1
|
Oh S, Kim S, Kim JP, Seo SW, Park BY, Park H. Association of APOC1 with cortical atrophy during conversion to Alzheimer's disease. GeroScience 2025:10.1007/s11357-025-01695-6. [PMID: 40369256 DOI: 10.1007/s11357-025-01695-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Accepted: 05/02/2025] [Indexed: 05/16/2025] Open
Abstract
Alzheimer's disease (AD) is a prevalent neurodegenerative disorder, with its progression influenced by aberrant gene expression and alterations in the brain network topology. Although APOE has been extensively studied in relation to AD, the role of APOC1 remains relatively underexplored. This study investigated the impact of APOC1 on changes in cortical thickness (CTh) during conversion to AD in a longitudinal setting. Using a normative modeling approach, we examined changes in CTh in patients with mild cognitive impairment (MCI). The spatial patterns of CTh changes were then correlated with APOC1 mRNA expression levels. We estimated the time to conversion to AD and compared progression rates between the low and high APOC1 expression groups. Finally, mediation analysis was performed to assess the indirect effects of APOC1 expression on memory function via CTh changes. In patients with MCI and AD, reduced CTh was observed in the limbic and default mode regions, with a notable impact on the entorhinal cortex, parahippocampus, and fusiform gyrus when comparing baseline and follow-up measurements. The degree of change in CTh was significantly associated with APOC1 expression, with the paralimbic regions identified as particularly vulnerable. Furthermore, the high APOC1 expression group demonstrated more rapid conversion to AD than that observed in the low expression group. Mediation analysis indicated a trend suggesting that APOC1 expression indirectly affected memory and cognitive function through its influence on CTh. These results highlight the potential of APOC1 as an additional focus of AD research, offering insights into the genetic influences on AD pathology.
Collapse
Affiliation(s)
- Sewook Oh
- Department of Electrical and Computer Engineering, Sungkyunkwan University, Jangan-gu, Suwon, Gyeonggi-do, 16419, Republic of Korea
- Center for Neuroscience Imaging Research, Institute for Basic Science, Jangan-gu, Suwon, Gyeonggi-do, 16419, Republic of Korea
| | - Sunghun Kim
- Department of Brain and Cognitive Engineering, Korea University, Seongbuk-gu, Seoul, 02841, Republic of Korea
- BK21 Four Institute of Precision Public Health, Seoul, Republic of Korea
| | - Jun Pyo Kim
- Alzheimer's Disease Convergence Research Center, Samsung Medical Center, Gangnam-gu, Seoul, 06351, Republic of Korea
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Gangnam-gu, Seoul, 06351, Republic of Korea
- Neuroscience Center, Samsung Medical Center, Gangnam-gu, Seoul, 06351, Republic of Korea
| | - Sang Won Seo
- Alzheimer's Disease Convergence Research Center, Samsung Medical Center, Gangnam-gu, Seoul, 06351, Republic of Korea
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Gangnam-gu, Seoul, 06351, Republic of Korea
- Neuroscience Center, Samsung Medical Center, Gangnam-gu, Seoul, 06351, Republic of Korea
| | - Bo-Yong Park
- Center for Neuroscience Imaging Research, Institute for Basic Science, Jangan-gu, Suwon, Gyeonggi-do, 16419, Republic of Korea.
- Department of Brain and Cognitive Engineering, Korea University, Seongbuk-gu, Seoul, 02841, Republic of Korea.
| | - Hyunjin Park
- Department of Electrical and Computer Engineering, Sungkyunkwan University, Jangan-gu, Suwon, Gyeonggi-do, 16419, Republic of Korea.
- Center for Neuroscience Imaging Research, Institute for Basic Science, Jangan-gu, Suwon, Gyeonggi-do, 16419, Republic of Korea.
| |
Collapse
|
2
|
Vogi V, Haschka D, Forer L, Schwendinger S, Petzer V, Coassin S, Tancevski I, Sonnweber T, Löffler-Ragg J, Puchhammer-Stöckl E, Graninger M, Wolf D, Kronenberg F, Zschocke J, Jukic E, Weiss G. Severe COVID-19 disease is associated with genetic factors affecting plasma ACE2 receptor and CRP concentrations. Sci Rep 2025; 15:4708. [PMID: 39922945 PMCID: PMC11807156 DOI: 10.1038/s41598-025-89306-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 02/04/2025] [Indexed: 02/10/2025] Open
Abstract
A hyperinflammatory state with highly elevated concentrations of inflammatory biomarkers such as C-reactive protein (CRP) is a characteristic feature of severe coronavirus disease 2019 (COVID-19). To examine a potential role of common genetic factors that may influence COVID-19 outcomes, we investigated whether individuals with a polygenic predisposition for a pro-inflammatory response (in the form of Polygenic Scores) are more likely to develop severe COVID-19. The innovative approach of polygenic scores to investigate genetic factors in COVID-19 severity should provide a comprehensive approach beyond single-gene studies. In our cohort of 156 patients of European ancestry, two overlapping Polygenic Scores (PGS) predicting a genetic predisposition to basal CRP concentrations were significantly different between non-severe and severe COVID-19 cases and were associated with less severe COVID-19 outcomes. Furthermore, specific single nucleotide polymorphisms (SNPs) that contribute to either of the two Polygenic Scores predicting basal CRP levels are associated with different traits that represent risk factors for COVID-19 disease initiation (ACE2 receptor, viral replication) and progression (CRP). We suggest that genetically determined enforced CRP formation may contribute to strengthening of innate immune responses and better initial pathogen control thereby reducing the risk of subsequent hyperinflammation and adverse course of COVID-19.
