D, Li M, Li X, Liu C, Liu C, Liu L, Lupo J, Nguyen K, Pfleuger SE, Qian J, Qian W, Ramirez V, Russ KA, Seo EH, Song YE, Tartaglia MC, Tian L, Torres M, Vo N, Wong EC, Xie Y, Yau EB, Yi I, Yu V, Zeng X, St George‐Hyslop P, Au R, Schellenberg GD, Dage JL, Varma R, Hsiung GR, Rosen H, Henderson VW, Foroud T, Kukull WA, Peavy GM, Lee H, Feldman HH, Mayeux R, Chui H, Jun GR, Ta Park VM, Chow TW, Wang L. Asian Cohort for Alzheimer's Disease (ACAD) pilot study on genetic and non-genetic risk factors for Alzheimer's disease among Asian Americans and Canadians. Alzheimers Dement 2024; 20:2058-2071. [PMID: 38215053 PMCID: PMC10984480 DOI: 10.1002/alz.13611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 09/25/2023] [Accepted: 11/27/2023] [Indexed: 01/14/2024]
Abstract
INTRODUCTION Clinical research in Alzheimer's disease (AD) lacks cohort diversity despite being a global health crisis. The Asian Cohort for Alzheimer's Disease (ACAD) was formed to address underrepresentation of Asians in research, and limited understanding of how genetics and non-genetic/lifestyle factors impact this multi-ethnic population. METHODS The ACAD started fully recruiting in October 2021 with one central coordination site, eight recruitment sites, and two analysis sites. We developed a comprehensive study protocol for outreach and recruitment, an extensive data collection packet, and a centralized data management system, in English, Chinese, Korean, and Vietnamese. RESULTS ACAD has recruited 606 participants with an additional 900 expressing interest in enrollment since program inception. DISCUSSION ACAD's traction indicates the feasibility of recruiting Asians for clinical research to enhance understanding of AD risk factors. ACAD will recruit > 5000 participants to identify genetic and non-genetic/lifestyle AD risk factors, establish blood biomarker levels for AD diagnosis, and facilitate clinical trial readiness. HIGHLIGHTS The Asian Cohort for Alzheimer's Disease (ACAD) promotes awareness of under-investment in clinical research for Asians. We are recruiting Asian Americans and Canadians for novel insights into Alzheimer's disease. We describe culturally appropriate recruitment strategies and data collection protocol. ACAD addresses challenges of recruitment from heterogeneous Asian subcommunities. We aim to implement a successful recruitment program that enrolls across three Asian subcommunities.
Collapse
Affiliation(s)
- Pei‐Chuan Ho
- Penn Neurodegeneration Genomics CenterDepartment of PathologyPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- The Leonard Davis Institute of Health EconomicsUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Wai Haung Yu
- Brain Health and Imaging Center and Geriatric Mental Health ServicesCentre for Addiction and Mental HealthTorontoOntarioCanada
- Department of Pharmacology and ToxicologyUniversity of TorontoTorontoOntarioCanada
| | - Boon Lead Tee
- Memory and Aging CenterDepartment of NeurologyWeill Institute for NeurosciencesUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
- Global Brain Health InstituteUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
| | - Wan‐Ping Lee
- Penn Neurodegeneration Genomics CenterDepartment of PathologyPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Clara Li
- Alzheimer's Disease Research CenterDepartment of PsychiatryIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Yian Gu
- Department of NeurologyColumbia University Medical CenterNew YorkNew YorkUSA
| | - Jennifer S. Yokoyama
- Memory and Aging CenterDepartment of NeurologyWeill Institute for NeurosciencesUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
- Department of Radiology and Biomedical ImagingUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
| | - Dolly Reyes‐Dumeyer
- Gertrude H. Sergievsky CenterTaub Institute of Aging Brain and Department of Neurology at Columbia UniversityNew YorkNew YorkUSA
| | - Yun‐Beom Choi
- Englewood HealthEnglewoodNew JerseyUSA
- Department of NeurologyRutgers New Jersey Medical SchoolNewarkNew JerseyUSA
| | - Hyun‐Sik Yang
- Center for Alzheimer Research and TreatmentDepartment of NeurologyBrigham and Women's HospitalBostonMassachusettsUSA
- Harvard Medical SchoolBostonMassachusettsUSA
- Broad Institute of MIT and HarvardCambridgeMassachusettsUSA
| | - Badri N. Vardarajan
- Gertrude H. Sergievsky CenterTaub Institute of Aging Brain and Department of Neurology at Columbia UniversityNew YorkNew YorkUSA
| | - Marian Tzuang
- Department of Community Health SystemsUniversity of California San Francisco School of NursingSan FranciscoCaliforniaUSA
| | - Kevin Lieu
- Memory and Aging CenterDepartment of NeurologyWeill Institute for NeurosciencesUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
| | - Anna Lu
- Alzheimer's Disease Cooperative StudyUniversity of California, San DiegoLa JollaCaliforniaUSA
| | - Kelley M. Faber
- Department of Medical and Molecular GeneticsIndiana University School of MedicineIndianapolisIndianaUSA
| | - Zoë D. Potter
- Department of Medical and Molecular GeneticsIndiana University School of MedicineIndianapolisIndianaUSA
| | - Carolyn Revta
- Alzheimer's Disease Cooperative StudyUniversity of California, San DiegoLa JollaCaliforniaUSA
| | - Maureen Kirsch
- Penn Neurodegeneration Genomics CenterDepartment of PathologyPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Jake McCallum
- Geriatric Mental Health Services, Centre for Addiction and Mental HealthTorontoOntarioCanada
| | - Diana Mei
- Memory and Aging CenterDepartment of NeurologyWeill Institute for NeurosciencesUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
| | - Briana Booth
- Penn Neurodegeneration Genomics CenterDepartment of PathologyPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Laura B. Cantwell
- Penn Neurodegeneration Genomics CenterDepartment of PathologyPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Fangcong Chen
- Department of NeurologyColumbia University Medical CenterNew YorkNew YorkUSA
| | - Sephera Chou
- Geriatric Mental Health Services, Centre for Addiction and Mental HealthTorontoOntarioCanada
| | - Dewi Clark
- Geriatric Mental Health Services, Centre for Addiction and Mental HealthTorontoOntarioCanada
| | - Michelle Deng
- Geriatric Mental Health Services, Centre for Addiction and Mental HealthTorontoOntarioCanada
| | - Ting Hei Hong
- University of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Ling‐Jen Hwang
- Stanford Alzheimer's Disease Research CenterStanfordCaliforniaUSA
| | - Lilly Jiang
- University of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Yoonmee Joo
- Department of Community Health SystemsUniversity of California San Francisco School of NursingSan FranciscoCaliforniaUSA
| | - Younhee Kang
- College of NursingGraduate Program in System Health Science and EngineeringEwha Womans UniversitySeoulRepublic of Korea
| | - Ellen S. Kim
- Division of NeurologyUniversity of British ColumbiaVancouverBritish ColumbiaCanada
| | - Hoowon Kim
- Department of NeurologyChosun University Hospital, Dong‐guGwangjuRepublic of Korea
| | - Kyungmin Kim
- Department of Child Development and Family StudiesCollege of Human EcologySeoul National UniversityJongno‐guSeoulRepublic of Korea
| | - Amanda B. Kuzma
- Penn Neurodegeneration Genomics CenterDepartment of PathologyPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Eleanor Lam
- Geriatric Mental Health Services, Centre for Addiction and Mental HealthTorontoOntarioCanada
| | - Serggio C. Lanata
- Memory and Aging CenterDepartment of NeurologyWeill Institute for NeurosciencesUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
| | - Kunho Lee
- Biomedical Science, College of Natural SciencesChosun UniversityGwanak‐guSeoulRepublic of Korea
| | - Donghe Li
- Department of Medicine (Biomedical Genetics)Boston University School of MedicineBostonMassachusettsUSA
| | - Mingyao Li
- Department of BiostatisticsEpidemiology and InformaticsPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Xiang Li
- Geriatric Mental Health Services, Centre for Addiction and Mental HealthTorontoOntarioCanada
| | - Chia‐Lun Liu
- Penn Neurodegeneration Genomics CenterDepartment of PathologyPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Collin Liu
- Department of NeurologyKeck School of Medicine at University of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Linghsi Liu
- Alzheimer's Disease Research CenterDepartment of PsychiatryIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Jody‐Lynn Lupo
- Alzheimer's Disease Cooperative StudyUniversity of California, San DiegoLa JollaCaliforniaUSA
| | - Khai Nguyen
- Department of MedicineUniversity of California at San DiegoLa JollaCaliforniaUSA
| | - Shannon E. Pfleuger
- Division of NeurologyUniversity of British ColumbiaVancouverBritish ColumbiaCanada
| | - James Qian
- Memory and Aging CenterDepartment of NeurologyWeill Institute for NeurosciencesUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
| | - Winnie Qian
- Geriatric Mental Health Services, Centre for Addiction and Mental HealthTorontoOntarioCanada
| | - Veronica Ramirez
- Stanford Alzheimer's Disease Research CenterStanfordCaliforniaUSA
| | - Kristen A. Russ
- Department of Medical and Molecular GeneticsIndiana University School of MedicineIndianapolisIndianaUSA
| | - Eun Hyun Seo
- Premedical Science, College of MedicineChosun University, Dong‐guGwangjuRepublic of Korea
| | - Yeunjoo E. Song
- Department of Population & Quantitative Health SciencesSchool of MedicineCase Western Reserve UniversityClevelandOhioUSA
| | - Maria Carmela Tartaglia
- Tanz Centre for Research in Neurodegenerative DiseasesUniversity of TorontoTorontoOntarioCanada
| | - Lu Tian
- Department of Biomedical Data ScienceStanford University School of MedicineStanfordCaliforniaUSA
| | - Mina Torres
- Southern California Eye Institute, CHA Hollywood Presbyterian Medical CenterLos AngelesCaliforniaUSA
| | - Namkhue Vo
- Alzheimer's Disease Cooperative StudyUniversity of California, San DiegoLa JollaCaliforniaUSA
| | - Ellen C. Wong
- Department of NeurologyKeck School of Medicine at University of Southern CaliforniaLos AngelesCaliforniaUSA
- Department of NeurologyRancho Los Amigos National Rehabilitation CenterDowneyCaliforniaUSA
| | - Yuan Xie
- Department of NeurologyColumbia University Medical CenterNew YorkNew YorkUSA
| | - Eugene B. Yau
- Division of NeurologyUniversity of British ColumbiaVancouverBritish ColumbiaCanada
| | - Isabelle Yi
- Stanford Alzheimer's Disease Research CenterStanfordCaliforniaUSA
| | - Victoria Yu
- Department of OphthalmologyKeck School of Medicine at University of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Xiaoyi Zeng
- Alzheimer's Disease Research CenterDepartment of PsychiatryIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Peter St George‐Hyslop
- Tanz Centre for Research in Neurodegenerative DiseasesUniversity of TorontoTorontoOntarioCanada
- Department of Neurology and the Taub Institute for Research on Alzheimer's Disease and the Aging BrainColumbia UniversityNew YorkNew YorkUSA
| | - Rhoda Au
- Department of Anatomy and NeurobiologySlone Epidemiology CenterBoston University Chobanian & Avedisian School of MedicineBostonMassachusettsUSA
- Department of EpidemiologyBoston University School of Public HealthBostonMassachusettsUSA
| | - Gerard D. Schellenberg
- Penn Neurodegeneration Genomics CenterDepartment of PathologyPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Jeffrey L. Dage
- Department of Medical and Molecular GeneticsIndiana University School of MedicineIndianapolisIndianaUSA
- Department of NeurologyIndiana University School of MedicineIndianapolisIndianaUSA
| | - Rohit Varma
- Southern California Eye Institute, CHA Hollywood Presbyterian Medical CenterLos AngelesCaliforniaUSA
| | - Ging‐Yuek R. Hsiung
- Division of NeurologyUniversity of British ColumbiaVancouverBritish ColumbiaCanada
| | - Howard Rosen
- Memory and Aging CenterDepartment of NeurologyWeill Institute for NeurosciencesUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
| | - Victor W. Henderson
- Department of Epidemiology and Population HealthStanford UniversityStanfordCaliforniaUSA
- Department of Neurology & Neurological SciencesStanford UniversityStanfordCaliforniaUSA
| | - Tatiana Foroud
- Department of Medical and Molecular GeneticsIndiana University School of MedicineIndianapolisIndianaUSA
| | - Walter A. Kukull
- Department of EpidemiologyUniversity of WashingtonSeattleWashingtonUSA
| | - Guerry M. Peavy
- Department of NeurosciencesUniversity of California San DiegoLa JollaCaliforniaUSA
| | - Haeok Lee
- Rory Meyers College of NursingNew York UniversityNew YorkNew YorkUSA
| | - Howard H. Feldman
- Alzheimer's Disease Cooperative StudyUniversity of California, San DiegoLa JollaCaliforniaUSA
- Department of NeurosciencesUniversity of California San DiegoLa JollaCaliforniaUSA
| | - Richard Mayeux
