1
|
Foncea A, Franchini N, Tobar I, Thienel S, Retamal IN, Cancino GI, Cornejo F. Ptprd deficiency promotes tau hyperphosphorylation and impairs cognitive function in aged mice. Biol Res 2025; 58:26. [PMID: 40329347 PMCID: PMC12054186 DOI: 10.1186/s40659-025-00607-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Accepted: 04/16/2025] [Indexed: 05/08/2025] Open
Abstract
BACKGROUND Tau phosphorylation is a tightly regulated process that ensures proper neuronal function. Indeed, hyperphosphorylation of tau closely contributes to neuronal dysfunction leading to neurodegenerative diseases, including tauopathies, which are characterized by excessive and aberrant tau phosphorylation and cognitive decline. Therefore, it is important to understand how to regulate its phosphorylation. In this regard, the protein tyrosine phosphatase receptor delta (PTPRD) has been genetically implicated in tau pathology in humans, but the mechanisms underlying its role in tau regulation remain unclear. This study investigates the impact of Ptprd deficiency on tau phosphorylation, cognitive function, neuroinflammation, and synaptic markers in aging mice. RESULTS Mice lacking Ptprd showed increased tau phosphorylation at multiple sites associated with its pathological aggregation. This effect was accompanied by the activation of the tau-related kinase Abl1, particularly in the hippocampus. Behavioral assessments revealed significant impairments in learning and memory, demonstrating the functional impact of these alterations. Moreover, Ptprd knockout mice showed increased microgliosis in both the entorhinal cortex and the hippocampus, suggesting a pro-inflammatory response. Furthermore, the synaptic protein PSD95 was also reduced in the cortex, indicating potential synaptic dysfunction. CONCLUSIONS The loss of Ptprd leads to increased tau phosphorylation, cognitive impairments, microgliosis, and synaptic alterations in older mice. Our findings also suggest that Ptprd plays a critical role in maintaining tau homeostasis through the Abl1 kinase. This indicates a new potential therapeutic approach for tauopathies, where PTPRD could serve a protective role against tau-related pathologies and may act as a key modulator in disease progression.
Collapse
Affiliation(s)
- Analía Foncea
- Centro de Biología Integrativa, Facultad de Ciencias, Universidad Mayor, Santiago, Chile
| | - Nayhara Franchini
- Centro de Biología Integrativa, Facultad de Ciencias, Universidad Mayor, Santiago, Chile
| | - Isidora Tobar
- Centro de Biología Integrativa, Facultad de Ciencias, Universidad Mayor, Santiago, Chile
| | - Sebastián Thienel
- Centro de Biología Integrativa, Facultad de Ciencias, Universidad Mayor, Santiago, Chile
| | - Ignacio N Retamal
- Centro de Oncología de Precisión, Escuela de Medicina, Facultad de Medicina y Ciencias de la Salud, Universidad Mayor, Santiago, Chile
| | - Gonzalo I Cancino
- Laboratorio de Neurobiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile.
| | - Francisca Cornejo
- Centro de Biología Integrativa, Facultad de Ciencias, Universidad Mayor, Santiago, Chile.
| |
Collapse
|
2
|
Puerta R, de Rojas I, García-González P, Olivé C, Sotolongo-Grau O, García-Sánchez A, García-Gutiérrez F, Montrreal L, Tartari JP, 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, López de Munain A, García-Alberca JM, Royo JL, Bullido MJ, Álvarez V, Real LM, Corbatón Anchuelo A, Gómez-Garre D, Martínez Larrad MT, 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, Sánchez Juan P, Cavazos JE, Cabrera-Socorro A, Cano A, Ruiz A. Linking genomic and proteomic signatures to brain amyloid burden: insights from GR@ACE/DEGESCO. Funct Integr Genomics 2025; 25:73. [PMID: 40133566 PMCID: PMC11937198 DOI: 10.1007/s10142-025-01581-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 03/12/2025] [Accepted: 03/13/2025] [Indexed: 03/27/2025]
Abstract
Alzheimer's disease (AD) is a complex disease with a strong genetic component, yet many 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 provide insights into the underlying biology of the disease. We performed a meta-GWAS of CSF Aβ42 and PET measures combining six independent cohorts (n = 2,076). Given the opposite beta direction of Aβ phenotypes in CSF and PET measures, only genetic signals showing opposite directions were considered for analysis (n = 376,599). We explored the amyloidosis signature in the CSF proteome using SOMAscan proteomics (ACE cohort, n = 1,008), connected it with GWAS loci modulating amyloidosis and performed an enrichment analysis of overlapping hits. Finally, we compared our results with a large meta-analysis using publicly available datasets in CSF (n = 13,409) and PET (n = 13,116). After filtering the meta-GWAS, we observed genome-wide significance in the rs429358-APOE locus and annotated nine suggestive hits. We replicated the APOE loci using the large CSF-PET meta-GWAS, identifying multiple AD-associated genes including the novel GADL1 locus. Additionally, we found 1,387 FDR-significant SOMAscan proteins associated with CSF Aβ42 levels. The overlap among GWAS loci and proteins associated with amyloid burden was minimal (n = 35). The enrichment analysis revealed mechanisms connecting amyloidosis with the plasma membrane's anchored component, synapse physiology and mental disorders that were replicated in the large CSF-PET meta-analysis. Combining CSF and PET amyloid GWAS with CSF proteome analyses may effectively elucidate causative molecular mechanisms behind amyloid mobilization and AD physiopathology.
Collapse
Affiliation(s)
- Raquel Puerta
- Ace Alzheimer Center Barcelona - Universitat Internacional de Catalunya, C/ Marquès de Sentmenat, 57, 08029, Barcelona, Spain
- Doctorate in Biotechnology, Facultat de Farmàcia i Ciències de l'Alimentació, Universitat de Barcelona, Avda. Diagonal 643, 08028, Barcelona, Spain
| | - Itziar de Rojas
- Ace Alzheimer Center Barcelona - Universitat Internacional de Catalunya, C/ Marquès de Sentmenat, 57, 08029, Barcelona, 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, C/ Marquès de Sentmenat, 57, 08029, Barcelona, 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, C/ Marquès de Sentmenat, 57, 08029, Barcelona, Spain
| | - Oscar Sotolongo-Grau
- Ace Alzheimer Center Barcelona - Universitat Internacional de Catalunya, C/ Marquès de Sentmenat, 57, 08029, Barcelona, Spain
| | - Ainhoa García-Sánchez
- Ace Alzheimer Center Barcelona - Universitat Internacional de Catalunya, C/ Marquès de Sentmenat, 57, 08029, Barcelona, Spain
| | - Fernando García-Gutiérrez
- Ace Alzheimer Center Barcelona - Universitat Internacional de Catalunya, C/ Marquès de Sentmenat, 57, 08029, Barcelona, Spain
| | - Laura Montrreal
- Ace Alzheimer Center Barcelona - Universitat Internacional de Catalunya, C/ Marquès de Sentmenat, 57, 08029, Barcelona, Spain
| | - Juan Pablo Tartari
- Ace Alzheimer Center Barcelona - Universitat Internacional de Catalunya, C/ Marquès de Sentmenat, 57, 08029, Barcelona, Spain
| | - Ángela Sanabria
- Ace Alzheimer Center Barcelona - Universitat Internacional de Catalunya, C/ Marquès de Sentmenat, 57, 08029, Barcelona, 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, C/ Marquès de Sentmenat, 57, 08029, Barcelona, 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
- Marqués de Valdecilla University Hospital- IDIVAL-Universidad de Cantabria, 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, C/ Marquès de Sentmenat, 57, 08029, Barcelona, Spain
| | - Eloy Rodriguez-Rodriguez
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
- Marqués de Valdecilla University Hospital- IDIVAL-Universidad de Cantabria, Santander, Spain
| | - Emilio Alarcón-Martín
- Ace Alzheimer Center Barcelona - Universitat Internacional de Catalunya, C/ Marquès de Sentmenat, 57, 08029, Barcelona, Spain
| | - Adelina Orellana
- Ace Alzheimer Center Barcelona - Universitat Internacional de Catalunya, C/ Marquès de Sentmenat, 57, 08029, Barcelona, 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 Munain
- 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 J Bullido
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
- Centro de Biología Molecular Severo Ochoa (UAM-CSIC), Madrid, Spain
- Instituto de Investigación Sanitaria 'Hospital La Paz' (IdIPaz), Madrid, Spain
- Universidad Autónoma de Madrid, Madrid, Spain
| | - Victoria Álvarez
- Laboratorio de Genética, Hospital Universitario Central de Asturias, Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
| | - Luis Miguel Real
- Instituto de Biomedicina de Sevilla/Hospital Universitario Virgen de Valme/CSIC/Dpto de Bioquímica Médica, Biología Molecular e Inmunología/Universidad de Sevilla, Sevilla, Spain
- CIBERINFEC, Instituto de Salud Carlos III, Madrid, Spain
| | - Arturo Corbatón Anchuelo
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Hospital Clínico San Carlos, Madrid, Spain
| | - Dulcenombre Gómez-Garre
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Hospital Clínico San Carlos, Madrid, Spain
- Laboratorio de Riesgo Cardiovascular y Microbiota, Hospital Clínico San Carlos; Departamento de Fisiología, Facultad de Medicina, Universidad Complutense de Madrid (UCM), Madrid, Spain
- 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, Madrid, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain
| | - Emilio Franco-Macías
- Dementia Unit, Department of Neurology, Hospital Universitario Virgen del Rocío, Instituto de Biomedicina de Sevilla (IBiS), Seville, 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, Madrid, Spain
| | - Raquel Sánchez-Valle
- Alzheimer'S Disease and Other Cognitive Disorders Unit. Service of Neurology. Hospital Clínic of Barcelona. Institut d'Investigacions Biomèdiques August Pi I Sunyer, University of Barcelona, Barcelona, Spain
| | - 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, C/ Marquès de Sentmenat, 57, 08029, Barcelona, 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, C/ Marquès de Sentmenat, 57, 08029, Barcelona, 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, C/ Marquès de Sentmenat, 57, 08029, Barcelona, 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, C/ Marquès de Sentmenat, 57, 08029, Barcelona, 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, C/ Marquès de Sentmenat, 57, 08029, Barcelona, 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
- Reina Sofia Alzheimer Centre, CIEN Foundation, ISCIII, Madrid, Spain
| | - Jose Enrique Cavazos
- South Texas Alzheimer's Disease Research Center, San Antonio, TX, USA
- Glenn Biggs Institute for Alzheimer's & Neurodegenerative Diseases, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Dr, San Antonio, TX, 78229, USA
| | - Alfredo Cabrera-Socorro
- Neuroscience Therapeutic Area, Janssen Research & Development, Turnhoutseweg 30, 2340, Beerse, Belgium
| | - Amanda Cano
- Ace Alzheimer Center Barcelona - Universitat Internacional de Catalunya, C/ Marquès de Sentmenat, 57, 08029, Barcelona, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
| | - Agustín Ruiz
- Ace Alzheimer Center Barcelona - Universitat Internacional de Catalunya, C/ Marquès de Sentmenat, 57, 08029, Barcelona, 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.
- Department of Microbiology, Immunology and Molecular Genetics. Long School of Medicine, University of Texas Health Science Center, San Antonio, TX, USA.
| |
Collapse
|
3
|
Cui Y, Arnold FJ, Li JS, Wu J, Wang D, Philippe J, Colwin MR, Michels S, Chen C, Sallam T, Thompson LM, La Spada AR, Li W. Multi-omic quantitative trait loci link tandem repeat size variation to gene regulation in human brain. Nat Genet 2025; 57:369-378. [PMID: 39809899 DOI: 10.1038/s41588-024-02057-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 12/10/2024] [Indexed: 01/16/2025]
Abstract
Tandem repeat (TR) size variation is implicated in ~50 neurological disorders, yet its impact on gene regulation in the human brain remains largely unknown. In the present study, we quantified the impact of TR size variation on brain gene regulation across distinct molecular phenotypes, based on 4,412 multi-omics samples from 1,597 donors, including 1,586 newly sequenced ones. We identified ~2.2 million TR molecular quantitative trait loci (TR-xQTLs), linking ~139,000 unique TRs to nearby molecular phenotypes, including many known disease-risk TRs, such as the G2C4 expansion in C9orf72 associated with amyotrophic lateral sclerosis. Fine-mapping revealed ~18,700 TRs as potential causal variants. Our in vitro experiments further confirmed the causal and independent regulatory effects of three TRs. Additional colocalization analysis indicated the potential causal role of TR variation in brain-related phenotypes, highlighted by a 3'-UTR TR in NUDT14 linked to cortical surface area and a TG repeat in PLEKHA1, associated with Alzheimer's disease.
Collapse
Affiliation(s)
- Ya Cui
- Division of Computational Biomedicine, Department of Biological Chemistry, University of California, Irvine, Irvine, CA, USA.
| | - Frederick J Arnold
- Departments of Pathology & Laboratory Medicine, Neurology, Biological Chemistry, and Neurobiology & Behavior, University of California, Irvine, Irvine, CA, USA
| | - Jason Sheng Li
- Division of Computational Biomedicine, Department of Biological Chemistry, University of California, Irvine, Irvine, CA, USA
| | - Jie Wu
- Departments of Psychiatry and Human Behavior, Neurobiology and Behavior, and Biological Chemistry, University of California, Irvine, Irvine, CA, USA
| | - Dan Wang
- Division of Cardiology, Department of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Julien Philippe
- Departments of Pathology & Laboratory Medicine, Neurology, Biological Chemistry, and Neurobiology & Behavior, University of California, Irvine, Irvine, CA, USA
| | - Michael R Colwin
- Departments of Pathology & Laboratory Medicine, Neurology, Biological Chemistry, and Neurobiology & Behavior, University of California, Irvine, Irvine, CA, USA
| | - Sebastian Michels
- Departments of Pathology & Laboratory Medicine, Neurology, Biological Chemistry, and Neurobiology & Behavior, University of California, Irvine, Irvine, CA, USA
- Department of Neurology, University of Ulm, Oberer Eselsberg, Ulm, Germany
| | - Chaorong Chen
- Division of Computational Biomedicine, Department of Biological Chemistry, University of California, Irvine, Irvine, CA, USA
| | - Tamer Sallam
- Division of Cardiology, Department of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Leslie M Thompson
- Departments of Psychiatry and Human Behavior, Neurobiology and Behavior, and Biological Chemistry, University of California, Irvine, Irvine, CA, USA.
| | - Albert R La Spada
- Departments of Pathology & Laboratory Medicine, Neurology, Biological Chemistry, and Neurobiology & Behavior, University of California, Irvine, Irvine, CA, USA.
- UCI Center for Neurotherapeutics, University of California, Irvine, Irvine, CA, USA.
| | - Wei Li
- Division of Computational Biomedicine, Department of Biological Chemistry, University of California, Irvine, Irvine, CA, USA.
| |
Collapse
|
4
|
Leventhal MJ, Zanella CA, Kang B, Peng J, Gritsch D, Liao Z, Bukhari H, Wang T, Pao PC, Danquah S, Benetatos J, Nehme R, Farhi S, Tsai LH, Dong X, Scherzer CR, Feany MB, Fraenkel E. An integrative systems-biology approach defines mechanisms of Alzheimer's disease neurodegeneration. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.17.585262. [PMID: 38559190 PMCID: PMC10980014 DOI: 10.1101/2024.03.17.585262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Despite years of intense investigation, the mechanisms underlying neuronal death in Alzheimer's disease, the most common neurodegenerative disorder, remain incompletely understood. To define relevant pathways, we integrated the results of an unbiased, genome-scale forward genetic screen for age-associated neurodegeneration in Drosophila with human and Drosophila Alzheimer's disease-associated multi-omics. We measured proteomics, phosphoproteomics, and metabolomics in Drosophila models of Alzheimer's disease and identified Alzheimer's disease human genetic variants that modify expression in disease-vulnerable neurons. We used a network optimization approach to integrate these data with previously published Alzheimer's disease multi-omic data. We computationally predicted and experimentally demonstrated how HNRNPA2B1 and MEPCE enhance tau-mediated neurotoxicity. Furthermore, we demonstrated that the screen hits CSNK2A1 and NOTCH1 regulate DNA damage in Drosophila and human iPSC-derived neural progenitor cells. Our work identifies candidate pathways that could be targeted to ameliorate neurodegeneration in Alzheimer's disease.