Collapse
Affiliation(s)
- Verena Vogi
- Institute of Human Genetics, Medical University Innsbruck, Innsbruck, 6020, Austria
| | - David Haschka
- Department of Internal Medicine II (Infectious Diseases, Immunology, Pneumology and Rheumatology), Medical University Innsbruck, Innsbruck, 6020, Austria
| | - Lukas Forer
- Institute of Genetic Epidemiology, Medical University Innsbruck, Innsbruck, 6020, Austria
| | - Simon Schwendinger
- Institute of Human Genetics, Medical University Innsbruck, Innsbruck, 6020, Austria
| | - Verena Petzer
- Department of Internal Medicine V (Hematology and Internistic Oncology), Medical University Innsbruck, Innsbruck, 6020, Austria
| | - Stefan Coassin
- Institute of Genetic Epidemiology, Medical University Innsbruck, Innsbruck, 6020, Austria
| | - Ivan Tancevski
- Department of Internal Medicine II (Infectious Diseases, Immunology, Pneumology and Rheumatology), Medical University Innsbruck, Innsbruck, 6020, Austria
| | - Thomas Sonnweber
- Department of Internal Medicine II (Infectious Diseases, Immunology, Pneumology and Rheumatology), Medical University Innsbruck, Innsbruck, 6020, Austria
| | - Judith Löffler-Ragg
- Department of Internal Medicine II (Infectious Diseases, Immunology, Pneumology and Rheumatology), Medical University Innsbruck, Innsbruck, 6020, Austria
| | | | - Marianne Graninger
- Department of Virology, Medical University Vienna, Vienna, 1090, Austria
| | - Dominik Wolf
- Department of Internal Medicine V (Hematology and Internistic Oncology), Medical University Innsbruck, Innsbruck, 6020, Austria
| | - Florian Kronenberg
- Institute of Genetic Epidemiology, Medical University Innsbruck, Innsbruck, 6020, Austria
| | - Johannes Zschocke
- Institute of Human Genetics, Medical University Innsbruck, Innsbruck, 6020, Austria
| | - Emina Jukic
- Institute of Human Genetics, Medical University Innsbruck, Innsbruck, 6020, Austria.
| | - Günter Weiss
- Department of Internal Medicine II (Infectious Diseases, Immunology, Pneumology and Rheumatology), Medical University Innsbruck, Innsbruck, 6020, Austria.
| |
Collapse
|
3
|
Kulminski AM, Jain‐Washburn E, Nazarian A, Wilkins HM, Veatch O, Swerdlow RH, Honea RA. Association of APOE alleles and polygenic profiles comprising APOE-TOMM40-APOC1 variants with Alzheimer's disease neuroimaging markers. Alzheimers Dement 2025; 21:e14445. [PMID: 39713891 PMCID: PMC11848341 DOI: 10.1002/alz.14445] [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: 09/13/2024] [Revised: 10/28/2024] [Accepted: 11/10/2024] [Indexed: 12/24/2024]
Abstract
INTRODUCTION TOMM40 and APOC1 variants can modulate the APOE-ε4-related Alzheimer's disease (AD) risk by up to fourfold. We aim to investigate whether the genetic modulation of ε4-related AD risk is reflected in brain morphology. METHODS We tested whether 27 magnetic resonance imaging-derived neuroimaging markers of neurodegeneration (volume and thickness in temporo-limbic regions) are associated with APOE-TOMM40-APOC1 polygenic profiles using the National Alzheimer's Coordinating Center Uniform Data Set linked to the AD Genetic Consortium data. RESULTS All brain regions studied using structural phenotypes were smaller in individuals with AD. The ε4 allele was associated with smaller limbic (entorhinal, hippocampus, parahippocampus) brain volume and cortical thickness in AD cases than controls. There were significant differences in the associations for the higher-risk and lower-risk ε4-bearing APOE-TOMM40-APOC1 profiles with temporo-limbic region markers. DISCUSSION The APOE-AD heterogeneity may be partly attributed to the modulating role of the TOMM40 and APOC1 genes in the APOE cluster. HIGHLIGHTS The ε4 allele is associated with smaller values of neuroimaging markers in AD cases. Larger values of neuroimaging markers may protect against AD in the ε4 carriers. TOMM40 and APOC1 variants differentiate AD risk in the ε4 carriers. The same variants can differentiate the links between ε4 and neuroimaging markers.