- Department of Neurology and the Taub Institute for Research on Alzheimer's Disease and the Aging BrainColumbia University, Vagelos College of Physicians and SurgeonsNew YorkNew YorkUSA
| | - Helena Chui
- Department of NeurologyKeck School of Medicine at University of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Gyungah R. Jun
- Department of Medicine (Biomedical Genetics)Boston University School of MedicineBostonMassachusettsUSA
- Department of OphthalmologyBoston University School of MedicineBostonMassachusettsUSA
- Department of BiostatisticsBoston University School of Public HealthBostonMassachusettsUSA
| | - Van M. Ta Park
- Department of Community Health SystemsUniversity of California San Francisco School of NursingSan FranciscoCaliforniaUSA
- Asian American Research Center on Health (ARCH)University of California San Francisco School of NursingSan FranciscoCaliforniaUSA
| | - Tiffany W. Chow
- Penn Neurodegeneration Genomics CenterDepartment of PathologyPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Alector Inc.South San FranciscoCaliforniaUSA
| | - Li‐San Wang
- Penn Neurodegeneration Genomics CenterDepartment of PathologyPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| |
Collapse
|
26
|
Yang HS, Teng L, Kang D, Menon V, Ge T, Finucane HK, Schultz AP, Properzi M, Klein HU, Chibnik LB, Schneider JA, Bennett DA, Hohman TJ, Mayeux RP, Johnson KA, De Jager PL, Sperling RA. Cell-type-specific Alzheimer's disease polygenic risk scores are associated with distinct disease processes in Alzheimer's disease. Nat Commun 2023; 14:7659. [PMID: 38036535 PMCID: PMC10689816 DOI: 10.1038/s41467-023-43132-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 11/01/2023] [Indexed: 12/02/2023] Open
Abstract
Many of the Alzheimer's disease (AD) risk genes are specifically expressed in microglia and astrocytes, but how and when the genetic risk localizing to these cell types contributes to AD pathophysiology remains unclear. Here, we derive cell-type-specific AD polygenic risk scores (ADPRS) from two extensively characterized datasets and uncover the impact of cell-type-specific genetic risk on AD endophenotypes. In an autopsy dataset spanning all stages of AD (n = 1457), the astrocytic ADPRS affected diffuse and neuritic plaques (amyloid-β), while microglial ADPRS affected neuritic plaques, microglial activation, neurofibrillary tangles (tau), and cognitive decline. In an independent neuroimaging dataset of cognitively unimpaired elderly (n = 2921), astrocytic ADPRS was associated with amyloid-β, and microglial ADPRS was associated with amyloid-β and tau, connecting cell-type-specific genetic risk with AD pathology even before symptom onset. Together, our study provides human genetic evidence implicating multiple glial cell types in AD pathophysiology, starting from the preclinical stage.
Collapse
Affiliation(s)
- Hyun-Sik Yang
- Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women's Hospital, Boston, MA, USA.
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| | - Ling Teng
- Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women's Hospital, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Daniel Kang
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| | - Vilas Menon
- Center for Translational & Computational Neuroimmunology, Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
- Department of Neurology and the Taub Institute for the Study of Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY, USA
| | - Tian Ge
- Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Center for Precision Psychiatry, Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA
| | - Hilary K Finucane
- Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Aaron P Schultz
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Michael Properzi
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| | - Hans-Ulrich Klein
- Center for Translational & Computational Neuroimmunology, Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
- Department of Neurology and the Taub Institute for the Study of Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY, USA
| | - Lori B Chibnik
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Julie A Schneider
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Timothy J Hohman
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Richard P Mayeux
- Department of Neurology and the Taub Institute for the Study of Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY, USA
| | - Keith A Johnson
- Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women's Hospital, Boston, MA, USA
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital, Boston, MA, USA
| | - Philip L De Jager
- Center for Translational & Computational Neuroimmunology, Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
- Department of Neurology and the Taub Institute for the Study of Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY, USA
| | - Reisa A Sperling
- Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women's Hospital, Boston, MA, USA
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| |
Collapse
|
27
|
Chung J, Sahelijo N, Maruyama T, Hu J, Panitch R, Xia W, Mez J, Stein TD, Alzheimer’s Disease Neuroimaging Initiative, Saykin AJ, Takeyama H, Farrer LA, Crane PK, Nho K, Jun GR. Alzheimer's disease heterogeneity explained by polygenic risk scores derived from brain transcriptomic profiles. Alzheimers Dement 2023; 19:5173-5184. [PMID: 37166019 PMCID: PMC10638468 DOI: 10.1002/alz.13069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 03/03/2023] [Accepted: 03/08/2023] [Indexed: 05/12/2023]
Abstract
INTRODUCTION Alzheimer's disease (AD) is heterogeneous, both clinically and neuropathologically. We investigated whether polygenic risk scores (PRSs) integrated with transcriptome profiles from AD brains can explain AD clinical heterogeneity. METHODS We conducted co-expression network analysis and identified gene sets (modules) that were preserved in three AD transcriptome datasets and associated with AD-related neuropathological traits including neuritic plaques (NPs) and neurofibrillary tangles (NFTs). We computed the module-based PRSs (mbPRSs) for each module and tested associations with mbPRSs for cognitive test scores, cognitively defined AD subgroups, and brain imaging data. RESULTS Of the modules significantly associated with NPs and/or NFTs, the mbPRSs from two modules (M6 and M9) showed distinct associations with language and visuospatial functioning, respectively. They matched clinical subtypes and brain atrophy at specific regions. DISCUSSION Our findings demonstrate that polygenic profiling based on co-expressed gene sets can explain heterogeneity in AD patients, enabling genetically informed patient stratification and precision medicine in AD. HIGHLIGHTS Co-expression gene-network analysis in Alzheimer's disease (AD) brains identified gene sets (modules) associated with AD heterogeneity. AD-associated modules were selected when genes in each module were enriched for neuritic plaques and neurofibrillary tangles. Polygenic risk scores from two selected modules were linked to the matching cognitively defined AD subgroups (language and visuospatial subgroups). Polygenic risk scores from the two modules were associated with cognitive performance in language and visuospatial domains and the associations were confirmed in regional-specific brain atrophy data.
Collapse
Affiliation(s)
- Jaeyoon Chung
- Department of Medicine (Biomedical Genetics), Boston University School of Medicine, 72 East Concord Street, Boston, MA 02118, USA
| | - Nathan Sahelijo
- Department of Medicine (Biomedical Genetics), Boston University School of Medicine, 72 East Concord Street, Boston, MA 02118, USA
| | - Toru Maruyama
- Department of Life Science and Medical Bioscience, Waseda University, 2-2 Wakamatsu-cho, Shinjuku-ku, Tokyo 162-8480, Japan
| | - Junming Hu
- Department of Medicine (Biomedical Genetics), Boston University School of Medicine, 72 East Concord Street, Boston, MA 02118, USA
| | - Rebecca Panitch
- Department of Medicine (Biomedical Genetics), Boston University School of Medicine, 72 East Concord Street, Boston, MA 02118, USA
| | - Weiming Xia
- Department of Pharmacology & Experimental Therapeutics, Boston University School of Medicine, 72 East Concord Street, Boston, MA 02118, USA
- Department of Veterans Affairs Medical Center, Bedford, MA 01730, USA
| | - Jesse Mez
- Department of Neurology, Boston University School of Medicine, 72 East Concord Street, Boston, MA 02118, USA
| | - Thor D. Stein
- Department of Veterans Affairs Medical Center, Bedford, MA 01730, USA
- Department of Pathology & Laboratory Medicine, Boston University School of Medicine, 72 East Concord Street, Boston, MA 02118, USA
- Boston VA Healthcare Center, Boston, MA 02130, USA
| | | | - Andrew J. Saykin
- Department of Radiology and Imaging Sciences and Indiana Alzheimer’s Disease Research Center, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Haruko Takeyama
- Department of Life Science and Medical Bioscience, Waseda University, 2-2 Wakamatsu-cho, Shinjuku-ku, Tokyo 162-8480, Japan
- Computational Bio Big-Data Open Innovation Laboratory, AIST-Waseda University, Japan, 3-4-1 Okubo, Shinjuku-ku, Tokyo 169-8555, Japan
- Research Organization for Nano and Life Innovations, Waseda University, 513, Wasedatsurumaki-cho, Shinjuku-ku, Tokyo 162-0041, Japan
- Institute for Advanced Research of Biosystem Dynamics, Waseda Research Institute for Science and Engineering, Graduate School of Advanced Science and Engineering, Waseda University, 3-4-1 Okubo, Shinjuku-ku, Tokyo 169-8555, Japan
| | - Lindsay A. Farrer
- Department of Medicine (Biomedical Genetics), Boston University School of Medicine, 72 East Concord Street, Boston, MA 02118, USA
- Department of Neurology, Boston University School of Medicine, 72 East Concord Street, Boston, MA 02118, USA
- Department of Ophthalmology, Boston University School of Medicine, 72 East Concord Street, Boston, MA 02118, USA
- Department of Biostatistics, Boston University School of Public Health, 715 Albany Street, Boston, MA 02118, USA
- Department of Epidemiology, Boston University School of Public Health, 715 Albany Street, Boston, MA 02118, USA
| | - Paul K. Crane
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - Kwangsik Nho
- Department of Radiology and Imaging Sciences and Indiana Alzheimer’s Disease Research Center, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Gyungah R. Jun
- Department of Medicine (Biomedical Genetics), Boston University School of Medicine, 72 East Concord Street, Boston, MA 02118, USA
- Department of Ophthalmology, Boston University School of Medicine, 72 East Concord Street, Boston, MA 02118, USA
- Department of Biostatistics, Boston University School of Public Health, 715 Albany Street, Boston, MA 02118, USA
| |
Collapse
|
28
|
Hoogland AI, Gonzalez BD, Park JY, Small BJ, Sutton SK, Pidala JA, Smith KS, Bower JE, Jacobsen PB, Jim HS. Associations of Germline Genetic Variants With Depression and Fatigue Among Hematologic Cancer Patients Treated With Allogeneic Hematopoietic Cell Transplantation. Psychosom Med 2023; 85:813-819. [PMID: 37678326 PMCID: PMC10915106 DOI: 10.1097/psy.0000000000001251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/09/2023]
Abstract
OBJECTIVE Depression and fatigue are common among cancer patients and are associated with germline genetic variation. The goal of this pilot study was to examine genetic associations with depression and fatigue in the year after allogeneic hematopoietic cell transplant (HCT). METHODS Blood was collected from patients and their donors before HCT. Patients completed self-report measures of depression and fatigue before HCT (T1), 90 days post-HCT (T2), and 1 year post-HCT (T3). Of the 384 genetic variants genotyped on a custom Illumina BeadChip microarray, 267 were retained for analysis based on quality control. Main effects of patient and donor variants as well as their interaction were examined using regression analyses. Significant variants were defined as those with a false discovery rate-adjusted p value of <.05. RESULTS The sample consisted of 59 patient-donor pairs. Mean levels of depression and fatigue did not change significantly over time ( p values of > .41). Increases in depression from T1 to T2 were associated with patient-donor interactions at rs1928040 ( p = 3.0 × 10 -4 ) and rs6311 ( p = 2.0 × 10 -4 ) in HTR2A . Increases in fatigue from T1 to T2 were associated with patient rs689021 in SORL1 ( p = 6.0 × 10 -5 ) and a patient-donor interaction at rs1885884 in HTR2A ( p < 1.0 × 10 -4 ). CONCLUSIONS Data suggest that variants in genes regulating the serotonergic system ( HTR2A ) and lipid metabolism ( SORL1 ) are associated with changes in depression and fatigue in allogeneic HCT patients, implicating patients' own genetic inheritance as well as that of donors. Additional studies are warranted to confirm these findings.
