1
|
Singh S, Khan S, Shahid M, Sardar M, Hassan MI, Islam A. Targeting tau in Alzheimer's and beyond: Insights into pathology and therapeutic strategies. Ageing Res Rev 2025; 104:102639. [PMID: 39674375 DOI: 10.1016/j.arr.2024.102639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 12/04/2024] [Accepted: 12/08/2024] [Indexed: 12/16/2024]
Abstract
Tauopathies encompass a group of approximately 20 neurodegenerative diseases characterized by the accumulation of the microtubule-associated protein tau in brain neurons. The pathogenesis of intracellular neurofibrillary tangles, a hallmark of tauopathies, is initiated by hyperphosphorylated tau protein isoforms that cause neuronal death and lead to diseases like Alzheimer's, Parkinson's disease, frontotemporal dementia, and other complex neurodegenerative diseases. Current applications of tau biomarkers, including imaging, cerebrospinal fluid, and blood-based assays, assist in the evaluation and diagnosis of tauopathies. Emerging research is providing various potential strategies to prevent cellular toxicity caused by tau aggregation such as: 1) suppressing toxic tau aggregation, 2) preventing post-translational modifications of tau, 3) stabilizing microtubules and 4) designing tau-directed immunogens. This review aims to discuss the role of tau in tauopathies along with neuropathological features of the different tauopathies and the new developments in treating tau aggregation with the therapeutics for treating and possibly preventing tauopathies.
Collapse
Affiliation(s)
- Sunidhi Singh
- Department of Biosciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India
| | - Sumaiya Khan
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India
| | - Mohammad Shahid
- Department of Basic Medical Sciences, College of Medicine, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
| | - Meryam Sardar
- Department of Biosciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India
| | - Md Imtaiyaz Hassan
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India
| | - Asimul Islam
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India.
| |
Collapse
|
2
|
Ferrazzoli D, Ortelli P, Versace V, Stolz J, Dezi S, Vos P, Giladi N, Saltuari L, Sebastianelli L. Post-traumatic parkinsonism: The intricate twist between trauma, inflammation and neurodegeneration. A narrative review. J Neurol Sci 2024; 466:123242. [PMID: 39303348 DOI: 10.1016/j.jns.2024.123242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 09/10/2024] [Accepted: 09/15/2024] [Indexed: 09/22/2024]
Abstract
Post-traumatic Parkinsonism (PTP) is a complex neurological disorder that is often associated with the occurrence of a traumatic brain injury (TBI). PTP can occur either in the acute or chronic phase of TBI. There is still uncertainty about the mechanisms provoking PTP, which can be the result of the acute blast itself or secondary neurodegenerative process occurring months to years post the acute trauma. Currently there is an underestimation of the clinical importance of PTP and lack of specific and proven therapeutic interventions, both in the pharmacological and the neurorehabilitation field. This narrative review aims to summarize the actual knowledge about PTP in terms of its pathophysiology, clinical aspects, treatments and perspective of care in the neurorehabilitative setting.
Collapse
Affiliation(s)
- Davide Ferrazzoli
- Department of Neurorehabilitation, Hospital of Vipiteno (SABES-ASDAA), Teaching Hospital of the Paracelsus Medical Private University (PMU), Vipiteno-Sterzing, Italy.
| | - Paola Ortelli
- Department of Neurorehabilitation, Hospital of Vipiteno (SABES-ASDAA), Teaching Hospital of the Paracelsus Medical Private University (PMU), Vipiteno-Sterzing, Italy
| | - Viviana Versace
- Department of Neurorehabilitation, Hospital of Vipiteno (SABES-ASDAA), Teaching Hospital of the Paracelsus Medical Private University (PMU), Vipiteno-Sterzing, Italy; Department of Neurology, Neurocritical Care and Neurorehabilitation, Christian Doppler University Hospital, Centre for Cognitive Neuroscience, Paracelsus Medical University (PMU), Salzburg, Austria
| | - Jakob Stolz
- Department of Neurorehabilitation, Hospital of Vipiteno (SABES-ASDAA), Teaching Hospital of the Paracelsus Medical Private University (PMU), Vipiteno-Sterzing, Italy
| | - Sabrina Dezi
- Department of Neurorehabilitation, Hospital of Vipiteno (SABES-ASDAA), Teaching Hospital of the Paracelsus Medical Private University (PMU), Vipiteno-Sterzing, Italy
| | - Pieter Vos
- Department of Neurology, Slingeland Hospital, Doetinchem, the Netherlands
| | - Nir Giladi
- Center for the Study of Movement, Cognition and Mobility, Neurological Institute, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel; Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel; Department of Neurology, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Leopold Saltuari
- Department of Neurorehabilitation, Hospital of Vipiteno (SABES-ASDAA), Teaching Hospital of the Paracelsus Medical Private University (PMU), Vipiteno-Sterzing, Italy
| | - Luca Sebastianelli
- Department of Neurorehabilitation, Hospital of Vipiteno (SABES-ASDAA), Teaching Hospital of the Paracelsus Medical Private University (PMU), Vipiteno-Sterzing, Italy
| |
Collapse
|
3
|
Arienti F, Casazza G, Franco G, Lazzeri G, Monfrini E, Di Maio A, Erro R, Barone P, Tamma F, Caputo E, Volontè MA, Cacciaguerra L, Pilotto A, Padovani A, Comi C, Magistrelli L, Valzania F, Cavallieri F, Avanzino L, Marchese R, Sensi M, Carroli G, Eleopra R, Cilia R, Spagnolo F, Tessitore A, De Micco R, Ceravolo R, Palermo G, Malaguti MC, Lopiano L, Tocco P, Sorbera C, Tinazzi M, Ciammola A, Ottaviani D, Valente EM, Albanese A, Blandini F, Canesi M, Antonini A, Carecchio M, Fetoni V, Colosimo C, Volpe D, Tambasco N, Cossu G, Zappia M, Di Fonzo A. Family History in Parkinson's Disease: A National Cross-Sectional Study. Mov Disord Clin Pract 2024; 11:1434-1440. [PMID: 39269187 PMCID: PMC11542285 DOI: 10.1002/mdc3.14206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 07/24/2024] [Accepted: 08/17/2024] [Indexed: 09/15/2024] Open
Abstract
BACKGROUND Family history of Parkinson's disease (PD) is a common finding in PD patients. However, a few studies have systematically examined this aspect. OBJECTIVES We investigated the family history of PD patients, comparing demographic and clinical features between familial PD (fPD) and sporadic PD (sPD). METHODS A cross-sectional study enrolling 2035 PD patients was conducted in 28 Italian centers. Clinical data and family history up to the third degree of kinship were collected. RESULTS Family history of PD was determined in 21.9% of patients. fPD patients had earlier age at onset than sporadic patients. No relevant differences in the prevalence of motor and nonmotor symptoms were detected. Family history of mood disorders resulted more prevalently in the fPD group. CONCLUSIONS fPD was found to recur more frequently than previously reported. Family history collection beyond the core family is essential to discover disease clusters and identify novel risk factors for PD.
Collapse
Affiliation(s)
- Federica Arienti
- Foundation IRCCS Ca'Granda Ospedale Maggiore Policlinico, Neurology UnitMilanItaly
- Dino Ferrari Center, Neuroscience Section, Department of Pathophysiology and TransplantationUniversity of MilanMilanItaly
| | - Giovanni Casazza
- Department of Clinical Sciences and Community HealthUniversità degli Studi di MilanoMilanItaly
- Fondazione IRCCS Ca' Granda Ospedale Maggiore PoliclinicoMilanItaly
| | - Giulia Franco
- Foundation IRCCS Ca'Granda Ospedale Maggiore Policlinico, Neurology UnitMilanItaly
- Dino Ferrari Center, Neuroscience Section, Department of Pathophysiology and TransplantationUniversity of MilanMilanItaly
| | - Giulia Lazzeri
- Dino Ferrari Center, Neuroscience Section, Department of Pathophysiology and TransplantationUniversity of MilanMilanItaly
- Gaetano Pini‐CTO, Parkinson InstituteMilanItaly
| | - Edoardo Monfrini
- Foundation IRCCS Ca'Granda Ospedale Maggiore Policlinico, Neurology UnitMilanItaly
- Dino Ferrari Center, Neuroscience Section, Department of Pathophysiology and TransplantationUniversity of MilanMilanItaly
| | - Alessandro Di Maio
- Foundation IRCCS Ca'Granda Ospedale Maggiore Policlinico, Neurology UnitMilanItaly
- Dino Ferrari Center, Neuroscience Section, Department of Pathophysiology and TransplantationUniversity of MilanMilanItaly
| | - Roberto Erro
- Neuroscience Section, Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana,"University of SalernoBaronissi (SA)Italy
| | - Paolo Barone
- Neuroscience Section, Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana,"University of SalernoBaronissi (SA)Italy
| | - Filippo Tamma
- Department of Neurology"F. Miulli" General HospitalAcquaviva delle FontiItaly
| | - Elena Caputo
- Department of Neurology"F. Miulli" General HospitalAcquaviva delle FontiItaly
| | | | - Laura Cacciaguerra
- Department of NeurologyIRCCS San Raffaele Scientific InstituteMilanItaly
| | - Andrea Pilotto
- Neurology Unit and Neurobiorepository and Laboratory of Advanced Biological Markers, Department of Clinical and Experimental Sciences, University of Brescia, and Department of Continuity of Care and FrailtyASST Spedali Civili Brescia HospitalBresciaItaly
| | - Alessandro Padovani
- Neurology Unit and Neurobiorepository and Laboratory of Advanced Biological Markers, Department of Clinical and Experimental Sciences, University of Brescia, and Department of Continuity of Care and FrailtyASST Spedali Civili Brescia HospitalBresciaItaly
| | - Cristoforo Comi
- Department of Translational Medicine, Section of NeurologyUniversity of Piemonte OrientaleNovaraItaly
| | | | - Franco Valzania
- Neurology Unit, Neuromotor and Rehabilitation DepartmentAzienda USL‐IRCCS di Reggio EmiliaReggio EmiliaItaly
| | - Francesco Cavallieri
- Neurology Unit, Neuromotor and Rehabilitation DepartmentAzienda USL‐IRCCS di Reggio EmiliaReggio EmiliaItaly
| | - Laura Avanzino
- Department of Experimental Medicine, Section of Human PhysiologyUniversity of GenoaGenoaItaly
- IRCCS Ospedale Policlinico San Martino—UOC Genetica MedicaGenoaItaly
| | - Roberta Marchese
- IRCCS Ospedale Policlinico San Martino—UOC Genetica MedicaGenoaItaly
| | - Mariachiara Sensi
- Department of Neuroscience and RehabilitationAzienda Ospedaliera‐Universitaria S. AnnaFerraraItaly
| | - Giorgia Carroli
- Department of Neuroscience and RehabilitationAzienda Ospedaliera‐Universitaria S. AnnaFerraraItaly
| | - Roberto Eleopra
- Fondazione IRCCS Istituto Neurologico Carlo Besta, Department of Clinical NeurosciencesMovement Disorders UnitMilanItaly
| | - Roberto Cilia
- Fondazione IRCCS Istituto Neurologico Carlo Besta, Department of Clinical NeurosciencesMovement Disorders UnitMilanItaly
| | | | - Alessandro Tessitore
- Department of Advanced Medical and Surgical SciencesUniversity of Campania “Luigi Vanvitelli”CasertaItaly
| | - Rosa De Micco
- Department of Advanced Medical and Surgical SciencesUniversity of Campania “Luigi Vanvitelli”CasertaItaly
| | - Roberto Ceravolo
- Unit of Neurology, Department of Clinical and Experimental MedicineCenter for Neurodegenerative Diseases—Parkinson's Disease and Movement DisordersUniversity of PisaPisaItaly
| | - Giovanni Palermo
- Unit of Neurology, Department of Clinical and Experimental MedicineCenter for Neurodegenerative Diseases—Parkinson's Disease and Movement DisordersUniversity of PisaPisaItaly
| | - Maria Chiara Malaguti
- Neurology Unit, Trento HospitalAzienda Provinciale per i Servizi Sanitari (APSS) di TrentoTrentoItaly
| | - Leonardo Lopiano
- Department of Neuroscience "Rita Levi Montalcini"University of TurinTurinItaly
- Neurology 2 Unit, A.O.U. Città della Salute e della Scienza di TorinoTurinItaly
| | | | - Chiara Sorbera
- Neurorehabilitation Unit IRCCS Centro Neurolesi "Bonino Pulejo," MessinaMessinaItaly
| | - Michele Tinazzi
- Department of Neurosciences, Biomedicine and Movement SciencesUniversity of VeronaVeronaItaly
| | - Andrea Ciammola
- Department of Neurology and Laboratory of NeuroscienceIstituto Auxologico Italiano IRCCSMilanItaly
| | - Donatella Ottaviani
- Neurology Unit, Rovereto HospitalAzienda Provinciale per i Servizi Sanitari (APSS) di TrentoTrentoItaly
| | - Enza Maria Valente
- Department of Molecular MedicineUniversity of PaviaPaviaItaly
- Neurogenetics Research CenterIRCCS Mondino FoundationPaviaItaly
| | - Alberto Albanese
- Department of NeurologyIRCCS Humanitas Research HospitalRozzanoItaly
| | - Fabio Blandini
- Fondazione IRCCS Ca' Granda Ospedale Maggiore PoliclinicoMilanItaly
| | - Margherita Canesi
- Department of Parkinson's DiseaseMovement Disorders and Brain Injury Rehabilitation, Moriggia Pelascini HospitalGravedonaItaly
| | - Angelo Antonini
- Parkinson and Movement Disorders Unit, Center for Neurodegenerative Diseases (CENSE), Department of NeuroscienceUniversity of PaduaPaduaItaly
| | - Miryam Carecchio
- Parkinson and Movement Disorders Unit, Center for Neurodegenerative Diseases (CENSE), Department of NeuroscienceUniversity of PaduaPaduaItaly
| | | | | | - Daniele Volpe
- Department of NeurorehabilitationParkinson's Disease Excellence Center, Casa di Cura Villa Margherita via Costacolonna n 1 ArcugnanoVicenzaItaly
| | - Nicola Tambasco
- Movement Disorders Center, Neurology DepartmentPerugia General Hospital and University of PerugiaPerugiaItaly
| | | | - Mario Zappia
- Department GF IngrassiaUniversity of CataniaCataniaItaly
| | | | - Alessio Di Fonzo
- Foundation IRCCS Ca'Granda Ospedale Maggiore Policlinico, Neurology UnitMilanItaly
| |
Collapse
|
4
|
Li W, Wu H, Li J, Wang Z, Cai M, Liu X, Liu G. Transcriptomic analysis reveals associations of blood-based A-to-I editing with Parkinson's disease. J Neurol 2024; 271:976-985. [PMID: 37902879 DOI: 10.1007/s00415-023-12053-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 10/07/2023] [Accepted: 10/09/2023] [Indexed: 11/01/2023]
Abstract
BACKGROUND Adenosine-to-inosine (A-to-I) editing is the most common type of RNA editing in humans and the role of A-to-I RNA editing remains unclear in Parkinson's disease (PD). OBJECTIVE We aimed to explore the potential causal association between A-to-I editing and PD, and to assess whether changes in A-to-I editing were associated with cognitive progression in PD. METHODS The RNA-seq data from 380 PD patients and 178 healthy controls in the Parkinson's Progression Marker Initiative cohort was used to quantify A-to-I editing sites. We performed cis-RNA editing quantitative trait loci analysis and a two-sample Mendelian Randomization (MR) study by integrating genome-wide association studies to infer the potential causality between A-to-I editing and PD pathogenesis. The potential causal A-to-I editing sites were further confirmed by Summary-data-based MR analysis. Spearman's correlation analysis was performed to characterize the association between longitudinal A-to-I editing and cognitive progression in patients with PD. RESULTS We identified 17 potential causal A-to-I editing sites for PD and indicated that genetic risk variants may contribute to the risk of PD through A-to-I editing. These A-to-I editing sites were located in genes NCOR1, KANSL1 and BST1. Moreover, we observed 57 sites whose longitudinal A-to-I editing levels correlated with cognitive progression in PD. CONCLUSIONS We found potential causal A-to-I editing sites for PD onset and longitudinal changes of A-to-I editing were associated with cognitive progression in PD. We anticipate this study will provide new biological insights and drive the discovery of the epitranscriptomic role underlying Parkinson's disease.
Collapse
Affiliation(s)
- Weimin Li
- Shenzhen Key Laboratory of Systems Medicine in Inflammatory Diseases, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, No.66, Gongchang Road, Guangming District, Shenzhen, 518107, Guangdong, People's Republic of China
- Neurobiology Research Center, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, No.66, Gongchang Road, Guangming District, Shenzhen, 518107, Guangdong, People's Republic of China
| | - Hao Wu
- Shenzhen Key Laboratory of Systems Medicine in Inflammatory Diseases, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, No.66, Gongchang Road, Guangming District, Shenzhen, 518107, Guangdong, People's Republic of China
- Neurobiology Research Center, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, No.66, Gongchang Road, Guangming District, Shenzhen, 518107, Guangdong, People's Republic of China
| | - Jinxia Li
- Shenzhen Key Laboratory of Systems Medicine in Inflammatory Diseases, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, No.66, Gongchang Road, Guangming District, Shenzhen, 518107, Guangdong, People's Republic of China
- Neurobiology Research Center, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, No.66, Gongchang Road, Guangming District, Shenzhen, 518107, Guangdong, People's Republic of China
| | - Zhuo Wang
- Warshel Institute for Computational Biology, School of Medicine, The Chinese University of Hong Kong, Shenzhen, 518172, Guangdong, People's Republic of China
| | - Miao Cai
- Neurology Department, Zhejiang Hospital, Hangzhou, 310013, People's Republic of China
| | - Xiaoli Liu
- Neurology Department, Zhejiang Hospital, Hangzhou, 310013, People's Republic of China
| | - Ganqiang Liu
- Shenzhen Key Laboratory of Systems Medicine in Inflammatory Diseases, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, No.66, Gongchang Road, Guangming District, Shenzhen, 518107, Guangdong, People's Republic of China.