Collapse
Affiliation(s)
- Matthew J Leventhal
- MIT Ph.D. Program in Computational and Systems Biology, Cambridge, MA, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Camila A Zanella
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Byunguk Kang
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Spatial Technology Platform, Broad Institute of Harvard and MIT, Cambridge, MA USA
| | - Jiajie Peng
- Precision Neurology Program, Brigham and Women's Hospital and Harvard Medical school, Boston, MA, USA
- APDA Center for Advanced Parkinson's Disease Research, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - David Gritsch
- Precision Neurology Program, Brigham and Women's Hospital and Harvard Medical school, Boston, MA, USA
- APDA Center for Advanced Parkinson's Disease Research, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Zhixiang Liao
- Precision Neurology Program, Brigham and Women's Hospital and Harvard Medical school, Boston, MA, USA
- APDA Center for Advanced Parkinson's Disease Research, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Hassan Bukhari
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Tao Wang
- Precision Neurology Program, Brigham and Women's Hospital and Harvard Medical school, Boston, MA, USA
- APDA Center for Advanced Parkinson's Disease Research, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Present address: School of Computer Science, Northwestern Polytechnical University, Xi'an, China
| | - Ping-Chieh Pao
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Serwah Danquah
- Spatial Technology Platform, Broad Institute of Harvard and MIT, Cambridge, MA USA
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Joseph Benetatos
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Ralda Nehme
- Spatial Technology Platform, Broad Institute of Harvard and MIT, Cambridge, MA USA
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Samouil Farhi
- Spatial Technology Platform, Broad Institute of Harvard and MIT, Cambridge, MA USA
| | - Li-Huei Tsai
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Xianjun Dong
- Precision Neurology Program, Brigham and Women's Hospital and Harvard Medical school, Boston, MA, USA
- APDA Center for Advanced Parkinson's Disease Research, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Clemens R Scherzer
- Precision Neurology Program, Brigham and Women's Hospital and Harvard Medical school, Boston, MA, USA
- APDA Center for Advanced Parkinson's Disease Research, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Present address: Stephen and Denise Adams Center of Yale School of Medicine, CT, USA
| | - Mel B Feany
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Ernest Fraenkel
- MIT Ph.D. Program in Computational and Systems Biology, Cambridge, MA, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Lead contact
| |
Collapse
|
5
|
Puerta R, de Rojas I, García-González P, Olivé C, Sotolongo-Grau O, García-Sánchez A, García-Gutiérrez F, Montrreal L, Pablo Tartari J, Sanabria Á, Pytel V, Lage C, Quintela I, Aguilera N, Rodriguez-Rodriguez E, Alarcón-Martín E, Orellana A, Pastor P, Pérez-Tur J, Piñol-Ripoll G, de Munian AL, García-Alberca JM, Royo JL, Bullido MJ, Álvarez V, Real LM, Anchuelo AC, Gómez-Garre D, Larrad MTM, Franco-Macías E, Mir P, Medina M, Sánchez-Valle R, Dols-Icardo O, Sáez ME, Carracedo Á, Tárraga L, Alegret M, Valero S, Marquié M, Boada M, Juan PS, Cavazos JE, Cabrera A, Cano A. Connecting genomic and proteomic signatures of amyloid burden in the brain. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.09.06.24313124. [PMID: 39281766 PMCID: PMC11398581 DOI: 10.1101/2024.09.06.24313124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/18/2024]
Abstract
Background Alzheimer's disease (AD) has a high heritable component characteristic of complex diseases, yet many of the genetic risk factors remain unknown. We combined genome-wide association studies (GWAS) on amyloid endophenotypes measured in cerebrospinal fluid (CSF) and positron emission tomography (PET) as surrogates of amyloid pathology, which may be helpful to understand the underlying biology of the disease. Methods We performed a meta-analysis of GWAS of CSF Aβ42 and PET measures combining six independent cohorts (n=2,076). Due to the opposite effect direction of Aβ phenotypes in CSF and PET measures, only genetic signals in the opposite direction were considered for analysis (n=376,599). Polygenic risk scores (PRS) were calculated and evaluated for AD status and amyloid endophenotypes. We then searched the CSF proteome signature of brain amyloidosis using SOMAscan proteomic data (Ace cohort, n=1,008) and connected it with GWAS results of loci modulating amyloidosis. Finally, we compared our results with a large meta-analysis using publicly available datasets in CSF (n=13,409) and PET (n=13,116). This combined approach enabled the identification of overlapping genes and proteins associated with amyloid burden and the assessment of their biological significance using enrichment analyses. Results After filtering the meta-GWAS, we observed genome-wide significance in the rs429358-APOE locus and nine suggestive hits were annotated. We replicated the APOE loci using the large CSF-PET meta-GWAS and identified multiple AD-associated genes as well as the novel GADL1 locus. Additionally, we found a significant association between the AD PRS and amyloid levels, whereas no significant association was found between any Aβ PRS with AD risk. CSF SOMAscan analysis identified 1,387 FDR-significant proteins associated with CSF Aβ42 levels. The overlap among GWAS loci and proteins associated with amyloid burden was very poor (n=35). The enrichment analysis of overlapping hits strongly suggested several signalling pathways connecting amyloidosis with the anchored component of the plasma membrane, synapse physiology and mental disorders that were replicated in the large CSF-PET meta-analysis. Conclusions The strategy of combining CSF and PET amyloid endophenotypes GWAS with CSF proteome analyses might be effective for identifying signals associated with the AD pathological process and elucidate causative molecular mechanisms behind the amyloid mobilization in AD.
Collapse
Affiliation(s)
- Raquel Puerta
- Ace Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Spain
- Universitat de Barcelona (UB)
| | - Itziar de Rojas
- Ace Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
| | - Pablo García-González
- Ace Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
| | - Clàudia Olivé
- Ace Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Spain
| | | | | | | | - Laura Montrreal
- Ace Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Spain
| | - Juan Pablo Tartari
- Ace Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Spain
| | - Ángela Sanabria
- Ace Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
| | - Vanesa Pytel
- Ace Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
| | - Carmen Lage
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
- Neurology Service, Marqués de Valdecilla University Hospital (University of Cantabria and IDIVAL), Santander, Spain
| | - Inés Quintela
- Grupo de Medicina Xenómica, Centro Nacional de Genotipado (CEGEN-PRB3-ISCIII). Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Nuria Aguilera
- Ace Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Spain
| | - Eloy Rodriguez-Rodriguez
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
- Neurology Service, Marqués de Valdecilla University Hospital (University of Cantabria and IDIVAL), Santander, Spain
| | | | - Adelina Orellana
- Ace Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
| | - Pau Pastor
- Unit of Neurodegenerative diseases, Department of Neurology, University Hospital Germans Trias i Pujol, Badalona, Barcelona, Spain
- The Germans Trias i Pujol Research Institute (IGTP), Badalona, Barcelona, Spain
| | - Jordi Pérez-Tur
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
- Unitat de Genètica Molecular, Institut de Biomedicina de València-CSIC, Valencia, Spain
- Unidad Mixta de Neurologia Genètica, Instituto de Investigación Sanitaria La Fe, Valencia, Spain
| | - Gerard Piñol-Ripoll
- Unitat Trastorns Cognitius, Hospital Universitari Santa Maria de Lleida, Lleida, Spain
- Institut de Recerca Biomedica de Lleida (IRBLLeida), Lleida, Spain
| | - Adolfo López de Munian
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
- Department of Neurology. Hospital Universitario Donostia. San Sebastian, Spain
- Department of Neurosciences. Faculty of Medicine and Nursery. University of the Basque Country, San Sebastián, Spain
- Neurosciences Area. Instituto Biodonostia. San Sebastian, Spain
| | - Jose María García-Alberca
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
- Alzheimer Research Center & Memory Clinic, Andalusian Institute for Neuroscience, Málaga, Spain
| | - Jose Luís Royo
- Departamento de Especialidades Quirúrgicas, Bioquímica e Inmunología. School of Medicine. University of Malaga. Málaga, Spain
| | - María Jesús Bullido
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
- Centro de Biología Molecular Severo Ochoa (UAM-CSIC)
- Instituto de Investigacion Sanitaria ‘Hospital la Paz’ (IdIPaz), Madrid, Spain
- Universidad Autónoma de Madrid
| | - Victoria Álvarez
- Laboratorio de Genética. Hospital Universitario Central de Asturias, Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA)
| | - Luis Miguel Real
- Departamento de Especialidades Quirúrgicas, Bioquímica e Inmunología. School of Medicine. University of Malaga. Málaga, Spain
- Unidad Clínica de Enfermedades Infecciosas y Microbiología.Hospital Universitario de Valme, Sevilla, Spain
| | - Arturo Corbatón Anchuelo
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Hospital Clínico San Carlos
| | - Dulcenombre Gómez-Garre
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Hospital Clínico San Carlos
- Laboratorio de Riesgo Cardiovascular y Microbiota, Hospital Clínico San Carlos; Departamento de Fisiología, Facultad de Medicina, Universidad Complutense de Madrid (UCM)
- Biomedical Research Networking Center in Cardiovascular Diseases (CIBERCV), Madrid, Spain
| | - María Teresa Martínez Larrad
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Hospital Clínico San Carlos
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM)
| | - Emilio Franco-Macías
- Dementia Unit, Department of Neurology, Hospital Universitario Virgen del Rocío, Instituto de Biomedicina de Sevilla (IBiS), Sevilla, Spain
| | - Pablo Mir
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
- Unidad de Trastornos del Movimiento, Servicio de Neurología y Neurofisiología. Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
| | - Miguel Medina
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
- CIEN Foundation/Queen Sofia Foundation Alzheimer Center
| | - Raquel Sánchez-Valle
- Alzheimer’s disease and other cognitive disorders unit. Service of Neurology. Hospital Clínic of Barcelona. Institut d’Investigacions Biomèdiques August Pi i Sunyer, University of Barcelona, Barcelona, Spain
| | - Oriol Dols-Icardo
- Department of Neurology, Sant Pau Memory Unit, Sant Pau Biomedical Research Institute, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
| | | | - Ángel Carracedo
- Grupo de Medicina Xenómica, Centro Nacional de Genotipado (CEGEN-PRB3-ISCIII). Universidade de Santiago de Compostela, Santiago de Compostela, Spain
- Fundación Pública Galega de Medicina Xenómica – CIBERER-IDIS, Santiago de Compostela, Spain
| | - Lluís Tárraga
- Ace Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
| | - Montse Alegret
- Ace Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
| | - Sergi Valero
- Ace Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
| | - Marta Marquié
- Ace Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
| | - Mercè Boada
- Ace Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
| | - Pascual Sánchez Juan
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
- Neurology Service, Marqués de Valdecilla University Hospital (University of Cantabria and IDIVAL), Santander, Spain
| | - Jose Enrique Cavazos
- South Texas Medical Science Training Program, University of Texas Health San Antonio, San Antonio
- Glenn Biggs Institute for Alzheimer’s & Neurodegenerative Diseases, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Dr, San Antonio, TX 78229 USA
| | - Alfredo Cabrera
- Neuroscience Therapeutic Area, Janssen Research & Development, Turnhoutseweg 30, 2340 Beerse, Belgium
| | - Amanda Cano
- Ace Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
| | - Alzheimer’s Disease Neuroimaging Initiative.
- Ace Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
- Glenn Biggs Institute for Alzheimer’s & Neurodegenerative Diseases, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Dr, San Antonio, TX 78229 USA
| |
Collapse
|
6
|
Poirier A, Picard C, Labonté A, Aubry I, Auld D, Zetterberg H, Blennow K, Tremblay ML, Poirier J. PTPRS is a novel marker for early Tau pathology and synaptic integrity in Alzheimer's disease. Sci Rep 2024; 14:14718. [PMID: 38926456 PMCID: PMC11208446 DOI: 10.1038/s41598-024-65104-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 06/17/2024] [Indexed: 06/28/2024] Open
Abstract
We examined the role of protein tyrosine phosphatase receptor sigma (PTPRS) in the context of Alzheimer's disease and synaptic integrity. Publicly available datasets (BRAINEAC, ROSMAP, ADC1) and a cohort of asymptomatic but "at risk" individuals (PREVENT-AD) were used to explore the relationship between PTPRS and various Alzheimer's disease biomarkers. We identified that PTPRS rs10415488 variant C shows features of neuroprotection against early Tau pathology and synaptic degeneration in Alzheimer's disease. This single nucleotide polymorphism correlated with higher PTPRS transcript abundance and lower p(181)Tau and GAP-43 levels in the CSF. In the brain, PTPRS protein abundance was significantly correlated with the quantity of two markers of synaptic integrity: SNAP25 and SYT-1. We also found the presence of sexual dimorphism for PTPRS, with higher CSF concentrations in males than females. Male carriers for variant C were found to have a 10-month delay in the onset of AD. We thus conclude that PTPRS acts as a neuroprotective receptor in Alzheimer's disease. Its protective effect is most important in males, in whom it postpones the age of onset of the disease.
Collapse
Affiliation(s)
- Alexandre Poirier
- Division of Experimental Medicine, Faculty of Medicine and Health Science, McGill University, Montréal, QC, Canada
- Goodman Cancer Institute, McGill University, Montréal, Canada
| | - Cynthia Picard
- Douglas Mental Health University Institute, Montréal, QC, Canada
- Centre for the Studies in the Prevention of Alzheimer's Disease, Montréal, QC, Canada
| | - Anne Labonté
- Douglas Mental Health University Institute, Montréal, QC, Canada
- Centre for the Studies in the Prevention of Alzheimer's Disease, Montréal, QC, Canada
| | - Isabelle Aubry
- Goodman Cancer Institute, McGill University, Montréal, Canada
- McGill University, Montréal, QC, Canada
| | - Daniel Auld
- McGill University, Montréal, QC, Canada
- Victor Phillip Dahdaleh Institute of Genomic Medicine, McGill University, Montréal, QC, Canada
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK
- UK Dementia Research Institute at UCL, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, SAR, People's Republic of China
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
- University of Science and Technology of China, Hefei, Anhui, People's Republic of China
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK
- University of Science and Technology of China, Hefei, Anhui, People's Republic of China
- Institut du Cerveau et de la Moelle épinière (ICM), Pitié-Salpêtrière Hospital, Sorbonne Université, Paris, France
| | - Michel L Tremblay
- Division of Experimental Medicine, Faculty of Medicine and Health Science, McGill University, Montréal, QC, Canada.
- Goodman Cancer Institute, McGill University, Montréal, Canada.
- McGill University, Montréal, QC, Canada.
- Department of Biochemistry, McGill University, Montréal, Canada.
| | - Judes Poirier
- Douglas Mental Health University Institute, Montréal, QC, Canada.
- Centre for the Studies in the Prevention of Alzheimer's Disease, Montréal, QC, Canada.
- McGill University, Montréal, QC, Canada.
| |
Collapse
|
7
|
Poirier A, Picard C, Labonté A, Aubry I, Auld D, Zetterberg H, Blennow K, Tremblay ML, Poirier J. PTPRS is a novel marker for early tau pathology and synaptic integrity in Alzheimer's disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.12.593733. [PMID: 38766183 PMCID: PMC11100782 DOI: 10.1101/2024.05.12.593733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
We examined the role of protein tyrosine phosphatase receptor sigma (PTPRS) in the context of Alzheimer's disease and synaptic integrity. Publicly available datasets (BRAINEAC, ROSMAP, ADC1) and a cohort of asymptomatic but "at risk" individuals (PREVENT-AD) were used to explore the relationship between PTPRS and various Alzheimer's disease biomarkers. We identified that PTPRS rs10415488 variant C shows features of neuroprotection against early tau pathology and synaptic degeneration in Alzheimer's disease. This single nucleotide polymorphism correlated with higher PTPRS transcript abundance and lower P-tau181 and GAP-43 levels in the CSF. In the brain, PTPRS protein abundance was significantly correlated with the quantity of two markers of synaptic integrity: SNAP25 and SYT-1. We also found the presence of sexual dimorphism for PTPRS, with higher CSF concentrations in males than females. Male carriers for variant C were found to have a 10-month delay in the onset of AD. We thus conclude that PTPRS acts as a neuroprotective receptor in Alzheimer's disease. Its protective effect is most important in males, in whom it postpones the age of onset of the disease.
Collapse
|
8
|
Eulalio T, Sun MW, Gevaert O, Greicius MD, Montine TJ, Nachun D, Montgomery SB. regionalpcs: improved discovery of DNA methylation associations with complex traits. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.01.590171. [PMID: 38746367 PMCID: PMC11092597 DOI: 10.1101/2024.05.01.590171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
We have developed the regional principal components (rPCs) method, a novel approach for summarizing gene-level methylation. rPCs address the challenge of deciphering complex epigenetic mechanisms in diseases like Alzheimer's disease (AD). In contrast to traditional averaging, rPCs leverage principal components analysis to capture complex methylation patterns across gene regions. Our method demonstrated a 54% improvement in sensitivity over averaging in simulations, offering a robust framework for identifying subtle epigenetic variations. Applying rPCs to the AD brain methylation data in ROSMAP, combined with cell type deconvolution, we uncovered 838 differentially methylated genes associated with neuritic plaque burden-significantly outperforming conventional methods. Integrating methylation quantitative trait loci (meQTL) with genome-wide association studies (GWAS) identified 17 genes with potential causal roles in AD, including MS4A4A and PICALM. Our approach is available in the Bioconductor package regionalpcs, opening avenues for research and facilitating a deeper understanding of the epigenetic landscape in complex diseases.