Collapse
Affiliation(s)
- Aasha I. Hoogland
- Department of Health Outcomes and Behavior, Moffitt Cancer Center, Tampa, FL 33612 USA
| | - Brian D. Gonzalez
- Department of Health Outcomes and Behavior, Moffitt Cancer Center, Tampa, FL 33612 USA
| | - Jong Y. Park
- Department of Cancer Epidemiology, Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Brent J. Small
- College of Aging, University of South Florida, Tampa, FL 33620 USA
- Department of Biostatistics and Bioinformatics, Moffitt Cancer Center, Tampa, FL 33612 USA
| | - Steven K. Sutton
- Department of Biostatistics and Bioinformatics, Moffitt Cancer Center, Tampa, FL 33612 USA
| | - Joseph A. Pidala
- Department of Blood and Marrow Transplantation and Cellular Immunotherapy, Moffitt Cancer Center, Tampa, FL 33612 USA
| | - Kristen S. Smith
- Department of Health Outcomes and Behavior, Moffitt Cancer Center, Tampa, FL 33612 USA
| | - Julienne E. Bower
- Department of Psychology, University of California-Los Angeles, Los Angeles, CA 90095 USA
| | - Paul B. Jacobsen
- Department of Health Outcomes and Behavior, Moffitt Cancer Center, Tampa, FL 33612 USA
| | - Heather S.L. Jim
- Department of Health Outcomes and Behavior, Moffitt Cancer Center, Tampa, FL 33612 USA
| |
Collapse
|
29
|
Talyansky S, Le Guen Y, Kasireddy N, Belloy ME, Greicius MD. APOE-ε4 and BIN1 increase risk of Alzheimer's disease pathology but not specifically of Lewy body pathology. Acta Neuropathol Commun 2023; 11:149. [PMID: 37700353 PMCID: PMC10496176 DOI: 10.1186/s40478-023-01626-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 07/22/2023] [Indexed: 09/14/2023] Open
Abstract
Lewy body (LB) pathology commonly occurs in individuals with Alzheimer's disease (AD) pathology. However, it remains unclear which genetic risk factors underlie AD pathology, LB pathology, or AD-LB co-pathology. Notably, whether APOE-ε4 affects risk of LB pathology independently from AD pathology is controversial. We adapted criteria from the literature to classify 4,985 subjects from the National Alzheimer's Coordinating Center (NACC) and the Rush University Medical Center as AD-LB co-pathology (AD+LB+), sole AD pathology (AD+LB-), sole LB pathology (AD-LB+), or no pathology (AD-LB-). We performed a meta-analysis of a genome-wide association study (GWAS) per subpopulation (NACC/Rush) for each disease phenotype compared to the control group (AD-LB-), and compared the AD+LB+ to AD+LB- groups. APOE-ε4 was significantly associated with risk of AD+LB- and AD+LB+ compared to AD-LB-. However, APOE-ε4 was not associated with risk of AD-LB+ compared to AD-LB- or risk of AD+LB+ compared to AD+LB-. Associations at the BIN1 locus exhibited qualitatively similar results. These results suggest that APOE-ε4 is a risk factor for AD pathology, but not for LB pathology when decoupled from AD pathology. The same holds for BIN1 risk variants. These findings, in the largest AD-LB neuropathology GWAS to date, distinguish the genetic risk factors for sole and dual AD-LB pathology phenotypes. Our GWAS meta-analysis summary statistics, derived from phenotypes based on postmortem pathologic evaluation, may provide more accurate disease-specific polygenic risk scores compared to GWAS based on clinical diagnoses, which are likely confounded by undetected dual pathology and clinical misdiagnoses of dementia type.
Collapse
Affiliation(s)
- Seth Talyansky
- Department of Neurology and Neurological Sciences, Stanford University, 290 Jane Stanford Way, E265, Stanford, CA, 94305-5090, USA
| | - Yann Le Guen
- Department of Neurology and Neurological Sciences, Stanford University, 290 Jane Stanford Way, E265, Stanford, CA, 94305-5090, USA.
- Institut du Cerveau, Paris Brain Institute - ICM, Paris, France.
| | - Nandita Kasireddy
- Department of Neurology and Neurological Sciences, Stanford University, 290 Jane Stanford Way, E265, Stanford, CA, 94305-5090, USA
| | - Michael E Belloy
- Department of Neurology and Neurological Sciences, Stanford University, 290 Jane Stanford Way, E265, Stanford, CA, 94305-5090, USA
| | - Michael D Greicius
- Department of Neurology and Neurological Sciences, Stanford University, 290 Jane Stanford Way, E265, Stanford, CA, 94305-5090, USA
| |
Collapse
|
30
|
Nag S, Schneider JA. Limbic-predominant age-related TDP43 encephalopathy (LATE) neuropathological change in neurodegenerative diseases. Nat Rev Neurol 2023; 19:525-541. [PMID: 37563264 PMCID: PMC10964248 DOI: 10.1038/s41582-023-00846-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/26/2023] [Indexed: 08/12/2023]
Abstract
TAR DNA-binding protein 43 (TDP43) is a focus of research in late-onset dementias. TDP43 pathology in the brain was initially identified in amyotrophic lateral sclerosis and frontotemporal lobar degeneration, and later in Alzheimer disease (AD), other neurodegenerative diseases and ageing. Limbic-predominant age-related TDP43 encephalopathy (LATE), recognized as a clinical entity in 2019, is characterized by amnestic dementia resembling AD dementia and occurring most commonly in adults over 80 years of age. Neuropathological findings in LATE, referred to as LATE neuropathological change (LATE-NC), consist of neuronal and glial cytoplasmic TDP43 localized predominantly in limbic areas with or without coexisting hippocampal sclerosis and/or AD neuropathological change and without frontotemporal lobar degeneration or amyotrophic lateral sclerosis pathology. LATE-NC is frequently associated with one or more coexisting pathologies, mainly AD neuropathological change. The focus of this Review is the pathology, genetic risk factors and nature of the cognitive impairments and dementia in pure LATE-NC and in LATE-NC associated with coexisting pathologies. As the clinical and cognitive profile of LATE is currently not easily distinguishable from AD dementia, it is important to develop biomarkers to aid in the diagnosis of this condition in the clinic. The pathogenesis of LATE-NC should be a focus of future research to form the basis for the development of preventive and therapeutic strategies.
Collapse
Affiliation(s)
- Sukriti Nag
- Rush Alzheimer's Disease Center, Department of Pathology (Neuropathology), Rush University Medical Center, Chicago, IL, USA.
| | - Julie A Schneider
- Rush Alzheimer's Disease Center, Department of Pathology (Neuropathology), Rush University Medical Center, Chicago, IL, USA.
| |
Collapse
|
31
|
Cochran JN, Acosta-Uribe J, Esposito BT, Madrigal L, Aguillón D, Giraldo MM, Taylor JW, Bradley J, Fulton-Howard B, Andrews SJ, Acosta-Baena N, Alzate D, Garcia GP, Piedrahita F, Lopez HE, Anderson AG, Rodriguez-Nunez I, Roberts K, Dominantly Inherited Alzheimer Network, Absher D, Myers RM, Beecham GW, Reitz C, Rizzardi LF, Fernandez MV, Goate AM, Cruchaga C, Renton AE, Lopera F, Kosik KS. Genetic associations with age at dementia onset in the PSEN1 E280A Colombian kindred. Alzheimers Dement 2023; 19:3835-3847. [PMID: 36951251 PMCID: PMC10514237 DOI: 10.1002/alz.13021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 02/03/2023] [Accepted: 02/07/2023] [Indexed: 03/24/2023]
Abstract
INTRODUCTION Genetic associations with Alzheimer's disease (AD) age at onset (AAO) could reveal genetic variants with therapeutic applications. We present a large Colombian kindred with autosomal dominant AD (ADAD) as a unique opportunity to discover AAO genetic associations. METHODS A genetic association study was conducted to examine ADAD AAO in 340 individuals with the PSEN1 E280A mutation via TOPMed array imputation. Replication was assessed in two ADAD cohorts, one sporadic early-onset AD study and four late-onset AD studies. RESULTS 13 variants had p<1×10-7 or p<1×10-5 with replication including three independent loci with candidate associations with clusterin including near CLU. Other suggestive associations were identified in or near HS3ST1, HSPG2, ACE, LRP1B, TSPAN10, and TSPAN14. DISCUSSION Variants with suggestive associations with AAO were associated with biological processes including clusterin, heparin sulfate, and amyloid processing. The detection of these effects in the presence of a strong mutation for ADAD reinforces their potentially impactful role.