- Neurobiology Research Center, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, No.66, Gongchang Road, Guangming District, Shenzhen, 518107, Guangdong, People's Republic of China.
| |
Collapse
|
5
|
Alvarado CX, Makarious MB, Weller CA, Vitale D, Koretsky MJ, Bandres-Ciga S, Iwaki H, Levine K, Singleton A, Faghri F, Nalls MA, Leonard HL. omicSynth: An open multi-omic community resource for identifying druggable targets across neurodegenerative diseases. Am J Hum Genet 2024; 111:150-164. [PMID: 38181731 PMCID: PMC10806756 DOI: 10.1016/j.ajhg.2023.12.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 12/04/2023] [Accepted: 12/04/2023] [Indexed: 01/07/2024] Open
Abstract
Treatments for neurodegenerative disorders remain rare, but recent FDA approvals, such as lecanemab and aducanumab for Alzheimer disease (MIM: 607822), highlight the importance of the underlying biological mechanisms in driving discovery and creating disease modifying therapies. The global population is aging, driving an urgent need for therapeutics that stop disease progression and eliminate symptoms. In this study, we create an open framework and resource for evidence-based identification of therapeutic targets for neurodegenerative disease. We use summary-data-based Mendelian randomization to identify genetic targets for drug discovery and repurposing. In parallel, we provide mechanistic insights into disease processes and potential network-level consequences of gene-based therapeutics. We identify 116 Alzheimer disease, 3 amyotrophic lateral sclerosis (MIM: 105400), 5 Lewy body dementia (MIM: 127750), 46 Parkinson disease (MIM: 605909), and 9 progressive supranuclear palsy (MIM: 601104) target genes passing multiple test corrections (pSMR_multi < 2.95 × 10-6 and pHEIDI > 0.01). We created a therapeutic scheme to classify our identified target genes into strata based on druggability and approved therapeutics, classifying 41 novel targets, 3 known targets, and 115 difficult targets (of these, 69.8% are expressed in the disease-relevant cell type from single-nucleus experiments). Our novel class of genes provides a springboard for new opportunities in drug discovery, development, and repurposing in the pre-competitive space. In addition, looking at drug-gene interaction networks, we identify previous trials that may require further follow-up such as riluzole in Alzheimer disease. We also provide a user-friendly web platform to help users explore potential therapeutic targets for neurodegenerative diseases, decreasing activation energy for the community.
Collapse
Affiliation(s)
- Chelsea X Alvarado
- Center for Alzheimer's and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20814, USA; Data Tecnica LLC, Washington, DC 20037, USA
| | - Mary B Makarious
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD 20814, USA; Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London WC1N 3BG, UK; UCL Movement Disorders Centre, University College London, London WC1N 3BG, UK
| | - Cory A Weller
- Center for Alzheimer's and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20814, USA; Data Tecnica LLC, Washington, DC 20037, USA
| | - Dan Vitale
- Center for Alzheimer's and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20814, USA; Data Tecnica LLC, Washington, DC 20037, USA
| | - Mathew J Koretsky
- Center for Alzheimer's and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20814, USA
| | - Sara Bandres-Ciga
- Center for Alzheimer's and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20814, USA
| | - Hirotaka Iwaki
- Center for Alzheimer's and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20814, USA; Data Tecnica LLC, Washington, DC 20037, USA; Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD 20814, USA
| | - Kristin Levine
- Center for Alzheimer's and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20814, USA; Data Tecnica LLC, Washington, DC 20037, USA
| | - Andrew Singleton
- Center for Alzheimer's and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20814, USA; Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD 20814, USA
| | - Faraz Faghri
- Center for Alzheimer's and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20814, USA; Data Tecnica LLC, Washington, DC 20037, USA; Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD 20814, USA
| | - Mike A Nalls
- Center for Alzheimer's and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20814, USA; Data Tecnica LLC, Washington, DC 20037, USA; Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD 20814, USA
| | - Hampton L Leonard
- Center for Alzheimer's and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20814, USA; Data Tecnica LLC, Washington, DC 20037, USA; Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD 20814, USA; German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany.
| |
Collapse
|
6
|
Subramaniyan S, Kuriakose BB, Mushfiq S, Prabhu NM, Muthusamy K. Gene Signals and SNPs Associated with Parkinson's Disease: A Nutrigenomics and Computational Prospective Insights. Neuroscience 2023; 533:77-95. [PMID: 37858629 DOI: 10.1016/j.neuroscience.2023.10.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 05/05/2023] [Accepted: 10/11/2023] [Indexed: 10/21/2023]
Abstract
Parkinson's disease is the most prevalent chronic neurodegenerative disease. Neurological conditions for PD were influenced by a variety of epigenetic factors and SNPs in some of the coexisting genes that were expressed. This article focused on nutrigenomics of PD and the prospective highlighting of how these genes are regulated in terms of nutritive factors and the genetic basis of PD risk, onset, and progression. Multigenetic associations of the following genetic alterations in the genes of SNCA, LRRK2, UCHL1, PARK2,PINK1, DJ-1, and ATP13A2 have been reported with the familial and de novo genetic origins of PD. Over the past two decades, significant attempts have been made to understand the biological mechanisms that are potential causes for this disease, as well as to identify therapeutic substances for the prevention and management of PD. Nutrigenomics has sparked considerable interest due to its nutritional, safe, and therapeutic effects on a variety of chronic diseases. In this study, we summarise some of the nutritive supplements that have an impact on PD.
Collapse
Affiliation(s)
- Swetha Subramaniyan
- Department of Bioinformatics, Alagappa University, Karaikudi, Tamil Nadu, India
| | - Beena Briget Kuriakose
- Department of Basic Medical Sciences, College of Applied Medical Sciences, King Khalid University, Khamis Mushayt, Saudi Arabia
| | - Sakeena Mushfiq
- Department of Public Health, College of Applied Medical Sciences, King Khalid University, Khamis Mushayt, Saudi Arabia
| | | | | |
Collapse
|
7
|
Alvarado CX, Makarious MB, Weller CA, Vitale D, Koretsky MJ, Bandres-Ciga S, Iwaki H, Levine K, Singleton A, Faghri F, Nalls MA, Leonard HL. omicSynth: an Open Multi-omic Community Resource for Identifying Druggable Targets across Neurodegenerative Diseases. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.04.06.23288266. [PMID: 37090611 PMCID: PMC10120805 DOI: 10.1101/2023.04.06.23288266] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/25/2023]
Abstract
Treatments for neurodegenerative disorders remain rare, although recent FDA approvals, such as Lecanemab and Aducanumab for Alzheimer's Disease, highlight the importance of the underlying biological mechanisms in driving discovery and creating disease modifying therapies. The global population is aging, driving an urgent need for therapeutics that stop disease progression and eliminate symptoms. In this study, we create an open framework and resource for evidence-based identification of therapeutic targets for neurodegenerative disease. We use Summary-data-based Mendelian Randomization to identify genetic targets for drug discovery and repurposing. In parallel, we provide mechanistic insights into disease processes and potential network-level consequences of gene-based therapeutics. We identify 116 Alzheimer's disease, 3 amyotrophic lateral sclerosis, 5 Lewy body dementia, 46 Parkinson's disease, and 9 Progressive supranuclear palsy target genes passing multiple test corrections (pSMR_multi < 2.95×10-6 and pHEIDI > 0.01). We created a therapeutic scheme to classify our identified target genes into strata based on druggability and approved therapeutics - classifying 41 novel targets, 3 known targets, and 115 difficult targets (of these 69.8% are expressed in the disease relevant cell type from single nucleus experiments). Our novel class of genes provides a springboard for new opportunities in drug discovery, development and repurposing in the pre-competitive space. In addition, looking at drug-gene interaction networks, we identify previous trials that may require further follow-up such as Riluzole in AD. We also provide a user-friendly web platform to help users explore potential therapeutic targets for neurodegenerative diseases, decreasing activation energy for the community [https://nih-card-ndd-smr-home-syboky.streamlit.app/].
Collapse
Affiliation(s)
- Chelsea X. Alvarado
- Center for Alzheimer’s and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA, 20814
- Data Tecnica International, Washington, DC, USA, 20037
| | - Mary B. Makarious
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA, 20814
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, UK, WC1N 3BG
- UCL Movement Disorders Centre, University College London, London, UK, WC1N 3BG
| | - Cory A. Weller
- Center for Alzheimer’s and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA, 20814
- Data Tecnica International, Washington, DC, USA, 20037
| | - Dan Vitale
- Center for Alzheimer’s and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA, 20814
- Data Tecnica International, Washington, DC, USA, 20037
| | - Mathew J. Koretsky
- Center for Alzheimer’s and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA, 20814
| | - Sara Bandres-Ciga
- Center for Alzheimer’s and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA, 20814
| | - Hirotaka Iwaki
- Center for Alzheimer’s and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA, 20814
- Data Tecnica International, Washington, DC, USA, 20037
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA, 20814
| | - Kristin Levine
- Center for Alzheimer’s and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA, 20814
- Data Tecnica International, Washington, DC, USA, 20037
| | - Andrew Singleton
- Center for Alzheimer’s and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA, 20814
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA, 20814
| | - Faraz Faghri
- Center for Alzheimer’s and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA, 20814
- Data Tecnica International, Washington, DC, USA, 20037
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA, 20814
| | - Mike A. Nalls
- Center for Alzheimer’s and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA, 20814
- Data Tecnica International, Washington, DC, USA, 20037
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA, 20814
| | - Hampton L. Leonard
- Center for Alzheimer’s and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA, 20814
- Data Tecnica International, Washington, DC, USA, 20037
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA, 20814
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
| |
Collapse
|
8
|
Tauber CV, Schwarz SC, Rösler TW, Arzberger T, Gentleman S, Windl O, Krumbiegel M, Reis A, Ruf VC, Herms J, Höglinger GU. Different MAPT haplotypes influence expression of total MAPT in postmortem brain tissue. Acta Neuropathol Commun 2023; 11:40. [PMID: 36906636 PMCID: PMC10008602 DOI: 10.1186/s40478-023-01534-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Accepted: 02/21/2023] [Indexed: 03/13/2023] Open
Abstract
The MAPT gene, encoding the microtubule-associated protein tau on chromosome 17q21.31, is result of an inversion polymorphism, leading to two allelic variants (H1 and H2). Homozygosity for the more common haplotype H1 is associated with an increased risk for several tauopathies, but also for the synucleinopathy Parkinson's disease (PD). In the present study, we aimed to clarify whether the MAPT haplotype influences expression of MAPT and SNCA, encoding the protein α-synuclein (α-syn), on mRNA and protein levels in postmortem brains of PD patients and controls. We also investigated mRNA expression of several other MAPT haplotype-encoded genes. Postmortem tissues from cortex of fusiform gyrus (ctx-fg) and of the cerebellar hemisphere (ctx-cbl) of neuropathologically confirmed PD patients (n = 95) and age- and sex-matched controls (n = 81) were MAPT haplotype genotyped to identify cases homozygous for either H1 or H2. Relative expression of genes was quantified using real-time qPCR; soluble and insoluble protein levels of tau and α-syn were determined by Western blotting. Homozygosity for H1 versus H2 was associated with increased total MAPT mRNA expression in ctx-fg regardless of disease state. Inversely, H2 homozygosity was associated with markedly increased expression of the corresponding antisense MAPT-AS1 in ctx-cbl. PD patients had higher levels of insoluble 0N3R and 1N4R tau isoforms regardless of the MAPT genotype. The increased presence of insoluble α-syn in PD patients in ctx-fg validated the selected postmortem brain tissue. Our findings in this small, but well controlled cohort of PD and controls support a putative biological relevance of tau in PD. However, we did not identify any link between the disease-predisposing H1/H1 associated overexpression of MAPT with PD status. Further studies are required to gain a deeper understanding of the potential regulatory role of MAPT-AS1 and its association to the disease-protective H2/H2 condition in the context of PD.
Collapse
Affiliation(s)
- Christina V Tauber
- Department of Translational Neurodegeneration, German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,Department of Neurology, School of Medicine, Technical University Munich, Munich, Germany.,Department of Obstetrics and Gynecology, Ludiwgs-Maximilians University of Munich, Munich, Germany
| | - Sigrid C Schwarz
- Department of Translational Neurodegeneration, German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Thomas W Rösler
- Department of Translational Neurodegeneration, German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,Department of Neurology, School of Medicine, Technical University Munich, Munich, Germany
| | - Thomas Arzberger
- Center for Neuropathology and Prion Research, Ludwig-Maximilians University of Munich, Munich, Germany.,Department of Psychiatry and Psychotherapy, University Hospital, Ludwig-Maximilians-University, Munich, Germany
| | - Steve Gentleman
- Parkinson's UK Brain Bank, Department of Brain Sciences, Imperial College London, London, UK.,Neuropathology Unit, Department of Brain Sciences, Department of Medicine, Imperial College London, London, UK
| | - Otto Windl
- Center for Neuropathology and Prion Research, Ludwig-Maximilians University of Munich, Munich, Germany
| | - Mandy Krumbiegel
- Institute of Human Genetics, Friedrich-Alexander University of Erlangen-Nuremberg, Erlangen, Germany
| | - André Reis
- Institute of Human Genetics, Friedrich-Alexander University of Erlangen-Nuremberg, Erlangen, Germany
| | - Viktoria C Ruf
- Center for Neuropathology and Prion Research, Ludwig-Maximilians University of Munich, Munich, Germany
| | - Jochen Herms
- Center for Neuropathology and Prion Research, Ludwig-Maximilians University of Munich, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Günter U Höglinger
- Department of Translational Neurodegeneration, German Center for Neurodegenerative Diseases (DZNE), Munich, Germany. .,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany. .,Department of Neurology, Ludwig-Maximilians University of Munich, Munich, Germany.
| |
Collapse
|
9
|
Chu Y, Hirst WD, Kordower JH. Mixed pathology as a rule, not exception: Time to reconsider disease nosology. HANDBOOK OF CLINICAL NEUROLOGY 2023; 192:57-71. [PMID: 36796948 DOI: 10.1016/b978-0-323-85538-9.00012-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
Abstract
Parkinson's disease is a progressive neurodegenerative disorder that is associated with motor and nonmotor symptoms. Accumulation of misfolded α-synuclein is considered a key pathological feature during disease initiation and progression. While clearly deemed a synucleinopathy, the development of amyloid-β plaques, tau-containing neurofibrillary tangles, and even TDP-43 protein inclusions occur within the nigrostriatal system and in other brain regions. In addition, inflammatory responses, manifested by glial reactivity, T-cell infiltration, and increased expression of inflammatory cytokines, plus other toxic mediators derived from activated glial cells, are currently recognized as prominent drivers of Parkinson's disease pathology. However, copathologies have increasingly been recognized as the rule (>90%) and not the exception, with Parkinson's disease cases on average exhibiting three different copathologies. While microinfarcts, atherosclerosis, arteriolosclerosis, and cerebral amyloid angiopathy may have an impact on disease progression, α-synuclein, amyloid-β, and TDP-43 pathology do not seem to contribute to progression.
Collapse
Affiliation(s)
- Yaping Chu
- ASU-Banner Neurodegenerative Disease Research Center, Arizona State University, Tempe, AZ, United States
| | - Warren D Hirst
- Neurodegenerative Diseases Research Unit, Biogen, Boston, MA, United States
| | - Jeffrey H Kordower
- ASU-Banner Neurodegenerative Disease Research Center, Arizona State University, Tempe, AZ, United States.
| |
Collapse
|
10
|
Campoy E, Puig M, Yakymenko I, Lerga-Jaso J, Cáceres M. Genomic architecture and functional effects of potential human inversion supergenes. Philos Trans R Soc Lond B Biol Sci 2022; 377:20210209. [PMID: 35694745 PMCID: PMC9189494 DOI: 10.1098/rstb.2021.0209] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Supergenes are involved in adaptation in multiple organisms, but they are little known in humans. Genomic inversions are the most common mechanism of supergene generation and maintenance. Here, we review the information about two large inversions that are the best examples of potential human supergenes. In addition, we do an integrative analysis of the newest data to understand better their functional effects and underlying genetic changes. We have found that the highly divergent haplotypes of the 17q21.31 inversion of approximately 1.5 Mb have multiple phenotypic associations, with consistent effects in brain-related traits, red and white blood cells, lung function, male and female characteristics and disease risk. By combining gene expression and nucleotide variation data, we also analysed the molecular differences between haplotypes, including gene duplications, amino acid substitutions and regulatory changes, and identify CRHR1, KANLS1 and MAPT as good candidates to be responsible for these phenotypes. The situation is more complex for the 8p23.1 inversion, where there is no clear genetic differentiation. However, the inversion is associated with several related phenotypes and gene expression differences that could be linked to haplotypes specific of one orientation. Our work, therefore, contributes to the characterization of both exceptional variants and illustrates the important role of inversions. This article is part of the theme issue 'Genomic architecture of supergenes: causes and evolutionary consequences'.