Collapse
Affiliation(s)
- Alexander M. Kulminski
- Biodemography of Aging Research UnitSocial Science Research InstituteDuke UniversityDurhamNorth CarolinaUSA
| | - Ethan Jain‐Washburn
- Biodemography of Aging Research UnitSocial Science Research InstituteDuke UniversityDurhamNorth CarolinaUSA
| | - Alireza Nazarian
- Biodemography of Aging Research UnitSocial Science Research InstituteDuke UniversityDurhamNorth CarolinaUSA
| | - Heather M. Wilkins
- Department of NeurologyUniversity of Kansas School of MedicineKansas CityKansasUSA
- University of Kansas Alzheimer's Disease CenterUniversity of Kansas School of MedicineKansas CityKansasUSA
| | - Olivia Veatch
- University of Kansas Alzheimer's Disease CenterUniversity of Kansas School of MedicineKansas CityKansasUSA
- Department of PsychiatryUniversity of Kansas School of MedicineKansas CityKansasUSA
| | - Russell H. Swerdlow
- Department of NeurologyUniversity of Kansas School of MedicineKansas CityKansasUSA
- University of Kansas Alzheimer's Disease CenterUniversity of Kansas School of MedicineKansas CityKansasUSA
| | - Robyn A. Honea
- Department of NeurologyUniversity of Kansas School of MedicineKansas CityKansasUSA
- University of Kansas Alzheimer's Disease CenterUniversity of Kansas School of MedicineKansas CityKansasUSA
| |
Collapse
|
4
|
Keresztes A, Bankó ÉM, Báthori N, Tomacsek V, Varga VA, Nárai Á, Nemecz Z, Dénes Á, Gál V, Hermann P, Simor P, Vidnyánszky Z. Multi-Night Electroencephalography Reveals Positive Association Between Sleep Efficiency and Hippocampal Subfield and Entorhinal Cortex Volumes in Healthy Aging. Hum Brain Mapp 2024; 45:e70090. [PMID: 39720895 PMCID: PMC11669120 DOI: 10.1002/hbm.70090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 11/08/2024] [Accepted: 11/18/2024] [Indexed: 12/26/2024] Open
Abstract
Age-related atrophy of the human hippocampus and the enthorinal cortex starts accelerating at around age 60. Due to the contributions of these regions to many cognitive functions seamlessly used in everyday life, this can heavily impact the lives of elderly people. The hippocampus is not a unitary structure, and mechanisms of its age-related decline appear to differentially affect its subfields. Human and animal studies have suggested that altered sleep is associated with hippocampal atrophy. Yet, we know little about subfield specific effects of altered sleep in healthy aging and their effect on cognition. Here, in a sample of 118 older middle-aged and older adults (Mage = 63.25 y, range: 50-80 y), we examined the association between highly reliable hippocampal subfield and entorhinal cortex volumetry (n = 112), sleep measures derived from multi-night recordings of portable electroencephalography (n = 61) and episodic memory (n = 117). Objective sleep efficiency-but not self-report measures of sleep-was associated with entorhinal cortex volume when controlling for age. Age-related differences in subfield volumes were associated with objective sleep efficiency, but not with self-report measures of sleep. Moreover, participants characterized by a common multivariate pattern of subfield volumes that contributed to positive sleep-subfield volume associations, showed lower rates of forgetting. Our results showcase the benefit of objective sleep measures in identifying potential contributors of age-related differences in brain-behavior couplings.
Collapse
Affiliation(s)
- Attila Keresztes
- Brain Imaging CentreHUN‐REN Research Centre for Natural SciencesBudapestHungary
- Institute of PsychologyELTE Eötvös Loránd UniversityBudapestHungary
| | - Éva M. Bankó
- Brain Imaging CentreHUN‐REN Research Centre for Natural SciencesBudapestHungary
| | - Noémi Báthori
- Brain Imaging CentreHUN‐REN Research Centre for Natural SciencesBudapestHungary
- Department of Cognitive ScienceBudapest University of Technology and EconomicsBudapestHungary
| | - Vivien Tomacsek
- Institute of PsychologyELTE Eötvös Loránd UniversityBudapestHungary
- Doctoral School of PsychologyELTE Eötvös Loránd UniversityBudapestHungary
| | - Virág Anna Varga
- Brain Imaging CentreHUN‐REN Research Centre for Natural SciencesBudapestHungary
| | - Ádám Nárai
- Brain Imaging CentreHUN‐REN Research Centre for Natural SciencesBudapestHungary
- Doctoral School of Biology and Sportbiology, Institute of Biology, Faculty of SciencesUniversity of PécsPécsHungary
| | - Zsuzsanna Nemecz
- Brain Imaging CentreHUN‐REN Research Centre for Natural SciencesBudapestHungary
- Institute of PsychologyELTE Eötvös Loránd UniversityBudapestHungary
- Doctoral School of PsychologyELTE Eötvös Loránd UniversityBudapestHungary
| | - Ádám Dénes
- Momentum Laboratory of NeuroimmunologyHUN‐REN Institute of Experimental MedicineBudapestHungary
| | - Viktor Gál
- Brain Imaging CentreHUN‐REN Research Centre for Natural SciencesBudapestHungary
| | - Petra Hermann
- Brain Imaging CentreHUN‐REN Research Centre for Natural SciencesBudapestHungary
| | - Péter Simor
- Institute of PsychologyELTE Eötvös Loránd UniversityBudapestHungary
- Institute of Behavioral SciencesSemmelweis UniversityBudapestHungary
| | - Zoltán Vidnyánszky
- Brain Imaging CentreHUN‐REN Research Centre for Natural SciencesBudapestHungary
| |
Collapse
|
5
|
Yu C, Zhang S, Shang M, Guo L, Han J, Du L. A Multi-Task Deep Feature Selection Method for Brain Imaging Genetics. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2024; 21:1613-1622. [PMID: 37432805 DOI: 10.1109/tcbb.2023.3294413] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/13/2023]
Abstract
Using brain imaging quantitative traits (QTs) for identifying genetic risk factors is an important research topic in brain imaging genetics. Many efforts have been made for this task via building linear models between imaging QTs and genetic factors such as single nucleotide polymorphisms (SNPs). To the best of our knowledge, linear models could not fully uncover the complicated relationship due to the loci's elusive and diverse influences on imaging QTs. In this paper, we propose a novel multi-task deep feature selection (MTDFS) method for brain imaging genetics. MTDFS first builds a multi-task deep neural network to model the complicated associations between imaging QTs and SNPs. And then designs a multi-task one-to-one layer and imposes a combined penalty to identify SNPs that make significant contributions. MTDFS can not only extract the nonlinear relationship but also arms the deep neural network with feature selection. We compared MTDFS to multi-task linear regression (MTLR) and single-task DFS (DFS) methods on the real neuroimaging genetic data. The experimental results showed that MTDFS performed better than MTLR and DFS on the QT-SNP relationship identification and feature selection. Thus, MTDFS is powerful for identifying risk loci and could be a great supplement to brain imaging genetics.