Collapse
Affiliation(s)
- Tiffany Eulalio
- Department of Biomedical Data Science, Stanford University, Stanford, CA, 94305, USA
| | - Min Woo Sun
- Department of Biomedical Data Science, Stanford University, Stanford, CA, 94305, USA
| | - Olivier Gevaert
- Department of Biomedical Data Science, Stanford University, Stanford, CA, 94305, USA
| | - Michael D Greicius
- Department of Neurology & Neurological Sciences, Stanford University, Stanford, CA, 94305, USA
| | - Thomas J Montine
- Department of Pathology, Stanford University, Stanford, CA, 94305, USA
| | - Daniel Nachun
- Department of Pathology, Stanford University, Stanford, CA, 94305, USA
| | | |
Collapse
|
9
|
Dulski J, Uitti RJ, Beasley A, Hernandez D, Ramanan VK, Cahn EJ, Ren Y, Johnson PW, Quicksall ZS, Wszolek ZK, Ross OA, Heckman MG. Genetics of Parkinson's disease heterogeneity: A genome-wide association study of clinical subtypes. Parkinsonism Relat Disord 2024; 119:105935. [PMID: 38072719 PMCID: PMC10872335 DOI: 10.1016/j.parkreldis.2023.105935] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 11/06/2023] [Accepted: 11/18/2023] [Indexed: 12/25/2023]
Abstract
INTRODUCTION Substantial heterogeneity between individual patients in the clinical presentation of Parkinson's disease (PD) has led to the classification of distinct PD subtypes. However, genetic susceptibility factors for specific PD subtypes are not well understood. Therefore, the present study aimed to investigate the genetics of PD heterogeneity by performing a genome-wide association study (GWAS) of PD subtypes. METHODS A total of 799 PD patients were included and classified into tremor-dominant (TD) (N = 345), akinetic-rigid (AR) (N = 227), gait-difficulty (GD) (N = 82), and mixed (MX) (N = 145) phenotypic subtypes. After array genotyping and subsequent imputation, a total of 7,918,344 variants were assessed for association with each PD subtype using logistic regression models that were adjusted for age, sex, and the top five principal components of GWAS data. RESULTS We identified one genome-wide significant association (P < 5 × 10-8), which was between the MIR3976HG rs7504760 variant and the AR subtype (Odds ratio [OR] = 6.12, P = 2.57 × 10-8). Suggestive associations (P < 1 × 10-6) were observed regarding TD for RP11-497G19.3/RP11-497G19.1 rs7304254 (OR = 3.33, P = 3.89 × 10-7), regarding GD for HES2 rs111473931 (OR = 3.18, P = 6.85 × 10-7), RP11-400D2.3/CTD-2012I17.1 rs149082205 (OR = 8.96, P = 9.08 × 10-7), and RN7SL408P/SGK1 rs56161738 (OR = 2.97, P = 6.19 × 10-7), and regarding MX for MMRN2 rs112991171 (OR = 4.98, P = 1.02 × 10-7). CONCLUSION Our findings indicate that genetic variation may account for part of the clinical heterogeneity of PD. In particular, we found a novel genome-wide significant association between MIR3976HG variation and the AR PD subtype. Replication of these findings will be important in order to better define the genetic architecture of clinical variability in PD disease presentation.
Collapse
Affiliation(s)
- Jarosław Dulski
- Department of Neurology, Mayo Clinic, Jacksonville, FL, USA; Division of Neurological and Psychiatric Nursing, Faculty of Health Sciences, Medical University of Gdansk, Gdansk, Poland; Neurology Department, St Adalbert Hospital, Copernicus PL Ltd., Gdansk, Poland; Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA.
| | - Ryan J Uitti
- Department of Neurology, Mayo Clinic, Jacksonville, FL, USA
| | | | - Dena Hernandez
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | | | - Elliot J Cahn
- Division of Clinical Trials and Biostatistics, Mayo Clinic, Rochester, MN, USA
| | - Yingxue Ren
- Division of Clinical Trials and Biostatistics, Mayo Clinic, Jacksonville, FL, USA
| | - Patrick W Johnson
- Division of Clinical Trials and Biostatistics, Mayo Clinic, Jacksonville, FL, USA
| | - Zachary S Quicksall
- Division of Clinical Trials and Biostatistics, Mayo Clinic, Jacksonville, FL, USA
| | | | - Owen A Ross
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA; Department of Clinical Genomics, Mayo Clinic, Jacksonville, FL, USA
| | - Michael G Heckman
- Division of Clinical Trials and Biostatistics, Mayo Clinic, Jacksonville, FL, USA.
| |
Collapse
|
10
|
Xu F, Chen A, Pan S, Wu Y, He H, Han Z, Lu L, Orgil B, Chi X, Yang C, Jia S, Yu C, Mi J. Systems genetics analysis reveals the common genetic basis for pain sensitivity and cognitive function. CNS Neurosci Ther 2024; 30:e14557. [PMID: 38421132 PMCID: PMC10850811 DOI: 10.1111/cns.14557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 10/31/2023] [Accepted: 11/25/2023] [Indexed: 03/02/2024] Open
Abstract
BACKGROUND There is growing evidence of a strong correlation between pain sensitivity and cognitive function under both physiological and pathological conditions. However, the detailed mechanisms remain largely unknown. In the current study, we sought to explore candidate genes and common molecular mechanisms underlying pain sensitivity and cognitive function with a transcriptome-wide association study using recombinant inbred mice from the BXD family. METHODS The pain sensitivity determined by Hargreaves' paw withdrawal test and cognition-related phenotypes were systematically analyzed in 60 strains of BXD mice and correlated with hippocampus transcriptomes, followed by quantitative trait locus (QTL) mapping and systems genetics analysis. RESULTS The pain sensitivity showed significant variability across the BXD strains and co-varies with cognitive traits. Pain sensitivity correlated hippocampual genes showed a significant involvement in cognition-related pathways, including glutamatergic synapse, and PI3K-Akt signaling pathway. Moreover, QTL mapping identified a genomic region on chromosome 4, potentially regulating the variation of pain sensitivity. Integrative analysis of expression QTL mapping, correlation analysis, and Bayesian network modeling identified Ring finger protein 20 (Rnf20) as the best candidate. Further pathway analysis indicated that Rnf20 may regulate the expression of pain sensitivity and cognitive function through the PI3K-Akt signaling pathway, particularly through interactions with genes Ppp2r2b, Ppp2r5c, Col9a3, Met, Rps6, Tnc, and Kras. CONCLUSIONS Our study demonstrated that pain sensitivity is associated with genetic background and Rnf20-mediated PI3K-Akt signaling may involve in the regulation of pain sensitivity and cognitive functions.
Collapse
Affiliation(s)
- Fuyi Xu
- Shandong Technology Innovation Center of Molecular Targeting and Intelligent Diagnosis and TreatmentBinzhou Medical UniversityYantaiChina
| | - Anran Chen
- The Affiliated Yantai Yuhuangding Hospital of Qingdao UniversityYantaiChina
| | - Shuijing Pan
- Shandong Technology Innovation Center of Molecular Targeting and Intelligent Diagnosis and TreatmentBinzhou Medical UniversityYantaiChina
| | - Yingying Wu
- Shandong Technology Innovation Center of Molecular Targeting and Intelligent Diagnosis and TreatmentBinzhou Medical UniversityYantaiChina
| | - Hongjie He
- Shandong Technology Innovation Center of Molecular Targeting and Intelligent Diagnosis and TreatmentBinzhou Medical UniversityYantaiChina
| | - Zhe Han
- Shandong Technology Innovation Center of Molecular Targeting and Intelligent Diagnosis and TreatmentBinzhou Medical UniversityYantaiChina
| | - Lu Lu
- University of Tennessee Health Science CenterMemphisTennesseeUSA
| | | | - XiaoDong Chi
- Shandong Technology Innovation Center of Molecular Targeting and Intelligent Diagnosis and TreatmentBinzhou Medical UniversityYantaiChina
| | - Cunhua Yang
- Shandong Technology Innovation Center of Molecular Targeting and Intelligent Diagnosis and TreatmentBinzhou Medical UniversityYantaiChina
| | - Shushan Jia
- Department of AnesthesiologyYanTai Affiliated Hospital of BinZhou Medical UniversityYantaiChina
| | - Cuicui Yu
- The Affiliated Yantai Yuhuangding Hospital of Qingdao UniversityYantaiChina
| | - Jia Mi
- Shandong Technology Innovation Center of Molecular Targeting and Intelligent Diagnosis and TreatmentBinzhou Medical UniversityYantaiChina
| |
Collapse
|
11
|
Yakovchik AY, Tolynyova DV, Kashtanova DA, Sutulova ER, Ivanov MV, Mamchur AA, Erema VV, Matkava LR, Terekhov MV, Rumyantseva AM, Blinova OI, Akinshina AI, Mitrofanov SI, Yudin VS, Makarov VV, Keskinov AА, Kraevoy SA, Yudin SM. Genetics of psycho-emotional well-being: genome-wide association study and polygenic risk score analysis. Front Psychiatry 2024; 14:1188427. [PMID: 38328521 PMCID: PMC10847277 DOI: 10.3389/fpsyt.2023.1188427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 07/28/2023] [Indexed: 02/09/2024] Open
Abstract
Background Psycho-emotional well-being is essential for living a life of satisfaction and fulfillment. However, depression and anxiety have become the leading mental health issues worldwide, according to the World Health Organization. Both disorders have been linked to stress and other psychological factors. Their genetic basis remains understudied. Methods In 2020-2021, the psycho-emotional well-being of 30,063 Russians with no known psychiatric history was assessed using the Hospital Anxiety and Depression Scale (HADS) for general mental health and the HADS subscale A (anxiety) for anxiety. Following the original instructions, an anxiety score of ≥11 points was used as the anxiety threshold. A genome-wide association study was performed to find associations between anxiety and HADS/HADS-A scores using linear and logistic regressions based on HADS/HADS-A scores as binary and continuous variables, respectively. In addition, the links between anxiety, sociodemographic factors (such as age, sex, and employment), lifestyle (such as physical activity, sleep duration, and smoking), and markers of caffeine and alcohol metabolism were analyzed. To assess the risk of anxiety, polygenic risk score modeling was carried out using open-access software and principal component analysis (PCA) to simplify the calculations (ROC AUC = 89.4 ± 2.2% on the test set). Results There was a strong positive association between HADS/HADS-A scores and sociodemographic factors and lifestyle. New single-nucleotide polymorphisms (SNPs) with genome-wide significance were discovered, which had not been associated with anxiety or other stress-related conditions but were located in genes previously associated with bipolar disorder, schizophrenia, or emotional instability. The CACNA1C variant rs1205787230 was associated with clinical anxiety (a HADS-A score of ≥11 points). There was an association between anxiety levels (HADS-A scores) and genes involved in the activity of excitatory neurotransmitters: PTPRN2 (rs3857647), DLGAP4 (rs8114927), and STK24 (rs9517326). Conclusion Our results suggest that calcium channels and monoamine neurotransmitters, as well as SNPs in genes directly or indirectly affecting neurogenesis and synaptic functions, may be involved in the development of increased anxiety. The role of some non-genetic factors and the clinical significance of physiological markers such as lifestyle were also demonstrated.
Collapse
Affiliation(s)
- Anna Yurievna Yakovchik
- Federal State Budgetary Institution Centre for Strategic Planning and Management of Biomedical Health Risks of the Federal Medical Biological Agency, Moscow, Russia
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Hao Y, Li C, Wang H, Ming C. Effects of copy number variations on longevity in late-onset Alzheimer's disease patients: insights from a causality network analysis. Front Aging Neurosci 2023; 15:1241412. [PMID: 38020759 PMCID: PMC10652415 DOI: 10.3389/fnagi.2023.1241412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 10/12/2023] [Indexed: 12/01/2023] Open
Abstract
Alzheimer's disease (AD), particularly late-onset Alzheimer's disease (LOAD), is a prevalent form of dementia that significantly affects patients' cognitive and behavioral capacities and longevity. Although approximately 70 genetic risk factors linked with AD have been identified, their influence on patient longevity remains unclear. Further, recent studies have associated copy number variations (CNVs) with the longevity of healthy individuals and immune-related pathways in AD patients. This study aims to investigate the role of CNVs on the longevity of AD patients by integrating the Whole Genome Sequencing (WGS) and transcriptomics data from the Religious Orders Study/Memory and Aging Project (ROSMAP) cohort through causality network inference. Our comprehensive analysis led to the construction of a CNV-Gene-Age of Death (AOD) causality network. We successfully identified three key CNVs (DEL5006, mCNV14192, and DUP42180) and seven AD-longevity causal genes (PLGRKT, TLR1, PLAU, CALB2, SYTL2, OTOF, and NT5DC1) impacting AD patient longevity, independent of disease severity. This outcome emphasizes the potential role of plasminogen activation and chemotaxis in longevity. We propose several hypotheses regarding the role of identified CNVs and the plasminogen system on patient longevity. However, experimental validation is required to further corroborate these findings and uncover precise mechanisms. Despite these limitations, our study offers promising insights into the genetic influence on AD patient longevity and contributes to paving the way for potential therapeutic interventions.
Collapse
Affiliation(s)
- Yanan Hao
- Department of Public Health and Medicinal Administration, Faculty of Health Sciences, University of Macau, Macau, Macao SAR, China
- Department of Biomedical Sciences, Faculty of Health Sciences, University of Macau, Macau, Macao SAR, China
| | - Chuhao Li
- Department of Public Health and Medicinal Administration, Faculty of Health Sciences, University of Macau, Macau, Macao SAR, China
| | - He Wang
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China
| | - Chen Ming
- Department of Public Health and Medicinal Administration, Faculty of Health Sciences, University of Macau, Macau, Macao SAR, China
| |
Collapse
|
13
|
Kang M, Ang TFA, Devine SA, Sherva R, Mukherjee S, Trittschuh EH, Gibbons LE, Scollard P, Lee M, Choi SE, Klinedinst B, Nakano C, Dumitrescu LC, Durant A, Hohman TJ, Cuccaro ML, Saykin AJ, Kukull WA, Bennett DA, Wang LS, Mayeux RP, Haines JL, Pericak-Vance MA, Schellenberg GD, Crane PK, Au R, Lunetta KL, Mez JB, Farrer LA. A genome-wide search for pleiotropy in more than 100,000 harmonized longitudinal cognitive domain scores. Mol Neurodegener 2023; 18:40. [PMID: 37349795 PMCID: PMC10286470 DOI: 10.1186/s13024-023-00633-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Accepted: 06/06/2023] [Indexed: 06/24/2023] Open
Abstract
BACKGROUND More than 75 common variant loci account for only a portion of the heritability for Alzheimer's disease (AD). A more complete understanding of the genetic basis of AD can be deduced by exploring associations with AD-related endophenotypes. METHODS We conducted genome-wide scans for cognitive domain performance using harmonized and co-calibrated scores derived by confirmatory factor analyses for executive function, language, and memory. We analyzed 103,796 longitudinal observations from 23,066 members of community-based (FHS, ACT, and ROSMAP) and clinic-based (ADRCs and ADNI) cohorts using generalized linear mixed models including terms for SNP, age, SNP × age interaction, sex, education, and five ancestry principal components. Significance was determined based on a joint test of the SNP's main effect and interaction with age. Results across datasets were combined using inverse-variance meta-analysis. Genome-wide tests of pleiotropy for each domain pair as the outcome were performed using PLACO software. RESULTS Individual domain and pleiotropy analyses revealed genome-wide significant (GWS) associations with five established loci for AD and AD-related disorders (BIN1, CR1, GRN, MS4A6A, and APOE) and eight novel loci. ULK2 was associated with executive function in the community-based cohorts (rs157405, P = 2.19 × 10-9). GWS associations for language were identified with CDK14 in the clinic-based cohorts (rs705353, P = 1.73 × 10-8) and LINC02712 in the total sample (rs145012974, P = 3.66 × 10-8). GRN (rs5848, P = 4.21 × 10-8) and PURG (rs117523305, P = 1.73 × 10-8) were associated with memory in the total and community-based cohorts, respectively. GWS pleiotropy was observed for language and memory with LOC107984373 (rs73005629, P = 3.12 × 10-8) in the clinic-based cohorts, and with NCALD (rs56162098, P = 1.23 × 10-9) and PTPRD (rs145989094, P = 8.34 × 10-9) in the community-based cohorts. GWS pleiotropy was also found for executive function and memory with OSGIN1 (rs12447050, P = 4.09 × 10-8) and PTPRD (rs145989094, P = 3.85 × 10-8) in the community-based cohorts. Functional studies have previously linked AD to ULK2, NCALD, and PTPRD. CONCLUSION Our results provide some insight into biological pathways underlying processes leading to domain-specific cognitive impairment and AD, as well as a conduit toward a syndrome-specific precision medicine approach to AD. Increasing the number of participants with harmonized cognitive domain scores will enhance the discovery of additional genetic factors of cognitive decline leading to AD and related dementias.