Collapse
Affiliation(s)
| | - Juliana Acosta-Uribe
- Neuroscience Research Institute, University of California, Santa Barbara, California, and Department of Molecular Cellular and Developmental Biology University of California, Santa Barbara, California, USA
- Grupo de Neurociencias de Antioquia. School of Medicine. Universidad de Antioquia, Medellín, Antioquia, Colombia
| | - Bianca T Esposito
- Department of Genetics & Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Lucia Madrigal
- Grupo de Neurociencias de Antioquia. School of Medicine. Universidad de Antioquia, Medellín, Antioquia, Colombia
| | - David Aguillón
- Grupo de Neurociencias de Antioquia. School of Medicine. Universidad de Antioquia, Medellín, Antioquia, Colombia
| | - Margarita M Giraldo
- Grupo de Neurociencias de Antioquia. School of Medicine. Universidad de Antioquia, Medellín, Antioquia, Colombia
| | - Jared W Taylor
- HudsonAlpha Institute for Biotechnology, Huntsville, Alabama, USA
| | - Joseph Bradley
- Washington University School of Medicine, St. Louis, Missouri, USA
| | - Brian Fulton-Howard
- Department of Genetics & Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Shea J Andrews
- Department of Genetics & Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Natalia Acosta-Baena
- Grupo de Neurociencias de Antioquia. School of Medicine. Universidad de Antioquia, Medellín, Antioquia, Colombia
| | - Diana Alzate
- Grupo de Neurociencias de Antioquia. School of Medicine. Universidad de Antioquia, Medellín, Antioquia, Colombia
| | - Gloria P Garcia
- Grupo de Neurociencias de Antioquia. School of Medicine. Universidad de Antioquia, Medellín, Antioquia, Colombia
| | - Francisco Piedrahita
- Grupo de Neurociencias de Antioquia. School of Medicine. Universidad de Antioquia, Medellín, Antioquia, Colombia
| | - Hugo E Lopez
- Grupo de Neurociencias de Antioquia. School of Medicine. Universidad de Antioquia, Medellín, Antioquia, Colombia
| | | | | | - Kevin Roberts
- HudsonAlpha Institute for Biotechnology, Huntsville, Alabama, USA
| | | | - Devin Absher
- HudsonAlpha Institute for Biotechnology, Huntsville, Alabama, USA
| | - Richard M Myers
- HudsonAlpha Institute for Biotechnology, Huntsville, Alabama, USA
| | - Gary W Beecham
- The John P. Hussman Institute for Human Genomics, University of Miami, Miami, Florida, USA
| | - Christiane Reitz
- Department of Epidemiology, Sergievsky Center, Taub Institute for Research on the Aging Brain, Columbia University, New York, New York, USA
| | | | | | - Alison M Goate
- Department of Genetics & Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Carlos Cruchaga
- Washington University School of Medicine, St. Louis, Missouri, USA
| | - Alan E Renton
- Department of Genetics & Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Francisco Lopera
- Grupo de Neurociencias de Antioquia. School of Medicine. Universidad de Antioquia, Medellín, Antioquia, Colombia
| | - Kenneth S Kosik
- Neuroscience Research Institute, University of California, Santa Barbara, California, and Department of Molecular Cellular and Developmental Biology University of California, Santa Barbara, California, USA
| |
Collapse
|
32
|
Katsumata Y, Fardo DW, Shade LMP, Nelson PT. LATE-NC risk alleles (in TMEM106B, GRN, and ABCC9 genes) among persons with African ancestry. J Neuropathol Exp Neurol 2023; 82:760-768. [PMID: 37528055 PMCID: PMC10440720 DOI: 10.1093/jnen/nlad059] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/03/2023] Open
Abstract
Limbic-predominant age-related TDP-43 encephalopathy (LATE) affects approximately one-third of older individuals and is associated with cognitive impairment. However, there is a highly incomplete understanding of the genetic determinants of LATE neuropathologic changes (LATE-NC) in diverse populations. The defining neuropathologic feature of LATE-NC is TDP-43 proteinopathy, often with comorbid hippocampal sclerosis (HS). In terms of genetic risk factors, LATE-NC and/or HS are associated with single nucleotide variants (SNVs) in 3 genes-TMEM106B (rs1990622), GRN (rs5848), and ABCC9 (rs1914361 and rs701478). We evaluated these 3 genes in convenience samples of individuals of African ancestry. The allele frequencies of the LATE-associated alleles were significantly different between persons of primarily African (versus European) ancestry: In persons of African ancestry, the risk-associated alleles for TMEM106B and ABCC9 were less frequent, whereas the risk allele in GRN was more frequent. We performed an exploratory analysis of data from African-American subjects processed by the Alzheimer's Disease Genomics Consortium, with a subset of African-American participants (n = 166) having corroborating neuropathologic data through the National Alzheimer's Coordinating Center (NACC). In this limited-size sample, the ABCC9/rs1914361 SNV was associated with HS pathology. More work is required concerning the genetic factors influencing non-Alzheimer disease pathology such as LATE-NC in diverse cohorts.
Collapse
Affiliation(s)
- Yuriko Katsumata
- University of Kentucky Sanders-Brown Center on Aging, Lexington, Kentucky, USA
- University of Kentucky Department of Biostatistics, Lexington, Kentucky, USA
| | - David W Fardo
- University of Kentucky Sanders-Brown Center on Aging, Lexington, Kentucky, USA
- University of Kentucky Department of Biostatistics, Lexington, Kentucky, USA
| | - Lincoln M P Shade
- University of Kentucky Department of Biostatistics, Lexington, Kentucky, USA
| | - Peter T Nelson
- University of Kentucky Sanders-Brown Center on Aging, Lexington, Kentucky, USA
- University of Kentucky Department of Pathology and Laboratory Medicine, Lexington, Kentucky, USA
| |
Collapse
|
33
|
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: 34] [Impact Index Per Article: 17.0] [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
|
34
|
Yang HS, Teng L, Kang D, Menon V, Ge T, Finucane HK, Schultz AP, Properzi M, Klein HU, Chibnik LB, Schneider JA, Bennett DA, Hohman TJ, Mayeux RP, Johnson KA, De Jager PL, Sperling RA. Cell-type-specific Alzheimer's disease polygenic risk scores are associated with distinct disease processes in Alzheimer's disease. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.06.01.23290850. [PMID: 37333223 PMCID: PMC10274993 DOI: 10.1101/2023.06.01.23290850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
Alzheimer's disease (AD) heritability is enriched in glial genes, but how and when cell-type-specific genetic risk contributes to AD remains unclear. Here, we derive cell-type-specific AD polygenic risk scores (ADPRS) from two extensively characterized datasets. In an autopsy dataset spanning all stages of AD (n=1,457), astrocytic (Ast) ADPRS was associated with both diffuse and neuritic Aβ plaques, while microglial (Mic) ADPRS was associated with neuritic Aβ plaques, microglial activation, tau, and cognitive decline. Causal modeling analyses further clarified these relationships. In an independent neuroimaging dataset of cognitively unimpaired elderly (n=2,921), Ast-ADPRS were associated with Aβ, and Mic-ADPRS was associated with Aβ and tau, showing a consistent pattern with the autopsy dataset. Oligodendrocytic and excitatory neuronal ADPRSs were associated with tau, but only in the autopsy dataset including symptomatic AD cases. Together, our study provides human genetic evidence implicating multiple glial cell types in AD pathophysiology, starting from the preclinical stage.
Collapse
Affiliation(s)
- Hyun-Sik Yang
- Department of Neurology, Brigham and Women’s Hospital, Boston, MA, USA
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA
- Broad Institute of MIT and Harvard, Cambridge, MA
| | - Ling Teng
- Department of Neurology, Brigham and Women’s Hospital, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA
| | - Daniel Kang
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| | - Vilas Menon
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
| | - Tian Ge
- Harvard Medical School, Boston, MA
- Broad Institute of MIT and Harvard, Cambridge, MA
- Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA
| | - Hilary K. Finucane
- Harvard Medical School, Boston, MA
- Broad Institute of MIT and Harvard, Cambridge, MA
- Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Aaron P. Schultz
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA
| | - Michael Properzi
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| | - Hans-Ulrich Klein
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
| | - Lori B. Chibnik
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Julie A. Schneider
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - David A. Bennett
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Timothy J. Hohman
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Richard P. Mayeux
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
| | - Keith A. Johnson
- Department of Neurology, Brigham and Women’s Hospital, Boston, MA, USA
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA
- Department of Radiology, Massachusetts General Hospital, Boston, MA, USA
| | - Philip L. De Jager
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
| | - Reisa A. Sperling
- Department of Neurology, Brigham and Women’s Hospital, Boston, MA, USA
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA
| |
Collapse
|
35
|
Wang T, Chen X, Zhang J, Feng Q, Huang M. Deep multimodality-disentangled association analysis network for imaging genetics in neurodegenerative diseases. Med Image Anal 2023; 88:102842. [PMID: 37247468 DOI: 10.1016/j.media.2023.102842] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 03/01/2023] [Accepted: 05/15/2023] [Indexed: 05/31/2023]
Abstract
Imaging genetics is a crucial tool that is applied to explore potentially disease-related biomarkers, particularly for neurodegenerative diseases (NDs). With the development of imaging technology, the association analysis between multimodal imaging data and genetic data is gradually being concerned by a wide range of imaging genetics studies. However, multimodal data are fused first and then correlated with genetic data in traditional methods, which leads to an incomplete exploration of their common and complementary information. In addition, the inaccurate formulation in the complex relationships between imaging and genetic data and information loss caused by missing multimodal data are still open problems in imaging genetics studies. Therefore, in this study, a deep multimodality-disentangled association analysis network (DMAAN) is proposed to solve the aforementioned issues and detect the disease-related biomarkers of NDs simultaneously. First, the imaging data are nonlinearly projected into a latent space and imaging representations can be achieved. The imaging representations are further disentangled into common and specific parts by using a multimodal-disentangled module. Second, the genetic data are encoded to achieve genetic representations, and then, the achieved genetic representations are nonlinearly mapped to the common and specific imaging representations to build nonlinear associations between imaging and genetic data through an association analysis module. Moreover, modality mask vectors are synchronously synthesized to integrate the genetic and imaging data, which helps the following disease diagnosis. Finally, the proposed method achieves reasonable diagnosis performance via a disease diagnosis module and utilizes the label information to detect the disease-related modality-shared and modality-specific biomarkers. Furthermore, the genetic representation can be used to impute the missing multimodal data with our learning strategy. Two publicly available datasets with different NDs are used to demonstrate the effectiveness of the proposed DMAAN. The experimental results show that the proposed DMAAN can identify the disease-related biomarkers, which suggests the proposed DMAAN may provide new insights into the pathological mechanism and early diagnosis of NDs. The codes are publicly available at https://github.com/Meiyan88/DMAAN.
Collapse
Affiliation(s)
- Tao Wang
- School of Biomedical Engineering, Southern Medical University, Guangzhou 510515, China
| | - Xiumei Chen
- School of Biomedical Engineering, Southern Medical University, Guangzhou 510515, China
| | - Jiawei Zhang
- School of Biomedical Engineering, Southern Medical University, Guangzhou 510515, China
| | - Qianjin Feng
- School of Biomedical Engineering, Southern Medical University, Guangzhou 510515, China; Guangdong Provincial Key Laboratory of Medical Image Processing, Southern Medical University, Guangzhou 510515, China; Guangdong Province Engineering Laboratory for Medical Imaging and Diagnostic Technology, Southern Medical University, Guangzhou 510515, China.
| | - Meiyan Huang
- School of Biomedical Engineering, Southern Medical University, Guangzhou 510515, China; Guangdong Provincial Key Laboratory of Medical Image Processing, Southern Medical University, Guangzhou 510515, China; Guangdong Province Engineering Laboratory for Medical Imaging and Diagnostic Technology, Southern Medical University, Guangzhou 510515, China.
| |
Collapse
|
36
|
Merchant JP, Zhu K, Henrion MYR, Zaidi SSA, Lau B, Moein S, Alamprese ML, Pearse RV, Bennett DA, Ertekin-Taner N, Young-Pearse TL, Chang R. Predictive network analysis identifies JMJD6 and other potential key drivers in Alzheimer's disease. Commun Biol 2023; 6:503. [PMID: 37188718 PMCID: PMC10185548 DOI: 10.1038/s42003-023-04791-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Accepted: 03/31/2023] [Indexed: 05/17/2023] Open
Abstract
Despite decades of genetic studies on late-onset Alzheimer's disease, the underlying molecular mechanisms remain unclear. To better comprehend its complex etiology, we use an integrative approach to build robust predictive (causal) network models using two large human multi-omics datasets. We delineate bulk-tissue gene expression into single cell-type gene expression and integrate clinical and pathologic traits, single nucleotide variation, and deconvoluted gene expression for the construction of cell type-specific predictive network models. Here, we focus on neuron-specific network models and prioritize 19 predicted key drivers modulating Alzheimer's pathology, which we then validate by knockdown in human induced pluripotent stem cell-derived neurons. We find that neuronal knockdown of 10 of the 19 targets significantly modulates levels of amyloid-beta and/or phosphorylated tau peptides, most notably JMJD6. We also confirm our network structure by RNA sequencing in the neurons following knockdown of each of the 10 targets, which additionally predicts that they are upstream regulators of REST and VGF. Our work thus identifies robust neuronal key drivers of the Alzheimer's-associated network state which may represent therapeutic targets with relevance to both amyloid and tau pathology in Alzheimer's disease.
Collapse
Affiliation(s)
- Julie P Merchant
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Neuroscience Graduate Group, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Kuixi Zhu
- The Center for Innovation in Brain Sciences, University of Arizona, Tucson, AZ, USA
| | - Marc Y R Henrion
- Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, Pembroke Place, L3 5QA, UK
- Malawi-Liverpool-Wellcome Trust Clinical Research Programme, PO Box 30096, Blantyre, Malawi
| | - Syed S A Zaidi
- The Center for Innovation in Brain Sciences, University of Arizona, Tucson, AZ, USA
| | - Branden Lau
- The Center for Innovation in Brain Sciences, University of Arizona, Tucson, AZ, USA
- Arizona Research Labs, Genetics Core, University of Arizona, Tucson, AZ, USA
| | - Sara Moein
- The Center for Innovation in Brain Sciences, University of Arizona, Tucson, AZ, USA
| | - Melissa L Alamprese
- The Center for Innovation in Brain Sciences, University of Arizona, Tucson, AZ, USA
| | - Richard V Pearse
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Nilüfer Ertekin-Taner
- Department of Neuroscience, Mayo Clinic Florida, Jacksonville, FL, USA
- Department of Neurology, Mayo Clinic Florida, Jacksonville, FL, USA
| | - Tracy L Young-Pearse
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.