Collapse
|
6
|
Wu R, He B, Hou B, Saykin AJ, Yan J, Shen L. Cluster Analysis of Cortical Amyloid Burden for Identifying Imaging-driven Subtypes in Mild Cognitive Impairment. AMIA JOINT SUMMITS ON TRANSLATIONAL SCIENCE PROCEEDINGS. AMIA JOINT SUMMITS ON TRANSLATIONAL SCIENCE 2024; 2024:439-448. [PMID: 38827045 PMCID: PMC11141862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
Over the past decade, Alzheimer's disease (AD) has become increasingly severe and gained greater attention. Mild Cognitive Impairment (MCI) serves as an important prodromal stage of AD, highlighting the urgency of early diagnosis for timely treatment and control of the condition. Identifying the subtypes of MCI patients exhibits importance for dissecting the heterogeneity of this complex disorder and facilitating more effective target discovery and therapeutic development. Conventional method uses clinical measurements such as cognitive score and neurophysical assessment to stratify MCI patients into two groups with early MCI (EMCI) and late MCI (LMCI), which shows their progressive stages. However, such clinical method is not designed to de-convolute the heterogeneity of the disorder. This study uses a data-driven approach to divide MCI patients into a novel grouping of two subtypes based on an amyloid dataset of 68 cortical features from positron emission tomography (PET), where each subtype has a homogeneous cortical amyloid burden pattern. Experimental evaluation including visual two-dimensional cluster distribution, Kaplan-Meier plot, genetic association studies, and biomarker distribution analysis demonstrates that the identified subtypes performs better across all metrics than the conventional EMCI and LMCI grouping.
Collapse
Affiliation(s)
- Ruiming Wu
- University of Pennsylvania, Philadelphia, PA
| | - Bing He
- Indiana University, Indianapolis, IN
| | - Bojian Hou
- University of Pennsylvania, Philadelphia, PA
| | | | | | - Li Shen
- University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
7
|
Yang Z, Yang L, Sun Z, Rong Y, Bai C, Dong Q, Jian L. miRNA-660-3p inhibits malignancy in glioblastoma via negative regulation of APOC1-TGFβ2 signaling pathway. Cancer Biol Ther 2023; 24:2281459. [PMID: 37981873 PMCID: PMC10783846 DOI: 10.1080/15384047.2023.2281459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 11/06/2023] [Indexed: 11/21/2023] Open
Abstract
Glioblastoma as the most common and aggressive central nervous system tumor in adults. Its prognosis and therapeutic outcome are poor due to the limited understanding of its molecular mechanism. Apolipoprotein C-1 (APOC1) as a member of the apolipoprotein family that acts as a tumor promoter in various cancers. MicroRNA (miRNA) can silence gene expression and suppress tumor progression. However, the role of APOC1 and its upstream miRNA has not been explored in glioblastoma. Two glioblastoma cell lines (U87 and U251) were used to explore the role of APOC1 and its upstream miRNA-660-3p in glioblastoma tumorigenesis in vitro. Cells with APOC1/miRNA-660-3p overexpression or knockdown were assessed for their proliferation, migration, and invasion in vitro, and tumorigenesis in vivo. Gene and protein expression was assessed by qRT-PCR and western blot, respectively. Cell proliferation was assessed by the MTT assay and the EdU and Ki67 staining. Cell migration and invasion were assessed by the transwell assay. Tumorigenesis in vivo was assessed in U87 cells with a xenograft mouse model. APOC1 was overexpressed in glioblastoma compared with normal peritumoral tissue and was inversely related to patient prognosis. APOC1 overexpression promotes cell proliferation, migration, and invasion in vitro. APOC1 inhibition reduced tumor growth in vivo. miRNA-660-3p inhibits tumorigenesis by directly targeting APOC1. Mechanistically, APOC1 drives the malignancy of glioblastoma by activating the TGFβ2 signaling pathway. miRNA-660-3p suppresses tumorigenesis by targeting APOC1. Therefore, miRNA-660-3p/APOC1 axis can serve as potential intervention targets in managing glioblastoma progression.