Collapse
Affiliation(s)
- Moonil Kang
- Department of Medicine (Biomedical Genetics), Boston University Chobanian & Avedisian School of Medicine, 72 East Concord Street E200, Boston, MA 02118 USA
| | - Ting Fang Alvin Ang
- Department of Anatomy and Neurobiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA USA
- Framingham Heart Study, Boston University Chobanian & Avedisian School of Medicine, Boston, MA USA
- Slone Epidemiology Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA USA
| | - Sherral A. Devine
- Department of Anatomy and Neurobiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA USA
- Framingham Heart Study, Boston University Chobanian & Avedisian School of Medicine, Boston, MA USA
| | - Richard Sherva
- Department of Medicine (Biomedical Genetics), Boston University Chobanian & Avedisian School of Medicine, 72 East Concord Street E200, Boston, MA 02118 USA
| | - Shubhabrata Mukherjee
- Department of Medicine, University of Washington School of Medicine, Seattle, WA USA
| | - Emily H. Trittschuh
- Geriatric Research, Education, and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA USA
- Department of Psychiatry and Behavioral Sciences, University of Washington School of Medicine, Seattle, WA USA
| | - Laura E. Gibbons
- Department of Medicine, University of Washington School of Medicine, Seattle, WA USA
| | - Phoebe Scollard
- Department of Medicine, University of Washington School of Medicine, Seattle, WA USA
| | - Michael Lee
- Department of Medicine, University of Washington School of Medicine, Seattle, WA USA
| | - Seo-Eun Choi
- Department of Medicine, University of Washington School of Medicine, Seattle, WA USA
| | - Brandon Klinedinst
- Department of Medicine, University of Washington School of Medicine, Seattle, WA USA
| | - Connie Nakano
- Department of Medicine, University of Washington School of Medicine, Seattle, WA USA
| | - Logan C. Dumitrescu
- Vanderbilt Memory & Alzheimer’s Center, Vanderbilt University Medical Center, Nashville, TN USA
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN USA
| | - Alaina Durant
- Vanderbilt Memory & Alzheimer’s Center, Vanderbilt University Medical Center, Nashville, TN USA
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN USA
| | - Timothy J. Hohman
- Vanderbilt Memory & Alzheimer’s Center, Vanderbilt University Medical Center, Nashville, TN USA
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN USA
| | - Michael L. Cuccaro
- John P. Hussman Institute for Human Genomics, Miller School of Medicine, Miami, FL USA
| | - Andrew J. Saykin
- Indiana Alzheimer’s Disease Research Center, Indiana University School of Medicine, Indianapolis, IN USA
- Department of Radiology and Imaging Services, Indiana University School of Medicine, Indianapolis, IN USA
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN USA
| | - Walter A. Kukull
- Department of Epidemiology, University of Washington, Seattle, WA USA
| | - David A. Bennett
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL USA
| | - Li-San Wang
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA USA
| | - Richard P. Mayeux
- Department of Neurology, Columbia University School of Medicine, New York, NY USA
| | - Jonathan L. Haines
- Cleveland Institute for Computational Biology, Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, OH USA
| | | | - Gerard D. Schellenberg
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA USA
| | - Paul K. Crane
- Department of Medicine, University of Washington School of Medicine, Seattle, WA USA
| | - Rhoda Au
- Department of Anatomy and Neurobiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA USA
- Framingham Heart Study, Boston University Chobanian & Avedisian School of Medicine, Boston, MA USA
- Slone Epidemiology Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA USA
- Boston University Alzheimer’s Disease Research Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA USA
- Department of Epidemiology, Boston University School of Public Health, Boston, MA USA
| | - Kathryn L. Lunetta
- Framingham Heart Study, Boston University Chobanian & Avedisian School of Medicine, Boston, MA USA
- Department of Biostatistics, Boston University School of Public Health, Boston, MA USA
| | - Jesse B. Mez
- Framingham Heart Study, Boston University Chobanian & Avedisian School of Medicine, Boston, MA USA
- Boston University Alzheimer’s Disease Research Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA USA
- Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA USA
| | - Lindsay A. Farrer
- Department of Medicine (Biomedical Genetics), Boston University Chobanian & Avedisian School of Medicine, 72 East Concord Street E200, Boston, MA 02118 USA
- Framingham Heart Study, Boston University Chobanian & Avedisian School of Medicine, Boston, MA USA
- Boston University Alzheimer’s Disease Research Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA USA
- Department of Epidemiology, Boston University School of Public Health, Boston, MA USA
- Department of Biostatistics, Boston University School of Public Health, Boston, MA USA
- Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA USA
- Department of Ophthalmology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA USA
| |
Collapse
|
14
|
Ho EV, Welch A, Thompson SL, Knowles JA, Dulawa SC. Mice lacking Ptprd exhibit deficits in goal-directed behavior and female-specific impairments in sensorimotor gating. PLoS One 2023; 18:e0277446. [PMID: 37205689 PMCID: PMC10198499 DOI: 10.1371/journal.pone.0277446] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 02/16/2023] [Indexed: 05/21/2023] Open
Abstract
Protein Tyrosine Phosphatase receptor type D (PTPRD) is a member of the protein tyrosine phosphatase family that mediates cell adhesion and synaptic specification. Genetic studies have linked Ptprd to several neuropsychiatric phenotypes, including Restless Leg Syndrome (RLS), opioid abuse disorder, and antipsychotic-induced weight gain. Genome-wide association studies (GWAS) of either pediatric obsessive-compulsive traits, or Obsessive-Compulsive Disorder (OCD), have identified loci near PTPRD as genome-wide significant, or strongly suggestive for this trait. We assessed Ptprd wild-type (WT), heterozygous (HT), and knockout (KO) mice for behavioral dimensions that are altered in OCD, including anxiety and exploration (open field test, dig test), perseverative behavior (splash-induced grooming, spatial d), sensorimotor gating (prepulse inhibition), and home cage goal-directed behavior (nest building). No effect of genotype was observed in any measure of the open field test, dig test, or splash test. However, Ptprd KO mice of both sexes showed impairments in nest building behavior. Finally, female, but not male, Ptprd KO mice showed deficits in prepulse inhibition, an operational measure of sensorimotor gating that is reduced in female, but not male, OCD patients. Our results indicate that constitutive lack of Ptprd may contribute to the development of certain domains that are altered OCD, including goal-directed behavior, and reduced sensorimotor gating specifically in females.
Collapse
Affiliation(s)
- Emily V. Ho
- Neurosciences Graduate Program, University of California San Diego1, La Jolla, CA, United States of America
| | - Amanda Welch
- Department of Psychiatry, University of California San Diego, La Jolla, CA, United States of America
| | - Summer L. Thompson
- Department of Psychiatry, University of California San Diego, La Jolla, CA, United States of America
| | - James A. Knowles
- Department of Cell Biology, SUNY Downstate Medical Center College of Medicine, Brooklyn, NY, United States of America
| | - Stephanie C. Dulawa
- Neurosciences Graduate Program, University of California San Diego1, La Jolla, CA, United States of America
- Department of Psychiatry, University of California San Diego, La Jolla, CA, United States of America
| |
Collapse
|
15
|
Cui Y, Arnold FJ, Peng F, Wang D, Li JS, Michels S, Wagner EJ, La Spada AR, Li W. Alternative polyadenylation transcriptome-wide association study identifies APA-linked susceptibility genes in brain disorders. Nat Commun 2023; 14:583. [PMID: 36737438 PMCID: PMC9898543 DOI: 10.1038/s41467-023-36311-8] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 01/25/2023] [Indexed: 02/05/2023] Open
Abstract
Alternative polyadenylation (APA) plays an essential role in brain development; however, current transcriptome-wide association studies (TWAS) largely overlook APA in nominating susceptibility genes. Here, we performed a 3' untranslated region (3'UTR) APA TWAS (3'aTWAS) for 11 brain disorders by combining their genome-wide association studies data with 17,300 RNA-seq samples across 2,937 individuals. We identified 354 3'aTWAS-significant genes, including known APA-linked risk genes, such as SNCA in Parkinson's disease. Among these 354 genes, ~57% are not significant in traditional expression- and splicing-TWAS studies, since APA may regulate the translation, localization and protein-protein interaction of the target genes independent of mRNA level expression or splicing. Furthermore, we discovered ATXN3 as a 3'aTWAS-significant gene for amyotrophic lateral sclerosis, and its modulation substantially impacted pathological hallmarks of amyotrophic lateral sclerosis in vitro. Together, 3'aTWAS is a powerful strategy to nominate important APA-linked brain disorder susceptibility genes, most of which are largely overlooked by conventional expression and splicing analyses.
Collapse
Affiliation(s)
- Ya Cui
- Division of Computational Biomedicine, Department of Biological Chemistry, School of Medicine, University of California, Irvine, Irvine, CA, 92697, USA
| | - Frederick J Arnold
- Departments of Pathology & Laboratory Medicine, Neurology, and Biological Chemistry, School of Medicine, and the UCI Institute for Neurotherapeutics, University of California Irvine, Irvine, CA, 92697, USA
| | - Fanglue Peng
- Department of Molecular and Cellular Biology, University Baylor College of Medicine, Houston, TX, 77030, USA
| | - Dan Wang
- Department of Medicine, Division of Cardiology, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Jason Sheng Li
- Division of Computational Biomedicine, Department of Biological Chemistry, School of Medicine, University of California, Irvine, Irvine, CA, 92697, USA
| | - Sebastian Michels
- Departments of Pathology & Laboratory Medicine, Neurology, and Biological Chemistry, School of Medicine, and the UCI Institute for Neurotherapeutics, University of California Irvine, Irvine, CA, 92697, USA
| | - Eric J Wagner
- School of Medicine and Dentistry, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Albert R La Spada
- Departments of Pathology & Laboratory Medicine, Neurology, and Biological Chemistry, School of Medicine, and the UCI Institute for Neurotherapeutics, University of California Irvine, Irvine, CA, 92697, USA.
| | - Wei Li
- Division of Computational Biomedicine, Department of Biological Chemistry, School of Medicine, University of California, Irvine, Irvine, CA, 92697, USA.
| |
Collapse
|
16
|
Hendriks WJAJ, van Cruchten RTP, Pulido R. Hereditable variants of classical protein tyrosine phosphatase genes: Will they prove innocent or guilty? Front Cell Dev Biol 2023; 10:1051311. [PMID: 36755664 PMCID: PMC9900141 DOI: 10.3389/fcell.2022.1051311] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Accepted: 12/28/2022] [Indexed: 01/24/2023] Open
Abstract
Protein tyrosine phosphatases, together with protein tyrosine kinases, control many molecular signaling steps that control life at cellular and organismal levels. Impairing alterations in the genes encoding the involved proteins is expected to profoundly affect the quality of life-if compatible with life at all. Here, we review the current knowledge on the effects of germline variants that have been reported for genes encoding a subset of the protein tyrosine phosphatase superfamily; that of the thirty seven classical members. The conclusion must be that the newest genome research tools produced an avalanche of data that suggest 'guilt by association' for individual genes to specific disorders. Future research should face the challenge to investigate these accusations thoroughly and convincingly, to reach a mature genotype-phenotype map for this intriguing protein family.
Collapse
Affiliation(s)
- Wiljan J. A. J. Hendriks
- Department of Cell Biology, Radboud University Medical Centre, Nijmegen, The Netherlands,*Correspondence: Wiljan J. A. J. Hendriks,
| | | | - Rafael Pulido
- Biomarkers in Cancer Unit, Biocruces Bizkaia Health Research Institute, Barakaldo, Spain,Ikerbasque, Basque Foundation for Science, Bilbao, Spain
| |
Collapse
|
17
|
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, 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
|
18
|
Ren M, Yang Y, Heng KHY, Ng LY, Chong CYY, Ng YT, Gorur-Shandilya S, Lee RMQ, Lim KL, Zhang J, Koh TW. MED13 and glycolysis are conserved modifiers of α-synuclein-associated neurodegeneration. Cell Rep 2022; 41:111852. [PMID: 36543134 DOI: 10.1016/j.celrep.2022.111852] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 10/04/2022] [Accepted: 11/29/2022] [Indexed: 12/24/2022] Open
Abstract
α-Synuclein (α-syn) is important in synucleinopathies such as Parkinson's disease (PD). While genome-wide association studies (GWASs) of synucleinopathies have identified many risk loci, the underlying genes have not been shown for most loci. Using Drosophila, we screened 3,471 mutant chromosomes for genetic modifiers of α-synuclein and identified 12 genes. Eleven modifiers have human orthologs associated with diseases, including MED13 and CDC27, which lie within PD GWAS loci. Drosophila Skd/Med13 and glycolytic enzymes are co-upregulated by α-syn-associated neurodegeneration. While elevated α-syn compromises mitochondrial function, co-expressing skd/Med13 RNAi and α-syn synergistically increase the ratio of oxidized-to-reduced glutathione. The resulting neurodegeneration can be suppressed by overexpressing a glycolytic enzyme or treatment with deferoxamine, suggesting that compensatory glycolysis is neuroprotective. In addition, the functional relationship between α-synuclein, MED13, and glycolytic enzymes is conserved between flies and mice. We propose that hypoxia-inducible factor and MED13 are part of a druggable pathway for PD.
Collapse
Affiliation(s)
- Mengda Ren
- Temasek Life Sciences Laboratory, Singapore 117604, Singapore; Department of Biological Sciences, National University of Singapore, Singapore 117558, Singapore; Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308207, Singapore; National Neuroscience Institute, Singapore 308433, Singapore
| | - Ying Yang
- Department of Pathology, Zhejiang University First Affiliated Hospital and School of Medicine, Hangzhou, Zhejiang 310002, China
| | | | - Lu Yi Ng
- Temasek Life Sciences Laboratory, Singapore 117604, Singapore
| | | | - Yan Ting Ng
- Temasek Life Sciences Laboratory, Singapore 117604, Singapore
| | | | - Rachel Min Qi Lee
- Temasek Life Sciences Laboratory, Singapore 117604, Singapore; Department of Biological Sciences, National University of Singapore, Singapore 117558, Singapore
| | - Kah Leong Lim
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308207, Singapore; National Neuroscience Institute, Singapore 308433, Singapore
| | - Jing Zhang
- Department of Pathology, Zhejiang University First Affiliated Hospital and School of Medicine, Hangzhou, Zhejiang 310002, China; China National Health and Disease Human Brain Tissue Resource Center, Hangzhou, Zhejiang 310002, China
| | - Tong-Wey Koh
- Temasek Life Sciences Laboratory, Singapore 117604, Singapore; Department of Biological Sciences, National University of Singapore, Singapore 117558, Singapore.
| |
Collapse
|
19
|
Mew M, Caldwell KA, Caldwell GA. From bugs to bedside: functional annotation of human genetic variation for neurological disorders using invertebrate models. Hum Mol Genet 2022; 31:R37-R46. [PMID: 35994032 PMCID: PMC9585664 DOI: 10.1093/hmg/ddac203] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 08/11/2022] [Accepted: 08/17/2022] [Indexed: 02/02/2023] Open
Abstract
The exponential accumulation of DNA sequencing data has opened new avenues for discovering the causative roles of single-nucleotide polymorphisms (SNPs) in neurological diseases. The opportunities emerging from this are staggering, yet only as good as our abilities to glean insights from this surplus of information. Whereas computational biology continues to improve with respect to predictions and molecular modeling, the differences between in silico and in vivo analysis remain substantial. Invertebrate in vivo model systems represent technically advanced, experimentally mature, high-throughput, efficient and cost-effective resources for investigating a disease. With a decades-long track record of enabling investigators to discern function from DNA, fly (Drosophila) and worm (Caenorhabditis elegans) models have never been better poised to serve as living engines of discovery. Both of these animals have already proven useful in the classification of genetic variants as either pathogenic or benign across a range of neurodevelopmental and neurodegenerative disorders-including autism spectrum disorders, ciliopathies, amyotrophic lateral sclerosis, Alzheimer's and Parkinson's disease. Pathogenic SNPs typically display distinctive phenotypes in functional assays when compared with null alleles and frequently lead to protein products with gain-of-function or partial loss-of-function properties that contribute to neurological disease pathogenesis. The utility of invertebrates is logically limited by overt differences in anatomical and physiological characteristics, and also the evolutionary distance in genome structure. Nevertheless, functional annotation of disease-SNPs using invertebrate models can expedite the process of assigning cellular and organismal consequences to mutations, ascertain insights into mechanisms of action, and accelerate therapeutic target discovery and drug development for neurological conditions.