Collapse
Affiliation(s)
- Elena Campoy
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, Bellaterra (Barcelona), Spain
| | - Marta Puig
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, Bellaterra (Barcelona), Spain.,Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, Bellaterra (Barcelona), Spain
| | - Illya Yakymenko
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, Bellaterra (Barcelona), Spain
| | - Jon Lerga-Jaso
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, Bellaterra (Barcelona), Spain
| | - Mario Cáceres
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, Bellaterra (Barcelona), Spain.,ICREA, Barcelona, Spain
| |
Collapse
|
11
|
Ho D, Schierding W, Farrow SL, Cooper AA, Kempa-Liehr AW, O’Sullivan JM. Machine Learning Identifies Six Genetic Variants and Alterations in the Heart Atrial Appendage as Key Contributors to PD Risk Predictivity. Front Genet 2022; 12:785436. [PMID: 35047012 PMCID: PMC8762216 DOI: 10.3389/fgene.2021.785436] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 11/09/2021] [Indexed: 12/14/2022] Open
Abstract
Parkinson's disease (PD) is a complex neurodegenerative disease with a range of causes and clinical presentations. Over 76 genetic loci (comprising 90 SNPs) have been associated with PD by the most recent GWAS meta-analysis. Most of these PD-associated variants are located in non-coding regions of the genome and it is difficult to understand what they are doing and how they contribute to the aetiology of PD. We hypothesised that PD-associated genetic variants modulate disease risk through tissue-specific expression quantitative trait loci (eQTL) effects. We developed and validated a machine learning approach that integrated tissue-specific eQTL data on known PD-associated genetic variants with PD case and control genotypes from the Wellcome Trust Case Control Consortium. In so doing, our analysis ranked the tissue-specific transcription effects for PD-associated genetic variants and estimated their relative contributions to PD risk. We identified roles for SNPs that are connected with INPP5P, CNTN1, GBA and SNCA in PD. Ranking the variants and tissue-specific eQTL effects contributing most to the machine learning model suggested a key role in the risk of developing PD for two variants (rs7617877 and rs6808178) and eQTL associated transcriptional changes of EAF1-AS1 within the heart atrial appendage. Similarly, effects associated with eQTLs located within the Brain Cerebellum were also recognized to confer major PD risk. These findings were replicated in two additional, independent cohorts (the UK Biobank, and NeuroX) and thus warrant further mechanistic investigations to determine if these transcriptional changes could act as early contributors to PD risk and disease development.
Collapse
Affiliation(s)
- Daniel Ho
- Liggins Institute, The University of Auckland, Auckland, New Zealand
| | - William Schierding
- Liggins Institute, The University of Auckland, Auckland, New Zealand
- MRC Lifecourse Epidemiology Unit, University of Southampton, Southampton, United Kingdom
| | - Sophie L. Farrow
- Liggins Institute, The University of Auckland, Auckland, New Zealand
- MRC Lifecourse Epidemiology Unit, University of Southampton, Southampton, United Kingdom
| | - Antony A. Cooper
- Australian Parkinsons Mission, Garvan Institute of Medical Research, Sydney, NSW, Australia
- St Vincent’s Clinical School, UNSW Sydney, Sydney, NSW, Australia
| | | | - Justin M. O’Sullivan
- Liggins Institute, The University of Auckland, Auckland, New Zealand
- MRC Lifecourse Epidemiology Unit, University of Southampton, Southampton, United Kingdom
- Brain Research New Zealand, The University of Auckland, Auckland, New Zealand
- The Maurice Wilkins Centre, The University of Auckland, Auckland, New Zealand
| |
Collapse
|
12
|
Martínez-Iglesias O, Naidoo V, Cacabelos N, Cacabelos R. Epigenetic Biomarkers as Diagnostic Tools for Neurodegenerative Disorders. Int J Mol Sci 2021; 23:13. [PMID: 35008438 PMCID: PMC8745005 DOI: 10.3390/ijms23010013] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 12/03/2021] [Accepted: 12/17/2021] [Indexed: 12/12/2022] Open
Abstract
Epigenetics is the study of heritable changes in gene expression that occur without alterations to the DNA sequence, linking the genome to its surroundings. The accumulation of epigenetic alterations over the lifespan may contribute to neurodegeneration. The aim of the present study was to identify epigenetic biomarkers for improving diagnostic efficacy in patients with neurodegenerative diseases. We analyzed global DNA methylation, chromatin remodeling/histone modifications, sirtuin (SIRT) expression and activity, and the expression of several important neurodegeneration-related genes. DNA methylation, SIRT expression and activity and neuregulin 1 (NRG1), microtubule-associated protein tau (MAPT) and brain-derived neurotrophic factor (BDNF) expression were reduced in buffy coat samples from patients with neurodegenerative disorders. Our data suggest that these epigenetic biomarkers may be useful in clinical practical for the diagnosis, surveillance, and prognosis of disease activity in patients with neurodegenerative diseases.
Collapse
Affiliation(s)
- Olaia Martínez-Iglesias
- EuroEspes Biomedical Research Center, International Center of Neuroscience and Genomic Medicine, Bergondo, 15165 Corunna, Spain; (V.N.); (N.C.); (R.C.)
| | | | | | | |
Collapse
|
13
|
Yao S, Zhang X, Zou SC, Zhu Y, Li B, Kuang WP, Guo Y, Li XS, Li L, Wang XY. A transcriptome-wide association study identifies susceptibility genes for Parkinson's disease. NPJ Parkinsons Dis 2021; 7:79. [PMID: 34504106 PMCID: PMC8429416 DOI: 10.1038/s41531-021-00221-7] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 08/10/2021] [Indexed: 12/19/2022] Open
Abstract
Genome-wide association study (GWAS) has seen great strides in revealing initial insights into the genetic architecture of Parkinson's disease (PD). Since GWAS signals often reside in non-coding regions, relatively few of the associations have implicated specific biological mechanisms. Here, we aimed to integrate the GWAS results with large-scale expression quantitative trait loci (eQTL) in 13 brain tissues to identify candidate causal genes for PD. We conducted a transcriptome-wide association study (TWAS) for PD using the summary statistics of over 480,000 individuals from the most recent PD GWAS. We identified 18 genes significantly associated with PD after Bonferroni corrections. The most significant gene, LRRC37A2, was associated with PD in all 13 brain tissues, such as in the hypothalamus (P = 6.12 × 10-22) and nucleus accumbens basal ganglia (P = 5.62 × 10-21). We also identified eight conditionally independent genes, including four new genes at known PD loci: CD38, LRRC37A2, RNF40, and ZSWIM7. Through conditional analyses, we demonstrated that several of the GWAS significant signals on PD could be driven by genetically regulated gene expression. The most significant TWAS gene LRRC37A2 accounts for 0.855 of the GWAS signal at its loci, and ZSWIM7 accounts for all the GWAS signals at its loci. We further identified several phenotypes previously associated with PD by querying the single nucleotide polymorphisms (SNPs) in the final model of the identified genes in phenome databases. In conclusion, we prioritized genes that are likely to affect PD by using a TWAS approach and identified phenotypes associated with PD.
Collapse
Affiliation(s)
- Shi Yao
- Department of Neurosurgery, Hunan Brain Hospital, Clinical Medical School of Hunan University of Chinese Medicine, Changsha, Hunan, P. R. China
- National and Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, P. R. China
| | - Xi Zhang
- Department of Neurosurgery, Hunan Brain Hospital, Clinical Medical School of Hunan University of Chinese Medicine, Changsha, Hunan, P. R. China
| | - Shu-Cheng Zou
- Department of Neurosurgery, Hunan Brain Hospital, Clinical Medical School of Hunan University of Chinese Medicine, Changsha, Hunan, P. R. China
| | - Yong Zhu
- Department of Neurosurgery, Hunan Brain Hospital, Clinical Medical School of Hunan University of Chinese Medicine, Changsha, Hunan, P. R. China
| | - Bo Li
- Department of Neurosurgery, Hunan Brain Hospital, Clinical Medical School of Hunan University of Chinese Medicine, Changsha, Hunan, P. R. China
| | - Wei-Ping Kuang
- Department of Neurosurgery, Hunan Brain Hospital, Clinical Medical School of Hunan University of Chinese Medicine, Changsha, Hunan, P. R. China
| | - Yan Guo
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Biomedical Informatics & Genomics Center, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, P. R. China
| | - Xiao-Song Li
- Department of Neurosurgery, Hunan Brain Hospital, Clinical Medical School of Hunan University of Chinese Medicine, Changsha, Hunan, P. R. China.
| | - Liang Li
- Provincial Key Laboratory of TCM Diagnostics, Hunan University of Chinese Medicine, Changsha, Hunan, P. R. China.
| | - Xiao-Ye Wang
- Department of Neurosurgery, Hunan Brain Hospital, Clinical Medical School of Hunan University of Chinese Medicine, Changsha, Hunan, P. R. China.
| |
Collapse
|
14
|
Reference SVA insertion polymorphisms are associated with Parkinson's Disease progression and differential gene expression. NPJ Parkinsons Dis 2021; 7:44. [PMID: 34035310 PMCID: PMC8149882 DOI: 10.1038/s41531-021-00189-4] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 04/23/2021] [Indexed: 12/20/2022] Open
Abstract
The development of Parkinson's disease (PD) involves a complex interaction of genetic and environmental factors. Genome-wide association studies using extensive single nucleotide polymorphism datasets have identified many loci involved in disease. However much of the heritability of Parkinson's disease is still to be identified and the functional elements associated with the risk to be determined and understood. To investigate the component of PD that may involve complex genetic variants we characterised the hominid specific retrotransposon SINE-VNTR-Alus (SVAs) in the Parkinson's Progression Markers Initiative cohort utilising whole genome sequencing. We identified 81 reference SVAs polymorphic for their presence/absence, seven of which were associated with the progression of the disease and with differential gene expression in whole blood RNA sequencing data. This study highlights the importance of addressing SVA variants and potentially other types of retrotransposons in PD genetics, furthermore, these SVA elements should be considered as regulatory domains that could play a role in disease progression.
Collapse
|
15
|
Yang S, Lim KH, Kim SH, Joo JY. Molecular landscape of long noncoding RNAs in brain disorders. Mol Psychiatry 2021; 26:1060-1074. [PMID: 33173194 DOI: 10.1038/s41380-020-00947-5] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 09/28/2020] [Accepted: 10/27/2020] [Indexed: 02/08/2023]
Abstract
According to current paradigms, various risk factors, such as genetic mutations, oxidative stress, neural network dysfunction, and abnormal protein degradation, contribute to the progression of brain disorders. Through the cooperation of gene transcripts in biological processes, the study of noncoding RNAs can lead to insights into the cause and treatment of brain disorders. Recently, long noncoding RNAs (lncRNAs) which are longer than 200 nucleotides in length have been suggested as key factors in various brain disorders. Accumulating evidence suggests the potential of lncRNAs as diagnostic or prognostic biomarkers and therapeutic targets. High-throughput screening-based sequencing has been instrumental in identification of lncRNAs that demand new approaches to understanding the progression of brain disorders. In this review, we discuss the recent progress in the study of lncRNAs, and addresses the pathogenesis of brain disorders that involve lncRNAs and describes the associations of lncRNAs with neurodegenerative disorders such as Alzheimer disease (AD), Parkinson disease (PD), and neurodevelopmental disorders. We also discuss potential targets of lncRNAs and their promise as novel therapeutics and biomarkers in brain disorders.
Collapse
Affiliation(s)
- Sumin Yang
- Neurodegenerative Disease Research Group, Korea Brain Research Institute, Daegu, 41062, Republic of Korea
| | - Key-Hwan Lim
- Neurodegenerative Disease Research Group, Korea Brain Research Institute, Daegu, 41062, Republic of Korea
| | - Sung-Hyun Kim
- Neurodegenerative Disease Research Group, Korea Brain Research Institute, Daegu, 41062, Republic of Korea
| | - Jae-Yeol Joo
- Neurodegenerative Disease Research Group, Korea Brain Research Institute, Daegu, 41062, Republic of Korea.
| |
Collapse
|
16
|
A Specific Diplotype H1j/H2 of the MAPT Gene Could Be Responsible for Parkinson's Disease with Dementia. Case Rep Genet 2020; 2020:8813344. [PMID: 33343949 PMCID: PMC7732378 DOI: 10.1155/2020/8813344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 11/15/2020] [Accepted: 11/20/2020] [Indexed: 11/17/2022] Open
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disorder after Alzheimer disease. Five to ten percent of patients have monogenic form of the disease, while most of sporadic PD cases are caused by the combination of genetic and environmental factors. Microtubule-associated protein tau (MAPT) has been appointed as one of the most important risk factors for several neurodegenerative diseases including PD. MAPT is characterized by an inversion in chromosome 17 resulting in two distinct haplotypes H1 and H2. Studies described a significant association of MAPT H1j subhaplotype with PD risk, while H2 haplotype was associated with Parkinsonism, particularly to its bradykinetic component. We report here an isolated case displaying an akinetic-rigid form of PD, with age of onset of 41 years and a good response to levodopa, who developed dementia gradually during the seven years of disease progression. The patient does not carry the LRRK2 G2019S mutation, copy number variations, nor pathogenic and rare variants in known genes associated with PD. MAPT subhaplotype genotyping revealed that the patient has the H1j/H2 diplotype, his mother H1j/H1j, his two healthy brothers H1j/H1v and his deceased father was by deduction H1v/H2. The H1j/H2 diplotype was shown in a total of 3 PD patients among 80, who also did not have known PD-causing mutation and in 1 out of 92 healthy individual controls. The three patients with this diplotype all have a similar clinical phenotype. Our results suggest that haplotypes H1j and H2 are strong risk factor alleles, and their combination could be responsible for early onset of PD with dementia.
Collapse
|
17
|
Intranasal insulin ameliorates cognitive impairment in a rat model of Parkinson's disease through Akt/GSK3β signaling pathway. Life Sci 2020; 259:118159. [PMID: 32763288 DOI: 10.1016/j.lfs.2020.118159] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 07/24/2020] [Accepted: 07/24/2020] [Indexed: 12/22/2022]
Abstract
AIMS Parkinson's disease dementia (PDD) is one of the most common non-motor symptoms of advanced Parkinson's disease (PD). This study aimed to determine whether intranasal insulin has protective effects on cognition in the rat PD model induced by 6-hydroxylase dopamine (6-OHDA) through the insulin signaling pathway. MATERIALS AND METHODS The rats were given intranasal insulin administration for six weeks after unilateral medial forebrain bundle (MFB) injection of 6-OHDA. Then a series of cognitive-behavioral tests, immunofluorescence, and immunoblotting was performed on the rats. KEY FINDINGS The results demonstrated that the injection of 6-OHDA in the unilateral MFB damaged working memory and long-term habituation of rats in the T-maze rewarded alternation test and hole-board test. Besides, rats with unilateral 6-OHDA injury performed poorly in terms of escape latency and average speed during the hidden platform training phase rather than in the probe trial of the Morris Water Maze (MWM) test. Immunofluorescence results showed that unilateral 6-OHDA injury in MFB led to the massive death of ipsilateral-substantia nigra (SN) tyrosine hydroxylase (TH)-positive neurons. Western blot results further indicated that 6-OHDA-induced necrosis of ipsilateral-SN dopaminergic neurons reduced the levels of p-Akt (Ser473) and p-GSK3β (Ser9) in the ipsilateral-hippocampus. SIGNIFICANCE These findings provide a solid evidence base for the relationship between PD cognitive impairment and insulin signaling pathways.
Collapse
|
18
|
Cornblath EJ, Robinson JL, Irwin DJ, Lee EB, Lee VMY, Trojanowski JQ, Bassett DS. Defining and predicting transdiagnostic categories of neurodegenerative disease. Nat Biomed Eng 2020; 4:787-800. [PMID: 32747831 PMCID: PMC7946378 DOI: 10.1038/s41551-020-0593-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Accepted: 06/25/2020] [Indexed: 11/09/2022]
Abstract
The prevalence of concomitant proteinopathies and heterogeneous clinical symptoms in neurodegenerative diseases hinders the identification of individuals who might be candidates for a particular intervention. Here, by applying an unsupervised clustering algorithm to post-mortem histopathological data from 895 patients with degeneration in the central nervous system, we show that six non-overlapping disease clusters can simultaneously account for tau neurofibrillary tangles, α-synuclein inclusions, neuritic plaques, inclusions of the transcriptional repressor TDP-43, angiopathy, neuron loss and gliosis. We also show that membership to the six transdiagnostic disease clusters, which explains more variance in cognitive phenotypes than can be explained by individual diagnoses, can be accurately predicted from scores of the Mini-Mental Status Exam, protein levels in cerebrospinal fluid, and genotype at the APOE and MAPT loci, via cross-validated multiple logistic regression. This combination of unsupervised and supervised data-driven tools provides a framework that could be used to identify latent disease subtypes in other areas of medicine.
Collapse
Affiliation(s)
- Eli J Cornblath
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, USA
| | - John L Robinson
- Center for Neurodegenerative Disease Research, Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - David J Irwin
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Edward B Lee
- Translational Neuropathology Research Laboratory, Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Virginia M-Y Lee
- Center for Neurodegenerative Disease Research, Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - John Q Trojanowski
- Center for Neurodegenerative Disease Research, Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Danielle S Bassett
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, USA.
- Center for Neurodegenerative Disease Research, Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Department of Physics and Astronomy, College of Arts and Sciences, University of Pennsylvania, Philadelphia, PA, USA.
- Department of Electrical and Systems Engineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, USA.
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Santa Fe Institute, Santa Fe, NM, USA.
| |
Collapse
|
19
|
Delic V, Beck KD, Pang KCH, Citron BA. Biological links between traumatic brain injury and Parkinson's disease. Acta Neuropathol Commun 2020; 8:45. [PMID: 32264976 PMCID: PMC7137235 DOI: 10.1186/s40478-020-00924-7] [Citation(s) in RCA: 96] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 03/26/2020] [Indexed: 12/19/2022] Open
Abstract
Parkinson's Disease (PD) is a progressive neurodegenerative disorder with no cure. Clinical presentation is characterized by postural instability, resting tremors, and gait problems that result from progressive loss of A9 dopaminergic neurons in the substantia nigra pars compacta. Traumatic brain injury (TBI) has been implicated as a risk factor for several neurodegenerative diseases, but the strongest evidence is linked to development of PD. Mild TBI (mTBI), is the most common and is defined by minimal, if any, loss of consciousness and the absence of significant observable damage to the brain tissue. mTBI is responsible for a 56% higher risk of developing PD in U.S. Veterans and the risk increases with severity of injury. While the mounting evidence from human studies suggests a link between TBI and PD, fundamental questions as to whether TBI nucleates PD pathology or accelerates PD pathology in vulnerable populations remains unanswered. Several promising lines of research point to inflammation, metabolic dysregulation, and protein accumulation as potential mechanisms through which TBI can initiate or accelerate PD. Amyloid precursor protein (APP), alpha synuclein (α-syn), hyper-phosphorylated Tau, and TAR DNA-binding protein 43 (TDP-43), are some of the most frequently reported proteins upregulated following a TBI and are also closely linked to PD. Recently, upregulation of Leucine Rich Repeat Kinase 2 (LRRK2), has been found in the brain of mice following a TBI. Subset of Rab proteins were identified as biological substrates of LRRK2, a protein also extensively linked to late onset PD. Inhibition of LRRK2 was found to be neuroprotective in PD and TBI models. The goal of this review is to survey current literature concerning the mechanistic overlap between TBI and PD with a particular focus on inflammation, metabolic dysregulation, and aforementioned proteins. This review will also cover the application of rodent TBI models to further our understanding of the relationship between TBI and PD.