Collapse
Affiliation(s)
- Zelin Yang
- Department of Neurosurgery, The Second Affiliated Hospital and Yuying Children’s Hospital, Wenzhou Medical University, Wenzhou, China
- School of Public Health and Management, Wenzhou Medical University, Wenzhou, China
| | - Liang Yang
- Department of Neurosurgery, The Second Affiliated Hospital and Yuying Children’s Hospital, Wenzhou Medical University, Wenzhou, China
| | - Zhenkai Sun
- Department of Neurosurgery, The Second Affiliated Hospital and Yuying Children’s Hospital, Wenzhou Medical University, Wenzhou, China
| | - Yuxi Rong
- Department of Neurosurgery, The Second Affiliated Hospital and Yuying Children’s Hospital, Wenzhou Medical University, Wenzhou, China
| | - Chenglian Bai
- School of Public Health and Management, Wenzhou Medical University, Wenzhou, China
| | - Qiaoxiang Dong
- Department of Neurosurgery, The Second Affiliated Hospital and Yuying Children’s Hospital, Wenzhou Medical University, Wenzhou, China
- School of Public Health and Management, Wenzhou Medical University, Wenzhou, China
| | - Lin Jian
- Department of Neurosurgery, The Second Affiliated Hospital and Yuying Children’s Hospital, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
8
|
Bae J, Logan PE, Acri DJ, Bharthur A, Nho K, Saykin AJ, Risacher SL, Nudelman K, Polsinelli AJ, Pentchev V, Kim J, Hammers DB, Apostolova LG, Alzheimer’s Disease Neuroimaging Initiative. A simulative deep learning model of SNP interactions on chromosome 19 for predicting Alzheimer's disease risk and rates of disease progression. Alzheimers Dement 2023; 19:5690-5699. [PMID: 37409680 PMCID: PMC10770299 DOI: 10.1002/alz.13319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 04/25/2023] [Accepted: 05/12/2023] [Indexed: 07/07/2023]
Abstract
BACKGROUND Identifying genetic patterns that contribute to Alzheimer's disease (AD) is important not only for pre-symptomatic risk assessment but also for building personalized therapeutic strategies. METHODS We implemented a novel simulative deep learning model to chromosome 19 genetic data from the Alzheimer's Disease Neuroimaging Initiative and the Imaging and Genetic Biomarkers of Alzheimer's Disease datasets. The model quantified the contribution of each single nucleotide polymorphism (SNP) and their epistatic impact on the likelihood of AD using the occlusion method. The top 35 AD-risk SNPs in chromosome 19 were identified, and their ability to predict the rate of AD progression was analyzed. RESULTS Rs561311966 (APOC1) and rs2229918 (ERCC1/CD3EAP) were recognized as the most powerful factors influencing AD risk. The top 35 chromosome 19 AD-risk SNPs were significant predictors of AD progression. DISCUSSION The model successfully estimated the contribution of AD-risk SNPs that account for AD progression at the individual level. This can help in building preventive precision medicine.
Collapse
Affiliation(s)
- Jinhyeong Bae
- Department of Neurology, School of Medicine, Indiana University School of Medicine, Indianapolis, IN, 46202, United States
| | - Paige E. Logan
- Department of Neurology, School of Medicine, Indiana University School of Medicine, Indianapolis, IN, 46202, United States
| | - Dominic J. Acri
- Department of Medical and Molecular Genetics, School of Medicine, Indiana University School of Medicine, Indianapolis, IN, 46202, United States
| | - Apoorva Bharthur
- Department of Neurology, School of Medicine, Indiana University School of Medicine, Indianapolis, IN, 46202, United States
| | - Kwangsik Nho
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, 46202, United States
| | - Andrew J. Saykin
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, 46202, United States
| | - Shannon L. Risacher
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, 46202, United States
| | - Kelly Nudelman
- Department of Medical and Molecular Genetics, School of Medicine, Indiana University School of Medicine, Indianapolis, IN, 46202, United States
| | - Angelina J. Polsinelli
- Department of Neurology, School of Medicine, Indiana University School of Medicine, Indianapolis, IN, 46202, United States
| | - Valentin Pentchev
- Department of Information Technology, Indiana University Network Science Institute, Bloomington, IN, 47408, United States
| | - Jungsu Kim
- Department of Medical and Molecular Genetics, School of Medicine, Indiana University School of Medicine, Indianapolis, IN, 46202, United States
| | - Dustin B. Hammers
- Department of Neurology, School of Medicine, Indiana University School of Medicine, Indianapolis, IN, 46202, United States
| | - Liana G. Apostolova
- Department of Neurology, School of Medicine, Indiana University School of Medicine, Indianapolis, IN, 46202, United States
- Department of Medical and Molecular Genetics, School of Medicine, Indiana University School of Medicine, Indianapolis, IN, 46202, United States
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, 46202, United States
| | | |
Collapse
|
9
|
Yu D, Wang L, Kong D, Zhu H. Mapping the Genetic-Imaging-Clinical Pathway with Applications to Alzheimer’s Disease. J Am Stat Assoc 2022; 117:1656-1668. [PMID: 37009529 PMCID: PMC10062702 DOI: 10.1080/01621459.2022.2087658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Alzheimer's disease is a progressive form of dementia that results in problems with memory, thinking, and behavior. It often starts with abnormal aggregation and deposition of β amyloid and tau, followed by neuronal damage such as atrophy of the hippocampi, leading to Alzheimers Disease (AD). The aim of this paper is to map the genetic-imaging-clinical pathway for AD in order to delineate the genetically-regulated brain changes that drive disease progression based on the Alzheimers Disease Neuroimaging Initiative (ADNI) dataset. We develop a novel two-step approach to delineate the association between high-dimensional 2D hippocampal surface exposures and the Alzheimers Disease Assessment Scale (ADAS) cognitive score, while taking into account the ultra-high dimensional clinical and genetic covariates at baseline. Analysis results suggest that the radial distance of each pixel of both hippocampi is negatively associated with the severity of behavioral deficits conditional on observed clinical and genetic covariates. These associations are stronger in Cornu Ammonis region 1 (CA1) and subiculum subregions compared to Cornu Ammonis region 2 (CA2) and Cornu Ammonis region 3 (CA3) subregions. Supplementary materials for this article, including a standardized description of the materials available for reproducing the work, are available as an online supplement.