Collapse
Affiliation(s)
- Melanie Mew
- Department of Biological Sciences, The University of Alabama, Tuscaloosa, AL 35487, USA
| | - Kim A Caldwell
- Department of Biological Sciences, The University of Alabama, Tuscaloosa, AL 35487, USA
- Alabama Research Institute on Aging, The University of Alabama, Tuscaloosa, AL 35487, USA
- Center for Convergent Bioscience and Medicine, The University of Alabama, Tuscaloosa, AL 35487, USA
- Departments of Neurobiology and Neurology, Center for Neurodegeneration and Experimental Therapeutics, Nathan Shock Center of Excellence for Research in the Basic Biology of Aging, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Guy A Caldwell
- Department of Biological Sciences, The University of Alabama, Tuscaloosa, AL 35487, USA
- Center for Convergent Bioscience and Medicine, The University of Alabama, Tuscaloosa, AL 35487, USA
- Departments of Neurobiology and Neurology, Center for Neurodegeneration and Experimental Therapeutics, Nathan Shock Center of Excellence for Research in the Basic Biology of Aging, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| |
Collapse
|
20
|
Henderson IM, Marez C, Dokladny K, Smoake J, Martinez M, Johnson D, Uhl GR. Substrate-selective positive allosteric modulation of PTPRD’s phosphatase by flavonols. Biochem Pharmacol 2022; 202:115109. [PMID: 35636503 PMCID: PMC10184881 DOI: 10.1016/j.bcp.2022.115109] [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: 03/12/2022] [Revised: 05/23/2022] [Accepted: 05/23/2022] [Indexed: 11/30/2022]
Abstract
The receptor type protein tyrosine phosphatase D (PTPRD) is expressed by neurons and implicated in interesting phenotypes that include reward from addictive substances, restless leg syndrome and neurofibrillary tangle densities in Alzheimer's disease (AD-NFTs). However, the brain phosphotyrosine phosphoprotein (PTPP) substrates for PTPRD's phosphatase have not been clearly defined. Although we have identified small molecule inhibitors of PTPRD's phosphatase that are candidates for reducing reward from addictive substances, no positive allosteric modulators of this phosphatase that might be candidates for reducing AD-NFTs have been reported. We now report identification of candidate brain substrates for PTPRD based on their increased phosphorylation in knockout vs wildtype animals, coexpression with PTPRD in neuronal subtypes and brisk dephosphorylation by recombinant human PTPRD phosphatase. We also report discovery that quercetin and other flavonols, though not closely-related flavones, enhance rates of PTPRD's dephosphorylation of a group of these candidate substrate PTPPs but not others. This substrate-selective positive allosteric modulation provides a novel pharmacological action. Flavonol-mediated increases in PTPRD's dephosphorylation of the GSK3 β and α kinases that hyperphosphorylate tau, the major component of AD-NFTs, could help to explain recent data concerning genetic and dietary impacts on Alzheimer's disease.
Collapse
Affiliation(s)
- Ian M Henderson
- Biomedical Research Institute of New Mexico, Albuquerque, NM 87108, United States; New Mexico VA Healthcare System, Albuquerque, NM 87108, United States
| | - Carlissa Marez
- Biomedical Research Institute of New Mexico, Albuquerque, NM 87108, United States; New Mexico VA Healthcare System, Albuquerque, NM 87108, United States
| | - Karol Dokladny
- Department of Medicine, University of New Mexico, Albuquerque, NM 87131, United States
| | - Jane Smoake
- Biomedical Research Institute of New Mexico, Albuquerque, NM 87108, United States; New Mexico VA Healthcare System, Albuquerque, NM 87108, United States
| | - Maria Martinez
- Biomedical Research Institute of New Mexico, Albuquerque, NM 87108, United States; New Mexico VA Healthcare System, Albuquerque, NM 87108, United States
| | - David Johnson
- College of Pharmacy, University of Kansas, Lawrence, KS 66045, United States
| | - George R Uhl
- Biomedical Research Institute of New Mexico, Albuquerque, NM 87108, United States; New Mexico VA Healthcare System, Albuquerque, NM 87108, United States; Departments of Neurology, Neuroscience and Molecular Genetics and Microbiology, University of New Mexico, Albuquerque, NM 87131, United States; Departments of Neurology and Pharmacology, University of Maryland School of Medicine, Baltimore, MD 21201, United States; Maryland VA Healthcare System, Baltimore, MD 21201, United States.
| |
Collapse
|
21
|
Taylor HA, Simmons KJ, Clavane EM, Trevelyan CJ, Brown JM, Przemyłska L, Watt NT, Matthews LC, Meakin PJ. PTPRD and DCC Are Novel BACE1 Substrates Differentially Expressed in Alzheimer's Disease: A Data Mining and Bioinformatics Study. Int J Mol Sci 2022; 23:ijms23094568. [PMID: 35562959 PMCID: PMC9103286 DOI: 10.3390/ijms23094568] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 04/15/2022] [Accepted: 04/18/2022] [Indexed: 02/04/2023] Open
Abstract
The β-site Amyloid precursor protein Cleaving Enzyme 1 (BACE1) is an extensively studied therapeutic target for Alzheimer's disease (AD), owing to its role in the production of neurotoxic amyloid beta (Aβ) peptides. However, despite numerous BACE1 inhibitors entering clinical trials, none have successfully improved AD pathogenesis, despite effectively lowering Aβ concentrations. This can, in part, be attributed to an incomplete understanding of BACE1, including its physiological functions and substrate specificity. We propose that BACE1 has additional important physiological functions, mediated through substrates still to be identified. Thus, to address this, we computationally analysed a list of 533 BACE1 dependent proteins, identified from the literature, for potential BACE1 substrates, and compared them against proteins differentially expressed in AD. We identified 15 novel BACE1 substrates that were specifically altered in AD. To confirm our analysis, we validated Protein tyrosine phosphatase receptor type D (PTPRD) and Netrin receptor DCC (DCC) using Western blotting. These findings shed light on the BACE1 inhibitor failings and could enable the design of substrate-specific inhibitors to target alternative BACE1 substrates. Furthermore, it gives us a greater understanding of the roles of BACE1 and its dysfunction in AD.
Collapse
Affiliation(s)
- Hannah A. Taylor
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds LS2 9JT, UK; (H.A.T.); (K.J.S.); (E.M.C.); (C.J.T.); (J.M.B.); (L.P.); (N.T.W.)
| | - Katie J. Simmons
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds LS2 9JT, UK; (H.A.T.); (K.J.S.); (E.M.C.); (C.J.T.); (J.M.B.); (L.P.); (N.T.W.)
| | - Eva M. Clavane
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds LS2 9JT, UK; (H.A.T.); (K.J.S.); (E.M.C.); (C.J.T.); (J.M.B.); (L.P.); (N.T.W.)
| | - Christopher J. Trevelyan
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds LS2 9JT, UK; (H.A.T.); (K.J.S.); (E.M.C.); (C.J.T.); (J.M.B.); (L.P.); (N.T.W.)
| | - Jane M. Brown
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds LS2 9JT, UK; (H.A.T.); (K.J.S.); (E.M.C.); (C.J.T.); (J.M.B.); (L.P.); (N.T.W.)
| | - Lena Przemyłska
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds LS2 9JT, UK; (H.A.T.); (K.J.S.); (E.M.C.); (C.J.T.); (J.M.B.); (L.P.); (N.T.W.)
| | - Nicole T. Watt
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds LS2 9JT, UK; (H.A.T.); (K.J.S.); (E.M.C.); (C.J.T.); (J.M.B.); (L.P.); (N.T.W.)
| | - Laura C. Matthews
- Leeds Institute of Medical Research, Faculty of Medicine and Health, University of Leeds, Leeds LS2 9JT, UK;
| | - Paul J. Meakin
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds LS2 9JT, UK; (H.A.T.); (K.J.S.); (E.M.C.); (C.J.T.); (J.M.B.); (L.P.); (N.T.W.)
- Correspondence:
| |
Collapse
|
22
|
Meng X, Wu Y, Liu W, Wang Y, Xu Z, Jiao Z. Research on Voxel-Based Features Detection and Analysis of Alzheimer’s Disease Using Random Survey Support Vector Machine. Front Neuroinform 2022; 16:856295. [PMID: 35418845 PMCID: PMC8995748 DOI: 10.3389/fninf.2022.856295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 02/08/2022] [Indexed: 11/13/2022] Open
Abstract
Alzheimer’s disease (AD) is a degenerative disease of the central nervous system characterized by memory and cognitive dysfunction, as well as abnormal changes in behavior and personality. The research focused on how machine learning classified AD became a recent hotspot. In this study, we proposed a novel voxel-based feature detection framework for AD. Specifically, using 649 voxel-based morphometry (VBM) methods obtained from MRI in Alzheimer’s Disease Neuroimaging Initiative (ADNI), we proposed a feature detection method according to the Random Survey Support Vector Machines (RS-SVM) and combined the research process based on image-, gene-, and pathway-level analysis for AD prediction. Particularly, we constructed 136, 141, and 113 novel voxel-based features for EMCI (early mild cognitive impairment)-HC (healthy control), LMCI (late mild cognitive impairment)-HC, and AD-HC groups, respectively. We applied linear regression model, least absolute shrinkage and selection operator (Lasso), partial least squares (PLS), SVM, and RS-SVM five methods to test and compare the accuracy of these features in these three groups. The prediction accuracy of the AD-HC group using the RS-SVM method was higher than 90%. In addition, we performed functional analysis of the features to explain the biological significance. The experimental results using five machine learning indicate that the identified features are effective for AD and HC classification, the RS-SVM framework has the best classification accuracy, and our strategy can identify important brain regions for AD.
Collapse
Affiliation(s)
- Xianglian Meng
- School of Computer Information and Engineering, Changzhou Institute of Technology, Changzhou, China
| | - Yue Wu
- School of Computer Information and Engineering, Changzhou Institute of Technology, Changzhou, China
| | - Wenjie Liu
- School of Computer Information and Engineering, Changzhou Institute of Technology, Changzhou, China
| | - Ying Wang
- School of Computer Science and Engineering, Changshu Institute of Technology, Changshu, China
| | - Zhe Xu
- School of Computer Information and Engineering, Changzhou Institute of Technology, Changzhou, China
| | - Zhuqing Jiao
- School of Computer Science and Artificial Intelligence, Changzhou University, Changzhou, China
- *Correspondence: Zhuqing Jiao,
| |
Collapse
|
23
|
Finding New Ways How to Control BACE1. J Membr Biol 2022; 255:293-318. [PMID: 35305135 DOI: 10.1007/s00232-022-00225-1] [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: 02/02/2022] [Accepted: 02/24/2022] [Indexed: 01/18/2023]
Abstract
Recently, all applications of BACE1 inhibitors failed as therapeutical targets for Alzheimer´s disease (AD) due to severe side effects. Therefore, alternative ways for treatment development are a hot research topic. The present analysis investigates BACE1 protein-protein interaction networks and attempts to solve the absence of complete knowledge about pathways involving BACE1. A bioinformatics analysis matched the functions of the non-substrate interaction network with Voltage-gated potassium channels, which also appear as top priority protein nodes. Targeting BACE1 interactions with PS1 and GGA-s, blocking of BACE1 access to APP by BRI3 and RTN-s, activation of Wnt signaling and upregulation of β-catenin, and brain delivery of the extracellular domain of p75NTR, are the main alternatives to the use of BACE 1 inhibitors highlighted by the analysis. The pathway enrichment analysis also emphasized substrates and substrate candidates with essential biological functions, which cleavage must remain controlled. They include ephrin receptors, ROBO1, ROBO2, CNTN-s, CASPR-s, CD147, CypB, TTR, APLP1/APLP2, NRXN-s, and PTPR-s. The analysis of the interaction subnetwork of BACE1 functionally related to inflammation identified a connection to three cardiomyopathies, which supports the hypothesis of the common molecular mechanisms with AD. A lot of potential shows the regulation of BACE1 activity through post-translational modifications. The interaction network of BACE1 and its phosphorylation enzyme CSNK1D functionally match the Circadian clock, p53, and Hedgehog signaling pathways. The regulation of BACE1 glycosylation could be achieved through N-acetylglucosamine transferases, α-(1→6)-fucosyltransferase, β-galactoside α-(2→6)-sialyltransferases, galactosyltransferases, and mannosidases suggested by the interaction network analysis of BACE1-MGAT3. The present analysis proposes possibilities for the alternative control of AD pathology.
Collapse
|
24
|
Camacho J, Rábano A, Marazuela P, Bonaterra‐Pastra A, Serna G, Moliné T, Ramón y Cajal S, Martínez‐Sáez E, Hernández‐Guillamon M. Association of CD2AP neuronal deposits with Braak neurofibrillary stage in Alzheimer's disease. Brain Pathol 2022; 32:e13016. [PMID: 34514662 PMCID: PMC8713526 DOI: 10.1111/bpa.13016] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 06/25/2021] [Accepted: 07/29/2021] [Indexed: 01/22/2023] Open
Abstract
Genome-wide association studies have described several genes as genetic susceptibility loci for Alzheimer's disease (AD). Among them, CD2AP encodes CD2-associated protein, a scaffold protein implicated in dynamic actin remodeling and membrane trafficking during endocytosis and cytokinesis. Although a clear link between CD2AP defects and glomerular pathology has been described, little is known about the function of CD2AP in the brain. The aim of this study was to analyze the distribution of CD2AP in the AD brain and its potential associations with tau aggregation and β-amyloid (Aβ) deposition. First, we performed immunohistochemical analysis of CD2AP expression in brain tissue from AD patients and controls (N = 60). Our results showed granular CD2AP immunoreactivity in the human brain endothelium in all samples. In AD cases, no CD2AP was found to be associated with Aβ deposits in vessels or parenchymal plaques. CD2AP neuronal inclusions similar to neurofibrillary tangles (NFT) and neuropil thread-like deposits were found only in AD samples. Moreover, immunofluorescence analysis revealed that CD2AP colocalized with pTau. Regarding CD2AP neuronal distribution, a hierarchical progression from the entorhinal to the temporal and occipital cortex was detected. We found that CD2AP immunodetection in neurons was strongly and positively associated with Braak neurofibrillary stage, independent of age and other pathological hallmarks. To further investigate the association between pTau and CD2AP, we included samples from cases of primary tauopathies (corticobasal degeneration [CBD], progressive supranuclear palsy [PSP], and Pick's disease [PiD]) in our study. Among these cases, CD2AP positivity was only found in PiD samples as neurofibrillary tangle-like and Pick body-like deposits, whereas no neuronal CD2AP deposits were detected in PSP or CBD samples, which suggested an association of CD2AP neuronal expression with 3R-Tau-diseases. In conclusion, our findings open a new road to investigate the complex cellular mechanism underlying the tangle conformation and tau pathology in the brain.
Collapse
Affiliation(s)
- Jessica Camacho
- Pathology DepartmentVall d’Hebron University HospitalBarcelonaSpain
- Morphological Science DepartmentUniversitat Autonoma de BarcelonaBarcelonaSpain
| | - Alberto Rábano
- Neuropathology DepartmentCIEN FoundationAlzheimer’s Centre Queen Sofía FoundationMadridSpain
| | - Paula Marazuela
- Neurovascular Research LaboratoryVall d’Hebron Research InstituteUniversitat Autonoma de BarcelonaBarcelonaSpain
| | - Anna Bonaterra‐Pastra
- Neurovascular Research LaboratoryVall d’Hebron Research InstituteUniversitat Autonoma de BarcelonaBarcelonaSpain
| | - Garazi Serna
- Morphological Science DepartmentUniversitat Autonoma de BarcelonaBarcelonaSpain
- Molecular Oncology GroupVall d’Hebron Institute of Oncology (VHIO)BarcelonaSpain
| | - Teresa Moliné
- Pathology DepartmentVall d’Hebron University HospitalBarcelonaSpain
| | - Santiago Ramón y Cajal
- Pathology DepartmentVall d’Hebron University HospitalBarcelonaSpain
- Morphological Science DepartmentUniversitat Autonoma de BarcelonaBarcelonaSpain
| | - Elena Martínez‐Sáez
- Pathology DepartmentVall d’Hebron University HospitalBarcelonaSpain
- Morphological Science DepartmentUniversitat Autonoma de BarcelonaBarcelonaSpain
| | - Mar Hernández‐Guillamon
- Neurovascular Research LaboratoryVall d’Hebron Research InstituteUniversitat Autonoma de BarcelonaBarcelonaSpain
| |
Collapse
|
25
|
Weller AE, Ferraro TN, Doyle GA, Reiner BC, Crist RC, Berrettini WH. Single Nucleus Transcriptome Data from Alzheimer's Disease Mouse Models Yield New Insight into Pathophysiology. J Alzheimers Dis 2022; 90:1233-1247. [PMID: 36213995 DOI: 10.3233/jad-220391] [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: 11/23/2022]
Abstract
BACKGROUND 5XFAD humanized mutant mice and Trem2 knockout (T2KO) mice are two mouse models relevant to the study of Alzheimer's disease (AD)-related pathology. OBJECTIVE To determine hippocampal transcriptomic and polyadenylation site usage alterations caused by genetic mutations engineered in 5XFAD and T2KO mice. METHODS Employing a publicly available single-nucleus RNA sequencing dataset, we used Seurat and Sierra analytic programs to identify differentially expressed genes (DEGs) and differential transcript usage (DTU), respectively, in hippocampal cell types from each of the two mouse models. We analyzed cell type-specific DEGs further using Ingenuity Pathway Analysis (IPA). RESULTS We identified several DEGs in both neuronal and glial cell subtypes in comparisons of wild type (WT) versus 5XFAD and WT versus T2KO mice, including Ttr, Fth1, Pcsk1n, Malat1, Rpl37, Rtn1, Sepw1, Uba52, Mbp, Arl6ip5, Gm26917, Vwa1, and Pgrmc1. We also observed DTU in common between the two comparisons in neuronal and glial subtypes, specifically in the genes Prnp, Rbm4b, Pnisr, Opcml, Cpne7, Adgrb1, Gabarapl2, Ubb, Ndfip1, Car11, and Stmn4. IPA identified three statistically significant canonical pathways that appeared in multiple cell types and that overlapped between 5XFAD and T2KO comparisons to WT, including 'FXR/RXR Activation', 'LXR/RXR Activation', and 'Acute Phase Response Signaling'. CONCLUSION DEG, DTU, and IPA findings, derived from two different mouse models of AD, highlight the importance of energy imbalance and inflammatory processes in specific hippocampal cell types, including subtypes of neurons and glial cells, in the development of AD-related pathology. Additional studies are needed to further characterize these findings.