- Harvard Stem Cell Institute, Harvard University, Boston, MA, USA.
| | - Rui Chang
- The Center for Innovation in Brain Sciences, University of Arizona, Tucson, AZ, USA.
- Department of Neurology, University of Arizona, Tucson, AZ, USA.
- INTelico Therapeutics LLC, Tucson, AZ, USA.
- PATH Biotech LLC, Tucson, AZ, USA.
| |
Collapse
|
37
|
Udeochu JC, Amin S, Huang Y, Fan L, Torres ERS, Carling GK, Liu B, McGurran H, Coronas-Samano G, Kauwe G, Mousa GA, Wong MY, Ye P, Nagiri RK, Lo I, Holtzman J, Corona C, Yarahmady A, Gill MT, Raju RM, Mok SA, Gong S, Luo W, Zhao M, Tracy TE, Ratan RR, Tsai LH, Sinha SC, Gan L. Tau activation of microglial cGAS-IFN reduces MEF2C-mediated cognitive resilience. Nat Neurosci 2023; 26:737-750. [PMID: 37095396 PMCID: PMC10166855 DOI: 10.1038/s41593-023-01315-6] [Citation(s) in RCA: 128] [Impact Index Per Article: 64.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Accepted: 03/20/2023] [Indexed: 04/26/2023]
Abstract
Pathological hallmarks of Alzheimer's disease (AD) precede clinical symptoms by years, indicating a period of cognitive resilience before the onset of dementia. Here, we report that activation of cyclic GMP-AMP synthase (cGAS) diminishes cognitive resilience by decreasing the neuronal transcriptional network of myocyte enhancer factor 2c (MEF2C) through type I interferon (IFN-I) signaling. Pathogenic tau activates cGAS and IFN-I responses in microglia, in part mediated by cytosolic leakage of mitochondrial DNA. Genetic ablation of Cgas in mice with tauopathy diminished the microglial IFN-I response, preserved synapse integrity and plasticity and protected against cognitive impairment without affecting the pathogenic tau load. cGAS ablation increased, while activation of IFN-I decreased, the neuronal MEF2C expression network linked to cognitive resilience in AD. Pharmacological inhibition of cGAS in mice with tauopathy enhanced the neuronal MEF2C transcriptional network and restored synaptic integrity, plasticity and memory, supporting the therapeutic potential of targeting the cGAS-IFN-MEF2C axis to improve resilience against AD-related pathological insults.
Collapse
Affiliation(s)
- Joe C Udeochu
- Helen and Robert Appel Alzheimer's Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Sadaf Amin
- Helen and Robert Appel Alzheimer's Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA.
| | - Yige Huang
- Helen and Robert Appel Alzheimer's Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Li Fan
- Helen and Robert Appel Alzheimer's Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Eileen Ruth S Torres
- Helen and Robert Appel Alzheimer's Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Gillian K Carling
- Helen and Robert Appel Alzheimer's Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Bangyan Liu
- Helen and Robert Appel Alzheimer's Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Hugo McGurran
- The Gladstone Institute of Neurological Disease, San Francisco, CA, USA
| | - Guillermo Coronas-Samano
- Helen and Robert Appel Alzheimer's Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Grant Kauwe
- Buck Institute for Research on Aging, Novato, CA, USA
| | - Gergey Alzaem Mousa
- Helen and Robert Appel Alzheimer's Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Man Ying Wong
- Helen and Robert Appel Alzheimer's Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Pearly Ye
- Helen and Robert Appel Alzheimer's Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Ravi Kumar Nagiri
- Helen and Robert Appel Alzheimer's Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Iris Lo
- The Gladstone Institute of Neurological Disease, San Francisco, CA, USA
| | - Julia Holtzman
- The Gladstone Institute of Neurological Disease, San Francisco, CA, USA
| | - Carlo Corona
- Burke Neurological Institute at Weill Cornell Medicine, White Plains, NY, USA
| | - Allan Yarahmady
- Department of Biochemistry, University of Alberta, Edmonton, Alberta, Canada
| | - Michael T Gill
- The Gladstone Institute of Neurological Disease, San Francisco, CA, USA
| | - Ravikiran M Raju
- The Picower Institute of Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
- Division of Newborn Medicine, Boston Children's Hospital, Boston, MA, USA
| | - Sue-Ann Mok
- Department of Biochemistry, University of Alberta, Edmonton, Alberta, Canada
| | - Shiaoching Gong
- Helen and Robert Appel Alzheimer's Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Wenjie Luo
- Helen and Robert Appel Alzheimer's Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Mingrui Zhao
- Helen and Robert Appel Alzheimer's Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Tara E Tracy
- Buck Institute for Research on Aging, Novato, CA, USA
| | - Rajiv R Ratan
- Burke Neurological Institute at Weill Cornell Medicine, White Plains, NY, USA
| | - Li-Huei Tsai
- The Picower Institute of Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Subhash C Sinha
- Helen and Robert Appel Alzheimer's Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Li Gan
- Helen and Robert Appel Alzheimer's Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
38
|
Feng J, Zheng N, Fan X, Li S, Jiang Y, Yi X, Yang H. Association of laxatives use with incident dementia and modifying effect of genetic susceptibility: a population-based cohort study with propensity score matching. BMC Geriatr 2023; 23:122. [PMID: 36870957 PMCID: PMC9985868 DOI: 10.1186/s12877-023-03854-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 02/28/2023] [Indexed: 03/06/2023] Open
Abstract
BACKGROUND Constipation was associated with incidence of dementia and cognitive decline. Laxatives are the mainstay of constipation management and are commonly used among older populations for both treatment and prevention of constipation. However, the association between use of laxatives and incident dementia, and whether laxatives use may modify the effect of genetic predisposition on dementia remains unclear. METHODS We applied 1:3 propensity score matching to balance the baseline characteristics of the laxative users versus non-users and to reduce potential confounders using multi-variates adjusted Cox hazards regression models. We categorized genetic risk into three groups (low, middle, and high) through a genetic risk score of common genetic variants. Information on laxatives use was assessed at baseline and categories into four varieties, including bulk forming laxatives, softeners and emollients, osmotic laxatives, and stimulant laxatives. RESULTS Of 486,994 participants, there were 14,422 laxatives users in UK Biobank. After propensity score matching, participants with use of laxatives (n = 14,422) and matched non-laxative (n = 43,266) exposed individuals were enrolled. Over follow-up to 15 years, there were 1377 participants developed dementia (539 for Alzheimer's disease, and 343 for vascular dementia). The use of laxatives had greater risk of dementia (HR, 1.72; 95% CI:1.54-1.92), Alzheimer's disease (HR, 1.36; 95% CI: 1.13-1.63), and vascular dementia (HR, 1.53; 95% CI: 1.23-1.92). Compared to non-laxative exposed participants, those with use of softeners and emollients drugs, stimulant laxatives, and osmotic laxatives were associated with 96% (HR, 1.96; 95 CI: 1.23-3.12; P = 0.005), 80% (HR, 1.80; 95% CI: 1.37-2.37; P < 0.001), and 107% (HR, 2.07; 95% CI: 1.47-2.92; P < 0.001) higher risk of developed incident dementia, respectively. In joint effect analysis, compared to participants with low/middle genetic susceptibility and non-laxatives use, the HR (95% CIs) of dementia was 4.10 (3.49-4.81) for those with high genetic susceptibility plus use of laxatives. There was an additive interaction between laxatives use and genetic susceptibility on dementia (RERI: 0.736, 95% CI: 0.127 to 1.246; AP: 0.180, 95% CI: 0.047 to 0.312). CONCLUSIONS Use of laxatives was associated with higher risk of dementia and modify the effect of genetic susceptibility on dementia. Our findings suggested that attention should be paid to the relationship between laxatives use and dementia, especially in people at high genetic susceptibility.
Collapse
Affiliation(s)
- Jiangtao Feng
- Department of Orthopedics, Tianjin NanKai Hospital, Tianjin, 300100, China
| | - Nan Zheng
- Department of Network Security and Informatization, Tianjin Medical University, Tianjin, 300070, China
| | - Xutong Fan
- Department of Bioinformatics, School of Basic Medical Sciences, Tianjin Medical University, Qixiangtai Road 22, Heping District, Tianjin, 300070, China
| | - Shu Li
- School of Management, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Yuhan Jiang
- Department of Bioinformatics, School of Basic Medical Sciences, Tianjin Medical University, Qixiangtai Road 22, Heping District, Tianjin, 300070, China
| | - Xianfu Yi
- Department of Bioinformatics, School of Basic Medical Sciences, Tianjin Medical University, Qixiangtai Road 22, Heping District, Tianjin, 300070, China
| | - Hongxi Yang
- Department of Bioinformatics, School of Basic Medical Sciences, Tianjin Medical University, Qixiangtai Road 22, Heping District, Tianjin, 300070, China. .,Department of Pharmacology, Tianjin Key Laboratory of Inflammation Biology, School of Basic Medical Sciences, Tianjin Medical University, Qixiangtai Road 22, Heping District, Tianjin, 300070, China.
| |
Collapse
|
39
|
Sha Z, Schijven D, Fisher SE, Francks C. Genetic architecture of the white matter connectome of the human brain. SCIENCE ADVANCES 2023; 9:eadd2870. [PMID: 36800424 PMCID: PMC9937579 DOI: 10.1126/sciadv.add2870] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 01/13/2023] [Indexed: 06/18/2023]
Abstract
White matter tracts form the structural basis of large-scale brain networks. We applied brain-wide tractography to diffusion images from 30,810 adults (U.K. Biobank) and found significant heritability for 90 node-level and 851 edge-level network connectivity measures. Multivariate genome-wide association analyses identified 325 genetic loci, of which 80% had not been previously associated with brain metrics. Enrichment analyses implicated neurodevelopmental processes including neurogenesis, neural differentiation, neural migration, neural projection guidance, and axon development, as well as prenatal brain expression especially in stem cells, astrocytes, microglia, and neurons. The multivariate association profiles implicated 31 loci in connectivity between core regions of the left-hemisphere language network. Polygenic scores for psychiatric, neurological, and behavioral traits also showed significant multivariate associations with structural connectivity, each implicating distinct sets of brain regions with trait-relevant functional profiles. This large-scale mapping study revealed common genetic contributions to variation in the structural connectome of the human brain.
Collapse
Affiliation(s)
- Zhiqiang Sha
- Language and Genetics Department, Max Planck Institute for Psycholinguistics, Nijmegen, Netherlands
| | - Dick Schijven
- Language and Genetics Department, Max Planck Institute for Psycholinguistics, Nijmegen, Netherlands
| | - Simon E. Fisher
- Language and Genetics Department, Max Planck Institute for Psycholinguistics, Nijmegen, Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, Netherlands
| | - Clyde Francks
- Language and Genetics Department, Max Planck Institute for Psycholinguistics, Nijmegen, Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, Netherlands
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, Netherlands
| |
Collapse
|
40
|
Martinez AE, Weissberger G, Kuklenyik Z, He X, Meuret C, Parekh T, Rees JC, Parks BA, Gardner MS, King SM, Collier TS, Harrington MG, Sweeney MD, Wang X, Zlokovic BV, Joe E, Nation DA, Schneider LS, Chui HC, Barr JR, Han SD, Krauss RM, Yassine HN. The small HDL particle hypothesis of Alzheimer's disease. Alzheimers Dement 2023; 19:391-404. [PMID: 35416404 PMCID: PMC10563117 DOI: 10.1002/alz.12649] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 02/08/2022] [Accepted: 02/12/2022] [Indexed: 01/03/2023]
Abstract
We propose the hypothesis that small high-density lipoprotein (HDL) particles reduce the risk of Alzheimer's disease (AD) by virtue of their capacity to exchange lipids, affecting neuronal membrane composition and vascular and synaptic functions. Concentrations of small HDLs in cerebrospinal fluid (CSF) and plasma were measured in 180 individuals ≥60 years of age using ion mobility methodology. Small HDL concentrations in CSF were positively associated with performance in three domains of cognitive function independent of apolipoprotein E (APOE) ε4 status, age, sex, and years of education. Moreover, there was a significant correlation between levels of small HDLs in CSF and plasma. Further studies will be aimed at determining whether specific components of small HDL exchange across the blood, brain, and CSF barriers, and developing approaches to exploit small HDLs for therapeutic purposes.
