1
|
Wang Y, Zhang X, Ren M, He S, Bie H, Duan M, Chen Z, Jia Q, Chi B, Gan X, Li C, Fu Y, Zhou H, Zhang S, Zhang Q, An F, Chen X, Jia E. LncRNA LUCAT1 offers protection against human coronary artery endothelial cellular oxidative stress injury through modulating hsa-miR-6776-5p/LRRC25 axis and activating autophagy flux. J Transl Med 2024; 22:1171. [PMID: 39741278 DOI: 10.1186/s12967-024-05966-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 12/11/2024] [Indexed: 01/02/2025] Open
Abstract
BACKGROUND Coronary artery disease (CAD) has become a dominant economic and health burden worldwide, and the role of autophagy in CAD requires further clarification. In this study, we comprehensively revealed the association between autophagy flux and CAD from multiple hierarchies. We explored autophagy-associated long noncoding RNA (lncRNA) and the mechanisms underlying oxidative stress-induced human coronary artery endothelial cells (HCAECs) injury. METHODS (1) Autophagy-related proteins including LC3, p62, Beclin1, ATG5, and ATG7 were immunohistochemical stained in coronary specimens; (2) The levels and function of autophagy in the HCAEC oxidative stress model were evaluated using western blot (WB), transmission electron microscopy (TEM), and mRFP-GFP-LC3 adenovirus transfection experiments; (3) The competing endogenous RNA (ceRNA) network of lncRNA LUCAT1/hsa-miR-6776-5p/LRRC25 axis was constructed and validated; (4) The expression levels of above autophagy-related RNAs in peripheral blood mononuclear cells (PBMCs) were verified by qPCR, and their diagnostic performance was subsequently analyzed using receiver operating characteristic (ROC) analysis. RESULTS (1) The expression of LC3, Beclin1, ATG5, and ATG7 demonstrated a consistent decline whereas p62 expression exhibited an opposite increase as atherosclerosis progressed; (2) Autophagy levels was significantly elevated in HCAECs under oxidative stress, while inhibition of the initial stage of autophagy with 3-MA exacerbated cellular damage; (3) The lncRNA LUCAT1/hsa-miR-6776-5p/LRRC25 axis was established through bioinformatic prediction and validated by dual-luciferase reporter assay, which resulted in a significant decrease in autophagy levels in HCAECs; (4) In total, p62, ATG7, lncRNA LUCAT1 and LRRC25 were validated as robust diagnostic biomarkers for CAD. CONCLUSIONS Our results delineated the dynamic disruption of the autophagy landscape during the progression of human coronary atherosclerosis and identified the lncRNA LUCAT1/hsa-miR-6776-5p/LRRC25 axis, uncovered through transcriptomic profiling, as a protective mechanism against endothelial cell injury through autophagy activation. Furthermore, we recognized p62, ATG7, lncRNA LUCAT1, and LRRC25 as dependable autophagy-related diagnostic biomarkers in circulating PBMCs, correlating with CAD severity. Collectively, Our findings furnish novel insights into the intricate autophagy landscape at various levels of coronary atherosclerosis and propose potential diagnostic biomarkers, and a theoretical foundation for managing CAD patients.