Collapse
Affiliation(s)
- Vedad Delic
- Laboratory of Molecular Biology, VA New Jersey Health Care System, Research and Development (Mailstop 15), 385 Tremont Ave, East Orange, NJ, 07018, USA.
- NeuroBehavioral Research Laboratory, VA New Jersey Health Care System, Research and Development (Mailstop 15), 385 Tremont Ave, East Orange, NJ, 07018, USA.
| | - Kevin D Beck
- NeuroBehavioral Research Laboratory, VA New Jersey Health Care System, Research and Development (Mailstop 15), 385 Tremont Ave, East Orange, NJ, 07018, USA
- Department of Pharmacology, Physiology, and Neuroscience, Rutgers- New Jersey Medical School, Newark, NJ, 07103, USA
| | - Kevin C H Pang
- NeuroBehavioral Research Laboratory, VA New Jersey Health Care System, Research and Development (Mailstop 15), 385 Tremont Ave, East Orange, NJ, 07018, USA
- Department of Pharmacology, Physiology, and Neuroscience, Rutgers- New Jersey Medical School, Newark, NJ, 07103, USA
| | - Bruce A Citron
- Laboratory of Molecular Biology, VA New Jersey Health Care System, Research and Development (Mailstop 15), 385 Tremont Ave, East Orange, NJ, 07018, USA
- Department of Pharmacology, Physiology, and Neuroscience, Rutgers- New Jersey Medical School, Newark, NJ, 07103, USA
| |
Collapse
|
20
|
Feng ST, Wang ZZ, Yuan YH, Sun HM, Chen NH, Zhang Y. Update on the association between alpha-synuclein and tau with mitochondrial dysfunction: Implications for Parkinson's disease. Eur J Neurosci 2020; 53:2946-2959. [PMID: 32031280 DOI: 10.1111/ejn.14699] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 01/13/2020] [Accepted: 01/23/2020] [Indexed: 12/26/2022]
Abstract
The critical role of mitochondrial dysfunction in the pathological mechanisms of neurodegenerative disorders, particularly Parkinson's disease (PD), is well established. Compelling evidence indicates that Parkinson's proteins (e.g., α-synuclein, Parkin, PINK1, DJ-1, and LRRK2) are associated with mitochondrial dysfunction and oxidative stress in PD. Significantly, there is a possible central role of alpha-synuclein (α-Syn) in the occurrence of mitochondrial dysfunction and oxidative stress by the mediation of different signaling pathways. Also, tau, traditionally considered as the main component of neurofibrillary tangles, aggregates and amplifies the neurotoxic effects on mitochondria by interacting with α-Syn. Moreover, oxidative stress caused by mitochondrial dysfunction favors assembly of both α-Syn and tau and also plays a key role in the formation of protein aggregates. In this review, we provide an overview of the relationship between these two pathological proteins and mitochondrial dysfunction in PD, and also summarize the underlying mechanisms in the interplay of α-Syn aggregation and phosphorylated tau targeting the mitochondria, to find new strategies to prevent PD processing.
Collapse
Affiliation(s)
- Si-Tong Feng
- Department of Anatomy, School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Zhen-Zhen Wang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yu-He Yuan
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hong-Mei Sun
- Department of Anatomy, School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Nai-Hong Chen
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yi Zhang
- Department of Anatomy, School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
21
|
Ruiz-Arenas C, Cáceres A, Moreno V, González JR. Common polymorphic inversions at 17q21.31 and 8p23.1 associate with cancer prognosis. Hum Genomics 2019; 13:57. [PMID: 31753042 PMCID: PMC6873427 DOI: 10.1186/s40246-019-0242-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2019] [Accepted: 10/09/2019] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Chromosomal inversions are structural genetic variants where a chromosome segment changes its orientation. While sporadic de novo inversions are known genetic risk factors for cancer susceptibility, it is unknown if common polymorphic inversions are also associated with the prognosis of common tumors, as they have been linked to other complex diseases. We studied the association of two well-characterized human inversions at 17q21.31 and 8p23.1 with the prognosis of lung, liver, breast, colorectal, and stomach cancers. RESULTS Using data from The Cancer Genome Atlas (TCGA), we observed that inv8p23.1 was associated with overall survival in breast cancer and that inv17q21.31 was associated with overall survival in stomach cancer. In the meta-analysis of two independent studies, inv17q21.31 heterozygosity was significantly associated with colorectal disease-free survival. We found that the association was mediated by the de-methylation of cg08283464 and cg03999934, also linked to lower disease-free survival. CONCLUSIONS Our results suggest that chromosomal inversions are important genetic factors of tumor prognosis, likely affecting changes in methylation patterns.
Collapse
Affiliation(s)
- Carlos Ruiz-Arenas
- Barcelona Institute for Global Health, ISGlobal, Doctor Aiguader 88, 08003, Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
- CIBER Epidemiología y Salud Pública (CIBERESP), Barcelona, Spain
| | - Alejandro Cáceres
- Barcelona Institute for Global Health, ISGlobal, Doctor Aiguader 88, 08003, Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
- CIBER Epidemiología y Salud Pública (CIBERESP), Barcelona, Spain
| | - Victor Moreno
- Programa de Prevención y Control del Cáncer, Instituto Catalán de Oncología, L'Hospitalet, Barcelona, Spain
| | - Juan R González
- Barcelona Institute for Global Health, ISGlobal, Doctor Aiguader 88, 08003, Barcelona, Spain.
- Universitat Pompeu Fabra (UPF), Barcelona, Spain.
- CIBER Epidemiología y Salud Pública (CIBERESP), Barcelona, Spain.
| |
Collapse
|
22
|
Trochet H, Pirinen M, Band G, Jostins L, McVean G, Spencer CCA. Bayesian meta-analysis across genome-wide association studies of diverse phenotypes. Genet Epidemiol 2019; 43:532-547. [PMID: 30920090 DOI: 10.1002/gepi.22202] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 03/01/2019] [Accepted: 03/05/2019] [Indexed: 11/09/2022]
Abstract
Genome-wide association studies (GWAS) are a powerful tool for understanding the genetic basis of diseases and traits, but most studies have been conducted in isolation, with a focus on either a single or a set of closely related phenotypes. We describe MetABF, a simple Bayesian framework for performing integrative meta-analysis across multiple GWAS using summary statistics. The approach is applicable across a wide range of study designs and can increase the power by 50% compared with standard frequentist tests when only a subset of studies have a true effect. We demonstrate its utility in a meta-analysis of 20 diverse GWAS which were part of the Wellcome Trust Case Control Consortium 2. The novelty of the approach is its ability to explore, and assess the evidence for a range of possible true patterns of association across studies in a computationally efficient framework.
Collapse
Affiliation(s)
- Holly Trochet
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK.,Institut de Cardiologie de Montréal (Centre de Recherche), Université de Montréal, Montréal, Québec, Canada
| | - Matti Pirinen
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland.,Department of Mathematics and Statistics, Helsinki Institute for Information Technology HIIT, University of Helsinki, Helsinki, Finland.,Department of Public Health, University of Helsinki, Helsinki, Finland
| | - Gavin Band
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Luke Jostins
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK.,Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford, UK.,Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK.,Christ Church, University of Oxford, Oxford, UK
| | - Gilean McVean
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK.,Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford, UK
| | - Chris C A Spencer
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
| |
Collapse
|
23
|
Kumar S, Yadav N, Pandey S, Thelma BK. Advances in the discovery of genetic risk factors for complex forms of neurodegenerative disorders: contemporary approaches, success, challenges and prospects. J Genet 2018. [DOI: 10.1007/s12041-018-0953-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
24
|
Kumar S, Yadav N, Pandey S, Thelma BK. Advances in the discovery of genetic risk factors for complex forms of neurodegenerative disorders: contemporary approaches, success, challenges and prospects. J Genet 2018; 97:625-648. [PMID: 30027900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Neurodegenerative diseases constitute a large proportion of disorders in elderly, majority being sporadic in occurrence with ∼5-10% familial. A strong genetic component underlies the Mendelian forms but nongenetic factors together with genetic vulnerability contributes to the complex sporadic forms. Several gene discoveries in the familial forms have provided novel insights into the pathogenesis of neurodegeneration with implications for treatment. Conversely, findings from genetic dissection of the sporadic forms, despite large genomewide association studies and more recently whole exome and whole genome sequencing, have been limited. This review provides a concise account of the genetics that we know, the pathways that they implicate, the challenges that are faced and the prospects that are envisaged for the sporadic, complex forms of neurodegenerative diseases, taking four most common conditions, namely Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis and Huntington disease as examples. Poor replication across studies, inability to establish genotype-phenotype correlations and the overall failure to predict risk and/or prevent disease in this group poses a continuing challenge. Among others, clinical heterogeneity emerges as the most important impediment warranting newer approaches. Advanced computational and system biology tools to analyse the big data are being generated and the alternate strategy such as subgrouping of case-control cohorts based on deep phenotyping using the principles of Ayurveda to overcome current limitation of phenotype heterogeneity seem to hold promise. However, at this point, with advances in discovery genomics and functional analysis of putative determinants with translation potential for the complex forms being minimal, stem cell therapies are being attempted as potential interventions. In this context, the possibility to generate patient derived induced pluripotent stem cells, mutant/gene/genome correction through CRISPR/Cas9 technology and repopulating the specific brain regions with corrected neurons, which may fulfil the dream of personalized medicine have been mentioned briefly. Understanding disease pathways/biology using this technology, with implications for development of novel therapeutics are optimistic expectations in the near future.
Collapse
Affiliation(s)
- Sumeet Kumar
- Department of Genetics, University of Delhi South Campus, New Delhi 110 021, India.
| | | | | | | |
Collapse
|
25
|
Catacchio CR, Maggiolini FAM, D'Addabbo P, Bitonto M, Capozzi O, Lepore Signorile M, Miroballo M, Archidiacono N, Eichler EE, Ventura M, Antonacci F. Inversion variants in human and primate genomes. Genome Res 2018; 28:910-920. [PMID: 29776991 PMCID: PMC5991517 DOI: 10.1101/gr.234831.118] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Accepted: 04/26/2018] [Indexed: 02/06/2023]
Abstract
For many years, inversions have been proposed to be a direct driving force in speciation since they suppress recombination when heterozygous. Inversions are the most common large-scale differences among humans and great apes. Nevertheless, they represent large events easily distinguishable by classical cytogenetics, whose resolution, however, is limited. Here, we performed a genome-wide comparison between human, great ape, and macaque genomes using the net alignments for the most recent releases of genome assemblies. We identified a total of 156 putative inversions, between 103 kb and 91 Mb, corresponding to 136 human loci. Combining literature, sequence, and experimental analyses, we analyzed 109 of these loci and found 67 regions inverted in one or multiple primates, including 28 newly identified inversions. These events overlap with 81 human genes at their breakpoints, and seven correspond to sites of recurrent rearrangements associated with human disease. This work doubles the number of validated primate inversions larger than 100 kb, beyond what was previously documented. We identified 74 sites of errors, where the sequence has been assembled in the wrong orientation, in the reference genomes analyzed. Our data serve two purposes: First, we generated a map of evolutionary inversions in these genomes representing a resource for interrogating differences among these species at a functional level; second, we provide a list of misassembled regions in these primate genomes, involving over 300 Mb of DNA and 1978 human genes. Accurately annotating these regions in the genome references has immediate applications for evolutionary and biomedical studies on primates.
Collapse
Affiliation(s)
| | | | - Pietro D'Addabbo
- Dipartimento di Biologia, Università degli Studi di Bari "Aldo Moro," Bari 70125, Italy
| | - Miriana Bitonto
- Dipartimento di Biologia, Università degli Studi di Bari "Aldo Moro," Bari 70125, Italy
| | - Oronzo Capozzi
- Dipartimento di Biologia, Università degli Studi di Bari "Aldo Moro," Bari 70125, Italy
| | | | - Mattia Miroballo
- Dipartimento di Biologia, Università degli Studi di Bari "Aldo Moro," Bari 70125, Italy
| | | | - Evan E Eichler
- Department of Genome Sciences, University of Washington School of Medicine, Seattle, Washington 98195, USA.,Howard Hughes Medical Institute, University of Washington, Seattle, Washington 98195, USA
| | - Mario Ventura
- Dipartimento di Biologia, Università degli Studi di Bari "Aldo Moro," Bari 70125, Italy
| | - Francesca Antonacci
- Dipartimento di Biologia, Università degli Studi di Bari "Aldo Moro," Bari 70125, Italy
| |
Collapse
|
26
|
Zhang CC, Zhu JX, Wan Y, Tan L, Wang HF, Yu JT, Tan L. Meta-analysis of the association between variants in MAPT and neurodegenerative diseases. Oncotarget 2018; 8:44994-45007. [PMID: 28402959 PMCID: PMC5546535 DOI: 10.18632/oncotarget.16690] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Accepted: 03/16/2017] [Indexed: 01/11/2023] Open
Abstract
Microtubule-associated protein tau (MAPT) gene is compelling among the susceptibility genes of neurodegenerative diseases which include Alzheimer’s disease (AD), Parkinson’s disease (PD), progressive supranuclear palsy (PSP), corticobasal degeneration (CBD), frontotemporal dementia (FTD) and amyotrophic lateral sclerosis (ALS). Our meta-analysis aimed to find the association between MAPT and the risk of these diseases. Published literatures were retrieved from MEDLINE and other databases, and 82 case-control studies were recruited. Six haplotype tagging single-nucleotide polymorphisms (rs1467967, rs242557, rs3785883, rs2471738, del-In9 and rs7521) and haplotypes (H2 and H1c) were significantly associated with the above diseases. The odds ratios (ORs) and 95 % confidence intervals (CIs) were evaluated by comparison in minor and major allele frequency using the R software. This study demonstrated that different variants in MAPT were associated with AD (rs2471738: OR= 1.04, 95%CI = 1.00 - 1.09; H2: OR = 0.94, 95% CI = 0.91 - 0.97), PD (H2: OR = 0.76, 95% CI = 0.74 - 0.79), PSP (rs242557: OR = 1. 96, 95% CI = 1. 71 - 2.25; rs2471738: OR = 1. 85, 95% CI = 1. 48 - 2.31; H2: OR = 0.20, 95% CI = 0.18 - 0.23), CBD (rs242557: OR = 2.51, 95%CI = 1. 66 -3.78; rs2471738: OR = 2.07, 95%CI = 1. 32 -3.23; H2: OR = OR = 0.30, 95% CI = 0.23 - 0.41) and ALS (H2: OR = 0.92, 95% CI = 0.86 - 0.98) instead of FTD (H2: OR = 1.02, 95% CI = 0.78 - 1.32). In conclusion, MAPT is associated with risk of neurodegenerative diseases, suggesting crucial roles of tau in neurodegenerative processes.
Collapse
Affiliation(s)
- Cheng-Cheng Zhang
- Department of Neurology, Qingdao Municipal Hospital, Dalian Medical University, PR China
| | - Jun-Xia Zhu
- Clinical Skills Training Center, Qingdao Municipal Hospital, Qingdao University, PR China
| | - Yu Wan
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, PR China
| | - Lin Tan
- College of Medicine and Pharmaceutics, Ocean University of China, Qingdao, China
| | - Hui-Fu Wang
- Clinical Skills Training Center, Qingdao Municipal Hospital, Qingdao University, PR China
| | - Jin-Tai Yu
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, PR China
| | - Lan Tan
- Department of Neurology, Qingdao Municipal Hospital, Dalian Medical University, PR China.,Department of Neurology, Qingdao Municipal Hospital, Qingdao University, PR China.,College of Medicine and Pharmaceutics, Ocean University of China, Qingdao, China
| |
Collapse
|
27
|
Abstract
Parkinson's disease is a progressive neurodegenerative disease characterized by Lewy body pathology of which the primary constituent is aggregated misfolded alpha-synuclein protein. Currently, there are no clinical therapies for treatment of the underlying alpha-synuclein dysfunction and accumulation, and the standard of care for patients with Parkinson's disease focuses only on symptom management, creating an immense therapeutic gap that needs to be filled. Defects in autophagy have been strongly implicated in Parkinson's disease. Here, we review evidence from human, mouse, and cell culture studies to briefly explain these defects in autophagy in Parkinson's disease and the necessity for autophagy to be carefully and precisely tuned to maintain neuron survival. We summarize recent experimental agents for treating alpha-synuclein accumulation in α-synuclein Parkinson's disease and related synucleinopathies. Most of the efforts for developing experimental agents have focused on immunotherapeutic strategies, but we discuss why those efforts are misplaced. Finally, we emphasize why increasing autophagy flux for alpha-synuclein clearance is the most promising therapeutic strategy. Activating autophagy has been successful in preclinical models of Parkinson's disease and yields promising results in clinical trials.