Collapse
Affiliation(s)
- Dengdeng Yu
- Department of Mathematics, University of Texas at Arlington
| | - Linbo Wang
- Department of Statistical Sciences, University of Toronto
| | - Dehan Kong
- Department of Statistical Sciences, University of Toronto
| | - Hongtu Zhu
- Department of Biostatistics, University of North Carolina, Chapel Hill for the Alzheimer’s Disease Neuroimaging Initiative*
| |
Collapse
|
10
|
Kok FK, van Leerdam SL, de Lange ECM. Potential Mechanisms Underlying Resistance to Dementia in Non-Demented Individuals with Alzheimer's Disease Neuropathology. J Alzheimers Dis 2022; 87:51-81. [PMID: 35275527 PMCID: PMC9198800 DOI: 10.3233/jad-210607] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Alzheimer’s disease (AD) is the most common form of dementia and typically characterized by the accumulation of amyloid-β plaques and tau tangles. Intriguingly, there also exists a group of elderly which do not develop dementia during their life, despite the AD neuropathology, the so-called non-demented individuals with AD neuropathology (NDAN). In this review, we provide extensive background on AD pathology and normal aging and discuss potential mechanisms that enable these NDAN individuals to remain cognitively intact. Studies presented in this review show that NDAN subjects are generally higher educated and have a larger cognitive reserve. Furthermore, enhanced neural hypertrophy could compensate for hippocampal and cingulate neural atrophy in NDAN individuals. On a cellular level, these individuals show increased levels of neural stem cells and ‘von Economo neurons’. Furthermore, in NDAN brains, binding of Aβ oligomers to synapses is prevented, resulting in decreased glial activation and reduced neuroinflammation. Overall, the evidence stated here strengthens the idea that some individuals are more resistant to AD pathology, or at least show an elongation of the asymptomatic state of the disease compared to others. Insights into the mechanisms underlying this resistance could provide new insight in understanding normal aging and AD itself. Further research should focus on factors and mechanisms that govern the NDAN cognitive resilience in order to find clues on novel biomarkers, targets, and better treatments of AD.
Collapse
Affiliation(s)
- Frédérique K Kok
- Predictive Pharmacology, Division of Systems Biomedicine and Pharmacology, Leiden Academic Centre of Drug Research, Leiden University, Leiden, The Netherlands
| | - Suzanne L van Leerdam
- Predictive Pharmacology, Division of Systems Biomedicine and Pharmacology, Leiden Academic Centre of Drug Research, Leiden University, Leiden, The Netherlands
| | - Elizabeth C M de Lange
- Predictive Pharmacology, Division of Systems Biomedicine and Pharmacology, Leiden Academic Centre of Drug Research, Leiden University, Leiden, The Netherlands
| |
Collapse
|
11
|
Genome-wide association study identifies APOE locus influencing plasma p-tau181 levels. J Hum Genet 2022; 67:459-463. [DOI: 10.1038/s10038-022-01026-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 02/10/2022] [Accepted: 02/15/2022] [Indexed: 11/09/2022]
|
12
|
PCBP1 regulates the transcription and alternative splicing of metastasis‑related genes and pathways in hepatocellular carcinoma. Sci Rep 2021; 11:23356. [PMID: 34857818 PMCID: PMC8640068 DOI: 10.1038/s41598-021-02642-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 11/03/2021] [Indexed: 02/08/2023] Open
Abstract
PCBP1 is a multifunctional RNA-binding protein (RBP) expressed in most human cells and is involved in posttranscriptional gene regulation. PCBP1 regulates the alternative splicing, translation and RNA stability of many cancer-related genes and has been identified as a potential tumour suppressor gene. PCBP1 inhibits the invasion of hepatocellular carcinoma (HCC) cells, but there are few studies on the specific regulatory target and mechanism of RBPs in HCC, and it is unclear whether PCBP1 plays a role in tumour metastasis as a splicing factor. We analysed the regulation of gene expression by PCBP1 at the transcriptional level. We obtained and analysed PCBP1-knockdown RNA-seq data and eCLIP-seq data of PCBP1 in HepG2 cells and found that PCBP1 widely regulates the alternative splicing and expression of genes enriched in cancer-related pathways, including extracellular matrix, cell adhesion, small molecule metabolic process and apoptosis. We validated five regulated alternative splicing events affected by PCBP1 using RT-qPCR and found that there was a significant difference in the expression of APOC1 and SPHK1 between tumour and normal tissues. In this study, we provided convincing evidence that human PCBP1 profoundly regulates the splicing of genes associated with tumour metastasis. These findings provide new insight into potential markers or therapeutic targets for HCC treatment.