Collapse
Affiliation(s)
- Yanjun Wang
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Nanjing Medical University, Guangzhou Road 300, Nanjing, 210029, Jiangsu, China
| | - Xin Zhang
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Nanjing Medical University, Guangzhou Road 300, Nanjing, 210029, Jiangsu, China
| | - Mengmeng Ren
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Nanjing Medical University, Guangzhou Road 300, Nanjing, 210029, Jiangsu, China
| | - Shu He
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Nanjing Medical University, Guangzhou Road 300, Nanjing, 210029, Jiangsu, China
| | - Hengjie Bie
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Nanjing Medical University, Guangzhou Road 300, Nanjing, 210029, Jiangsu, China
| | - Mengyang Duan
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Nanjing Medical University, Guangzhou Road 300, Nanjing, 210029, Jiangsu, China
| | - Zhiyuan Chen
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Nanjing Medical University, Guangzhou Road 300, Nanjing, 210029, Jiangsu, China
| | - Qiaowei Jia
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Nanjing Medical University, Guangzhou Road 300, Nanjing, 210029, Jiangsu, China
| | - Boyu Chi
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Nanjing Medical University, Guangzhou Road 300, Nanjing, 210029, Jiangsu, China
| | - Xiongkang Gan
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Nanjing Medical University, Guangzhou Road 300, Nanjing, 210029, Jiangsu, China
| | - Chengcheng Li
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Nanjing Medical University, Guangzhou Road 300, Nanjing, 210029, Jiangsu, China
| | - Yahong Fu
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Nanjing Medical University, Guangzhou Road 300, Nanjing, 210029, Jiangsu, China
| | - Hanxiao Zhou
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Nanjing Medical University, Guangzhou Road 300, Nanjing, 210029, Jiangsu, China
| | - Sheng Zhang
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Nanjing Medical University, Guangzhou Road 300, Nanjing, 210029, Jiangsu, China
| | - Qian Zhang
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Nanjing Medical University, Guangzhou Road 300, Nanjing, 210029, Jiangsu, China
| | - Fenghui An
- Department of Critical Care Medicine, The Friendship Hospital of Ili Kazakh Autonomous Prefecture, Yining, 835000, Xinjiang, China.
| | - Xiumei Chen
- Department of Geriatric, The First Affiliated Hospital of Nanjing Medical University, Guangzhou Road 300, Nanjing, 210029, Jiangsu, China.
| | - Enzhi Jia
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Nanjing Medical University, Guangzhou Road 300, Nanjing, 210029, Jiangsu, China.
| |
Collapse
|
2
|
Guo Y, Li G, Xia F, Li C. Upregulation of RCN2 accelerates tumor progression and indicates poor prognosis in OSCC. Oral Surg Oral Med Oral Pathol Oral Radiol 2024:S2212-4403(24)00933-7. [PMID: 39730259 DOI: 10.1016/j.oooo.2024.12.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Revised: 11/30/2024] [Accepted: 12/03/2024] [Indexed: 12/29/2024]
Abstract
OBJECTIVE Oral squamous cell carcinoma (OSCC) is a prevalent cancer of the head and neck region. However, the potential role of RCN2 in OSCC is currently not well understood. STUDY DESIGN A series of molecular biology experiments were conducted to explore the mechanism by which RCN2 promotes OSCC growth through protein kinase A (PKA). RESULTS Our results revealed a significant increase in RCN2 levels in OSCC tissues. Moreover, OSCC patients with high RCN2 expression had a significantly worse prognosis than those with lower RCN2 expression. Interestingly, PKA activity was increased in RCN2-overexpressing YD-10B cells but reduced in RCN2-knockout Ca9-22 cells. These findings suggest that RCN2-mediated PKA activity is activated in OSCC cells. Moreover, the specific PKA inhibitor H89 significantly reduced the proliferation ability of RCN2-overexpressing Ca9-22 cells. Furthermore, we identified AKT/mTORC as a downstream pathway through which PKA promotes OSCC cell proliferation. The Tumor Immune Estimation Resource database revealed that the expression level of RCN2 was correlated with the infiltration levels of B cells, CD8+ T cells, CD4+ T cells, and neutrophils in the microenvironment of OSCC. CONCLUSIONS Our study revealed that RCN2 promotes tumor progression by activating PKA/AKT/mTORC signaling, which suggests that RCN2 may serve as a potential target for OSCC treatment.