Collapse
Affiliation(s)
- Andrew E Weller
- Department of Psychiatry, Center for Neurobiology and Behavior, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Thomas N Ferraro
- Department of Psychiatry, Center for Neurobiology and Behavior, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ, USA
| | - Glenn A Doyle
- Department of Psychiatry, Center for Neurobiology and Behavior, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Benjamin C Reiner
- Department of Psychiatry, Center for Neurobiology and Behavior, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Richard C Crist
- Department of Psychiatry, Center for Neurobiology and Behavior, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Wade H Berrettini
- Department of Psychiatry, Center for Neurobiology and Behavior, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
26
|
Cornejo F, Cortés BI, Findlay GM, Cancino GI. LAR Receptor Tyrosine Phosphatase Family in Healthy and Diseased Brain. Front Cell Dev Biol 2021; 9:659951. [PMID: 34966732 PMCID: PMC8711739 DOI: 10.3389/fcell.2021.659951] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 11/17/2021] [Indexed: 11/23/2022] Open
Abstract
Protein phosphatases are major regulators of signal transduction and they are involved in key cellular mechanisms such as proliferation, differentiation, and cell survival. Here we focus on one class of protein phosphatases, the type IIA Receptor-type Protein Tyrosine Phosphatases (RPTPs), or LAR-RPTP subfamily. In the last decade, LAR-RPTPs have been demonstrated to have great importance in neurobiology, from neurodevelopment to brain disorders. In vertebrates, the LAR-RPTP subfamily is composed of three members: PTPRF (LAR), PTPRD (PTPδ) and PTPRS (PTPσ), and all participate in several brain functions. In this review we describe the structure and proteolytic processing of the LAR-RPTP subfamily, their alternative splicing and enzymatic regulation. Also, we review the role of the LAR-RPTP subfamily in neural function such as dendrite and axon growth and guidance, synapse formation and differentiation, their participation in synaptic activity, and in brain development, discussing controversial findings and commenting on the most recent studies in the field. Finally, we discuss the clinical outcomes of LAR-RPTP mutations, which are associated with several brain disorders.
Collapse
Affiliation(s)
- Francisca Cornejo
- Center for Integrative Biology, Facultad de Ciencias, Universidad Mayor, Santiago, Chile
| | - Bastián I Cortés
- Center for Integrative Biology, Facultad de Ciencias, Universidad Mayor, Santiago, Chile
| | - Greg M Findlay
- MRC Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Gonzalo I Cancino
- Center for Integrative Biology, Facultad de Ciencias, Universidad Mayor, Santiago, Chile.,Escuela de Biotecnología, Facultad de Ciencias, Universidad Mayor, Santiago, Chile
| |
Collapse
|
27
|
Bi XA, Li L, Xu R, Xing Z. Pathogenic Factors Identification of Brain Imaging and Gene in Late Mild Cognitive Impairment. Interdiscip Sci 2021; 13:511-520. [PMID: 34106420 DOI: 10.1007/s12539-021-00449-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 06/01/2021] [Accepted: 06/04/2021] [Indexed: 11/28/2022]
Abstract
Mild cognitive impairment (MCI) is a dangerous signal of severe cognitive decline. It can be separated into two steps: early MCI (EMCI) and late MCI (LMCI). As the post-state of MCI and pre-state of Alzheimer's disease (AD), LMCI receives insufficient attention in the field of brain science, causing the internal mechanism of LMCI has not been well understood. To better explore the focus and pathological mechanism of LMCI, a method called genetic evolved random forest (GERF) is applied. Resting functional magnetic resonance imaging (rfMRI) and gene data are obtained from 62 subjects (36 LMCI and 26 normal controls), and Pearson correlation analysis is adopted to perform the multimodal fusion of two types of data to construct fusion features. We identified pathogenic brain regions and genes that are highly related to LMCI using GERF and achieves a good effect. Compared with the normal control (NC) group, the abnormal brain regions of LMCI are PUT.L, PreCG.L, IFGtriang.R, REC.R, DCG.R, PoCG.L, and HES.L, and the pathogenic genes are FHIT, RF00019, FRMD4A, PTPRD, and RBFOX1. More importantly, most of these risk genes and abnormal brain regions have been confirmed to be related to AD and MCI in previous studies. In this study, we mapped them to LMCI with higher accuracies, so as to provide a more robust understanding of the physiological mechanism of MCI.
Collapse
Affiliation(s)
- Xia-An Bi
- Hunan Provincial Key Laboratory of Intelligent Computing and Language Information Processing, Hunan Normal University, Changsha, People's Republic of China. .,College of Information Science and Engineering, Hunan Normal University, Changsha, People's Republic of China.
| | - Lou Li
- Hunan Provincial Key Laboratory of Intelligent Computing and Language Information Processing, Hunan Normal University, Changsha, People's Republic of China.,College of Information Science and Engineering, Hunan Normal University, Changsha, People's Republic of China
| | - Ruihui Xu
- Hunan Provincial Key Laboratory of Intelligent Computing and Language Information Processing, Hunan Normal University, Changsha, People's Republic of China.,College of Information Science and Engineering, Hunan Normal University, Changsha, People's Republic of China
| | - Zhaoxu Xing
- Hunan Provincial Key Laboratory of Intelligent Computing and Language Information Processing, Hunan Normal University, Changsha, People's Republic of China.,College of Information Science and Engineering, Hunan Normal University, Changsha, People's Republic of China
| |
Collapse
|
28
|
Ji G, Li S, Ye L, Guan J. Gene Module Analysis Reveals Cell-Type Specificity and Potential Target Genes in Autism's Pathogenesis. Biomedicines 2021; 9:biomedicines9040410. [PMID: 33920310 PMCID: PMC8069308 DOI: 10.3390/biomedicines9040410] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 03/28/2021] [Accepted: 04/01/2021] [Indexed: 11/16/2022] Open
Abstract
Multiple genetic factors contribute to the pathogenesis of autism spectrum disorder (ASD), a kind of neurodevelopmental disorder. Genes were usually studied separately for their associations with ASD. However, genes associated with ASD do not act alone but interact with each other in a network module. The identification of these modules is the basis for the systematic understanding of the pathogenesis of ASD. Moreover, ASD is characterized by highly pathogenic heterogeneity, and gene modules associated with ASD are cell-type-specific. In this study, based on the single-nucleus RNA sequencing data of 41 post-mortem tissue samples from the prefrontal cortex and anterior cingulate cortex of 19 ASD patients and 16 control individuals, we applied sparse module activity factorization, a matrix decomposition method consistent with the multi-factor and heterogeneous characteristics of ASD pathogenesis, to identify cell-type-specific gene modules. Then, statistical procedures were performed to detect highly reproducible cell-type-specific ASD-associated gene modules. Through the enrichment analysis of cell markers, 31 cell-type-specific gene modules related to ASD were further screened out. These 31 gene modules are all enriched with curated ASD risk genes. Finally, we utilized the expression patterns of these cell-type-specific ASD-associated gene modules to build predictive models for ASD. The excellent predictive performance also proved the associations between these gene modules and ASD. Our study confirmed the multifactorial and cell-type-specific characteristics of ASD pathogeneses. The results showed that excitatory neurons such as L2/3, L4, and L5/6-CC play essential roles in ASD's pathogenic processes. We identified the potential ASD target genes that act together in cell-type-specific modules, such as NRG3, KCNIP4, BAI3, PTPRD, LRRTM4, and LINGO2 in the L2/3 gene modules. Our study offers new potential genomic targets for ASD and provides a novel method to study gene modules involved in the pathogenesis of ASD.
Collapse
Affiliation(s)
- Guoli Ji
- Department of Automation, Xiamen University, Xiamen 361102, China; (G.J.); (S.L.)
- National Institute for Data Science in Health and Medicine, Xiamen University, Xiamen 361102, China
| | - Shuchao Li
- Department of Automation, Xiamen University, Xiamen 361102, China; (G.J.); (S.L.)
| | - Lishan Ye
- Xiamen Health and Medical Big Data Center, Xiamen 361008, China
- Correspondence: (L.Y.); (J.G.)
| | - Jinting Guan
- Department of Automation, Xiamen University, Xiamen 361102, China; (G.J.); (S.L.)
- National Institute for Data Science in Health and Medicine, Xiamen University, Xiamen 361102, China
- Correspondence: (L.Y.); (J.G.)
| |
Collapse
|
29
|
Knockdown of long non-coding RNA SOX21-AS1 attenuates amyloid-β-induced neuronal damage by sponging miR-107. Biosci Rep 2021; 40:222277. [PMID: 32124921 PMCID: PMC7103586 DOI: 10.1042/bsr20194295] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 02/16/2020] [Accepted: 03/02/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Alzheimer's disease (AD), which has no effective drugs to delay or prevent its progression, is a multifactorial complex neurodegenerative disease. Long non-coding RNA SOX21 antisense RNA1 (SOX21-AS1) is associated with the development of AD, but the underlying molecular mechanism of SOX21-AS1 in AD is still largely unclear. METHODS To construct the AD model, SH-SY5Y and SK-N-SH cells were treated with amyloid-β1-42 (Aβ1-42). Quantitative real-time polymerase chain reaction (qRT-PCR) was executed to detect the expression of SOX21-AS1 and miRNA-107. Western blot analysis was utilized to assess the levels of phosphorylated Tau (p-Tau). 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) or flow cytometry assay was employed to determine the viability and apoptosis of SH-SY5Y and SK-N-SH cells. The relationship between SOX21-AS1 and miRNA-107 was verified with the dual-luciferase reporter assay. RESULTS SOX21-AS1 expression was augmented while miR-107 expression was decreased in Aβ1-42-treated SH-SY5Y and SK-N-SH cells. Moreover, Aβ1-42 elevated the levels of p-Tau and impeded viability and induced apoptosis of SH-SY5Y and SK-N-SH cells. Also, SOX21-AS1 silencing attenuated Aβ1-42 mediated the levels of p-Tau, viability, and apoptosis of SH-SY5Y and SK-N-SH cells. Importantly, SOX21-AS1 acted as a sponge for miR-107 in SH-SY5Y and SK-N-SH cells. Furthermore, the increase in p-Tau levels and apoptosis and the repression of viability of Aβ1-42-treated SH-SY5Y and SK-N-SH cells mediated by miR-107 inhibition were partly recovered by SOX21-AS1 depletion. CONCLUSION SOX21-AS1 silencing could attenuate Aβ1-42-induced neuronal damage by sponging miR-107, which provided a possible strategy for the treatment of AD.
Collapse
|
30
|
Sobue A, Komine O, Hara Y, Endo F, Mizoguchi H, Watanabe S, Murayama S, Saito T, Saido TC, Sahara N, Higuchi M, Ogi T, Yamanaka K. Microglial gene signature reveals loss of homeostatic microglia associated with neurodegeneration of Alzheimer's disease. Acta Neuropathol Commun 2021; 9:1. [PMID: 33402227 PMCID: PMC7786928 DOI: 10.1186/s40478-020-01099-x] [Citation(s) in RCA: 143] [Impact Index Per Article: 35.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 12/06/2020] [Indexed: 02/08/2023] Open
Abstract
Microglia-mediated neuroinflammation has been implicated in the pathogenesis of Alzheimer’s disease (AD). Although microglia in aging and neurodegenerative disease model mice show a loss of homeostatic phenotype and activation of disease-associated microglia (DAM), a correlation between those phenotypes and the degree of neuronal cell loss has not been clarified. In this study, we performed RNA sequencing of microglia isolated from three representative neurodegenerative mouse models, AppNL-G-F/NL-G-F with amyloid pathology, rTg4510 with tauopathy, and SOD1G93A with motor neuron disease by magnetic activated cell sorting. In parallel, gene expression patterns of the human precuneus with early Alzheimer’s change (n = 11) and control brain (n = 14) were also analyzed by RNA sequencing. We found that a substantial reduction of homeostatic microglial genes in rTg4510 and SOD1G93A microglia, whereas DAM genes were uniformly upregulated in all mouse models. The reduction of homeostatic microglial genes was correlated with the degree of neuronal cell loss. In human precuneus with early AD pathology, reduced expression of genes related to microglia- and oligodendrocyte-specific markers was observed, although the expression of DAM genes was not upregulated. Our results implicate a loss of homeostatic microglial function in the progression of AD and other neurodegenerative diseases. Moreover, analyses of human precuneus also suggest loss of microglia and oligodendrocyte functions induced by early amyloid pathology in human.
Collapse
|
31
|
Mukherjee S, Heath L, Preuss C, Jayadev S, Garden GA, Greenwood AK, Sieberts SK, De Jager PL, Ertekin-Taner N, Carter GW, Mangravite LM, Logsdon BA. Molecular estimation of neurodegeneration pseudotime in older brains. Nat Commun 2020; 11:5781. [PMID: 33188183 PMCID: PMC7666177 DOI: 10.1038/s41467-020-19622-y] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Accepted: 10/23/2020] [Indexed: 01/15/2023] Open
Abstract
The temporal molecular changes that lead to disease onset and progression in Alzheimer's disease (AD) are still unknown. Here we develop a temporal model for these unobserved molecular changes with a manifold learning method applied to RNA-Seq data collected from human postmortem brain samples collected within the ROS/MAP and Mayo Clinic RNA-Seq studies. We define an ordering across samples based on their similarity in gene expression and use this ordering to estimate the molecular disease stage-or disease pseudotime-for each sample. Disease pseudotime is strongly concordant with the burden of tau (Braak score, P = 1.0 × 10-5), Aβ (CERAD score, P = 1.8 × 10-5), and cognitive diagnosis (P = 3.5 × 10-7) of late-onset (LO) AD. Early stage disease pseudotime samples are enriched for controls and show changes in basic cellular functions. Late stage disease pseudotime samples are enriched for late stage AD cases and show changes in neuroinflammation and amyloid pathologic processes. We also identify a set of late stage pseudotime samples that are controls and show changes in genes enriched for protein trafficking, splicing, regulation of apoptosis, and prevention of amyloid cleavage pathways. In summary, we present a method for ordering patients along a trajectory of LOAD disease progression from brain transcriptomic data.
Collapse
Affiliation(s)
- Sumit Mukherjee
- Sage Bionetworks, Seattle, WA, USA
- Microsoft, Redmond, WA, USA
| | | | | | - Suman Jayadev
- Department of Neurology, University of Washington, Seattle, WA, USA
| | - Gwenn A Garden
- Department of Neurology, University of Washington, Seattle, WA, USA
| | | | | | - Philip L De Jager
- Center for Translational & Computational Neuroimmunology, Department of Neurology, Columbia University Irving Medical Center, New York City, NY, USA
- Taub Institute, Columbia University Irving Medical Center, New York City, NY, USA
| | - Nilüfer Ertekin-Taner
- Department of Neurology, Mayo Clinic Florid, Jacksonville, FL, USA
- Department of Neuroscience, Mayo Clinic Florida, Jacksonville, FL, USA
| | | | | | - Benjamin A Logsdon
- Sage Bionetworks, Seattle, WA, USA.
- Cajal Neuroscience, Seattle, WA, USA.
| |
Collapse
|
32
|
Sieberts SK, Perumal TM, Carrasquillo MM, Allen M, Reddy JS, Hoffman GE, Dang KK, Calley J, Ebert PJ, Eddy J, Wang X, Greenwood AK, Mostafavi S, Omberg L, Peters MA, Logsdon BA, De Jager PL, Ertekin-Taner N, Mangravite LM. Large eQTL meta-analysis reveals differing patterns between cerebral cortical and cerebellar brain regions. Sci Data 2020; 7:340. [PMID: 33046718 PMCID: PMC7550587 DOI: 10.1038/s41597-020-00642-8] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Accepted: 08/24/2020] [Indexed: 12/27/2022] Open
Abstract
The availability of high-quality RNA-sequencing and genotyping data of post-mortem brain collections from consortia such as CommonMind Consortium (CMC) and the Accelerating Medicines Partnership for Alzheimer's Disease (AMP-AD) Consortium enable the generation of a large-scale brain cis-eQTL meta-analysis. Here we generate cerebral cortical eQTL from 1433 samples available from four cohorts (identifying >4.1 million significant eQTL for >18,000 genes), as well as cerebellar eQTL from 261 samples (identifying 874,836 significant eQTL for >10,000 genes). We find substantially improved power in the meta-analysis over individual cohort analyses, particularly in comparison to the Genotype-Tissue Expression (GTEx) Project eQTL. Additionally, we observed differences in eQTL patterns between cerebral and cerebellar brain regions. We provide these brain eQTL as a resource for use by the research community. As a proof of principle for their utility, we apply a colocalization analysis to identify genes underlying the GWAS association peaks for schizophrenia and identify a potentially novel gene colocalization with lncRNA RP11-677M14.2 (posterior probability of colocalization 0.975).