Collapse
|
13
|
Leal-Calvo T, Martins BL, Bertoluci DF, Rosa PS, de Camargo RM, Germano GV, Brito de Souza VN, Pereira Latini AC, Moraes MO. Large-Scale Gene Expression Signatures Reveal a Microbicidal Pattern of Activation in Mycobacterium leprae-Infected Monocyte-Derived Macrophages With Low Multiplicity of Infection. Front Immunol 2021; 12:647832. [PMID: 33936067 PMCID: PMC8085500 DOI: 10.3389/fimmu.2021.647832] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Accepted: 03/31/2021] [Indexed: 12/17/2022] Open
Abstract
Leprosy is a disease with a clinical spectrum of presentations that is also manifested in diverse histological features. At one pole, lepromatous lesions (L-pole) have phagocytic foamy macrophages heavily parasitized with freely multiplying intracellular Mycobacterium leprae. At the other pole, the presence of epithelioid giant cells and granulomatous formation in tuberculoid lesions (T-pole) lead to the control of M. leprae replication and the containment of its spread. The mechanism that triggers this polarization is unknown, but macrophages are central in this process. Over the past few years, leprosy has been studied using large scale techniques to shed light on the basic pathways that, upon infection, rewire the host cellular metabolism and gene expression. M. leprae is particularly peculiar as it invades Schwann cells in the nerves, reprogramming their gene expression leading to a stem-like cell phenotype. This modulatory behavior exerted by M. leprae is also observed in skin macrophages. Here, we used live M. leprae to infect (10:1 multiplicity of infection) monocyte-derived macrophages (MDMs) for 48 h and analyzed the whole gene expression profile using microarrays. In this model, we observe an intense upregulation of genes consistent with a cellular immune response, with enriched pathways including peptide and protein secretion, leukocyte activation, inflammation, and cellular divalent inorganic cation homeostasis. Among the most differentially expressed genes (DEGs) are CCL5/RANTES and CYP27B1, and several members of the metallothionein and metalloproteinase families. This is consistent with a proinflammatory state that would resemble macrophage rewiring toward granulomatous formation observed at the T-pole. Furthermore, a comparison with a dataset retrieved from the Gene Expression Omnibus of M. leprae-infected Schwann cells (MOI 100:1) showed that the patterns among the DEGs are highly distinct, as the Schwann cells under these conditions had a scavenging and phagocytic gene profile similar to M2-like macrophages, with enriched pathways rearrangements in the cytoskeleton, lipid and cholesterol metabolism and upregulated genes including MVK, MSMO1, and LACC1/FAMIN. In summary, macrophages may have a central role in defining the paradigmatic cellular (T-pole) vs. humoral (L-pole) responses and it is likely that the multiplicity of infection and genetic polymorphisms in key genes are gearing this polarization.
Collapse
Affiliation(s)
- Thyago Leal-Calvo
- Laboratório de Hanseníase, Instituto Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, Brazil
| | - Bruna Leticia Martins
- Divisão de Pesquisa e Ensino, Instituto Lauro de Souza Lima, Bauru, Brazil.,Departamento de Doenças Tropicais, Faculdade de Medicina de Botucatu, Universidade Estadual Paulista, Botucatu, Brazil
| | - Daniele Ferreira Bertoluci
- Divisão de Pesquisa e Ensino, Instituto Lauro de Souza Lima, Bauru, Brazil.,Departamento de Doenças Tropicais, Faculdade de Medicina de Botucatu, Universidade Estadual Paulista, Botucatu, Brazil
| | | | - Rodrigo Mendes de Camargo
- Divisão de Pesquisa e Ensino, Instituto Lauro de Souza Lima, Bauru, Brazil.,Departamento de Doenças Tropicais, Faculdade de Medicina de Botucatu, Universidade Estadual Paulista, Botucatu, Brazil
| | - Giovanna Vale Germano
- Divisão de Pesquisa e Ensino, Instituto Lauro de Souza Lima, Bauru, Brazil.,Departamento de Doenças Tropicais, Faculdade de Medicina de Botucatu, Universidade Estadual Paulista, Botucatu, Brazil
| | - Vania Nieto Brito de Souza
- Divisão de Pesquisa e Ensino, Instituto Lauro de Souza Lima, Bauru, Brazil.,Departamento de Doenças Tropicais, Faculdade de Medicina de Botucatu, Universidade Estadual Paulista, Botucatu, Brazil
| | - Ana Carla Pereira Latini
- Divisão de Pesquisa e Ensino, Instituto Lauro de Souza Lima, Bauru, Brazil.,Departamento de Doenças Tropicais, Faculdade de Medicina de Botucatu, Universidade Estadual Paulista, Botucatu, Brazil
| | - Milton Ozório Moraes
- Laboratório de Hanseníase, Instituto Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, Brazil
| |
Collapse
|
14
|
Farias FHG, Benitez BA, Cruchaga C. Quantitative endophenotypes as an alternative approach to understanding genetic risk in neurodegenerative diseases. Neurobiol Dis 2021; 151:105247. [PMID: 33429041 DOI: 10.1016/j.nbd.2020.105247] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 12/24/2020] [Accepted: 12/30/2020] [Indexed: 01/02/2023] Open
Abstract
Endophenotypes, as measurable intermediate features of human diseases, reflect underlying molecular mechanisms. The use of quantitative endophenotypes in genetic studies has improved our understanding of pathophysiological changes associated with diseases. The main advantage of the quantitative endophenotypes approach to study human diseases over a classic case-control study design is the inferred biological context that can enable the development of effective disease-modifying treatments. Here, we summarize recent progress on biomarkers for neurodegenerative diseases, including cerebrospinal fluid and blood-based, neuroimaging, neuropathological, and clinical studies. This review focuses on how endophenotypic studies have successfully linked genetic modifiers to disease risk, disease onset, or progression rate and provided biological context to genes identified in genome-wide association studies. Finally, we review critical methodological considerations for implementing this approach and future directions.