Collapse
Affiliation(s)
- Ashley E. Martinez
- Department of Medicine, University of Southern California, Los Angeles, California, USA
| | - Gali Weissberger
- The Interdisciplinary Department of Social Sciences, Bar Ilan University, Israel
| | - Zsuzsanna Kuklenyik
- Clinical Chemistry Branch, Division of Laboratory Sciences, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Xulei He
- Department of Medicine, University of Southern California, Los Angeles, California, USA
| | - Cristiana Meuret
- Department of Medicine, University of Southern California, Los Angeles, California, USA
| | - Trusha Parekh
- Department of Medicine, University of Southern California, Los Angeles, California, USA
| | - Jon C. Rees
- Clinical Chemistry Branch, Division of Laboratory Sciences, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Bryan A. Parks
- Clinical Chemistry Branch, Division of Laboratory Sciences, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Michael S. Gardner
- Clinical Chemistry Branch, Division of Laboratory Sciences, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Sarah M. King
- Departments of Pediatrics and Medicine, University of California, San Francisco, California, USA
| | | | - Michael G. Harrington
- Department of Neurology, University of Southern California, Los Angeles, California, USA
| | - Melanie D. Sweeney
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, California, USA
| | - Xinhui Wang
- Department of Neurology, University of Southern California, Los Angeles, California, USA
| | - Berislav V. Zlokovic
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, California, USA
| | - Elizabeth Joe
- Department of Neurology, University of Southern California, Los Angeles, California, USA
| | - Daniel A. Nation
- Irvine, Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, California, USA
| | - Lon S. Schneider
- Department of Neurology, University of Southern California, Los Angeles, California, USA
- Department of Psychiatry and Behavioral Sciences, University of Southern California, Los Angeles, California, USA
| | - Helena C. Chui
- Department of Neurology, University of Southern California, Los Angeles, California, USA
| | - John R. Barr
- Clinical Chemistry Branch, Division of Laboratory Sciences, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - S. Duke Han
- Department of Family Medicine, University of Southern California, Los Angeles, California, USA
| | - Ronald M. Krauss
- Departments of Pediatrics and Medicine, University of California, San Francisco, California, USA
| | - Hussein N. Yassine
- Department of Medicine, University of Southern California, Los Angeles, California, USA
- Department of Neurology, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
41
|
Moore JM, Bell EL, Hughes RO, Garfield AS. ABC transporters: human disease and pharmacotherapeutic potential. Trends Mol Med 2023; 29:152-172. [PMID: 36503994 DOI: 10.1016/j.molmed.2022.11.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 10/24/2022] [Accepted: 11/01/2022] [Indexed: 12/12/2022]
Abstract
Adenosine triphosphate (ATP)-binding cassette (ABC) transporters are a 48-member superfamily of membrane proteins that actively transport a variety of biological substrates across lipid membranes. Their functional diversity defines an expansive involvement in myriad aspects of human biology. At least 21 ABC transporters underlie rare monogenic disorders, with even more implicated in the predisposition to and symptomology of common and complex diseases. Such broad (patho)physiological relevance places this class of proteins at the intersection of disease causation and therapeutic potential, underlining them as promising targets for drug discovery, as exemplified by the transformative CFTR (ABCC7) modulator therapies for cystic fibrosis. This review will explore the growing relevance of ABC transporters to human disease and their potential as small-molecule drug targets.
Collapse
|
42
|
Oatman SR, Reddy JS, Quicksall Z, Carrasquillo MM, Wang X, Liu CC, Yamazaki Y, Nguyen TT, Malphrus K, Heckman M, Biswas K, Nho K, Baker M, Martens YA, Zhao N, Kim JP, Risacher SL, Rademakers R, Saykin AJ, DeTure M, Murray ME, Kanekiyo T, for the Alzheimer’s Disease Neuroimaging Initiative, Dickson DW, Bu G, Allen M, Ertekin-Taner N. Genome-wide association study of brain biochemical phenotypes reveals distinct genetic architecture of Alzheimer's disease related proteins. Mol Neurodegener 2023; 18:2. [PMID: 36609403 PMCID: PMC9825010 DOI: 10.1186/s13024-022-00592-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 12/19/2022] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is neuropathologically characterized by amyloid-beta (Aβ) plaques and neurofibrillary tangles. The main protein components of these hallmarks include Aβ40, Aβ42, tau, phosphor-tau, and APOE. We hypothesize that genetic variants influence the levels and solubility of these AD-related proteins in the brain; identifying these may provide key insights into disease pathogenesis. METHODS Genome-wide genotypes were collected from 441 AD cases, imputed to the haplotype reference consortium (HRC) panel, and filtered for quality and frequency. Temporal cortex levels of five AD-related proteins from three fractions, buffer-soluble (TBS), detergent-soluble (Triton-X = TX), and insoluble (Formic acid = FA), were available for these same individuals. Variants were tested for association with each quantitative biochemical measure using linear regression, and GSA-SNP2 was used to identify enriched Gene Ontology (GO) terms. Implicated variants and genes were further assessed for association with other relevant variables. RESULTS We identified genome-wide significant associations at seven novel loci and the APOE locus. Genes and variants at these loci also associate with multiple AD-related measures, regulate gene expression, have cell-type specific enrichment, and roles in brain health and other neuropsychiatric diseases. Pathway analysis identified significant enrichment of shared and distinct biological pathways. CONCLUSIONS Although all biochemical measures tested reflect proteins core to AD pathology, our results strongly suggest that each have unique genetic architecture and biological pathways that influence their specific biochemical states in the brain. Our novel approach of deep brain biochemical endophenotype GWAS has implications for pathophysiology of proteostasis in AD that can guide therapeutic discovery efforts focused on these proteins.
Collapse
Affiliation(s)
- Stephanie R. Oatman
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL 32224 USA
| | - Joseph S. Reddy
- Department of Quantitative Health Sciences, Mayo Clinic, Jacksonville, FL USA
| | - Zachary Quicksall
- Department of Quantitative Health Sciences, Mayo Clinic, Jacksonville, FL USA
| | | | - Xue Wang
- Department of Quantitative Health Sciences, Mayo Clinic, Jacksonville, FL USA
| | - Chia-Chen Liu
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL 32224 USA
| | - Yu Yamazaki
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL 32224 USA
| | - Thuy T. Nguyen
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL 32224 USA
| | - Kimberly Malphrus
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL 32224 USA
| | - Michael Heckman
- Department of Quantitative Health Sciences, Mayo Clinic, Jacksonville, FL USA
| | - Kristi Biswas
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL 32224 USA
| | - Kwangsik Nho
- Indiana Alzheimer Disease Center, Indiana University School of Medicine, Indianapolis, IN USA
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN USA
- School of Informatics and Computing, Indiana University School of Medicine, Indianapolis, IN USA
| | - Matthew Baker
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL 32224 USA
| | - Yuka A. Martens
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL 32224 USA
| | - Na Zhao
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL 32224 USA
| | - Jun Pyo Kim
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN USA
| | - Shannon L. Risacher
- Indiana Alzheimer Disease Center, Indiana University School of Medicine, Indianapolis, IN USA
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN USA
| | - Rosa Rademakers
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL 32224 USA
- VIB-UA Center for Molecular Neurology, VIB, University of Antwerp, Antwerp, Belgium
| | - Andrew J. Saykin
- Indiana Alzheimer Disease Center, Indiana University School of Medicine, Indianapolis, IN USA
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN USA
| | - Michael DeTure
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL 32224 USA
| | - Melissa E. Murray
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL 32224 USA
| | - Takahisa Kanekiyo
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL 32224 USA
| | - for the Alzheimer’s Disease Neuroimaging Initiative
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL 32224 USA
- Department of Quantitative Health Sciences, Mayo Clinic, Jacksonville, FL USA
- Indiana Alzheimer Disease Center, Indiana University School of Medicine, Indianapolis, IN USA
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN USA
- School of Informatics and Computing, Indiana University School of Medicine, Indianapolis, IN USA
- VIB-UA Center for Molecular Neurology, VIB, University of Antwerp, Antwerp, Belgium
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN USA
- Department of Neurology, Mayo Clinic, 4500 San Pablo Road, Birdsall 3, Jacksonville, FL 32224 USA
| | - Dennis W. Dickson
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL 32224 USA
| | - Guojun Bu
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL 32224 USA
| | - Mariet Allen
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL 32224 USA
| | - Nilüfer Ertekin-Taner
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL 32224 USA
- Department of Neurology, Mayo Clinic, 4500 San Pablo Road, Birdsall 3, Jacksonville, FL 32224 USA
| |
Collapse
|
43
|
Sinha N, Fausto BA, Mander B, Gluck MA. High-Quality Sleep Mitigates ABCA7-Related Generalization Deficits in Healthy Older African Americans. J Alzheimers Dis 2023; 94:281-290. [PMID: 37212111 PMCID: PMC10357211 DOI: 10.3233/jad-230043] [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] [Accepted: 04/22/2023] [Indexed: 05/23/2023]
Abstract
BACKGROUND Both sleep deficiencies and Alzheimer's disease (AD) disproportionately affect older African Americans. Genetic susceptibility to AD further compounds risk for cognitive decline in this population. Aside from APOE ɛ4, ABCA7 rs115550680 is the strongest genetic locus associated with late-onset AD in African Americans. While sleep and ABCA7 rs115550680 independently influence late-life cognitive outcomes, we know too little about the interplay between these two factors on cognitive function. OBJECTIVE We investigated the interaction between sleep and ABCA7 rs115550680 on hippocampal-dependent cognitive function in older African Americans. METHODS One-hundred fourteen cognitively healthy older African Americans were genotyped for ABCA7 risk (n = 57 carriers of risk "G" allele; n = 57 non-carriers), responded to lifestyle questionnaires, and completed a cognitive battery. Sleep was assessed via a self-reported rating of sleep quality (poor, average, good). Covariates included age and years of education. RESULTS Using ANCOVA, we found that carriers of the risk genotype who reported poor or average sleep quality demonstrated significantly poorer generalization of prior learning-a cognitive marker of AD-compared to their non-risk counterparts. Conversely, there was no genotype-related difference in generalization performance in individuals who reported good sleep quality. CONCLUSION These results indicate that sleep quality may be neuroprotective against genetic risk for AD. Future studies employing more rigorous methodology should investigate the mechanistic role of sleep neurophysiology in the pathogenesis and progression of AD associated with ABCA7. There is also need for the continued development of non-invasive sleep interventions tailored to racial groups with specific AD genetic risk profiles.