Collapse
Affiliation(s)
| | | | | | - Mariet Allen
- Department of Neuroscience, Mayo Clinic Florida, Jacksonville, FL, 32224, USA
| | - Joseph S Reddy
- Department of Neuroscience, Mayo Clinic Florida, Jacksonville, FL, 32224, USA
| | - Gabriel E Hoffman
- Pamela Sklar Division of Psychiatric Genomics, Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Icahn Institute for Data Science and Genomic Technology, Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | | | - John Calley
- Lilly Research Labs, Eli Lilly and Company, Indianapolis, IN, 46225, USA
| | - Philip J Ebert
- Lilly Research Labs, Eli Lilly and Company, Indianapolis, IN, 46225, USA
| | - James Eddy
- Sage Bionetworks, Seattle, WA, 98121, USA
| | - Xue Wang
- Department of Neuroscience, Mayo Clinic Florida, Jacksonville, FL, 32224, USA
| | | | - Sara Mostafavi
- Departments of Statistics and Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada
- Centre for Molecular Medicine and Therapeutics, Vancouver, British Columbia, Canada
- Canadian Institute for Advanced Research, CIFAR Program in Child and Brain Development, Toronto, Ontario, Canada
| | | | | | | | - Philip L De Jager
- Center for Translational & Computational Neuroimmunology, Department of Neurology, Columbia University Medical Center, New York, NY, 10032, USA
| | - Nilüfer Ertekin-Taner
- Department of Neuroscience, Mayo Clinic Florida, Jacksonville, FL, 32224, USA
- Department of Neurology, Mayo Clinic Florida, Jacksonville, FL, 32224, USA
| | | |
Collapse
|
33
|
Mangleburg CG, Wu T, Yalamanchili HK, Guo C, Hsieh YC, Duong DM, Dammer EB, De Jager PL, Seyfried NT, Liu Z, Shulman JM. Integrated analysis of the aging brain transcriptome and proteome in tauopathy. Mol Neurodegener 2020; 15:56. [PMID: 32993812 PMCID: PMC7526226 DOI: 10.1186/s13024-020-00405-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Accepted: 09/18/2020] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Tau neurofibrillary tangle pathology characterizes Alzheimer's disease and other neurodegenerative tauopathies. Brain gene expression profiles can reveal mechanisms; however, few studies have systematically examined both the transcriptome and proteome or differentiated Tau- versus age-dependent changes. METHODS Paired, longitudinal RNA-sequencing and mass-spectrometry were performed in a Drosophila model of tauopathy, based on pan-neuronal expression of human wildtype Tau (TauWT) or a mutant form causing frontotemporal dementia (TauR406W). Tau-induced, differentially expressed transcripts and proteins were examined cross-sectionally or using linear regression and adjusting for age. Hierarchical clustering was performed to highlight network perturbations, and we examined overlaps with human brain gene expression profiles in tauopathy. RESULTS TauWT induced 1514 and 213 differentially expressed transcripts and proteins, respectively. TauR406W had a substantially greater impact, causing changes in 5494 transcripts and 697 proteins. There was a ~ 70% overlap between age- and Tau-induced changes and our analyses reveal pervasive bi-directional interactions. Strikingly, 42% of Tau-induced transcripts were discordant in the proteome, showing opposite direction of change. Tau-responsive gene expression networks strongly implicate innate immune activation. Cross-species analyses pinpoint human brain gene perturbations specifically triggered by Tau pathology and/or aging, and further differentiate between disease amplifying and protective changes. CONCLUSIONS Our results comprise a powerful, cross-species functional genomics resource for tauopathy, revealing Tau-mediated disruption of gene expression, including dynamic, age-dependent interactions between the brain transcriptome and proteome.
Collapse
Affiliation(s)
- Carl Grant Mangleburg
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030 USA
- Medical Scientist Training Program, Baylor College of Medicine, Houston, TX 77030 USA
| | - Timothy Wu
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030 USA
- Medical Scientist Training Program, Baylor College of Medicine, Houston, TX 77030 USA
| | - Hari K. Yalamanchili
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030 USA
| | - Caiwei Guo
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030 USA
| | - Yi-Chen Hsieh
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030 USA
| | - Duc M. Duong
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA 30322 USA
| | - Eric B. Dammer
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA 30322 USA
| | - Philip L. De Jager
- Center for Translational & Computational Neuroimmunology, Department of Neurology and Taub Institute for the study of Alzheimer’s disease and the aging brain, Columbia University Medical Center, New York, NY 10032 USA
- Cell Circuits Program, Broad Institute, Cambridge, MA 02142 USA
| | - Nicholas T. Seyfried
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA 30322 USA
- Department of Neurology, Emory University School of Medicine, Atlanta, GA 30322 USA
| | - Zhandong Liu
- Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030 USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, 1250 Moursund St., Suite N.1150, Houston, TX 77030 USA
| | - Joshua M. Shulman
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030 USA
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030 USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, 1250 Moursund St., Suite N.1150, Houston, TX 77030 USA
- Department of Neurology, Baylor College of Medicine, Houston, TX 77030 USA
| |
Collapse
|
34
|
Selective Neuronal Vulnerability in Alzheimer's Disease: A Network-Based Analysis. Neuron 2020; 107:821-835.e12. [PMID: 32603655 DOI: 10.1016/j.neuron.2020.06.010] [Citation(s) in RCA: 113] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 04/23/2020] [Accepted: 06/05/2020] [Indexed: 12/17/2022]
Abstract
A major obstacle to treating Alzheimer's disease (AD) is our lack of understanding of the molecular mechanisms underlying selective neuronal vulnerability, a key characteristic of the disease. Here, we present a framework integrating high-quality neuron-type-specific molecular profiles across the lifetime of the healthy mouse, which we generated using bacTRAP, with postmortem human functional genomics and quantitative genetics data. We demonstrate human-mouse conservation of cellular taxonomy at the molecular level for neurons vulnerable and resistant in AD, identify specific genes and pathways associated with AD neuropathology, and pinpoint a specific functional gene module underlying selective vulnerability, enriched in processes associated with axonal remodeling, and affected by amyloid accumulation and aging. We have made all cell-type-specific profiles and functional networks available at http://alz.princeton.edu. Overall, our study provides a molecular framework for understanding the complex interplay between Aβ, aging, and neurodegeneration within the most vulnerable neurons in AD.
Collapse
|
35
|
Milind N, Preuss C, Haber A, Ananda G, Mukherjee S, John C, Shapley S, Logsdon BA, Crane PK, Carter GW. Transcriptomic stratification of late-onset Alzheimer's cases reveals novel genetic modifiers of disease pathology. PLoS Genet 2020; 16:e1008775. [PMID: 32492070 PMCID: PMC7295244 DOI: 10.1371/journal.pgen.1008775] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 06/15/2020] [Accepted: 04/09/2020] [Indexed: 11/18/2022] Open
Abstract
Late-Onset Alzheimer's disease (LOAD) is a common, complex genetic disorder well-known for its heterogeneous pathology. The genetic heterogeneity underlying common, complex diseases poses a major challenge for targeted therapies and the identification of novel disease-associated variants. Case-control approaches are often limited to examining a specific outcome in a group of heterogenous patients with different clinical characteristics. Here, we developed a novel approach to define relevant transcriptomic endophenotypes and stratify decedents based on molecular profiles in three independent human LOAD cohorts. By integrating post-mortem brain gene co-expression data from 2114 human samples with LOAD, we developed a novel quantitative, composite phenotype that can better account for the heterogeneity in genetic architecture underlying the disease. We used iterative weighted gene co-expression network analysis (WGCNA) to reduce data dimensionality and to isolate gene sets that are highly co-expressed within disease subtypes and represent specific molecular pathways. We then performed single variant association testing using whole genome-sequencing data for the novel composite phenotype in order to identify genetic loci that contribute to disease heterogeneity. Distinct LOAD subtypes were identified for all three study cohorts (two in ROSMAP, three in Mayo Clinic, and two in Mount Sinai Brain Bank). Single variant association analysis identified a genome-wide significant variant in TMEM106B (p-value < 5×10-8, rs1990620G) in the ROSMAP cohort that confers protection from the inflammatory LOAD subtype. Taken together, our novel approach can be used to stratify LOAD into distinct molecular subtypes based on affected disease pathways.
Collapse
Affiliation(s)
- Nikhil Milind
- The Jackson Laboratory, Bar Harbor, Maine, United States of America
- Program in Genetics, Department of Biological Sciences, North Carolina State University, Raleigh, North Carolina, United States of America
| | - Christoph Preuss
- The Jackson Laboratory, Bar Harbor, Maine, United States of America
| | - Annat Haber
- The Jackson Laboratory, Bar Harbor, Maine, United States of America
| | | | - Shubhabrata Mukherjee
- Department of Medicine, School of Medicine, University of Washington, Seattle, Washington, United States of America
| | - Cai John
- The Jackson Laboratory, Bar Harbor, Maine, United States of America
| | - Sarah Shapley
- The Jackson Laboratory, Bar Harbor, Maine, United States of America
- Program in Neuroscience, Department of Biology and Geology, Baldwin Wallace University, Berea, Ohio, United States of America
| | | | - Paul K. Crane
- Department of Medicine, School of Medicine, University of Washington, Seattle, Washington, United States of America
| | | |
Collapse
|
36
|
Brown AS, Meera P, Quinones G, Magri J, Otis TS, Pulst SM, Oro AE. Receptor protein tyrosine phosphatases control Purkinje neuron firing. Cell Cycle 2020; 19:153-159. [PMID: 31876231 PMCID: PMC6961678 DOI: 10.1080/15384101.2019.1695995] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 10/23/2019] [Accepted: 11/07/2019] [Indexed: 10/25/2022] Open
Abstract
Spinocerebellar ataxias (SCA) are a genetically heterogeneous family of cerebellar neurodegenerative diseases characterized by abnormal firing of Purkinje neurons and degeneration. We recently demonstrated the slowed firing rates seen in several SCAs share a common etiology of hyper-activation of the Src family of non-receptor tyrosine kinases (SFKs). However, the lack of clinically available neuroactive SFK inhibitors lead us to investigate alternative mechanisms to modulate SFK activity. Previous studies demonstrate that SFK activity can be enhanced by the removal of inhibitory phospho-marks by receptor-protein-tyrosine phosphatases (RPTPs). In this Extra View we show that MTSS1 inhibits SFK activity through the binding and inhibition of a subset of the RPTP family members, and lowering RPTP activity in cerebellar slices with peptide inhibitors increases the suppressed Purkinje neuron basal firing rates seen in two different SCA models. Together these results identify RPTPs as novel effectors of Purkinje neuron basal firing, extending the MTSS1/SFK regulatory circuit we previously described and expanding the therapeutic targets for SCA patients.
Collapse
Affiliation(s)
- Alexander S. Brown
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Pratap Meera
- Department of Neurobiology, University of California, Los Angeles, CA, USA
| | - Gabe Quinones
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Jessica Magri
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Thomas S. Otis
- Sainsbury Wellcome Centre for Neural Circuits and Behavior, University College London, London, UK
| | - Stefan M. Pulst
- Department of Neurology, University of Utah Medical Center, Salt Lake City, UT, USA
| | - Anthony E. Oro
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
37
|
Sun S, Jiao M, Han C, Zhang Q, Shi W, Shi J, Li X. Causal Effects of Genetically Determined Metabolites on Risk of Polycystic Ovary Syndrome: A Mendelian Randomization Study. Front Endocrinol (Lausanne) 2020; 11:621. [PMID: 33013699 PMCID: PMC7505923 DOI: 10.3389/fendo.2020.00621] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 07/30/2020] [Indexed: 12/16/2022] Open
Abstract
Background: Polycystic ovary syndrome (PCOS) is a heterogeneous endocrine disorder that is influenced by both genetic and environmental factors. However, the etiology of PCOS remains unclear. Methods: We conducted a two-sample Mendelian randomization (MR) analysis to assess the causal effects of genetically determined metabolites (GDMs) on the risk of PCOS. We used summary level data of a genome-wide association study (GWAS) on 486 metabolites (n = 7,824) as exposure and a PCOS GWAS consisting of 4,138 cases and 20,129 controls as the outcome. Both datasets were obtained from publicly published databases. For each metabolite, a genetic instrumental variable was generated to assess the relationship between the metabolite and PCOS. For MR analysis, we primarily used the standard inverse variance weighted (IVW) method, while three additional methods-the MR-Egger, weighted median, and MR-PRESSO (pleiotropy residual sum and outlier) methods-were performed as sensitivity analyses. Results: Using genetic variants as predictors, we observed a robust relationship between epiandrosterone sulfate (EPIA-S) and PCOS (PIVW = 0.0186, PMR-Egger = 0.0111; PWeighted-median = 0.0154, and PMR-PRESSO = 0.0290). Similarly, 3-dehydrocarnitine, 4-hydroxyhippurate, hexadecanedioate, and β-hydroxyisovalerate may also have causal effects on PCOS development. Conclusions: We identified metabolites that might have causal effects on PCOS development. Our study emphasizes the role of genetic factors underlying the causal relationships between metabolites and PCOS and provides novel insights through the integration of metabolomics and genomics to better understand the mechanisms involved in human disease pathogenesis.
Collapse
Affiliation(s)
- Shuliu Sun
- Department of Obstetrics and Gynecology, Northwest Women's and Children's Hospital, Xi'an, China
| | - Minjie Jiao
- Department of Obstetrics and Gynecology, Northwest Women's and Children's Hospital, Xi'an, China
| | - Chengcheng Han
- Department of Obstetrics and Gynecology, Northwest Women's and Children's Hospital, Xi'an, China
| | - Qian Zhang
- Department of Obstetrics and Gynecology, Northwest Women's and Children's Hospital, Xi'an, China
| | - Wenhao Shi
- The Assisted Reproductive Centre, Northwest Women's and Children's Hospital, Xi'an, China
| | - Juanzi Shi
- The Assisted Reproductive Centre, Northwest Women's and Children's Hospital, Xi'an, China
| | - Xiaojuan Li
- Department of Obstetrics and Gynecology, Northwest Women's and Children's Hospital, Xi'an, China
- *Correspondence: Xiaojuan Li
| |
Collapse
|
38
|
Ojelade SA, Lee TV, Giagtzoglou N, Yu L, Ugur B, Li Y, Duraine L, Zuo Z, Petyuk V, De Jager PL, Bennett DA, Arenkiel BR, Bellen HJ, Shulman JM. cindr, the Drosophila Homolog of the CD2AP Alzheimer's Disease Risk Gene, Is Required for Synaptic Transmission and Proteostasis. Cell Rep 2019; 28:1799-1813.e5. [PMID: 31412248 PMCID: PMC6703184 DOI: 10.1016/j.celrep.2019.07.041] [Citation(s) in RCA: 21] [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: 12/28/2018] [Revised: 05/30/2019] [Accepted: 07/15/2019] [Indexed: 12/11/2022] Open
Abstract
The Alzheimer's disease (AD) susceptibility gene, CD2-associated protein (CD2AP), encodes an actin binding adaptor protein, but its function in the nervous system is largely unknown. Loss of the Drosophila ortholog cindr enhances neurotoxicity of human Tau, which forms neurofibrillary tangle pathology in AD. We show that Cindr is expressed in neurons and present at synaptic terminals. cindr mutants show impairments in synapse maturation and both synaptic vesicle recycling and release. Cindr associates and genetically interacts with 14-3-3ζ, regulates the ubiquitin-proteasome system, and affects turnover of Synapsin and the plasma membrane calcium ATPase (PMCA). Loss of cindr elevates PMCA levels and reduces cytosolic calcium. Studies of Cd2ap null mice support a conserved role in synaptic proteostasis, and CD2AP protein levels are inversely related to Synapsin abundance in human postmortem brains. Our results reveal CD2AP neuronal requirements with relevance to AD susceptibility, including for proteostasis, calcium handling, and synaptic structure and function.