Collapse
Affiliation(s)
- Alan J Fowler
- Department of Neurology, Laboratory for Dementia and Parkinsonism, Translational Neurotherapeutics Program, Room 203-C, Building D, 4000 Reservoir Rd. NW, Washington, DC, USA
| | - Charbel E-H Moussa
- Department of Neurology, Laboratory for Dementia and Parkinsonism, Translational Neurotherapeutics Program, Room 203-C, Building D, 4000 Reservoir Rd. NW, Washington, DC, USA.
| |
Collapse
|
28
|
Effects of α-synuclein on axonal transport. Neurobiol Dis 2017; 105:321-327. [DOI: 10.1016/j.nbd.2016.12.008] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Revised: 12/05/2016] [Accepted: 12/07/2016] [Indexed: 11/22/2022] Open
|
29
|
Beevers JE, Lai MC, Collins E, Booth HDE, Zambon F, Parkkinen L, Vowles J, Cowley SA, Wade-Martins R, Caffrey TM. MAPT Genetic Variation and Neuronal Maturity Alter Isoform Expression Affecting Axonal Transport in iPSC-Derived Dopamine Neurons. Stem Cell Reports 2017; 9:587-599. [PMID: 28689993 PMCID: PMC5549835 DOI: 10.1016/j.stemcr.2017.06.005] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Revised: 06/06/2017] [Accepted: 06/06/2017] [Indexed: 12/24/2022] Open
Abstract
The H1 haplotype of the microtubule-associated protein tau (MAPT) locus is genetically associated with neurodegenerative diseases, including Parkinson's disease (PD), and affects gene expression and splicing. However, the functional impact on neurons of such expression differences has yet to be fully elucidated. Here, we employ extended maturation phases during differentiation of induced pluripotent stem cells (iPSCs) into mature dopaminergic neuronal cultures to obtain cultures expressing all six adult tau protein isoforms. After 6 months of maturation, levels of exon 3+ and exon 10+ transcripts approach those of adult brain. Mature dopaminergic neuronal cultures display haplotype differences in expression, with H1 expressing 22% higher levels of MAPT transcripts than H2 and H2 expressing 2-fold greater exon 3+ transcripts than H1. Furthermore, knocking down adult tau protein variants alters axonal transport velocities in mature iPSC-derived dopaminergic neuronal cultures. This work links haplotype-specific MAPT expression with a biologically functional outcome relevant for PD.
Collapse
Affiliation(s)
- Joel E Beevers
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford OX1 3QX, UK
| | - Mang Ching Lai
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford OX1 3QX, UK
| | - Emma Collins
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford OX1 3QX, UK
| | - Heather D E Booth
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford OX1 3QX, UK
| | - Federico Zambon
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford OX1 3QX, UK
| | - Laura Parkkinen
- Nuffield Department of Clinical Neurosciences, Academic Unit of Neuropathology, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, UK; The Oxford Parkinson's Disease Centre, University of Oxford, Oxford OX1 3QX, UK
| | - Jane Vowles
- The Oxford Parkinson's Disease Centre, University of Oxford, Oxford OX1 3QX, UK; Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford OX1 3RE, UK
| | - Sally A Cowley
- The Oxford Parkinson's Disease Centre, University of Oxford, Oxford OX1 3QX, UK; Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford OX1 3RE, UK
| | - Richard Wade-Martins
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford OX1 3QX, UK; The Oxford Parkinson's Disease Centre, University of Oxford, Oxford OX1 3QX, UK.
| | - Tara M Caffrey
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford OX1 3QX, UK.
| |
Collapse
|
30
|
Monfrini E, Di Fonzo A. Leucine-Rich Repeat Kinase (LRRK2) Genetics and Parkinson's Disease. ADVANCES IN NEUROBIOLOGY 2017; 14:3-30. [PMID: 28353276 DOI: 10.1007/978-3-319-49969-7_1] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The discovery of LRRK2 mutations as a cause of Parkinson's disease (PD), including the sporadic late-onset form, established the decisive role of genetics in the field of PD research. Among LRRK2 mutations, the G2019S, mostly lying in a haplotype originating from a common Middle Eastern ancestor, has been identified in different populations worldwide. The G2385R and R1628P variants represent validated risk factors for PD in Asian populations. Here, we describe in detail the origin, the present worldwide epidemiology, and the penetrance of LRRK2 mutations. Furthermore, this chapter aims to characterize other definitely/probably pathogenic mutations and risk variants of LRRK2. Finally, we provide some general guidelines for a LRRK2 genetic testing and counseling. In summary, LRRK2 discovery revolutionized the understanding of PD etiology and laid the foundation for a promising future of genetics in PD research.
Collapse
Affiliation(s)
- Edoardo Monfrini
- IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico, Dino Ferrari Center, Neuroscience Section, Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Alessio Di Fonzo
- IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico, Dino Ferrari Center, Neuroscience Section, Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy.
| |
Collapse
|
31
|
Back to the tubule: microtubule dynamics in Parkinson's disease. Cell Mol Life Sci 2016; 74:409-434. [PMID: 27600680 PMCID: PMC5241350 DOI: 10.1007/s00018-016-2351-6] [Citation(s) in RCA: 86] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2016] [Revised: 08/18/2016] [Accepted: 08/29/2016] [Indexed: 12/12/2022]
Abstract
Cytoskeletal homeostasis is essential for the development, survival and maintenance of an efficient nervous system. Microtubules are highly dynamic polymers important for neuronal growth, morphology, migration and polarity. In cooperation with several classes of binding proteins, microtubules regulate long-distance intracellular cargo trafficking along axons and dendrites. The importance of a delicate interplay between cytoskeletal components is reflected in several human neurodegenerative disorders linked to abnormal microtubule dynamics, including Parkinson’s disease (PD). Mounting evidence now suggests PD pathogenesis might be underlined by early cytoskeletal dysfunction. Advances in genetics have identified PD-associated mutations and variants in genes encoding various proteins affecting microtubule function including the microtubule-associated protein tau. In this review, we highlight the role of microtubules, their major posttranslational modifications and microtubule associated proteins in neuronal function. We then present key evidence on the contribution of microtubule dysfunction to PD. Finally, we discuss how regulation of microtubule dynamics with microtubule-targeting agents and deacetylase inhibitors represents a promising strategy for innovative therapeutic development.
Collapse
|
32
|
Pascale E, Di Battista ME, Rubino A, Purcaro C, Valente M, Fattapposta F, Ferraguti G, Meco G. Genetic Architecture of MAPT Gene Region in Parkinson Disease Subtypes. Front Cell Neurosci 2016; 10:96. [PMID: 27147968 PMCID: PMC4826864 DOI: 10.3389/fncel.2016.00096] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Accepted: 03/29/2016] [Indexed: 01/30/2023] Open
Abstract
The microtubule-associated protein tau (MAPT) region has been conceptualized as a model of the interaction between genetics and functional disease outcomes in neurodegenerative disorders, such as Parkinson disease (PD). Indeed, haplotype-specific differences in expression and alternative splicing of MAPT transcripts affect cellular functions at different levels, increasing susceptibility to a range of neurodegenerative processes. In order to evaluate a possible link between MAPT variants, PD risk and PD motor phenotype, we analyzed the genetic architecture of MAPT in a cohort of PD patients. We observed a statistically significant association between the H1 haplotype and PD risk (79.5 vs 69.5%; χ2 = 9.9; OR, 1.7; 95% CI, 1.2–2.4; p = 0.002). The effect was more evident in non tremor dominant (TD) PD subjects (NTD-PD) (82 vs 69.5%; χ2 = 13.6; OR, 2.03; 95% CI, 1.4–3; p = 0.0003), while no difference emerged between PD subgroup of tremor dominant patients (TD-PD) and control subjects. Examination of specific intra-H1 variations showed that the H1h subhaplotype was overrepresented in NTD-PD patients compared with controls (p = 0.007; OR, 2.9; 95% CI, 1.3–6.3). Although we cannot exclude that MAPT variation may be associated with ethnicity, our results may support the hypothesis that MAPT H1 clade and a specific H1 subhaplotype influence the risk of PD and modulate the clinical expression of the disease, including motor phenotype.
Collapse
Affiliation(s)
- Esterina Pascale
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University Rome, Italy
| | - Maria Elena Di Battista
- Department of Neurology and Psychiatry (Parkinson's Centre), Sapienza UniversityRome, Italy; Research Centre of Social Diseases (CIMS), Sapienza UniversityRome, Italy
| | - Alfonso Rubino
- Department of Neurology and Psychiatry (Parkinson's Centre), Sapienza UniversityRome, Italy; Research Centre of Social Diseases (CIMS), Sapienza UniversityRome, Italy
| | - Carlo Purcaro
- Department of Neurology and Psychiatry (Parkinson's Centre), Sapienza UniversityRome, Italy; Research Centre of Social Diseases (CIMS), Sapienza UniversityRome, Italy
| | - Marcella Valente
- Department of Neurology and Psychiatry (Parkinson's Centre), Sapienza UniversityRome, Italy; Research Centre of Social Diseases (CIMS), Sapienza UniversityRome, Italy
| | - Francesco Fattapposta
- Department of Neurology and Psychiatry (Parkinson's Centre), Sapienza University Rome, Italy
| | - Giampiero Ferraguti
- Department of Cellular Biotechnologies and Hematology, Sapienza University Rome, Italy
| | - Giuseppe Meco
- Department of Neurology and Psychiatry (Parkinson's Centre), Sapienza UniversityRome, Italy; Research Centre of Social Diseases (CIMS), Sapienza UniversityRome, Italy
| |
Collapse
|
33
|
Collins LM, Williams-Gray CH. The Genetic Basis of Cognitive Impairment and Dementia in Parkinson's Disease. Front Psychiatry 2016; 7:89. [PMID: 27242557 PMCID: PMC4873499 DOI: 10.3389/fpsyt.2016.00089] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Accepted: 05/09/2016] [Indexed: 01/01/2023] Open
Abstract
Cognitive dysfunction is a common feature of Parkinson's disease (PD) with mild cognitive impairment affecting around a quarter of patients in the early stages of their disease, and approximately half developing dementia by 10 years from diagnosis. However, the pattern of cognitive impairments and their speed of evolution vary markedly between individuals. While some of this variability may relate to extrinsic factors and comorbidities, inherited genetic heterogeneity is also known to play an important role. A number of common genetic variants have been identified, which contribute to cognitive function in PD, including variants in catechol-O-methyltransferase, microtubule-associated protein tau, and apolipoprotein E. Furthermore, rarer mutations in glucocerebrosidase and α-synuclein and are strongly associated with dementia risk in PD. This review explores the functional impact of these variants on cognition in PD and discusses how such genotype-phenotype associations provide a window into the mechanistic basis of cognitive heterogeneity in this disorder. This has consequent implications for the development of much more targeted therapeutic strategies for cognitive symptoms in PD.
Collapse
Affiliation(s)
- Lucy M Collins
- John Van Geest Centre for Brain Repair, University of Cambridge , Cambridge , UK
| | | |
Collapse
|
34
|
Abstract
Polymorphic inversions are a type of structural variants that are difficult to analyze owing to their balanced nature and the location of breakpoints within complex repeated regions. So far, only a handful of inversions have been studied in detail in humans and current knowledge about their possible functional effects is still limited. However, inversions have been related to phenotypic changes and adaptation in multiple species. In this review, we summarize the evidences of the functional impact of inversions in the human genome. First, given that inversions have been shown to inhibit recombination in heterokaryotes, chromosomes displaying different orientation are expected to evolve independently and this may lead to distinct gene-expression patterns. Second, inversions have a role as disease-causing mutations both by directly affecting gene structure or regulation in different ways, and by predisposing to other secondary arrangements in the offspring of inversion carriers. Finally, several inversions show signals of being selected during human evolution. These findings illustrate the potential of inversions to have phenotypic consequences also in humans and emphasize the importance of their inclusion in genome-wide association studies.
Collapse
|
35
|
Caillet-Boudin ML, Buée L, Sergeant N, Lefebvre B. Regulation of human MAPT gene expression. Mol Neurodegener 2015; 10:28. [PMID: 26170022 PMCID: PMC4499907 DOI: 10.1186/s13024-015-0025-8] [Citation(s) in RCA: 127] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Accepted: 06/30/2015] [Indexed: 12/12/2022] Open
Abstract
The number of known pathologies involving deregulated Tau expression/metabolism is increasing. Indeed, in addition to tauopathies, which comprise approximately 30 diseases characterized by neuronal aggregation of hyperphosphorylated Tau in brain neurons, this protein has also been associated with various other pathologies such as cancer, inclusion body myositis, and microdeletion/microduplication syndromes, suggesting its possible function in peripheral tissues. In addition to Tau aggregation, Tau deregulation can occur at the expression and/or splicing levels, as has been clearly demonstrated in some of these pathologies. Here, we aim to review current knowledge regarding the regulation of human MAPT gene expression at the DNA and RNA levels to provide a better understanding of its possible deregulation. Several aspects, including repeated motifs, CpG island/methylation, and haplotypes at the DNA level, as well as the key regions involved in mRNA expression and stability and the splicing patterns of different mRNA isoforms at the RNA level, will be discussed.
Collapse
Affiliation(s)
| | - Luc Buée
- Univ. Lille, UMR-S 1172, Inserm, CHU, 59000, Lille, France
| | | | - Bruno Lefebvre
- Univ. Lille, UMR-S 1172, Inserm, CHU, 59000, Lille, France
| |
Collapse
|
36
|
Golpich M, Amini E, Hemmati F, Ibrahim NM, Rahmani B, Mohamed Z, Raymond AA, Dargahi L, Ghasemi R, Ahmadiani A. Glycogen synthase kinase-3 beta (GSK-3β) signaling: Implications for Parkinson's disease. Pharmacol Res 2015; 97:16-26. [PMID: 25829335 DOI: 10.1016/j.phrs.2015.03.010] [Citation(s) in RCA: 194] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Revised: 03/05/2015] [Accepted: 03/16/2015] [Indexed: 01/02/2023]
Abstract
Glycogen synthase kinase 3 (GSK-3) dysregulation plays an important role in the pathogenesis of numerous disorders, affecting the central nervous system (CNS) encompassing both neuroinflammation and neurodegenerative diseases. Several lines of evidence have illustrated a key role of the GSK-3 and its cellular and molecular signaling cascades in the control of neuroinflammation. Glycogen synthase kinase 3 beta (GSK-3β), one of the GSK-3 isomers, plays a major role in neuronal apoptosis and its inhibition decreases expression of alpha-Synuclein (α-Synuclein), which make this kinase an attractive therapeutic target for neurodegenerative disorders. Parkinson's disease (PD) is a chronic neurodegenerative movement disorder characterized by the progressive and massive loss of dopaminergic neurons by neuronal apoptosis in the substantia nigra pars compacta and depletion of dopamine in the striatum, which lead to pathological and clinical abnormalities. Thus, understanding the role of GSK-3β in PD will enhance our knowledge of the basic mechanisms underlying the pathogenesis of this disorder and facilitate the identification of new therapeutic avenues. In recent years, GSK-3β has been shown to play essential roles in modulating a variety of cellular functions, which have prompted efforts to develop GSK-3β inhibitors as therapeutics. In this review, we summarize GSK-3 signaling pathways and its association with neuroinflammation. Moreover, we highlight the interaction between GSK-3β and several cellular processes involved in the pathogenesis of PD, including the accumulation of α-Synuclein aggregates, oxidative stress and mitochondrial dysfunction. Finally, we discuss about GSK-3β inhibitors as a potential therapeutic strategy in PD.
Collapse
Affiliation(s)
- Mojtaba Golpich
- Department of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Cheras, Kuala Lumpur, Malaysia
| | - Elham Amini
- Department of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Cheras, Kuala Lumpur, Malaysia
| | - Fatemeh Hemmati
- Department of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Cheras, Kuala Lumpur, Malaysia
| | - Norlinah Mohamed Ibrahim
- Department of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Cheras, Kuala Lumpur, Malaysia
| | - Behrouz Rahmani
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zahurin Mohamed
- Department of Pharmacology, Faculty of Medicine, University of Malaya, 50603, Kuala Lumpur, Malaysia
| | - Azman Ali Raymond
- Department of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Cheras, Kuala Lumpur, Malaysia
| | - Leila Dargahi
- NeuroBiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Rasoul Ghasemi
- Neurophysiology Research Center and Department of Physiology, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Abolhassan Ahmadiani
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Department of Pharmacology, Faculty of Medicine, University of Malaya, 50603, Kuala Lumpur, Malaysia.
| |
Collapse
|
37
|
Winder-Rhodes SE, Hampshire A, Rowe JB, Peelle JE, Robbins TW, Owen AM, Barker RA. Association between MAPT haplotype and memory function in patients with Parkinson's disease and healthy aging individuals. Neurobiol Aging 2015; 36:1519-28. [PMID: 25577413 PMCID: PMC4353560 DOI: 10.1016/j.neurobiolaging.2014.12.006] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2014] [Revised: 11/27/2014] [Accepted: 12/04/2014] [Indexed: 11/23/2022]
Abstract
Genetic variation is associated with differences in the function of the brain as well as its susceptibility to disease. The common H1 haplotypic variant of the microtubule-associated protein tau gene (MAPT) has been related to an increased risk for Parkinson's disease (PD). Furthermore, among PD patients, H1 homozygotes have an accelerated progression to dementia. We investigated the neurocognitive correlates of MAPT haplotypes using functional magnetic resonance imaging. Thirty-seven nondemented patients with PD (19 H1/H1, 18 H2 carriers) and 40 age-matched controls (21 H1/H1, 19 H2 carriers) were scanned during performance of a picture memory encoding task. Behaviorally, H1 homozygosity was associated with impaired picture recognition memory in PD patients and control subjects. These impairments in the H1 homozygotes were accompanied by an altered blood-oxygen level-dependent response in the medial temporal lobe during successful memory encoding. Additional age-related differences in blood-oxygen level-dependent response were observed in the medial temporal lobes of H1 homozygotes with PD. These results suggest that common variation in MAPT is not only associated with the dementia of PD but also differences in the neural circuitry underlying aspects of cognition in normal aging.