Collapse
Affiliation(s)
- Neha Sinha
- Aging & Brain Health Alliance, Center for Molecular & Behavioral Neuroscience, Rutgers University – Newark, Newark, NJ, USA
| | - Bernadette A. Fausto
- Aging & Brain Health Alliance, Center for Molecular & Behavioral Neuroscience, Rutgers University – Newark, Newark, NJ, USA
| | - Bryce Mander
- Center for the Neurobiology of Learning and Memory, University of California, Irvine, CA, USA
- Department of Psychiatry and Human Behavior, University of California, Irvine, CA, USA, USA
| | - Mark A. Gluck
- Aging & Brain Health Alliance, Center for Molecular & Behavioral Neuroscience, Rutgers University – Newark, Newark, NJ, USA
| |
Collapse
|
44
|
Guo Y, Sun CK, Tang L, Tan MS. Microglia PTK2B/Pyk2 in the Pathogenesis of Alzheimer's Disease. Curr Alzheimer Res 2023; 20:692-704. [PMID: 38321895 DOI: 10.2174/0115672050299004240129051655] [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: 12/16/2023] [Revised: 01/16/2024] [Accepted: 01/22/2024] [Indexed: 02/08/2024]
Abstract
Alzheimer's disease (AD) is a highly hereditary disease with complex genetic susceptibility factors. Extensive genome-wide association studies have established a distinct susceptibility link between the protein tyrosine kinase 2β (PTK2B) gene and late-onset Alzheimer's disease (LOAD), but the specific pathogenic mechanisms remain incompletely understood. PTK2B is known to be expressed in neurons, and recent research has revealed its more important significance in microglia. Elucidating the role of PTK2B high expression in microglia in AD's progression is crucial for uncovering novel pathogenic mechanisms of the disease. Our review of existing studies suggests a close relationship between PTK2B/proline-rich tyrosine kinase 2 (Pyk2) and tau pathology, and this process might be β-amyloid (Aβ) dependence. Pyk2 is hypothesized as a pivotal target linking Aβ and tau pathologies. Concurrently, Aβ-activated Pyk2 participates in the regulation of microglial activation and its proinflammatory functions. Consequently, it is reasonable to presume that Pyk2 in microglia contributes to amyloid-induced tau pathology in AD via a neuroinflammatory pathway. Furthermore, many things remain unclear, such as identifying the specific pathways that lead to the release of downstream inflammatory factors due to Pyk2 phosphorylation and whether all types of inflammatory factors can activate neuronal kinase pathways. Additionally, further in vivo experiments are essential to validate this hypothesized pathway. Considering PTK2B/Pyk2's potential role in AD pathogenesis, targeting this pathway may offer innovative and promising therapeutic approaches for AD.
Collapse
Affiliation(s)
- Yun Guo
- School of Clinical Medicine, Weifang Medical University, Weifang, China
| | - Cheng-Kun Sun
- Department of Neurology, Qingdao Municipal Hospital, Dalian Medical University, Qingdao, China
| | - Lian Tang
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Meng-Shan Tan
- School of Clinical Medicine, Weifang Medical University, Weifang, China
- Department of Neurology, Qingdao Municipal Hospital, Dalian Medical University, Qingdao, China
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
- Department of Neurology, Qingdao Municipal Hospital, University of Health and Rehabilitation Sciences, Qingdao, China
| |
Collapse
|
45
|
Zahra A, Liu R, Han W, Meng H, Wang Q, Wang Y, Campbell SL, Wu J. K Ca-Related Neurological Disorders: Phenotypic Spectrum and Therapeutic Indications. Curr Neuropharmacol 2023; 21:1504-1518. [PMID: 36503451 PMCID: PMC10472807 DOI: 10.2174/1570159x21666221208091805] [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: 04/21/2022] [Revised: 07/21/2022] [Accepted: 07/23/2022] [Indexed: 12/14/2022] Open
Abstract
Although potassium channelopathies have been linked to a wide range of neurological conditions, the underlying pathogenic mechanism is not always clear, and a systematic summary of clinical manifestation is absent. Several neurological disorders have been associated with alterations of calcium-activated potassium channels (KCa channels), such as loss- or gain-of-function mutations, post-transcriptional modification, etc. Here, we outlined the current understanding of the molecular and cellular properties of three subtypes of KCa channels, including big conductance KCa channels (BK), small conductance KCa channels (SK), and the intermediate conductance KCa channels (IK). Next, we comprehensively reviewed the loss- or gain-of-function mutations of each KCa channel and described the corresponding mutation sites in specific diseases to broaden the phenotypic-genotypic spectrum of KCa-related neurological disorders. Moreover, we reviewed the current pharmaceutical strategies targeting KCa channels in KCa-related neurological disorders to provide new directions for drug discovery in anti-seizure medication.
Collapse
Affiliation(s)
- Aqeela Zahra
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan 430070, China
- Department of Zoology, University of Sialkot, Sialkot 51310, Pakistan
| | - Ru Liu
- Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing 100070, China
- National Clinical Research Center for Neurological Diseases, Beijing 100070, China
| | - Wenzhe Han
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan 430070, China
| | - Hui Meng
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan 430070, China
| | - Qun Wang
- Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
- National Clinical Research Center for Neurological Diseases, Beijing 100070, China
| | - YunFu Wang
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan 430070, China
| | - Susan L. Campbell
- Animal and Poultry Sciences, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States
| | - Jianping Wu
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan 430070, China
- Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing 100070, China
- National Clinical Research Center for Neurological Diseases, Beijing 100070, China
| |
Collapse
|
46
|
Bonaterra-Pastra A, Benítez S, Pancorbo O, Rodríguez-Luna D, Vert C, Rovira A, Freijo MM, Tur S, Martínez-Zabaleta M, Cardona Portela P, Vera R, Lebrato-Hernández L, Arenillas JF, Pérez-Sánchez S, Domínguez-Mayoral A, Fàbregas JM, Mauri G, Montaner J, Sánchez-Quesada JL, Hernández-Guillamon M. Association of candidate genetic variants and circulating levels of ApoE/ApoJ with common neuroimaging features of cerebral amyloid angiopathy. Front Aging Neurosci 2023; 15:1134399. [PMID: 37113571 PMCID: PMC10126235 DOI: 10.3389/fnagi.2023.1134399] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 03/27/2023] [Indexed: 04/29/2023] Open
Abstract
Introduction Cerebral amyloid angiopathy (CAA) is characterized by the accumulation of amyloid-β (Aβ) in brain vessels and is a main cause of lobar intracerebral hemorrhage (ICH) in the elderly. CAA is associated with magnetic resonance imaging (MRI) markers of small vessel disease (SVD). Since Aβ is also accumulated in Alzheimer's disease (AD) in the brain parenchyma, we aimed to study if several single nucleotide polymorphisms (SNPs) previously associated with AD were also associated with CAA pathology. Furthermore, we also studied the influence of APOE and CLU genetic variants in apolipoprotein E (ApoE) and clusterin/apolipoprotein J (ApoJ) circulating levels and their distribution among lipoproteins. Methods The study was carried out in a multicentric cohort of 126 patients with lobar ICH and clinical suspicion of CAA. Results We observed several SNPs associated with CAA neuroimaging MRI markers [cortical superficial siderosis (cSS), enlarged perivascular spaces in the centrum semiovale (CSO-EPVS), lobar cerebral microbleeds (CMB), white matter hyperintensities (WMH), corticosubcortical atrophy and CAA-SVD burden score]. Concretely, ABCA7 (rs3764650), CLU (rs9331896 and rs933188), EPHA1 (rs11767557), and TREML2 (rs3747742) were significantly associated with a CAA-SVD burden score. Regarding circulating levels of apolipoproteins, protective AD SNPs of CLU [rs11136000 (T) and rs9331896 (C)] were significantly associated with higher HDL ApoJ content in the lobar ICH cohort. APOEε2 carriers presented higher plasma and LDL-associated ApoE levels whereas APOEε4 carriers presented lower plasma ApoE levels. Additionally, we observed that lower circulating ApoJ and ApoE levels were significantly associated with CAA-related MRI markers. More specifically, lower LDL-associated ApoJ and plasma and HDL-associated ApoE levels were significantly associated with CSO-EPVS, lower ApoJ content in HDL with brain atrophy and lower ApoE content in LDL with the extent of cSS. Discussion This study reinforces the relevance of lipid metabolism in CAA and cerebrovascular functionality. We propose that ApoJ and ApoE distribution among lipoproteins may be associated with pathological features related to CAA with higher ApoE and ApoJ levels in HDL possibly enhancing atheroprotective, antioxidative, and anti-inflammatory responses in cerebral β-amyloidosis.
Collapse
Affiliation(s)
- Anna Bonaterra-Pastra
- Neurovascular Research Laboratory, Vall d’Hebron Research Institute, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Sònia Benítez
- Cardiovascular Biochemistry Group, Research Institute of the Hospital de Sant Pau (IIB Sant Pau), Barcelona, Spain
- Center for Biomedical Research Network on Diabetes and Associated Metabolic Diseases (CIBERDEM), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Olalla Pancorbo
- Stroke Research Group, Vall d’Hebron Research Institute, Barcelona, Spain
| | | | - Carla Vert
- Section of Neuroradiology, Department of Radiology, Vall d’Hebron University Hospital, Vall d’Hebron Research Institute, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Alex Rovira
- Section of Neuroradiology, Department of Radiology, Vall d’Hebron University Hospital, Vall d’Hebron Research Institute, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - M. Mar Freijo
- Neurovascular Group, BioCruces Health Research Institute, Barakaldo, Spain
| | - Silvia Tur
- Department of Neurology, Son Espases University Hospital, Balearic Islands, Spain
| | | | - Pere Cardona Portela
- Department of Neurology, Bellvitge University Hospital, L’Hospitalet de Llobregat, Spain
| | - Rocío Vera
- Stroke Unit, Department of Neurology, Ramón y Cajal University Hospital, Madrid, Spain
| | - Lucia Lebrato-Hernández
- Stroke Unit, Department of Neurology and Neurophysiology, Virgen del Rocío University Hospital, Seville, Spain
| | - Juan F. Arenillas
- Stroke Program, Department of Neurology, Hospital Clínico Universitario, Valladolid, Spain
- Clinical Neurosciences Research Group, Department of Medicine, University of Valladolid, Valladolid, Spain
| | | | | | - Joan Martí Fàbregas
- Stroke Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Gerard Mauri
- Stroke Unit, Department of Neurology, Hospital Universitari Arnau de Vilanova de Lleida, Lleida, Spain
| | - Joan Montaner
- Neurovascular Research Laboratory, Vall d’Hebron Research Institute, Universitat Autònoma de Barcelona, Barcelona, Spain
- Stroke Research Program, Institute of Biomedicine of Seville (IBiS), Virgen del Rocío University Hospital, University of Seville, Seville, Spain
- Department of Neurology, Virgen Macarena University Hospital, Seville, Spain
| | - Jose Luis Sánchez-Quesada
- Cardiovascular Biochemistry Group, Research Institute of the Hospital de Sant Pau (IIB Sant Pau), Barcelona, Spain
- Center for Biomedical Research Network on Diabetes and Associated Metabolic Diseases (CIBERDEM), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Jose Luis Sánchez-Quesada,
| | - Mar Hernández-Guillamon
- Neurovascular Research Laboratory, Vall d’Hebron Research Institute, Universitat Autònoma de Barcelona, Barcelona, Spain
- *Correspondence: Mar Hernández-Guillamon,
| |
Collapse
|
47
|
Rodríguez-Giraldo M, González-Reyes RE, Ramírez-Guerrero S, Bonilla-Trilleras CE, Guardo-Maya S, Nava-Mesa MO. Astrocytes as a Therapeutic Target in Alzheimer's Disease-Comprehensive Review and Recent Developments. Int J Mol Sci 2022; 23:13630. [PMID: 36362415 PMCID: PMC9654484 DOI: 10.3390/ijms232113630] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 10/25/2022] [Accepted: 10/26/2022] [Indexed: 09/20/2023] Open
Abstract
Alzheimer's disease (AD) is a frequent and disabling neurodegenerative disorder, in which astrocytes participate in several pathophysiological processes including neuroinflammation, excitotoxicity, oxidative stress and lipid metabolism (along with a critical role in apolipoprotein E function). Current evidence shows that astrocytes have both neuroprotective and neurotoxic effects depending on the disease stage and microenvironmental factors. Furthermore, astrocytes appear to be affected by the presence of amyloid-beta (Aβ), with alterations in calcium levels, gliotransmission and proinflammatory activity via RAGE-NF-κB pathway. In addition, astrocytes play an important role in the metabolism of tau and clearance of Aβ through the glymphatic system. In this review, we will discuss novel pharmacological and non-pharmacological treatments focused on astrocytes as therapeutic targets for AD. These interventions include effects on anti-inflammatory/antioxidant systems, glutamate activity, lipid metabolism, neurovascular coupling and glymphatic system, calcium dysregulation, and in the release of peptides which affects glial and neuronal function. According to the AD stage, these therapies may be of benefit in either preventing or delaying the progression of the disease.