Collapse
Affiliation(s)
- Fabiana H G Farias
- Department of Psychiatry, Washington University, St. Louis, MO 63110, United States of America; NeuroGenomics and Informatics, Washington University, St. Louis, MO 63110, United States of America
| | - Bruno A Benitez
- Department of Psychiatry, Washington University, St. Louis, MO 63110, United States of America; NeuroGenomics and Informatics, Washington University, St. Louis, MO 63110, United States of America
| | - Carlos Cruchaga
- Department of Psychiatry, Washington University, St. Louis, MO 63110, United States of America; NeuroGenomics and Informatics, Washington University, St. Louis, MO 63110, United States of America; Hope Center for Neurologic Diseases, Washington University, St. Louis, MO 63110, United States of America; The Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University School of Medicine, St Louis, MO, 63110, United States of America; Department of Genetics, Washington University School of Medicine, St Louis, MO, 63110, United States of America.
| |
Collapse
|
15
|
Sabbagh MN, Pope E, Cordes L, Shi J, DeCourt B. Therapeutic considerations for APOE and TOMM40 in Alzheimers disease: A tribute to Allen Roses MD. Expert Opin Investig Drugs 2021; 30:39-44. [PMID: 33455481 PMCID: PMC9262379 DOI: 10.1080/13543784.2021.1849138] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 11/04/2020] [Indexed: 01/25/2023]
Abstract
Introduction: Four years ago this Autumn, pioneering neurologist Prof. Allen. D. Roses passed away. Hence, we have taken time to reflect on his work and legacy in Alzheimer's disease (AD) research. Prof. Roses rejected the widely accepted amyloid hypothesis, which identifies amyloid beta (Aβ) protein accumulation within the brain as the cause of AD. Instead, he proposed that the epsilon type 4 allele of apolipoprotein (APOE- Ɛ4) and translocase of outer mitochondrial membrane 40 homolog (TOMM40) were preeminent factors in the pathogenesis and progression of AD, particularly in late-onset AD (LOAD). This rejection of the amyloid hypothesis has generated new investigations into APOE and TOMM40 as risk factors for AD. Areas covered: We discuss the contributions of Prof. Roses to AD research, describe how APOE-Ɛ4 and TOMM40 have been posited to trigger neuropathological changes leading to AD, and explore paths to future clinical applications built on the foundations of his research. Expert opinion: The unconventional methodology of targeting APOE and TOMM40 offers great potential for the development of effective preventive and disease-modifying AD interventions. Future preclinical and clinical investigations will greatly benefit from the groundbreaking scientific discoveries of Prof. Roses.
Collapse
Affiliation(s)
| | - Evans Pope
- Cleveland Clinic, Lou Ruvo Center for Brain Health , Las Vegas, NV, USA
| | - Laura Cordes
- Cleveland Clinic, Lou Ruvo Center for Brain Health , Las Vegas, NV, USA
| | - Jiong Shi
- Cleveland Clinic, Lou Ruvo Center for Brain Health , Las Vegas, NV, USA
| | - Boris DeCourt
- Cleveland Clinic, Lou Ruvo Center for Brain Health , Las Vegas, NV, USA
| |
Collapse
|
16
|
Shi X, Wang J, Dai S, Qin L, Zhou J, Chen Y. Apolipoprotein C1 (APOC1): A Novel Diagnostic and Prognostic Biomarker for Cervical Cancer. Onco Targets Ther 2020; 13:12881-12891. [PMID: 33364782 PMCID: PMC7751697 DOI: 10.2147/ott.s280690] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 11/26/2020] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Previous reports showed that APOC1 was associated with several cancers but the function of APOC1 in cervical cancer was unknown. This study aimed to investigate the clinical effect and function of APOC1 in cervical cancer. MATERIALS AND METHODS In this study, the relative expression of APOC1 in cervical cancer was detected by RT-qPCR. In order to determine the cell proliferation and migration and invading ability and apoptosis more accurately, we used CCK8 assay, Edu assay, wound healing assay, migration and invasion assay, flow cytometry assay, co-immunoprecipitation, proteomics and Western blot by silencing and overexpressing APOC1, respectively. The role of APOC1 on tumor progression was explored in vitro and vivo. RESULTS The relative expression of APOC1 in cervical cancer tissues was up-regulated (P<0.05). In cervical cancer cell lines, silencing of APOC1 restrained cell progression and EMT, while over-expression of APOC1 accelerated cell progression and EMT in vivo and vitro (P<0.05). CONCLUSION APOC1 acts as an oncogene in cervical cancers and knockdown of APOC1 inhibited cervical cancer cells growth in vitro and in vivo. There is a close relationship between the relative expression of APOC1 and clinical outcome in cervical cancer patients.
Collapse
Affiliation(s)
- Xiu Shi
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, People’s Republic of China
- Clinical Research Center of Obstetrics and Gynecology, Jiangsu Key Laboratory of Clinical Immunology, Soochow University, Suzhou, Jiangsu Province, People’s Republic of China
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, People’s Republic of China
| | - Juan Wang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, People’s Republic of China
| | - Shouqian Dai
- Department of Emergency Medicine, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, People’s Republic of China
| | - Lingyan Qin
- Department of Pathology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, People’s Republic of China
| | - Jinhua Zhou
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, People’s Republic of China
| | - Youguo Chen
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, People’s Republic of China
| |
Collapse
|