Collapse
Affiliation(s)
- Yongshan Guo
- Department of Stomatology, Xinjiang Production and Construction Corps Hospital, Urumqi, China
| | - Guolong Li
- Department of Stomatology, Xinjiang Production and Construction Corps Hospital, Urumqi, China
| | - Feifei Xia
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Shihezi University, Shihezi, China
| | - Changxue Li
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Shihezi University, Shihezi, China.
| |
Collapse
|
3
|
Alur V, Vastrad B, Raju V, Vastrad C, Kotturshetti S. The identification of key genes and pathways in polycystic ovary syndrome by bioinformatics analysis of next-generation sequencing data. MIDDLE EAST FERTILITY SOCIETY JOURNAL 2024; 29:53. [DOI: 10.1186/s43043-024-00212-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 11/17/2024] [Indexed: 01/02/2025] Open
Abstract
Abstract
Background
Polycystic ovary syndrome (PCOS) is a reproductive endocrine disorder. The specific molecular mechanism of PCOS remains unclear. The aim of this study was to apply a bioinformatics approach to reveal related pathways or genes involved in the development of PCOS.
Methods
The next-generation sequencing (NGS) dataset GSE199225 was downloaded from the gene expression omnibus (GEO) database and NGS dataset analyzed is obtained from in vitro culture of PCOS patients’ muscle cells and muscle cells of healthy lean control women. Differentially expressed gene (DEG) analysis was performed using DESeq2. The g:Profiler was utilized to analyze the gene ontology (GO) and REACTOME pathways of the differentially expressed genes. A protein–protein interaction (PPI) network was constructed and module analysis was performed using HiPPIE and cytoscape. The miRNA-hub gene regulatory network and TF-hub gene regulatory network were constructed. The hub genes were validated by using receiver operating characteristic (ROC) curve analysis.
Results
We have identified 957 DEG in total, including 478 upregulated genes and 479 downregulated gene. GO terms and REACTOME pathways illustrated that DEG were significantly enriched in regulation of molecular function, developmental process, interferon signaling and platelet activation, signaling, and aggregation. The top 5 upregulated hub genes including HSPA5, PLK1, RIN3, DBN1, and CCDC85B and top 5 downregulated hub genes including DISC1, AR, MTUS2, LYN, and TCF4 might be associated with PCOS. The hub gens of HSPA5 and KMT2A, together with corresponding predicted miRNAs (e.g., hsa-mir-34b-5p and hsa-mir-378a-5p), and HSPA5 and TCF4 together with corresponding predicted TF (e.g., RCOR3 and TEAD4) were found to be significantly correlated with PCOS.
Conclusions
These study uses of bioinformatics analysis of NGS data to obtain hub genes and key signaling pathways related to PCOS and its associated complications. Also provides novel ideas for finding biomarkers and treatment methods for PCOS and its associated complications.
Collapse
|
4
|
Liu Y, Chen P, Hu B, Xiao Y, Su T, Luo X, Tu M, Cai G. Excessive mechanical loading promotes osteoarthritis development by upregulating Rcn2. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167251. [PMID: 38795835 DOI: 10.1016/j.bbadis.2024.167251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 05/10/2024] [Accepted: 05/14/2024] [Indexed: 05/28/2024]
Abstract
Exposure of articular cartilage to excessive mechanical loading is closely related to the pathogenesis of osteoarthritis (OA). However, the exact molecular mechanism by which excessive mechanical loading drives OA remains unclear. In vitro, primary chondrocytes were exposed to cyclic tensile strain at 0.5 Hz and 10 % elongation for 30 min to simulate excessive mechanical loading in OA. In vivo experiments involved mice undergoing anterior cruciate ligament transection (ACLT) to model OA, followed by interventions on Rcn2 expression through adeno-associated virus (AAV) injection and tamoxifen-induced gene deletion. 10 μL AAV2/5 containing AAV-Rcn2 or AAV-shRcn2 was administered to the mice by articular injection at 1 week post ACLT surgery, and Col2a1-creERT: Rcn2flox/flox mice were injected with tamoxifen intraperitoneally to obtain Rcn2-conditional knockout mice. Finally, we explored the mechanism of Rcn2 affecting OA. Here, we identified reticulocalbin-2 (Rcn2) as a mechanosensitive factor in chondrocytes, which was significantly elevated in chondrocytes under mechanical overloading. PIEZO type mechanosensitive ion channel component 1 (Piezo1) is a critical mechanosensitive ion channel, which mediates the effect of mechanical loading on chondrocytes, and we found that increased Rcn2 could be suppressed through knocking down Piezo1 under excessive mechanical loading. Furthermore, chondrocyte-specific deletion of Rcn2 in adult mice alleviated OA progression in the mice receiving the surgery of ACLT. On the contrary, articular injection of Rcn2-expressing adeno-associated virus (AAV) accelerated the progression of ACLT-induced OA in mice. Mechanistically, Rcn2 accelerated the progression of OA through promoting the phosphorylation and nuclear translocation of signal transducer and activator of transcription 3 (Stat3).