Collapse
Affiliation(s)
| | | | | | | | | | - Mauricio O. Nava-Mesa
- Grupo de Investigación en Neurociencias (NeURos), Centro de Neurociencias Neurovitae-UR, Instituto de Medicina Traslacional (IMT), Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Bogotá 111711, Colombia
| |
Collapse
|
48
|
Jun GR, You Y, Zhu C, Meng G, Chung J, Panitch R, Hu J, Xia W, The Alzheimer's Disease Genetics Consortium, Bennett DA, Foroud TM, Wang L, Haines JL, Mayeux R, Pericak‐Vance MA, Schellenberg GD, Au R, Lunetta KL, Ikezu T, Stein TD, Farrer LA. Protein phosphatase 2A and complement component 4 are linked to the protective effect of APOE ɛ2 for Alzheimer's disease. Alzheimers Dement 2022; 18:2042-2054. [PMID: 35142023 PMCID: PMC9360190 DOI: 10.1002/alz.12607] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 12/27/2021] [Accepted: 01/01/2022] [Indexed: 01/31/2023]
Abstract
INTRODUCTION The apolipoprotein E (APOE) ɛ2 allele reduces risk against Alzheimer's disease (AD) but mechanisms underlying this effect are largely unknown. METHODS We conducted a genome-wide association study for AD among 2096 ɛ2 carriers. The potential role of the top-ranked gene and complement 4 (C4) proteins, which were previously linked to AD in ɛ2 carriers, was investigated using human isogenic APOE allele-specific induced pluripotent stem cell (iPSC)-derived neurons and astrocytes and in 224 neuropathologically examined human brains. RESULTS PPP2CB rs117296832 was the second most significantly associated single nucleotide polymorphism among ɛ2 carriers (P = 1.1 × 10-7 ) and the AD risk allele increased PPP2CB expression in blood (P = 6.6 × 10-27 ). PPP2CB expression was correlated with phosphorylated tau231/total tau ratio (P = .01) and expression of C4 protein subunits C4A/B (P = 2.0 × 10-4 ) in the iPSCs. PPP2CB (subunit of protein phosphatase 2A) and C4b protein levels were correlated in brain (P = 3.3 × 10-7 ). DISCUSSION PP2A may be linked to classical complement activation leading to AD-related tau pathology.
Collapse
Affiliation(s)
- Gyungah R. Jun
- Department of Medicine (Biomedical Genetics), Boston University School of MedicineBostonMassachusettsUSA
- Department of Ophthalmology, Boston University School of MedicineBostonMassachusettsUSA
- Department of BiostatisticsBoston University School of Public HealthBostonMassachusettsUSA
| | - Yang You
- Department of Pharmacology & Experimental TherapeuticsBoston University School of MedicineBostonMassachusettsUSA
| | - Congcong Zhu
- Department of Medicine (Biomedical Genetics), Boston University School of MedicineBostonMassachusettsUSA
| | - Gaoyuan Meng
- Department of Veterans Affairs Medical CenterBedfordMassachusettsUSA
| | - Jaeyoon Chung
- Department of Medicine (Biomedical Genetics), Boston University School of MedicineBostonMassachusettsUSA
| | - Rebecca Panitch
- Department of Medicine (Biomedical Genetics), Boston University School of MedicineBostonMassachusettsUSA
| | - Junming Hu
- Department of Medicine (Biomedical Genetics), Boston University School of MedicineBostonMassachusettsUSA
| | - Weiming Xia
- Department of Pharmacology & Experimental TherapeuticsBoston University School of MedicineBostonMassachusettsUSA
- Department of Veterans Affairs Medical CenterBedfordMassachusettsUSA
| | | | - David A. Bennett
- Rush Alzheimer's Disease CenterRush University Medical CenterChicagoIllinoisUSA
| | - Tatiana M. Foroud
- Department of Medical and Molecular GeneticsIndiana UniversityIndianapolisIndianaUSA
| | - Li‐San Wang
- Penn Neurodegeneration Genomics Center, Department of Pathology and Laboratory MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Jonathan L. Haines
- Department of Population & Quantitative Health SciencesCase Western Reserve UniversityClevelandOhioUSA
| | - Richard Mayeux
- Taub Institute on Alzheimer's Disease and the Aging Brain, Gertrude H. Sergievsky Center Department of NeurologyColumbia UniversityNew YorkNew YorkUSA
| | - Margaret A. Pericak‐Vance
- John P. Hussman Institute for Human Genomics, Department of Human Genetics, and Dr. John T. Macdonald FoundationUniversity of MiamiMiamiFloridaUSA
| | - Gerard D. Schellenberg
- Penn Neurodegeneration Genomics Center, Department of Pathology and Laboratory MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Rhoda Au
- Department of Neurology, Boston University School of MedicineBostonMassachusettsUSA
- Department of Anatomy & Neurobiology, Boston University School of MedicineBostonMassachusettsUSA
- Department of EpidemiologyBoston University School of Public HealthBostonMassachusettsUSA
| | - Kathryn L. Lunetta
- Department of BiostatisticsBoston University School of Public HealthBostonMassachusettsUSA
| | - Tsuneya Ikezu
- Department of Pharmacology & Experimental TherapeuticsBoston University School of MedicineBostonMassachusettsUSA
- Department of Neurology, Boston University School of MedicineBostonMassachusettsUSA
- Center for Systems NeuroscienceBoston University School of MedicineBostonMassachusettsUSA
| | - Thor D. Stein
- Department of Veterans Affairs Medical CenterBedfordMassachusettsUSA
- Department of Pathology & Laboratory Medicine, Boston University School of MedicineBostonMassachusettsUSA
| | - Lindsay A. Farrer
- Department of Medicine (Biomedical Genetics), Boston University School of MedicineBostonMassachusettsUSA
- Department of Ophthalmology, Boston University School of MedicineBostonMassachusettsUSA
- Department of BiostatisticsBoston University School of Public HealthBostonMassachusettsUSA
- Department of Neurology, Boston University School of MedicineBostonMassachusettsUSA
- Department of EpidemiologyBoston University School of Public HealthBostonMassachusettsUSA
| |
Collapse
|
49
|
Kumar R, Tiwari V, Dey S. Role of proline-rich tyrosine kinase 2 (Pyk2) in the pathogenesis of Alzheimer's disease. Eur J Neurosci 2022; 56:5442-5452. [PMID: 34905657 DOI: 10.1111/ejn.15569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 12/02/2021] [Accepted: 12/03/2021] [Indexed: 12/14/2022]
Abstract
Alzheimer's disease (AD), a progressive neurodegenerative disorder, is the most common form of dementia in the elderly. Two major pathological hallmarks have been identified for AD: extracellular amyloid plaques and intracellular neurofibrillary tangles (NFT). Recently, proline-rich tyrosine kinase 2 (Pyk2), which belongs to the focal adhesion kinase (FAK) non-receptor tyrosine kinase family, was recognized to contribute significantly towards the pathogenesis of AD. Pyk2 can influence the formation of amyloid plaques as well as NFTs. The kinase can directly phosphorylate tau, which is a significant component of NFTs and enhances tau pathology. Several competitive inhibitors have been developed for Pyk2, tested in several cancer models, as Pyk2 is known to be overexpressed under those conditions. The current review article discusses the possible mechanistic pathways by which Pyk2 can influence the pathogenesis of AD. Besides, it describes various inhibitors for Pyk2 and their potential role as therapeutics for AD in the future.
Collapse
Affiliation(s)
- Rahul Kumar
- Department of Biotechnology, GITAM Institute of Sciences, GITAM University, Visakhapatnam, India
| | - Vishvanath Tiwari
- Department of Biochemistry, Central University of Rajasthan, Ajmer, India
| | - Sharmistha Dey
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
50
|
Katsumata Y, Shade LM, Hohman TJ, Schneider JA, Bennett DA, Farfel JM, Kukull WA, Fardo DW, Nelson PT. Multiple gene variants linked to Alzheimer's-type clinical dementia via GWAS are also associated with non-Alzheimer's neuropathologic entities. Neurobiol Dis 2022; 174:105880. [PMID: 36191742 PMCID: PMC9641973 DOI: 10.1016/j.nbd.2022.105880] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 09/27/2022] [Accepted: 09/29/2022] [Indexed: 11/26/2022] Open
Abstract
The classic pathologic hallmarks of Alzheimer's disease (AD) are amyloid plaques and neurofibrillary tangles (AD neuropathologic changes, or ADNC). However, brains from individuals clinically diagnosed with "AD-type" (amnestic) dementia usually harbor heterogeneous neuropathologies in addition to, or other than, ADNC. We hypothesized that some AD-type dementia associated genetic single nucleotide variants (SNVs) identified from large genomewide association studies (GWAS) were associated with non-ADNC neuropathologies. To test this hypothesis, we analyzed data from multiple studies with available genotype and neuropathologic phenotype information. Clinical AD/dementia risk alleles of interest were derived from the very large GWAS by Bellenguez et al. (2022) who reported 83 clinical AD/dementia-linked SNVs in addition to the APOE risk alleles. To query the pathologic phenotypes associated with variation of those SNVs, National Alzheimer's disease Coordinating Center (NACC) neuropathologic data were linked to AD Sequencing Project (ADSP) and AD Genomics Consortium (ADGC) data. Separate data were obtained from the harmonized Religious Orders Study and the Rush Memory and Aging Project (ROSMAP). A total of 4811 European participants had at least ADNC neuropathology data and also genotype data available; data were meta-analyzed across cohorts. As expected, a subset of dementia-associated SNVs were associated with ADNC risk in Europeans-e.g., BIN1, PICALM, CR1, MME, and COX7C. Other gene variants linked to (clinical) AD dementia were associated with non-ADNC pathologies. For example, the associations of GRN and TMEM106B SNVs with limbic-predominant age-related TDP-43 neuropathologic changes (LATE-NC) were replicated. In addition, SNVs in TNIP1 and WNT3 previously reported as AD-related were instead associated with hippocampal sclerosis pathology. Some genotype/neuropathology association trends were not statistically significant at P < 0.05 after correcting for multiple testing, but were intriguing. For example, variants in SORL1 and TPCN1 showed trends for association with LATE-NC whereas Lewy body pathology trended toward association with USP6NL and BIN1 gene variants. A smaller cohort of non-European subjects (n = 273, approximately one-half of whom were African-Americans) provided the basis for additional exploratory analyses. Overall, these findings were consistent with the hypothesis that some genetic variants linked to AD dementia risk exert their affect by influencing non-ADNC neuropathologies.
Collapse
Affiliation(s)
- Yuriko Katsumata
- Department of Biostatistics, University of Kentucky, Lexington, KY, USA; Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA
| | - Lincoln M Shade
- Department of Biostatistics, University of Kentucky, Lexington, KY, USA
| | - Timothy J Hohman
- Vanderbilt Memory and Alzheimer's Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Julie A Schneider
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA; Department of Pathology, Rush University Medical Center, Chicago, IL, USA; Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - David A Bennett
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA; Department of Pathology, Rush University Medical Center, Chicago, IL, USA; Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Jose M Farfel
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA; Department of Pathology, Rush University Medical Center, Chicago, IL, USA; Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Walter A Kukull
- Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - David W Fardo
- Department of Biostatistics, University of Kentucky, Lexington, KY, USA; Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA
| | - Peter T Nelson
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA; Department of Pathology, University of Kentucky, Lexington, KY 40536, USA.
| |
Collapse
|