Collapse
Affiliation(s)
- Sophie E Winder-Rhodes
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK; Department of Child and Adolescent Psychiatry, Institute of Psychiatry, King's College London, UK.
| | - Adam Hampshire
- MRC Cognition and Brain Sciences Unit, Cambridge, UK; The Division of Brain Sciences, Department of Medicine, Imperial College, London, UK
| | - James B Rowe
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK; MRC Cognition and Brain Sciences Unit, Cambridge, UK; Behavioural and Clinical Neuroscience Institute, University of Cambridge, Cambridge, UK
| | - Jonathan E Peelle
- Department of Otolaryngology, Washington University in St. Louis, St. Louis, MO, USA
| | - Trevor W Robbins
- Behavioural and Clinical Neuroscience Institute, University of Cambridge, Cambridge, UK
| | - Adrian M Owen
- MRC Cognition and Brain Sciences Unit, Cambridge, UK; The Brain and Mind Institute, The University of Western Ontario, London Ontario, Canada
| | - Roger A Barker
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK; Behavioural and Clinical Neuroscience Institute, University of Cambridge, Cambridge, UK.
| |
Collapse
|
38
|
Kanaan NM, Himmelstein DS, Ward SM, Combs B, Binder LI. Tau Protein. Mov Disord 2015. [DOI: 10.1016/b978-0-12-405195-9.00056-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
|
39
|
Sonmez E, Ozel MD, Islek EE, Sazci A, Idrisoglu HA. Association of rs62063857 variant of the saitohin gene with Parkinson's disease. Cell Mol Neurobiol 2015; 35:115-21. [PMID: 25168738 PMCID: PMC11486326 DOI: 10.1007/s10571-014-0102-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2014] [Accepted: 08/20/2014] [Indexed: 10/24/2022]
Abstract
Saitohin gene found within the tau gene is thought to play a role in the pathogenesis of neurodegenerative diseases. The rs62063857 polymorphism originally found in the saitohin gene seems to be the responsible SNP in this event. This polymorphism is studied mostly in patients with Alzheimer's disease. Data on Parkinson's disease are scarce. Therefore, we examined the rs62063857 polymorphism in 583 Parkinson's disease patients (347 male and 236 female) and 396 healthy controls (238 male and 158 female) by a polymerase chain reaction and restriction fragment length polymorphism method to see whether it was associated with Parkinson's disease from the City of Istanbul, Turkey. The G allele frequency was 22 % in overall controls and 16 % in Parkinson's disease patients. In this study, the saitohin rs62063857 polymorphism was associated with Parkinson's disease (χ2 = 16.765; P = 0.000). Individuals with the AA genotype showed 1.7-fold increased risk for Parkinson's disease (χ2 = 16.680; P = 0.000), whereas individuals with the AG genotype revealed protection against Parkinson's disease (χ2 = 14.554; P = 0.000). After the stratification analysis according to gender, both male and female PD patients showed association with the alleles and genotypes of the rs62063857 polymorphism of the saitohin gene (χ2 = 9.476, P = 0.009; χ2 = 7.593, P = 0.022, respectively). When the Parkinson's patients were divided into two groups with regard to onset of the disease, both groups showed association with the disease. The Parkinson's patients with disease onset below 65 years of age showed 1.8-fold increased risk for the disease. The Parkinson's patients with disease onset over 65 showed more robust association with a 2.051-fold increased risk for the disease. Consequently, the rs62063857 polymorphism of the saitohin gene is a genetic risk factor for Parkinson's disease. Hence, this polymorphism may play a role in the etiology of Parkinson's disease.
Collapse
Affiliation(s)
- Ezgi Sonmez
- Departments of Medical Biology and Genetics, Faculty of Medicine, University of Kocaeli, Kocaeli, Turkey
| | - Mavi Deniz Ozel
- Departments of Medical Biology and Genetics, Faculty of Medicine, University of Kocaeli, Kocaeli, Turkey
| | - Eylul Ece Islek
- Departments of Medical Biology and Genetics, Faculty of Medicine, University of Kocaeli, Kocaeli, Turkey
| | - Ali Sazci
- Departments of Medical Biology and Genetics, Faculty of Medicine, University of Kocaeli, Kocaeli, Turkey
| | - Halil Atilla Idrisoglu
- Department of Neurology, Istanbul Faculty of Medicine, University of Istanbul, Capa, Istanbul Turkey
| |
Collapse
|
40
|
Friesen DE, Craddock TJA, Kalra AP, Tuszynski JA. Biological wires, communication systems, and implications for disease. Biosystems 2014; 127:14-27. [PMID: 25448891 DOI: 10.1016/j.biosystems.2014.10.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Revised: 10/30/2014] [Accepted: 10/31/2014] [Indexed: 12/14/2022]
Abstract
Microtubules, actin, and collagen are macromolecular structures that compose a large percentage of the proteins in the human body, helping form and maintain both intracellular and extracellular structure. They are biological wires and are structurally connected through various other proteins. Microtubules (MTs) have been theorized to be involved in classical and quantum information processing, and evidence continues to suggest possible semiconduction through MTs. The previous Dendritic Cytoskeleton Information Processing Model has hypothesized how MTs and actin form a communication network in neurons. Here, we review information transfer possibilities involving MTs, actin, and collagen, and the evidence of an organism-wide high-speed communication network that may regulate morphogenesis and cellular proliferation. The direct and indirect evidence in support of this hypothesis, and implications for chronic diseases such as cancer and neurodegenerative diseases are discussed.
Collapse
Affiliation(s)
- Douglas E Friesen
- Department of Oncology, University of Alberta, Edmonton, Alberta T6G 1Z2, Canada
| | - Travis J A Craddock
- Center for Psychological Studies, Graduate School of Computer and Information Sciences, College of Osteopathic Medicine, Nova Southeastern University, Ft. Lauderdale, FL 33314, USA; Clinical Systems Biology Group, Institute for Neuro-Immune Medicine, Nova Southeastern University, Ft. Lauderdale, FL 33328, USA
| | - Aarat P Kalra
- Department of Chemistry, Dayalbagh Educational Institute, Agra 282005, India
| | - Jack A Tuszynski
- Department of Oncology, University of Alberta, Edmonton, Alberta T6G 1Z2, Canada; Department of Physics, University of Alberta, Edmonton, Alberta T6G 2E1, Canada.
| |
Collapse
|
41
|
Lu SS, Gong FF, Feng F, Hu CY, Qian ZZ, Wu YL, Yang HY, Sun YH. Association of microtubule associated protein tau/Saitohin (MAPT/STH) MAPT_238bp/STH Q7R polymorphisms and Parkinson’s disease: A meta-analysis. Biochem Biophys Res Commun 2014; 453:653-61. [DOI: 10.1016/j.bbrc.2014.10.013] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2014] [Accepted: 10/03/2014] [Indexed: 01/14/2023]
|
42
|
Zhang X, Moen EL, Liu C, Mu W, Gamazon ER, Delaney SM, Wing C, Godley LA, Dolan ME, Zhang W. Linking the genetic architecture of cytosine modifications with human complex traits. Hum Mol Genet 2014; 23:5893-905. [PMID: 24943591 DOI: 10.1093/hmg/ddu313] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Interindividual variation in cytosine modifications could contribute to heterogeneity in disease risks and other complex traits. We assessed the genetic architecture of cytosine modifications at 283,540 CpG sites in lymphoblastoid cell lines (LCLs) derived from independent samples of European and African descent. Our study suggests that cytosine modification variation was primarily controlled in local by single major modification quantitative trait locus (mQTL) and additional minor loci. Local genetic epistasis was detectable for a small proportion of CpG sites, which were enriched by more than 9-fold for CpG sites mapped to population-specific mQTL. Genetically dependent CpG sites whose modification levels negatively (repressive sites) or positively (facilitative sites) correlated with gene expression levels significantly co-localized with transcription factor binding, with the repressive sites predominantly associated with active promoters whereas the facilitative sites rarely at active promoters. Genetically independent repressive or facilitative sites preferentially modulated gene expression variation by influencing local chromatin accessibility, with the facilitative sites primarily antagonizing H3K27me3 and H3K9me3 deposition. In comparison with expression quantitative trait loci (eQTL), mQTL detected from LCLs were enriched in associations for a broader range of disease categories including chronic inflammatory, autoimmune and psychiatric disorders, suggesting that cytosine modification variation, while possesses a degree of cell linage specificity, is more stably inherited over development than gene expression variation. About 11% of unique single-nucleotide polymorphisms reported in the Genome-Wide Association Study Catalog were annotated, 78% as mQTL and 31% as eQTL in LCLs, which covered 37% of the investigated diseases/traits and provided insights to the biological mechanisms.
Collapse
Affiliation(s)
- Xu Zhang
- Section of Hematology/Oncology, Department of Medicine
| | | | | | | | | | | | - Claudia Wing
- Section of Hematology/Oncology, Department of Medicine and
| | - Lucy A Godley
- Section of Hematology/Oncology, Department of Medicine and Comprehensive Cancer Center, The University of Chicago, Chicago, IL 60637, USA
| | - M Eileen Dolan
- Section of Hematology/Oncology, Department of Medicine and Comprehensive Cancer Center, The University of Chicago, Chicago, IL 60637, USA
| | - Wei Zhang
- Department of Pediatrics, Institute of Human Genetics, The University of Illinois, Chicago, IL 60612, USA,
| |
Collapse
|
43
|
Grünblatt E. Commonalities in the genetics of Alzheimer’s disease and Parkinson’s disease. Expert Rev Neurother 2014; 8:1865-77. [DOI: 10.1586/14737175.8.12.1865] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
44
|
Coupland KG, Mellick GD, Silburn PA, Mather K, Armstrong NJ, Sachdev PS, Brodaty H, Huang Y, Halliday GM, Hallupp M, Kim WS, Dobson-Stone C, Kwok JBJ. DNA methylation of the MAPT gene in Parkinson's disease cohorts and modulation by vitamin E in vitro. Mov Disord 2013; 29:1606-14. [PMID: 24375821 DOI: 10.1002/mds.25784] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2013] [Revised: 10/10/2013] [Accepted: 10/21/2013] [Indexed: 01/08/2023] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder for which environmental factors influence disease risk and may act via an epigenetic mechanism. The microtubule-associated protein tau (MAPT) is a susceptibility gene for idiopathic PD. Methylation levels were determined by pyrosequencing of bisulfite-treated DNA in a leukocyte cohort (358 PD patients and 1084 controls) and in two brain cohorts (Brain1, comprising 69 cerebellum controls; and Brain2, comprising 3 brain regions from 28 PD patients and 12 controls). In vitro assays involved the transfection of methylated promoter-luciferase constructs or treatment with an exogenous micronutrient. In normal leukocytes, the MAPT H1/H2 diplotype and sex were predictors of MAPT methylation. Haplotype-specific pyrosequencing confirmed that the H1 haplotype had higher methylation than the H2 haplotype in normal leukocytes and brain tissues. MAPT methylation was negatively associated with MAPT expression in the Brain1 cohort and in transfected cells. Methylation levels differed between three normal brain regions (Brain2 cohort, putamen < cerebellum < anterior cingulate cortex). In PD samples, age at onset was positively associated with MAPT methylation in leukocytes. Moreover, there was hypermethylation in the cerebellum and hypomethylation in the putamen of PD patients compared with controls (Brain2 cohort). Finally, leukocyte methylation status was positively associated with blood vitamin E levels, and the effect was more significant in H2 haplotype carriers; this result was confirmed in cells that were exposed to 100 μM vitamin E. The significant effects of sex, diplotype, and brain region suggest that hypermethylation of the MAPT gene is neuroprotective by reducing MAPT expression. The effect of vitamin E on MAPT represents a possible gene-environment interaction.
Collapse
Affiliation(s)
- Kirsten G Coupland
- Neuroscience Research Australia, Sydney, New South Wales, Australia; School of Medical Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Orozco D, Edbauer D. FUS-mediated alternative splicing in the nervous system: consequences for ALS and FTLD. J Mol Med (Berl) 2013; 91:1343-54. [PMID: 23974990 DOI: 10.1007/s00109-013-1077-2] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2013] [Revised: 08/01/2013] [Accepted: 08/06/2013] [Indexed: 12/13/2022]
Abstract
Mutations in fused in sarcoma (FUS) in a subset of patients with amyotrophic lateral sclerosis (ALS) linked this DNA/RNA-binding protein to neurodegeneration. Most of the mutations disrupt the nuclear localization signal which strongly suggests a loss-of-function pathomechanism, supported by cytoplasmic inclusions. FUS-positive neuronal cytoplasmic inclusions are also found in a subset of patients with frontotemporal lobar degeneration (FTLD). Here, we discuss recent data on the role of alternative splicing in FUS-mediated pathology in the central nervous system. Several groups have shown that FUS binds broadly to many transcripts in the brain and have also identified a plethora of putative splice targets; however, only ABLIM1, BRAF, Ewing sarcoma protein R1 (EWSR1), microtubule-associated protein tau (MAPT), NgCAM cell adhesion molecule (NRCAM), and netrin G1 (NTNG1) have been identified in at least three of four studies. Gene ontology analysis of all putative targets unanimously suggests a role in axon growth and cytoskeletal organization, consistent with the altered morphology of dendritic spines and axonal growth cones reported upon loss of FUS. Among the axonal targets, MAPT/tau and NTNG1 have been further validated in biochemical studies. The next challenge will be to confirm changes of FUS-mediated alternative splicing in patients and define their precise role in the pathophysiology of ALS and FTLD.
Collapse
Affiliation(s)
- Denise Orozco
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | | |
Collapse
|
46
|
Heckman MG, Elbaz A, Soto-Ortolaza AI, Serie DJ, Aasly JO, Annesi G, Auburger G, Bacon JA, Boczarska-Jedynak M, Bozi M, Brighina L, Chartier-Harlin MC, Dardiotis E, Destée A, Ferrarese C, Ferraris A, Fiske B, Gispert S, Hadjigeorgiou GM, Hattori N, Ioannidis JPA, Jasinska-Myga B, Jeon BS, Kim YJ, Klein C, Kruger R, Kyratzi E, Lin CH, Lohmann K, Loriot MA, Lynch T, Mellick GD, Mutez E, Opala G, Park SS, Petrucci S, Quattrone A, Sharma M, Silburn PA, Sohn YH, Stefanis L, Tadic V, Tomiyama H, Uitti RJ, Valente EM, Vassilatis DK, Vilariño-Güell C, White LR, Wirdefeldt K, Wszolek ZK, Wu RM, Xiromerisiou G, Maraganore DM, Farrer MJ, Ross OA. Protective effect of LRRK2 p.R1398H on risk of Parkinson's disease is independent of MAPT and SNCA variants. Neurobiol Aging 2013; 35:266.e5-14. [PMID: 23962496 DOI: 10.1016/j.neurobiolaging.2013.07.013] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2013] [Revised: 07/10/2013] [Accepted: 07/15/2013] [Indexed: 11/19/2022]
Abstract
The best validated susceptibility variants for Parkinson's disease are located in the α-synuclein (SNCA) and microtubule-associated protein tau (MAPT) genes. Recently, a protective p.N551K-R1398H-K1423K haplotype in the leucine-rich repeat kinase 2 (LRRK2) gene was identified, with p.R1398H appearing to be the most likely functional variant. To date, the consistency of the protective effect of LRRK2 p.R1398H across MAPT and SNCA variant genotypes has not been assessed. To address this, we examined 4 SNCA variants (rs181489, rs356219, rs11931074, and rs2583988), the MAPT H1-haplotype-defining variant rs1052553, and LRRK2 p.R1398H (rs7133914) in Caucasian (n = 10,322) and Asian (n = 2289) series. There was no evidence of an interaction of LRRK2 p.R1398H with MAPT or SNCA variants (all p ≥ 0.10); the protective effect of p.R1398H was observed at similar magnitude across MAPT and SNCA genotypes, and the risk effects of MAPT and SNCA variants were observed consistently for LRRK2 p.R1398H genotypes. Our results indicate that the association of LRRK2 p.R1398H with Parkinson's disease is independent of SNCA and MAPT variants, and vice versa, in Caucasian and Asian populations.
Collapse
|
47
|
Agúndez JAG, García-Martín E, Martínez C, Benito-León J, Millán-Pascual J, Calleja P, Díaz-Sánchez M, Pisa D, Turpín-Fenoll L, Alonso-Navarro H, Ayuso-Peralta L, Torrecillas D, Plaza-Nieto JF, Jiménez-Jiménez FJ. MAPT gene rs1052553 variant is not associated with the risk for multiple sclerosis. Hum Immunol 2013; 74:1705-8. [PMID: 23911736 DOI: 10.1016/j.humimm.2013.07.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2013] [Revised: 07/08/2013] [Accepted: 07/19/2013] [Indexed: 02/08/2023]
Abstract
BACKGROUND/OBJECTIVES Some experimental data suggest a possible role of tau protein in the pathogenesis of multiple sclerosis (MS) and in experimental autoimmune encephalomyelitis. The aim of this study was to investigate a possible influence of the SNP rs1052553 in the MAPT gene in the risk for relapsing bout onset (relapsing-remitting and secondary progressive) MS. METHODS We analyzed the allelic and genotype frequency of MAPT rs1052553, which has been associated with some neurodegenerative diseases, in 259 patients with relapsing bout onset MS and 291 healthy controls, using TaqMan Assays. RESULTS MAPT rs1052553 allelic and genotype frequencies did not differ significantly between relapsing bout onset MS patients and controls, and were unrelated with the age of onset of MS or gender. CONCLUSIONS These results suggest that MAPT rs1052553 polymorphism is not related with the risk for relapsing bout onset MS.