Collapse
Affiliation(s)
- Yalin Liu
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, China
| | - Peng Chen
- Department of Orthopedic, Xiangya Hospital of Central South University, Changsha, China
| | - Biao Hu
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, China
| | - Ye Xiao
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, China
| | - Tian Su
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, China
| | - Xianghang Luo
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, China
| | - Manli Tu
- Department of Endocrinology and Metabolism, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, China; Jiangxi Clinical Research Center for Endocrine and Metabolic Disease, China; Jiangxi Branch of National Clinical Research Center for metabolic Disease, China.
| | - Guangping Cai
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, China.
| |
Collapse
|
5
|
Lin Y, Ni L, Yang L, Li H, Chen Z, Gao Y, Zhu K, Jia Y, Wu Z, Li S. Identification of Endoplasmic Reticulum Stress-Related Biomarkers in Coronary Artery Disease. Cardiovasc Ther 2024; 2024:4664731. [PMID: 39742022 PMCID: PMC11236471 DOI: 10.1155/2024/4664731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 06/05/2024] [Accepted: 06/08/2024] [Indexed: 01/03/2025] Open
Abstract
Coronary artery disease (CAD) is caused by atherosclerotic lesions in the coronary vessels. Endoplasmic reticulum stress (ERS) acts in cardiovascular disease, and its role in CAD is not clear. A total of 13 differentially expressed ERS-related genes (DEERSRGs) in CAD were identified. Functional enrichment analysis demonstrated the DEERSRGs were mainly enriched in endoplasmic reticulum (ER)-related pathways. Then, eight genes (RCN2, HRC, DERL2, RNF183, CRH, TMED2, PPP1R15A, and IL1A) were authenticated as ERS-related biomarkers in CAD by least absolute shrinkage and selection operator (LASSO). The receiver operating characteristic (ROC) analysis showed that the LASSO logistic model constructed based on biomarkers had a better diagnostic effect, which was confirmed by the ANN and GSE23561 datasets. Also, ROC results showed that seven of the eight biomarkers had better diagnostic effects. The nomogram model had good predictive power, and biomarkers were mostly enriched in pathways associated with CAD. The biomarkers were significantly associated with 10 immune cells, and RCN2, DERL2, TMED2, and RNF183 were negatively correlated with most chemokines. Eight biomarkers had significant correlations with both immunoinhibitors and immunostimulators. In addition, eight biomarkers were significantly different in both CAD and control samples, CRH and HRC were upregulated in CAD. The quantitative reverse transcription-polymerase chain reaction (qRT-PCR) showed that RCN2, HRC, DERL2, CRH, and IL1A were consistent with the bioinformatics analysis. RCN2, HRC, DERL2, RNF183, CRH, TMED2, PPP1R15A, and IL1A were identified as biomarkers of CAD. Functional enrichment analysis and immunoassays for biomarkers provide new ideas for the treatment of CAD.