Collapse
Affiliation(s)
- Shamsideen A Ojelade
- Department of Neurology, Baylor College of Medicine, Houston, TX 77030, USA; Jan and Dan Duncan Neurologic Research Institute, Texas Children's Hospital, Houston, TX 77030, USA
| | - Tom V Lee
- Department of Neurology, Baylor College of Medicine, Houston, TX 77030, USA; Jan and Dan Duncan Neurologic Research Institute, Texas Children's Hospital, Houston, TX 77030, USA
| | - Nikolaos Giagtzoglou
- Department of Neurology, Baylor College of Medicine, Houston, TX 77030, USA; Jan and Dan Duncan Neurologic Research Institute, Texas Children's Hospital, Houston, TX 77030, USA
| | - Lei Yu
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL 60612, USA
| | - Berrak Ugur
- Program in Developmental Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Yarong Li
- Department of Neurology, Baylor College of Medicine, Houston, TX 77030, USA; Jan and Dan Duncan Neurologic Research Institute, Texas Children's Hospital, Houston, TX 77030, USA
| | - Lita Duraine
- Department of Molecular & Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Zhongyuan Zuo
- Department of Molecular & Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Vlad Petyuk
- Pacific Northwest National Laboratory, Richland, WA 99354, USA
| | - Philip L De Jager
- Center for Translational & Computational Neuroimmunology, Department of Neurology and the Taub Institute, Columbia University Medical Center, New York, NY 10032, USA; Cell Circuits Program, Broad Institute, Cambridge, MA 02142, USA
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL 60612, USA
| | - Benjamin R Arenkiel
- Jan and Dan Duncan Neurologic Research Institute, Texas Children's Hospital, Houston, TX 77030, USA; Program in Developmental Biology, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular & Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA
| | - Hugo J Bellen
- Jan and Dan Duncan Neurologic Research Institute, Texas Children's Hospital, Houston, TX 77030, USA; Program in Developmental Biology, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular & Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA; Howard Hughes Medical Institute, Baylor College of Medicine, Houston, TX 77030, USA
| | - Joshua M Shulman
- Department of Neurology, Baylor College of Medicine, Houston, TX 77030, USA; Jan and Dan Duncan Neurologic Research Institute, Texas Children's Hospital, Houston, TX 77030, USA; Program in Developmental Biology, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular & Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA.
| |
Collapse
|
39
|
Bihlmeyer NA, Merrill E, Lambert Y, Srivastava GP, Clark TW, Hyman BT, Das S. Novel methods for integration and visualization of genomics and genetics data in Alzheimer's disease. Alzheimers Dement 2019; 15:788-798. [PMID: 30935898 PMCID: PMC6664293 DOI: 10.1016/j.jalz.2019.01.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Revised: 12/12/2018] [Accepted: 01/18/2019] [Indexed: 12/31/2022]
Abstract
INTRODUCTION Numerous omics studies have been conducted to understand the molecular networks involved in Alzheimer's disease (AD), but the pathophysiology is still not completely understood; new approaches that enable neuroscientists to better interpret the results of omics analysis are required. METHODS We have developed advanced methods to analyze and visualize publicly-available genomics and genetics data. The tools include a composite clinical-neuropathological score for defining AD, gene expression maps in the brain, and networks integrating omics data to understand the impact of polymorphisms on AD pathways. RESULTS We have analyzed over 50 public human gene expression data sets, spanning 19 different brain regions and encompassing three separate cohorts. We integrated genome-wide association studies with expression data to identify important genes in the pathophysiology of AD, which provides further insight into the calcium signaling and calcineurin pathways. DISCUSSION Biologists can use these freely-available tools to obtain a comprehensive, information-rich view of the pathways in AD.
Collapse
Affiliation(s)
- Nathan A Bihlmeyer
- MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Emily Merrill
- MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Charlestown, MA, USA
| | - Yann Lambert
- Centre d'Investigation Clinique Antilles-Guyane, Cayenne Hospital, Cayenne Cedex, French Guiana, France
| | | | - Timothy W Clark
- Data Science Institute, School of Medicine, University of Virginia, Charlottesville, VA, USA
| | - Bradley T Hyman
- MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Sudeshna Das
- MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA.
| |
Collapse
|
40
|
Uhl GR, Martinez MJ. PTPRD: neurobiology, genetics, and initial pharmacology of a pleiotropic contributor to brain phenotypes. Ann N Y Acad Sci 2019; 1451:112-129. [PMID: 30648269 PMCID: PMC6629525 DOI: 10.1111/nyas.14002] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Revised: 11/12/2018] [Accepted: 12/19/2018] [Indexed: 12/12/2022]
Abstract
Receptor-type protein tyrosine phosphatase, receptor type D (PTPRD) has likely roles as a neuronal cell adhesion molecule and synaptic specifier. Interest in its neurobiology and genomics has been stimulated by results from human genetics and mouse models for phenotypes related to addiction, restless leg syndrome, neurofibrillary pathology in Alzheimer's disease, cognitive impairment/intellectual disability, mood lability, and obsessive-compulsive disorder. We review PTPRD's discovery, gene family, candidate homomeric and heteromeric binding partners, phosphatase activities, brain distribution, human genetic associations with nervous system phenotypes, and mouse model data relevant to these phenotypes. We discuss the recently reported discovery of the first small molecule inhibitor of PTPRD phosphatase, the identification of its addiction-related effects, and the implications of these findings for the PTPRD-associated brain phenotypes. In assembling PTPRD neurobiology, human genetics, and mouse genetic and pharmacological datasets, we provide a compelling picture of the roles played by PTPRD, its variation, and its potential as a target for novel therapeutics.
Collapse
Affiliation(s)
- George R Uhl
- Neurology and Research Services, New Mexico VA Healthcare System, Albuquerque, New Mexico.,Departments of Neurology, Neuroscience, Molecular Genetics and Microbiology, University of New Mexico, Albuquerque, New Mexico.,Biomedical Research Institute of New Mexico, Albuquerque, New Mexico.,Departments of Neurology, Neuroscience and Mental Health, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Maria J Martinez
- Neurology and Research Services, New Mexico VA Healthcare System, Albuquerque, New Mexico.,Biomedical Research Institute of New Mexico, Albuquerque, New Mexico
| |
Collapse
|
41
|
Cocaine reward is reduced by decreased expression of receptor-type protein tyrosine phosphatase D (PTPRD) and by a novel PTPRD antagonist. Proc Natl Acad Sci U S A 2018; 115:11597-11602. [PMID: 30348770 DOI: 10.1073/pnas.1720446115] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Receptor-type protein tyrosine phosphatase D (PTPRD) is a neuronal cell-adhesion molecule/synaptic specifier that has been implicated in addiction vulnerability and stimulant reward by human genomewide association and mouse cocaine-conditioned place-preference data. However, there have been no reports of effects of reduced expression on cocaine self-administration. There have been no reports of PTPRD targeting by any small molecule. There are no data about behavioral effects of any PTPRD ligand. We now report (i) robust effects of heterozygous PTPRD KO on cocaine self-administration (These data substantially extend prior conditioned place-preference data and add to the rationale for PTPRD as a target for addiction therapeutics.); (ii) identification of 7-butoxy illudalic acid analog (7-BIA) as a small molecule that targets PTPRD and inhibits its phosphatase with some specificity; (iii) lack of toxicity when 7-BIA is administered to mice acutely or with repeated dosing; (iv) reduced cocaine-conditioned place preference when 7-BIA is administered before conditioning sessions; and (v) reductions in well-established cocaine self-administration when 7-BIA is administered before a session (in WT, not PTPRD heterozygous KOs). These results add to support for PTPRD as a target for medications to combat cocaine use disorders. 7-BIA provides a lead compound for addiction therapeutics.
Collapse
|
42
|
Tasaki S, Gaiteri C, Mostafavi S, De Jager PL, Bennett DA. The Molecular and Neuropathological Consequences of Genetic Risk for Alzheimer's Dementia. Front Neurosci 2018; 12:699. [PMID: 30349450 PMCID: PMC6187226 DOI: 10.3389/fnins.2018.00699] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 09/18/2018] [Indexed: 12/12/2022] Open
Abstract
Alzheimer's dementia commonly impacts the health of older adults and lacks any preventative therapy. While Alzheimer's dementia risk has a substantial genetic component, the specific molecular mechanisms and neuropathologies triggered by most of the known genetic variants are unclear. Resultantly, they have shown limited influence on drug development portfolios to date. To facilitate our understanding of the consequences of Alzheimer's dementia susceptibility variants, we examined their relationship to a wide range of clinical, molecular and neuropathological features. Because the effect size of individual variants is typically small, we utilized a polygenic (overall) risk approach to identify the global impact of Alzheimer's dementia susceptibility variants. Under this approach, each individual has a polygenic risk score (PRS) that we related to clinical, molecular and neuropathological phenotypes. Applying this approach to 1,272 individuals who came to autopsy from one of two longitudinal aging cohorts, we observed that an individual's PRS was associated with cognitive decline and brain pathologies including beta-amyloid, tau-tangles, hippocampal sclerosis, and TDP-43, MIR132, four proteins including VGF, IGFBP5, and STX1A, and many chromosomal regions decorated with acetylation on histone H3 lysine 9 (H3K9Ac). While excluding the APOE/TOMM40 region (containing the single largest genetic risk factor for late-onset Alzheimer's dementia) in the calculation of the PRS resulted in a slightly weaker association with the molecular signatures, results remained significant. These PRS-associated brain pathologies and molecular signatures appear to mediate genetic risk, as they attenuated the association of the PRS with cognitive decline. Notably, the PRS induced changes in H3K9Ac throughout the genome, implicating it in large-scale chromatin changes. Thus, the PRS for Alzheimer's dementia (AD-PRS) showed effects on diverse clinical, molecular, and pathological systems, ranging from the epigenome to specific proteins. These convergent targets of a large number of genetic risk factors for Alzheimer's dementia will help define the experimental systems and models needed to test therapeutic targets, which are expected to be broadly effective in the aging population that carries diverse genetic risks for Alzheimer's dementia.
Collapse
Affiliation(s)
- Shinya Tasaki
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, United States
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, United States
| | - Chris Gaiteri
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, United States
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, United States
| | - Sara Mostafavi
- Department of Statistics, Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| | - Philip L. De Jager
- Department of Neurology, Center for Translational and Computational Neuroimmunology, Columbia University Medical Center, New York, NY, United States
- Cell Circuits Program, Broad Institute, Cambridge, MA, United States
| | - David A. Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, United States
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, United States
| |
Collapse
|
43
|
Felsky D, Patrick E, Schneider JA, Mostafavi S, Gaiteri C, Patsopoulos N, Bennett DA, De Jager PL. Polygenic analysis of inflammatory disease variants and effects on microglia in the aging brain. Mol Neurodegener 2018; 13:38. [PMID: 30041668 PMCID: PMC6057096 DOI: 10.1186/s13024-018-0272-6] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Accepted: 07/13/2018] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND The role of the innate immune system in Alzheimer's disease (AD) and neurodegenerative disease susceptibility has recently been highlighted in genetic studies. However, we do not know whether risk for inflammatory disease predisposes unaffected individuals to late-life cognitive deficits or AD-related neuropathology. We investigated whether genetic risk scores for seven immune diseases and central nervous system traits were related to cognitive decline (nmax = 1601), classical AD neuropathology (nmax = 985), or microglial density (nmax = 184). METHODS Longitudinal cognitive decline, postmortem amyloid and tau neuropathology, microglial density, and gene module expression from bulk brain tissue were all measured in participants from two large cohorts (the Rush Religious Orders Study and Memory and Aging Project; ROS/MAP) of elderly subjects (mean age at entry 78 +/- 8.7 years). We analyzed data primarily using robust regression methods. Neuropathologists were blind to clinical data. RESULTS The AD genetic risk scores, including and excluding APOE effects, were strongly associated with cognitive decline in all domains (min Puncor = 3.2 × 10- 29). Multiple sclerosis (MS), Parkinson's disease, and schizophrenia risk did not influence cognitive decline in older age, but the rheumatoid arthritis (RA) risk score alone was significantly associated with microglial density after correction (t146 = - 3.88, Puncor = 1.6 × 10- 4). Post-hoc tests found significant effects of the RA genetic risk score in multiple regions and stages of microglial activation (min Puncor = 1.5 × 10- 6). However, these associations were driven by only one or two variants, rather than cumulative polygenicity. Further, individual MS (Pone-sided < 8.4 × 10- 4) and RA (Pone-sided = 3 × 10- 4) variants associated with higher microglial density were also associated with increased expression of brain immune gene modules. CONCLUSIONS Our results demonstrate that global risk of inflammatory disease does not strongly influence aging-related cognitive decline but that susceptibility variants that influence peripheral immune function also alter microglial density and immune gene expression in the aging brain, opening a new perspective on the control of microglial and immune responses within the central nervous system. Further study on the molecular mechanisms of peripheral immune disease risk influencing glial cell activation will be required to identify key regulators of these pathways.
Collapse
Affiliation(s)
- Daniel Felsky
- Center for Translational and Computational Neuroimmunology, Department of Neurology, Columbia University Medical Center, 630 West 168th Street, PH 19 – 302, New York, NY 10032 USA
- Department of Neurology, Brigham and Woman’s Hospital, 75 Francis Street, Boston, MA 02115 USA
- Department of Neurology, Harvard Medical School, 25 Shattuck Street, Boston, MA 02115 USA
- Program in Population and Medical Genetics, Broad Institute of MIT and Harvard, 415 Main St, Cambridge, MA 02142 USA
| | - Ellis Patrick
- Department of Statistics, University of Sydney, Camperdown, NSW 2006 Australia
| | - Julie A. Schneider
- Department of Neurology, Rush University Medical Center, 1653 West Congress Parkway, Chicago, IL 60612 USA
- Rush Alzheimer’s Disease Center, Rush University Medical Center, 600 South Paulina Street, Chicago, IL 60612 USA
| | - Sara Mostafavi
- Department of Statistics, University of British Columbia, 2329 West Mall, Vancouver, BC V6T 1Z4 Canada
| | - Chris Gaiteri
- Department of Neurology, Rush University Medical Center, 1653 West Congress Parkway, Chicago, IL 60612 USA
- Rush Alzheimer’s Disease Center, Rush University Medical Center, 600 South Paulina Street, Chicago, IL 60612 USA
| | - Nikolaos Patsopoulos
- Department of Neurology, Brigham and Woman’s Hospital, 75 Francis Street, Boston, MA 02115 USA
- Department of Neurology, Harvard Medical School, 25 Shattuck Street, Boston, MA 02115 USA
| | - David A. Bennett
- Department of Neurology, Rush University Medical Center, 1653 West Congress Parkway, Chicago, IL 60612 USA
- Rush Alzheimer’s Disease Center, Rush University Medical Center, 600 South Paulina Street, Chicago, IL 60612 USA
| | - Philip L. De Jager
- Center for Translational and Computational Neuroimmunology, Department of Neurology, Columbia University Medical Center, 630 West 168th Street, PH 19 – 302, New York, NY 10032 USA
- Department of Neurology, Brigham and Woman’s Hospital, 75 Francis Street, Boston, MA 02115 USA
- Department of Neurology, Harvard Medical School, 25 Shattuck Street, Boston, MA 02115 USA
- Program in Population and Medical Genetics, Broad Institute of MIT and Harvard, 415 Main St, Cambridge, MA 02142 USA
| |
Collapse
|
44
|
Bennett DA, Buchman AS, Boyle PA, Barnes LL, Wilson RS, Schneider JA. Religious Orders Study and Rush Memory and Aging Project. J Alzheimers Dis 2018; 64:S161-S189. [PMID: 29865057 PMCID: PMC6380522 DOI: 10.3233/jad-179939] [Citation(s) in RCA: 801] [Impact Index Per Article: 114.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND The Religious Orders Study and Rush Memory and Aging Project are both ongoing longitudinal clinical-pathologic cohort studies of aging and Alzheimer's disease (AD). OBJECTIVES To summarize progress over the past five years and its implications for understanding neurodegenerative diseases. METHODS Participants in both studies are older adults who enroll without dementia and agree to detailed longitudinal clinical evaluations and organ donation. The last review summarized findings through the end of 2011. Here we summarize progress and study findings over the past five years and discuss new directions for how these studies can inform on aging and AD in the future. RESULTS We summarize 1) findings on the relation of neurobiology to clinical AD; 2) neurobiologic pathways linking risk factors to clinical AD; 3) non-cognitive AD phenotypes including motor function and decision making; 4) the development of a novel drug discovery platform. CONCLUSION Complexity at multiple levels needs to be understood and overcome to develop effective treatments and preventions for cognitive decline and AD dementia.
Collapse
Affiliation(s)
- David A. Bennett
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL., USA
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL., USA
| | - Aron S. Buchman
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL., USA
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL., USA
| | - Patricia A. Boyle
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL., USA
- Department of Behavioral Sciences, Rush University Medical Center, Chicago, IL., USA
| | - Lisa L. Barnes
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL., USA
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL., USA
- Department of Behavioral Sciences, Rush University Medical Center, Chicago, IL., USA
| | - Robert S. Wilson
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL., USA
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL., USA
- Department of Behavioral Sciences, Rush University Medical Center, Chicago, IL., USA
| | - Julie A Schneider
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL., USA
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL., USA
- Department of Pathology (Neuropathology), Rush University Medical Center, Chicago, IL., USA
| |
Collapse
|