Collapse
Affiliation(s)
- José A G Agúndez
- Department of Pharmacology, University of Extremadura, Cáceres, Spain
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
MAPT H1 haplotype is associated with enhanced α-synuclein deposition in dementia with Lewy bodies. Neurobiol Aging 2013; 34:936-42. [DOI: 10.1016/j.neurobiolaging.2012.06.015] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2012] [Revised: 06/15/2012] [Accepted: 06/21/2012] [Indexed: 11/20/2022]
|
49
|
Hebron ML, Algarzae NK, Lonskaya I, Moussa C. Fractalkine signaling and Tau hyper-phosphorylation are associated with autophagic alterations in lentiviral Tau and Aβ1-42 gene transfer models. Exp Neurol 2013; 251:127-38. [PMID: 23333589 DOI: 10.1016/j.expneurol.2013.01.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2012] [Revised: 01/03/2013] [Accepted: 01/09/2013] [Indexed: 12/17/2022]
Abstract
Tau hyper-phosphorylation (p-Tau) and neuro-inflammation are hallmarks of neurodegeneration. Previous findings suggest that microglial activation via CX3CL1 promotes p-Tau. We examined inflammation and autophagic p-Tau clearance in lentiviral Tau and mutant P301L expressing rats and used lentiviral Aβ1-42 to induce p-Tau. Lentiviral Tau or P301L expression significantly increased caspase-3 activity and TNF-α, but CX3CL1 was significantly higher in animals expressing Tau compared to P301L. Lentiviral Aβ1-42 induced p-Tau 4 weeks post-injection, and increased caspase-3 activation (8-fold) and TNF-α levels. Increased levels of ADAM-10/17 were also detected with p-Tau. IL-6 levels were increased but CX3CL1 did not change in the absence of p-Tau (2 weeks); however, p-Tau reversed these effects, which were associated with increased microglial activity. We observed changes in autophagic markers, including accumulation of autophagic vacuoles (AVs) and p-Tau accumulation in autophagosomes but not lysosomes, suggesting alteration of autophagy. Taken together, microglial activation may promote p-Tau independent of total Tau levels via CX3CL1 signaling, which seems to depend on interaction with inflammatory markers, mainly IL-6. The simultaneous change in autophagy and CX3CL1 signaling suggests communication between microglia and neurons, raising the possibility that accumulation of intraneuronal amyloid, due to lack of autophagic clearance, may lead microglia activation to promote p-Tau as a tag for phagocytic degradation.
Collapse
Affiliation(s)
- Michaeline L Hebron
- Department of Neuroscience, Georgetown University Medical Center, Washington D.C. 20007, USA; Department of Biochemistry and Cell Biology, Georgetown University Medical Center, Washington D.C. 20007, USA
| | - Norah K Algarzae
- Department of Neuroscience, Georgetown University Medical Center, Washington D.C. 20007, USA; Department of Biochemistry and Cell Biology, Georgetown University Medical Center, Washington D.C. 20007, USA
| | - Irina Lonskaya
- Department of Neuroscience, Georgetown University Medical Center, Washington D.C. 20007, USA
| | - Charbel Moussa
- Department of Neuroscience, Georgetown University Medical Center, Washington D.C. 20007, USA.
| |
Collapse
|
50
|
Permuth-Wey J, Lawrenson K, Shen HC, Velkova A, Tyrer JP, Chen Z, Lin HY, Chen YA, Tsai YY, Qu X, Ramus SJ, Karevan R, Lee J, Lee N, Larson MC, Aben KK, Anton-Culver H, Antonenkova N, Antoniou A, Armasu SM, Bacot F, Baglietto L, Bandera EV, Barnholtz-Sloan J, Beckmann MW, Birrer MJ, Bloom G, Bogdanova N, Brinton LA, Brooks-Wilson A, Brown R, Butzow R, Cai Q, Campbell I, Chang-Claude J, Chanock S, Chenevix-Trench G, Cheng JQ, Cicek MS, Coetzee GA, Cook LS, Couch FJ, Cramer DW, Cunningham JM, Dansonka-Mieszkowska A, Despierre E, Doherty JA, Dörk T, du Bois A, Dürst M, Easton DF, Eccles D, Edwards R, Ekici AB, Fasching PA, Fenstermacher DA, Flanagan JM, Garcia-Closas M, Gentry-Maharaj A, Giles GG, Glasspool RM, Gonzalez-Bosquet J, Goodman MT, Gore M, Górski B, Gronwald J, Hall P, Halle MK, Harter P, Heitz F, Hillemanns P, Hoatlin M, Høgdall CK, Høgdall E, Hosono S, Jakubowska A, Jensen A, Jim H, Kalli KR, Karlan BY, Kaye SB, Kelemen LE, Kiemeney LA, Kikkawa F, Konecny GE, Krakstad C, Kjaer SK, Kupryjanczyk J, Lambrechts D, Lambrechts S, Lancaster JM, Le ND, Leminen A, Levine DA, Liang D, Lim BK, Lin J, Lissowska J, Lu KH, Lubiński J, et alPermuth-Wey J, Lawrenson K, Shen HC, Velkova A, Tyrer JP, Chen Z, Lin HY, Chen YA, Tsai YY, Qu X, Ramus SJ, Karevan R, Lee J, Lee N, Larson MC, Aben KK, Anton-Culver H, Antonenkova N, Antoniou A, Armasu SM, Bacot F, Baglietto L, Bandera EV, Barnholtz-Sloan J, Beckmann MW, Birrer MJ, Bloom G, Bogdanova N, Brinton LA, Brooks-Wilson A, Brown R, Butzow R, Cai Q, Campbell I, Chang-Claude J, Chanock S, Chenevix-Trench G, Cheng JQ, Cicek MS, Coetzee GA, Cook LS, Couch FJ, Cramer DW, Cunningham JM, Dansonka-Mieszkowska A, Despierre E, Doherty JA, Dörk T, du Bois A, Dürst M, Easton DF, Eccles D, Edwards R, Ekici AB, Fasching PA, Fenstermacher DA, Flanagan JM, Garcia-Closas M, Gentry-Maharaj A, Giles GG, Glasspool RM, Gonzalez-Bosquet J, Goodman MT, Gore M, Górski B, Gronwald J, Hall P, Halle MK, Harter P, Heitz F, Hillemanns P, Hoatlin M, Høgdall CK, Høgdall E, Hosono S, Jakubowska A, Jensen A, Jim H, Kalli KR, Karlan BY, Kaye SB, Kelemen LE, Kiemeney LA, Kikkawa F, Konecny GE, Krakstad C, Kjaer SK, Kupryjanczyk J, Lambrechts D, Lambrechts S, Lancaster JM, Le ND, Leminen A, Levine DA, Liang D, Lim BK, Lin J, Lissowska J, Lu KH, Lubiński J, Lurie G, Massuger LF, Matsuo K, McGuire V, McLaughlin JR, Menon U, Modugno F, Moysich KB, Nakanishi T, Narod SA, Nedergaard L, Ness RB, Nevanlinna H, Nickels S, Noushmehr H, Odunsi K, Olson SH, Orlow I, Paul J, Pearce CL, Pejovic T, Pelttari LM, Pike MC, Poole EM, Raska P, Renner SP, Risch HA, Rodriguez-Rodriguez L, Rossing MA, Rudolph A, Runnebaum IB, Rzepecka IK, Salvesen HB, Schwaab I, Severi G, Shridhar V, Shu XO, Shvetsov YB, Sieh W, Song H, Southey MC, Spiewankiewicz B, Stram D, Sutphen R, Teo SH, Terry KL, Tessier DC, Thompson PJ, Tworoger SS, van Altena AM, Vergote I, Vierkant RA, Vincent D, Vitonis AF, Wang-Gohrke S, Weber RP, Wentzensen N, Whittemore AS, Wik E, Wilkens LR, Winterhoff B, Woo YL, Wu AH, Xiang YB, Yang HP, Zheng W, Ziogas A, Zulkifli F, Phelan CM, Iversen E, Schildkraut JM, Berchuck A, Fridley BL, Goode EL, Pharoah PDP, Monteiro AN, Sellers TA, Gayther SA. Identification and molecular characterization of a new ovarian cancer susceptibility locus at 17q21.31. Nat Commun 2013; 4:1627. [PMID: 23535648 PMCID: PMC3709460 DOI: 10.1038/ncomms2613] [Show More Authors] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2012] [Accepted: 02/18/2013] [Indexed: 12/20/2022] Open
Abstract
Epithelial ovarian cancer (EOC) has a heritable component that remains to be fully characterized. Most identified common susceptibility variants lie in non-protein-coding sequences. We hypothesized that variants in the 3' untranslated region at putative microRNA (miRNA)-binding sites represent functional targets that influence EOC susceptibility. Here, we evaluate the association between 767 miRNA-related single-nucleotide polymorphisms (miRSNPs) and EOC risk in 18,174 EOC cases and 26,134 controls from 43 studies genotyped through the Collaborative Oncological Gene-environment Study. We identify several miRSNPs associated with invasive serous EOC risk (odds ratio=1.12, P=10(-8)) mapping to an inversion polymorphism at 17q21.31. Additional genotyping of non-miRSNPs at 17q21.31 reveals stronger signals outside the inversion (P=10(-10)). Variation at 17q21.31 is associated with neurological diseases, and our collaboration is the first to report an association with EOC susceptibility. An integrated molecular analysis in this region provides evidence for ARHGAP27 and PLEKHM1 as candidate EOC susceptibility genes.
Collapse
Affiliation(s)
- Jennifer Permuth-Wey
- Department of Cancer Epidemiology, Division of Population Sciences, Moffitt Cancer Center, Tampa, FL, USA, 33612
| | - Kate Lawrenson
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California Norris Comprehensive Cancer Center, Los Angeles, CA, USA, 90033
| | - Howard C. Shen
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California Norris Comprehensive Cancer Center, Los Angeles, CA, USA, 90033
| | - Aneliya Velkova
- Department of Cancer Epidemiology, Division of Population Sciences, Moffitt Cancer Center, Tampa, FL, USA, 33612
| | - Jonathan P. Tyrer
- Department of Oncology, University of Cambridge, Cambridge, CB1 8RN, UK
| | - Zhihua Chen
- Department of Biomedical Informatics, Moffitt Cancer Center, Tampa, FL, USA, 33612
| | - Hui-Yi Lin
- Department of Biostatistics, Moffitt Cancer Center, Tampa, FL, USA, 33612
| | - Y. Ann Chen
- Department of Biostatistics, Moffitt Cancer Center, Tampa, FL, USA, 33612
| | - Ya-Yu Tsai
- Department of Cancer Epidemiology, Division of Population Sciences, Moffitt Cancer Center, Tampa, FL, USA, 33612
| | - Xiaotao Qu
- Department of Biomedical Informatics, Moffitt Cancer Center, Tampa, FL, USA, 33612
| | - Susan J. Ramus
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California Norris Comprehensive Cancer Center, Los Angeles, CA, USA, 90033
| | - Rod Karevan
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California Norris Comprehensive Cancer Center, Los Angeles, CA, USA, 90033
| | - Janet Lee
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California Norris Comprehensive Cancer Center, Los Angeles, CA, USA, 90033
| | - Nathan Lee
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California Norris Comprehensive Cancer Center, Los Angeles, CA, USA, 90033
| | - Melissa C. Larson
- Department of Health Science Research, Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN, USA, 55905
| | - Katja K. Aben
- Department of Epidemiology, Biostatistics and HTA, Radboud University Medical Centre, Nijmegen, HB 6500, Netherlands
- Comprehensive Cancer Center, the Netherlands, Utrecht, Amsterdam, 1066CX, The Netherlands
| | - Hoda Anton-Culver
- Department of Epidemiology, Director of Genetic Epidemiology Research Institute, UCI Center of Medicine, University of California Irvine, Irvine, CA, USA, 92697
| | - Natalia Antonenkova
- Byelorussian Institute for Oncology and Medical Radiology Aleksandrov N.N., 223040, Minsk, Belarus
| | - Antonis Antoniou
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, CB1 8RN, UK
| | - Sebastian M. Armasu
- Department of Health Science Research, Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN, USA, 55905
| | | | - Australian Ovarian Cancer Study
- Queensland Institute of Medical Research, Brisbane QLD 4006, Australia
- Cancer Genetics Laboratory, Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC 3002, Australia
| | - François Bacot
- McGill University and Génome Québec Innovation Centre, Montréal (Québec) Canada, H3A 0G1
| | - Laura Baglietto
- Cancer Epidemiology Centre, The Cancer Council Victoria, Melbourne, Carlton VIC 3053, Australia
- Centre for Molecular, Environmental, Genetic and Analytical Epidemiology, University of Melbourne, Melbourne, VIC 3010, Australia
| | - Elisa V. Bandera
- The Cancer Institute of New Jersey, Robert Wood Johnson Medical School, New Brunswick, NJ, USA, 08901
| | - Jill Barnholtz-Sloan
- Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, OH, USA, 44195
| | - Matthias W. Beckmann
- University Hospital Erlangen, Department of Gynecology and Obstetrics, Friedrich-Alexander-University Erlangen-Nuremberg, Comprehensive Cancer Center, Erlangen, 91054, Germany
| | | | - Greg Bloom
- Department of Biomedical Informatics, Moffitt Cancer Center, Tampa, FL, USA, 33612
| | - Natalia Bogdanova
- Gynaecology Research Unit, Hannover Medical School, Hannover, 30625, Germany
| | - Louise A. Brinton
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda MD, USA, 20892
| | | | - Robert Brown
- Department of Surgery and Cancer, Imperial College London, London, SW7 2AZ, UK
| | - Ralf Butzow
- Department of Pathology, Helsinki University Central Hospital, Helsinki, Finland, 00530
- Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Central Hospital, Helsinki, Finland, 00530
| | - Qiuyin Cai
- Vanderbilt Epidemiology Center and Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN, USA, 37232
| | - Ian Campbell
- Cancer Genetics Laboratory, Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC 3002, Australia
- Department of Pathology, University of Melbourne, Parkville, VIC 3053, Australia
| | - Jenny Chang-Claude
- German Cancer Research Center, Division of Cancer Epidemiology, 69120, Heidelberg, Germany
| | - Stephen Chanock
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda MD, USA, 20892
| | | | - Jin Q. Cheng
- Department of Interdisciplinary Oncology, Moffitt Cancer Center, Tampa, FL, USA, 33612
| | - Mine S. Cicek
- Division of Epidemiology, Department of Health Sciences Research, Mayo Clinic College of Medicine, Rochester, MN, USA, 55905
| | - Gerhard A. Coetzee
- Department of Urology, Microbiology and Preventive Medicine, University of Southern California, Norris Comprehensive Cancer Center, Los Angeles, CA, USA, 90089
| | - Consortium of Investigators of Modifiers of BRCA1/2
- Cancer Research UK, Genetic Epidemiology Unit, Dept of Public Health & Primary Care, University of Cambridge, Strangeways Research Lab, Cambridge, CB1 8RN, UK
- Department of Laboratory of Medicine and Pathology, Mayo Clinic, Rochester, MN, USA, 55905
| | - Linda S. Cook
- Division Epidemiology and Biostatistics, University of New Mexico, Albuquerque, NM, USA, 87131
| | - Fergus J. Couch
- Department of Laboratory of Medicine and Pathology, Mayo Clinic, Rochester, MN, USA, 55905
| | - Daniel W. Cramer
- Obstetrics and Gynecology Epidemiology Center, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA, 02115
| | - Julie M. Cunningham
- Division of Epidemiology, Department of Health Sciences Research, Mayo Clinic College of Medicine, Rochester, MN, USA, 55905
| | - Agnieszka Dansonka-Mieszkowska
- Department of Molecular Pathology, The Maria Sklodowska-Curie Memorial Cancer Center and Institute of Oncology, Warsaw, Poland, 02-781
| | - Evelyn Despierre
- Division of Gynecologic Oncology, Department of Obstetrics and Gynaecology and Leuven Cancer Institute, University Hospitals Leuven, Leuven, Belgium, 3000
| | - Jennifer A Doherty
- Section of Biostatistics and Epidemiology, The Geisel School of Medicine at Dartmouth, Lebanon, NH, USA, 03755
| | - Thilo Dörk
- Gynaecology Research Unit, Hannover Medical School, Hannover, 30625, Germany
| | - Andreas du Bois
- Department of Gynecology and Gynecologic Oncology, Dr. Horst Schmidt Klinik Wiesbaden, 65199, Wiesbaden, Germany
- Department of Gynecology and Gynecologic Oncology, Kliniken Essen-Mitte, 45136, Essen, Germany
| | - Matthias Dürst
- Department of Gynecology and Obstetrics, Jena University Hospital, 07743, Jena, Germany
| | - Douglas F Easton
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, CB1 8RN, UK
- Centre for Cancer Genetic Epidemiology, Department of Oncology, University of Cambridge, Cambridge, CB1 8RN, UK
| | - Diana Eccles
- Faculty of Medicine, University of Southampton, University Hospital Southampton, SO17 1BJ, UK
| | | | - Arif B. Ekici
- Institute of Human Genetics, Friedrich-Alexander-University Erlangen-Nuremberg, 91054, Erlangen, Germany
| | - Peter A. Fasching
- University Hospital Erlangen, Department of Gynecology and Obstetrics, Friedrich-Alexander-University Erlangen-Nuremberg, Comprehensive Cancer Center, Erlangen, 91054, Germany
- Department of Medicine, Division of Hematology and Oncology, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA, USA, 90095
| | | | - James M. Flanagan
- Department of Surgery and Cancer, Imperial College London, London, SW7 2AZ, UK
| | - Montserrat Garcia-Closas
- Sections of Epidemiology and Genetics at the Institute of Cancer Research and Breakthrough Breast Cancer Research Centre, London, UK, SW7 3RP
| | - Aleksandra Gentry-Maharaj
- Gynaecological Cancer Research Centre, UCL EGA Institute for Women's Health, London, NW1 2BU, United Kingdom
| | - Graham G. Giles
- Cancer Epidemiology Centre, The Cancer Council Victoria, Melbourne, Carlton VIC 3053, Australia
- Centre for Molecular, Environmental, Genetic and Analytical Epidemiology, University of Melbourne, Melbourne, VIC 3010, Australia
- Department of Epidemiology and Preventive Medicine, Monash University, Melbourne, VIC 3806, Australia
| | | | | | - Marc T. Goodman
- Samuel Oschin Comprehensive Cancer Center Institute, Cedars Sinai Medical Center, Los Angeles, CA, USA, 90048
| | - Martin Gore
- Gynecological Oncology Unit, The Royal Marsden Hospital, London, SW3 6JJ, United Kingdom
| | - Bohdan Górski
- International Hereditary Cancer Center, Department of Genetics and Pathology, Pomeranian Medical University, Szczecin, Poland, 70-115