Collapse
Affiliation(s)
- Yuanyuan Lin
- Department of Nuclear MedicineFirst Hospital of Shanxi Medical UniversityShanxi Medical University, Taiyuan, Shanxi 030001, China
- Shanxi Bethune HospitalShanxi Academy of Medical SciencesTongji Shanxi HospitalThird Hospital of Shanxi Medical University, Taiyuan 030032, China
| | - Lin Ni
- Shanxi Bethune HospitalShanxi Academy of Medical SciencesTongji Shanxi HospitalThird Hospital of Shanxi Medical University, Taiyuan 030032, China
| | - Luqun Yang
- Shanxi Bethune HospitalShanxi Academy of Medical SciencesTongji Shanxi HospitalThird Hospital of Shanxi Medical University, Taiyuan 030032, China
| | - Hao Li
- Shanxi Bethune HospitalShanxi Academy of Medical SciencesTongji Shanxi HospitalThird Hospital of Shanxi Medical University, Taiyuan 030032, China
| | - Zelin Chen
- Shanxi Bethune HospitalShanxi Academy of Medical SciencesTongji Shanxi HospitalThird Hospital of Shanxi Medical University, Taiyuan 030032, China
| | - Yuping Gao
- Shanxi Bethune HospitalShanxi Academy of Medical SciencesTongji Shanxi HospitalThird Hospital of Shanxi Medical University, Taiyuan 030032, China
| | - Kaiyi Zhu
- Shanxi Bethune HospitalShanxi Academy of Medical SciencesTongji Shanxi HospitalThird Hospital of Shanxi Medical University, Taiyuan 030032, China
| | - Yanni Jia
- Department of Nuclear MedicineFirst Hospital of Shanxi Medical UniversityShanxi Medical University, Taiyuan, Shanxi 030001, China
| | - Zhifang Wu
- Department of Nuclear MedicineFirst Hospital of Shanxi Medical UniversityShanxi Medical University, Taiyuan, Shanxi 030001, China
| | - Sijin Li
- Department of Nuclear MedicineFirst Hospital of Shanxi Medical UniversityShanxi Medical University, Taiyuan, Shanxi 030001, China
| |
Collapse
|
6
|
Shi L, Xu J, Green R, Wretlind A, Homann J, Buckley NJ, Tijms BM, Vos SJB, Lill CM, Kate MT, Engelborghs S, Sleegers K, Frisoni GB, Wallin A, Lleó A, Popp J, Martinez-Lage P, Streffer J, Barkhof F, Zetterberg H, Visser PJ, Lovestone S, Bertram L, Nevado-Holgado AJ, Proitsi P, Legido-Quigley C. Multiomics profiling of human plasma and cerebrospinal fluid reveals ATN-derived networks and highlights causal links in Alzheimer's disease. Alzheimers Dement 2023; 19:3350-3364. [PMID: 36790009 DOI: 10.1002/alz.12961] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 12/07/2022] [Accepted: 12/20/2022] [Indexed: 02/16/2023]
Abstract
INTRODUCTION This study employed an integrative system and causal inference approach to explore molecular signatures in blood and CSF, the amyloid/tau/neurodegeneration [AT(N)] framework, mild cognitive impairment (MCI) conversion to Alzheimer's disease (AD), and genetic risk for AD. METHODS Using the European Medical Information Framework (EMIF)-AD cohort, we measured 696 proteins in cerebrospinal fluid (n = 371), 4001 proteins in plasma (n = 972), 611 metabolites in plasma (n = 696), and genotyped whole-blood (7,778,465 autosomal single nucleotide epolymorphisms, n = 936). We investigated associations: molecular modules to AT(N), module hubs with AD Polygenic Risk scores and APOE4 genotypes, molecular hubs to MCI conversion and probed for causality with AD using Mendelian randomization (MR). RESULTS AT(N) framework associated with protein and lipid hubs. In plasma, Proprotein Convertase Subtilisin/Kexin Type 7 showed evidence for causal associations with AD. AD was causally associated with Reticulocalbin 2 and sphingomyelins, an association driven by the APOE isoform. DISCUSSION This study reveals multi-omics networks associated with AT(N) and causal AD molecular candidates.
Collapse
Affiliation(s)
- Liu Shi
- Department of Psychiatry, University of Oxford, Oxford, UK
| | - Jin Xu
- Institute of Pharmaceutical Science, King's College London, London, UK
| | - Rebecca Green
- Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
- UK National Institute for Health Research (NIHR) Maudsley Biomedical Research Centre, South London and Maudsley Trust, London, UK
- MRC Unit for Lifelong Health & Ageing at UCL, University College London, London, UK
| | | | - Jan Homann
- Lübeck Interdisciplinary Platform for Genome Analytics, University of Lübeck, Lübeck, Germany
| | - Noel J Buckley
- Department of Psychiatry, University of Oxford, Oxford, UK
| | - Betty M Tijms
- Alzheimer Center, VU University Medical Center, Amsterdam, the Netherlands
| | - Stephanie J B Vos
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Alzheimer Centrum Limburg, Maastricht University, Maastricht, the Netherlands
| | - Christina M Lill
- Lübeck Interdisciplinary Platform for Genome Analytics, University of Lübeck, Lübeck, Germany
- Institute of Epidemiology and Social Medicine, University of Muenster, Muenster, Germany
- Ageing Epidemiology Research Unit (AGE), School of Public Health, Imperial College London, London, UK
| | - Mara Ten Kate
- Alzheimer Center, VU University Medical Center, Amsterdam, the Netherlands
| | - Sebastiaan Engelborghs
- Reference Center for Biological Markers of Dementia (BIODEM), Institute Born-Bunge, University of Antwerp, Antwerp, Belgium
- Department of Neurology, UZ Brussel and Center for Neurociences (C4N), Vrije Universiteit Brussel, Brussels, Belgium
| | - Kristel Sleegers
- Complex Genetics Group, VIB Center for Molecular Neurology, VIB, Antwerp, Belgium
- Institute Born-Bunge, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Giovanni B Frisoni
- University of Geneva, Geneva, Switzerland
- IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Anders Wallin
- Institute of Neuroscience and Physiology, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Alberto Lleó
- Neurology Department, Centro de Investigación en Red en enfermedades neurodegenerativas (CIBERNED), Hospital Sant Pau, Barcelona, Spain
| | - Julius Popp
- University Hospital of Lausanne, Lausanne, Switzerland
- Department of Geriatric Psychiatry, University Hospital of Psychiatry and University of Zürich, Zürich, Switzerland
| | | | - Johannes Streffer
- AC Immune SA, formerly Janssen R&D, LLC. Beerse, Belgium at the time of study conduct, Lausanne, Switzerland
| | - Frederik Barkhof
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, Vrije Universiteit, Amsterdam, The Netherland
- Queen Square Institute of Neurology and Centre for Medical Image Computing, University College London, London, UK
| | - Henrik Zetterberg
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- UK Dementia Research Institute at UCL, London, UK
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK
| | - Pieter Jelle Visser
- Alzheimer Center, VU University Medical Center, Amsterdam, the Netherlands
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Alzheimer Centrum Limburg, Maastricht University, Maastricht, the Netherlands
| | - Simon Lovestone
- Department of Psychiatry, University of Oxford, Oxford, UK
- Janssen Medical (UK), High Wycombe, UK
| | - Lars Bertram
- Lübeck Interdisciplinary Platform for Genome Analytics, University of Lübeck, Lübeck, Germany
- Department of Psychology, University of Oslo, Oslo, Norway
| | | | - Petroula Proitsi
- Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Cristina Legido-Quigley
- Institute of Pharmaceutical Science, King's College London, London, UK
- Steno Diabetes Center Copenhagen, Gentofte, Denmark
| |
Collapse
|