| | - Jacek Gronwald
- International Hereditary Cancer Center, Department of Genetics and Pathology, Pomeranian Medical University, Szczecin, Poland, 70-115
| | - Per Hall
- Department of Epidemiology and Biostatistics, Karolinska Istitutet, Stockholm, Sweden, 171-77
| | - Mari K. Halle
- Department of Gynecology and Obstetrics, Haukeland University Hospital, Bergen, HB 5006, Norway
- Department of Clinical Medicine, University of Bergen, 5006, Bergen, Norway
| | - Philipp Harter
- Department of Gynecology and Gynecologic Oncology, Dr. Horst Schmidt Klinik Wiesbaden, 65199, Wiesbaden, Germany
- Department of Gynecology and Gynecologic Oncology, Kliniken Essen-Mitte, 45136, Essen, Germany
| | - Florian Heitz
- Department of Gynecology and Gynecologic Oncology, Dr. Horst Schmidt Klinik Wiesbaden, 65199, Wiesbaden, Germany
- Department of Gynecology and Gynecologic Oncology, Kliniken Essen-Mitte, 45136, Essen, Germany
| | - Peter Hillemanns
- Clinics of Obstetrics and Gynaecology, Hannover Medical School, 30625, Hannover, Germany
| | - Maureen Hoatlin
- Department of Biochemistry and Molecular Biology, Oregon Health and Science University, Portland, OR, USA, 97239
| | - Claus K. Høgdall
- The Juliane Marie Centre, Department of Obstetrics and Gynecology, Rigshospitalet, Copenhagen, 2100, Denmark
| | - Estrid Høgdall
- Department of Pathology, Molecular Unit, Herlev Hospital, University of Copenhagen, Denmark, 2730
- Virus, Lifestyle and Genes, Danish Cancer Society Research Center, DK-2100, Copenhagen, Denmark
| | - Satoyo Hosono
- Division of Epidemiology and Prevention, Aichi Cancer Center Research Institute, Nagoya, Aichi, 464-8681, Japan
| | - Anna Jakubowska
- International Hereditary Cancer Center, Department of Genetics and Pathology, Pomeranian Medical University, Szczecin, Poland, 70-115
| | - Allan Jensen
- Virus, Lifestyle and Genes, Danish Cancer Society Research Center, DK-2100, Copenhagen, Denmark
| | - Heather Jim
- Department of Health Outcomes and Behavior, Moffitt Cancer Center, Tampa, FL, USA, 33612
| | - Kimberly R. Kalli
- Department of Medical Oncology, Mayo Clinic, Rochester, MN, USA, 55905
| | - Beth Y. Karlan
- Women's Cancer Program at the Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA, 90048
| | - Stanley B. Kaye
- Section of Medicine, Institute of Cancer Research, Sutton, SM2 5NG, UK
| | - Linda E. Kelemen
- Department of Popluation Health Research, Alberta Health Services-Cancer Care, Calgary, Alberta, Canada and Departments of Medical Genetics and Oncology, University of Calgary, Calgary, AB, Canada, T2N 2T9
| | - Lambertus A. Kiemeney
- Department of Epidemiology, Biostatistics and HTA, Radboud University Medical Centre, Nijmegen, HB 6500, Netherlands
- Comprehensive Cancer Center, the Netherlands, Utrecht, Amsterdam, 1066CX, The Netherlands
- Department of Urology, Radboud University Medical Centre, Nijmegen, HB 6500, Netherlands
| | - Fumitaka Kikkawa
- Department of Obsterics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, 466-8550, Japan
| | - Gottfried E. Konecny
- Department of Medicine, Division of Hematology and Oncology, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA, USA, 90095
| | - Camilla Krakstad
- Department of Gynecology and Obstetrics, Haukeland University Hospital, Bergen, HB 5006, Norway
- Department of Clinical Medicine, University of Bergen, 5006, Bergen, Norway
| | - Susanne Krüger Kjaer
- The Juliane Marie Centre, Department of Obstetrics and Gynecology, Rigshospitalet, Copenhagen, 2100, Denmark
- Virus, Lifestyle and Genes, Danish Cancer Society Research Center, DK-2100, Copenhagen, Denmark
| | - Jolanta Kupryjanczyk
- Department of Molecular Pathology, The Maria Sklodowska-Curie Memorial Cancer Center and Institute of Oncology, Warsaw, Poland, 02-781
| | - Diether Lambrechts
- Vesalius Research Center, VIB, 3000, Leuven, Belgium
- Laboratory for Translational Genetics, Department of Oncology, University of Leuven, 3000, Leuven, Belgium
| | - Sandrina Lambrechts
- Division of Gynecologic Oncology, Department of Obstetrics and Gynaecology and Leuven Cancer Institute, University Hospitals Leuven, Leuven, Belgium, 3000
| | | | - Nhu D. Le
- Cancer Control Research, BC Cancer Agency, Vancouver, BC, Canada, G12 0YN
| | - Arto Leminen
- Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Central Hospital, Helsinki, Finland, 00530
| | - Douglas A. Levine
- Gynecology Service, Department of Surgery, Memorial Sloan-Kettering Cancer Center, New York, NY, USA, 10021
| | - Dong Liang
- College of Pharmacy and Health Sciences, Texas Southern University, Houston, Texas, USA, 77044
| | - Boon Kiong Lim
- Department of Obstetrics and Gynaecology, University Malaya Medical Centre, University Malaya, 59100 Kuala Lumpur, Federal Territory of Kuala Lumpur, Malaysia
| | - Jie Lin
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA, 77030
| | - Jolanta Lissowska
- Department of Cancer Epidemiology and Prevention, The Maria Sklodowska-Curie Memorial Cancer Center, 02-781, Warsaw, Poland
| | - Karen H. Lu
- Department of Gynecologic Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA, 77030
| | - Jan Lubiński
- International Hereditary Cancer Center, Department of Genetics and Pathology, Pomeranian Medical University, Szczecin, Poland, 70-115
| | - Galina Lurie
- Cancer Epidemiology Program, University of Hawaii Cancer Center, Hawaii, USA, 96813
| | - Leon F.A.G. Massuger
- Department of Gynaecology, Radboud University Medical Centre, Nijmegen, HB 6500, Netherlands
| | - Keitaro Matsuo
- Division of Epidemiology and Prevention, Aichi Cancer Center Research Institute, Nagoya, Aichi, 464-8681, Japan
| | - Valerie McGuire
- Department of Health Research and Policy - Epidemiology, Stanford University School of Medicine, Stanford CA, USA, 94305
| | - John R McLaughlin
- Dalla Lana School of Public Health, Faculty of Medicine, University of Toronto, ON, M5T 3M7, Canada
- Samuel Lunenfeld Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada, M5G 1X5
| | - Usha Menon
- Gynaecological Cancer Research Centre, UCL EGA Institute for Women's Health, London, NW1 2BU, United Kingdom
| | - Francesmary Modugno
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA, 77030
- Department of Gynecologic Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA, 77030
- Women's Cancer Research Program, Magee-Womens Research Institute and University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA, 15213
| | - Kirsten B. Moysich
- Department of Cancer Prevention and Control, Roswell Park Cancer Institute, Buffalo, NY, USA, 14263
| | - Toru Nakanishi
- Department of Gynecologic Oncology, Aichi Cancer Center Central Hospital, Nagoya, Aichi, Nagoya, 464-8681, Japan
| | - Steven A. Narod
- Women's College Research Institute, University of Toronto, Toronto, Ontario, Canada, M5G 1N8
| | - Lotte Nedergaard
- Department of Pathology, Rigshospitalet, University of Copenhagen, 2100, Denmark
| | - Roberta B. Ness
- The University of Texas School of Public Health, Houston, TX, USA, 77030
| | - Heli Nevanlinna
- Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Central Hospital, Helsinki, Finland, 00530
| | - Stefan Nickels
- German Cancer Research Center, Division of Cancer Epidemiology, 69120, Heidelberg, Germany
| | - Houtan Noushmehr
- Department of Urology, Microbiology and Preventive Medicine, University of Southern California, Norris Comprehensive Cancer Center, Los Angeles, CA, USA, 90089
- USC Epigenome Center, Keck School of Medicine, University of Southern California, Norris Comprehensive Cancer Center, Los Angeles, CA, 90089
| | - Kunle Odunsi
- Department of Gynecologic Oncology, Roswell Park Cancer Institute, Buffalo, NY, USA, 14263
| | - Sara H. Olson
- Department of Epidemiology and Biostatistics, Memorial Sloan-Kettering Cancer Center, New York, NY, USA, 10065
| | - Irene Orlow
- Department of Epidemiology and Biostatistics, Memorial Sloan-Kettering Cancer Center, New York, NY, USA, 10065
| | - James Paul
- The Beatson West of Scotland Cancer Centre, Glasgow, G12 0YN, UK
| | - Celeste L Pearce
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California Norris Comprehensive Cancer Center, Los Angeles, CA, USA, 90033
| | - Tanja Pejovic
- Department of Obstetrics and Gynecology, Oregon Health and Science University, Portland, OR, USA, 97239
- Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA, 97239
| | - Liisa M. Pelttari
- Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Central Hospital, Helsinki, Finland, 00530
| | - Malcolm C. Pike
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California Norris Comprehensive Cancer Center, Los Angeles, CA, USA, 90033
- Department of Epidemiology and Biostatistics, Memorial Sloan-Kettering Cancer Center, New York, NY, USA, 10065
| | - Elizabeth M. Poole
- Department of Epidemiology, Harvard School of Public Health, Boston, Massachusetts, USA, 02115
- Channing Laboratory, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, USA, 02115
| | - Paola Raska
- Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, OH, USA, 44195
| | - Stefan P. Renner
- University Hospital Erlangen, Department of Gynecology and Obstetrics, Friedrich-Alexander-University Erlangen-Nuremberg, Comprehensive Cancer Center, Erlangen, 91054, Germany
| | - Harvey A. Risch
- Department of Epidemiology and Public Health, Yale University School of Public Health and School of Medicine, New Haven, CT, USA, 06520
| | - Lorna Rodriguez-Rodriguez
- The Cancer Institute of New Jersey, Robert Wood Johnson Medical School, New Brunswick, NJ, USA, 08901
| | - Mary Anne Rossing
- Program in Epidemiology, Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, USA, 98109
- Department of Epidemiology, University of Washington, Seattle, WA, USA, 98109
| | - Anja Rudolph
- German Cancer Research Center, Division of Cancer Epidemiology, 69120, Heidelberg, Germany
| | - Ingo B. Runnebaum
- Department of Gynecology and Obstetrics, Jena University Hospital, 07743, Jena, Germany
| | - Iwona K. Rzepecka
- Department of Molecular Pathology, The Maria Sklodowska-Curie Memorial Cancer Center and Institute of Oncology, Warsaw, Poland, 02-781
| | - Helga B. Salvesen
- Department of Gynecology and Obstetrics, Haukeland University Hospital, Bergen, HB 5006, Norway
- Department of Clinical Medicine, University of Bergen, 5006, Bergen, Norway
| | - Ira Schwaab
- Institut für Humangenetik Wiesbaden, 65187, Wiesbaden, Germany
| | - Gianluca Severi
- Cancer Epidemiology Centre, The Cancer Council Victoria, Melbourne, Carlton VIC 3053, Australia
- Centre for Molecular, Environmental, Genetic and Analytical Epidemiology, University of Melbourne, Melbourne, VIC 3010, Australia
| | - Vijayalakshmi Shridhar
- Department of Laboratory Medicine and Pathology, Division of Anatomic Pathology, Mayo Clinic, Rochester, MN, USA, 55905
| | - Xiao-Ou Shu
- Vanderbilt Epidemiology Center and Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN, USA, 37232
| | - Yurii B. Shvetsov
- Cancer Epidemiology Program, University of Hawaii Cancer Center, Hawaii, USA, 96813
| | - Weiva Sieh
- Department of Health Research and Policy - Epidemiology, Stanford University School of Medicine, Stanford CA, USA, 94305
| | - Honglin Song
- Department of Oncology, University of Cambridge, Cambridge, CB1 8RN, UK
| | - Melissa C. Southey
- Genetic Epidemiology Laboratory, Department of Pathology, The University of Melbourne, Melbourne, VIC 3053, Australia
| | - Beata Spiewankiewicz
- Department of Gynecologic Oncology, The Maria Sklodowska-Curie Memorial Cancer Center and Institute of Oncology, Warsaw, Poland, 02-781
| | - Daniel Stram
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California Norris Comprehensive Cancer Center, Los Angeles, CA, USA, 90033
| | - Rebecca Sutphen
- Pediatrics Epidemiology Center, College of Medicine, University of South Florida, Tampa, FL, USA, 33612
| | - Soo-Hwang Teo
- Department of Obstetrics and Gynaecology, University Malaya Medical Centre, University Malaya, 59100 Kuala Lumpur, Federal Territory of Kuala Lumpur, Malaysia
| | - Kathryn L. Terry
- Obstetrics and Gynecology Epidemiology Center, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA, 02115
| | - Daniel C. Tessier
- McGill University and Génome Québec Innovation Centre, Montréal (Québec) Canada, H3A 0G1
| | - Pamela J. Thompson
- Samuel Oschin Comprehensive Cancer Center Institute, Cedars Sinai Medical Center, Los Angeles, CA, USA, 90048
| | - Shelley S. Tworoger
- Department of Epidemiology, Harvard School of Public Health, Boston, Massachusetts, USA, 02115
- Channing Laboratory, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, USA, 02115
| | - Anne M. van Altena
- Department of Gynaecology, Radboud University Medical Centre, Nijmegen, HB 6500, Netherlands
| | - Ignace Vergote
- Division of Gynecologic Oncology, Department of Obstetrics and Gynaecology and Leuven Cancer Institute, University Hospitals Leuven, Leuven, Belgium, 3000
| | - Robert A. Vierkant
- Division of Epidemiology, Department of Health Sciences Research, Mayo Clinic College of Medicine, Rochester, MN, USA, 55905
| | - Daniel Vincent
- McGill University and Génome Québec Innovation Centre, Montréal (Québec) Canada, H3A 0G1
| | - Allison F. Vitonis
- Obstetrics and Gynecology Epidemiology Center, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA, 02115
| | - Shan Wang-Gohrke
- Department of Obstetrics and Gynecology, University of Ulm, Ulm, 89081, Germany
| | - Rachel Palmieri Weber
- Department of Community and Family Medicine, Duke University Medical Center, Durham, NC, USA, 27708
| | - Nicolas Wentzensen
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda MD, USA, 20892
| | - Alice S. Whittemore
- Department of Health Research and Policy - Epidemiology, Stanford University School of Medicine, Stanford CA, USA, 94305
| | - Elisabeth Wik
- Department of Gynecology and Obstetrics, Haukeland University Hospital, Bergen, HB 5006, Norway
- Department of Clinical Medicine, University of Bergen, 5006, Bergen, Norway
| | - Lynne R. Wilkens
- Cancer Epidemiology Program, University of Hawaii Cancer Center, Hawaii, USA, 96813
| | - Boris Winterhoff
- Department of Obstetrics and Gynecology, Mayo Clinic, Rochester, MN, USA, 55905
| | - Yin Ling Woo
- Department of Obstetrics and Gynaecology, University Malaya Medical Centre, University Malaya, 59100 Kuala Lumpur, Federal Territory of Kuala Lumpur, Malaysia
| | - Anna H. Wu
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California Norris Comprehensive Cancer Center, Los Angeles, CA, USA, 90033
| | | | - Hannah P. Yang
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda MD, USA, 20892
| | - Wei Zheng
- Vanderbilt Epidemiology Center and Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN, USA, 37232
| | - Argyrios Ziogas
- Department of Epidemiology, Center for Cancer Genetics Research and Prevention, School of Medicine, University of California Irvine, Irvine, California, USA, 92697
| | - Famida Zulkifli
- Department of Obstetrics and Gynaecology, University Malaya Medical Centre, University Malaya, 59100 Kuala Lumpur, Federal Territory of Kuala Lumpur, Malaysia
| | - Catherine M. Phelan
- Department of Cancer Epidemiology, Division of Population Sciences, Moffitt Cancer Center, Tampa, FL, USA, 33612
| | - Edwin Iversen
- Department of Statistical Science, Duke University, Durham, NC, USA, 27708
| | - Joellen M. Schildkraut
- Department of Community and Family Medicine, Duke University Medical Center, Durham, NC, USA, 27708
- Cancer Prevention, Detection and Control Research Program, Duke Cancer Institute, Durham, North Carolina, USA, 27708-0251
| | - Andrew Berchuck
- Department of Obstetrics and Gynecology, Duke Comprehensive Cancer Center, Durham, NC, USA, 27708
| | - Brooke L. Fridley
- Department of Biostatistics, University of Kansas Medical Center, Kansas City, KS, USA, 66160
| | - Ellen L. Goode
- Division of Epidemiology, Department of Health Sciences Research, Mayo Clinic College of Medicine, Rochester, MN, USA, 55905
| | - Paul D. P. Pharoah
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, CB1 8RN, UK
- Centre for Cancer Genetic Epidemiology, Department of Oncology, University of Cambridge, Cambridge, CB1 8RN, UK
| | - Alvaro N.A. Monteiro
- Department of Cancer Epidemiology, Division of Population Sciences, Moffitt Cancer Center, Tampa, FL, USA, 33612
| | - Thomas A. Sellers
- Department of Cancer Epidemiology, Division of Population Sciences, Moffitt Cancer Center, Tampa, FL, USA, 33612
| | - Simon A. Gayther
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California Norris Comprehensive Cancer Center, Los Angeles, CA, USA, 90033
| |
Collapse
|