1
|
İş Ö, Min Y, Wang X, Oatman SR, Abraham Daniel A, Ertekin‐Taner N. Multi Layered Omics Approaches Reveal Glia Specific Alterations in Alzheimer's Disease: A Systematic Review and Future Prospects. Glia 2025; 73:539-573. [PMID: 39652363 PMCID: PMC11784841 DOI: 10.1002/glia.24652] [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: 08/12/2024] [Revised: 11/11/2024] [Accepted: 11/16/2024] [Indexed: 02/01/2025]
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative dementia with multi-layered complexity in its molecular etiology. Multiple omics-based approaches, such as genomics, epigenomics, transcriptomics, proteomics, metabolomics, and lipidomics are enabling researchers to dissect this molecular complexity, and to uncover a plethora of alterations yielding insights into the pathophysiology of this disease. These approaches reveal multi-omics alterations essentially in all cell types of the brain, including glia. In this systematic review, we screen the literature for human studies implementing any omics approach within the last 10 years, to discover AD-associated molecular perturbations in brain glial cells. The findings from over 200 AD-related studies are reviewed under four different glial cell categories: microglia, oligodendrocytes, astrocytes and brain vascular cells. Under each category, we summarize the shared and unique molecular alterations identified in glial cells through complementary omics approaches. We discuss the implications of these findings for the development, progression and ultimately treatment of this complex disease as well as directions for future omics studies in glia cells.
Collapse
Affiliation(s)
- Özkan İş
- Department of NeuroscienceMayo ClinicJacksonvilleFloridaUSA
| | - Yuhao Min
- Department of NeuroscienceMayo ClinicJacksonvilleFloridaUSA
| | - Xue Wang
- Department of Quantitative Health SciencesMayo ClinicJacksonvilleFloridaUSA
| | | | | | - Nilüfer Ertekin‐Taner
- Department of NeuroscienceMayo ClinicJacksonvilleFloridaUSA
- Department of NeurologyMayo ClinicJacksonvilleFloridaUSA
| |
Collapse
|
2
|
Kumar S, Ramos E, Hidalgo A, Rodarte D, Sharma B, Torres MM, Devara D, Gadad SS. Integrated Multi-Omics Analyses of Synaptosomes Revealed Synapse-Centered Novel Targets in Alzheimer's Disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.09.631584. [PMID: 39868328 PMCID: PMC11761606 DOI: 10.1101/2025.01.09.631584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
Synapse dysfunction is an early event in Alzheimer's disease (AD) caused by various factors such as Amyloid beta, p-tau, inflammation, and aging. However, the exact molecular mechanism of synapse dysfunction in AD is largely unknown. To understand this, we comprehensively analyzed the synaptosome fraction in postmortem brain samples from AD patients and cognitively normal individuals. We conducted high-throughput transcriptomic analyses to identify changes in microRNA (miRNA) and mRNA levels in synaptosomes extracted from the brains of both unaffected individuals and those with Alzheimer's disease (AD). Additionally, we performed mass spectrometry analysis of synaptosomal proteins in the same sample group. These analyses revealed significant differences in the levels of miRNAs, mRNAs, and proteins between the groups. To further understand the pathways or molecules involved, we used an integrated omics approach and studied the molecular interactions of deregulated synapse miRNAs, mRNAs, and proteins in the samples from individuals with AD and the control group, which demonstrated the impact of deregulated miRNAs on their target mRNAs and proteins. Furthermore, the DIABLO analysis highlighted complex relationships between mRNAs, miRNAs, and proteins that could be key in understanding the pathophysiology of AD. Our study identified synapse-centered novel candidates that could be critical in restoring synapse dysfunction in AD.
Collapse
|
3
|
Tian S, Liu Y, Liu P, Nomura S, Wei Y, Huang T. Development and Validation of a Comprehensive Prognostic and Depression Risk Index for Gastric Adenocarcinoma. Int J Mol Sci 2024; 25:10776. [PMID: 39409106 PMCID: PMC11476876 DOI: 10.3390/ijms251910776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 09/24/2024] [Accepted: 09/26/2024] [Indexed: 10/20/2024] Open
Abstract
Depressive disorder contributes to the initiation and prognosis of patients with cancer, but the interaction between cancer and depressive disorder remains unclear. We generated a gastric adenocarcinoma patient-derived xenograft mice model, treated with chronic unpredictable mild stimulation. Based on the RNA-sequence from the mouse model, patient data from TCGA, and MDD-related (major depressive disorder) genes from the GEO database, 56 hub genes were identified by the intersection of differential expression genes from the three datasets. Molecular subtypes and a prognostic signature were generated based on the 56 genes. A depressive mouse model was constructed to test the key changes in the signatures. The signature was constructed based on the NDUFA4L2, ANKRD45, and AQP3 genes. Patients with high risk-score had a worse overall survival than the patients with low scores, consistent with the results from the two GEO cohorts. The comprehensive results showed that a higher risk-score was correlated with higher levels of tumor immune exclusion, higher infiltration of M0 macrophages, M2 macrophages, and neutrophils, higher angiogenetic activities, and more enriched epithelial-mesenchymal transition signaling pathways. A higher risk score was correlated to a higher MDD score, elevated MDD-related cytokines, and the dysfunction of neurogenesis-related genes, and parts of these changes showed similar trends in the animal model. With the Genomics of Drug Sensitivity in Cancer database, we found that the gastric adenocarcinoma patients with high risk-score may be sensitive to Pazopanib, XMD8.85, Midostaurin, HG.6.64.1, Elesclomol, Linifanib, AP.24534, Roscovitine, Cytarabine, and Axitinib. The gene signature consisting of the NDUFA4L2, ANKRD45, and AQP3 genes is a promising biomarker to distinguish the prognosis, the molecular and immune characteristics, the depressive risk, and the therapy candidates for gastric adenocarcinoma patients.
Collapse
Affiliation(s)
- Sheng Tian
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China; (S.T.); (Y.L.); (P.L.)
- Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China
- Hubei Cancer Clinical Study Center, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China
| | - Yixin Liu
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China; (S.T.); (Y.L.); (P.L.)
- Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China
- Hubei Cancer Clinical Study Center, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China
| | - Pan Liu
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China; (S.T.); (Y.L.); (P.L.)
- Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China
- Hubei Cancer Clinical Study Center, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China
| | - Sachiyo Nomura
- Department of Gastrointestinal Surgery, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan;
| | - Yongchang Wei
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China; (S.T.); (Y.L.); (P.L.)
- Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China
- Hubei Cancer Clinical Study Center, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China
| | - Tianhe Huang
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China; (S.T.); (Y.L.); (P.L.)
- Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China
- Hubei Cancer Clinical Study Center, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China
| |
Collapse
|
4
|
Johnston KG, Grieco SF, Nie Q, Theis FJ, Xu X. Small data methods in omics: the power of one. Nat Methods 2024; 21:1597-1602. [PMID: 39174710 DOI: 10.1038/s41592-024-02390-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Accepted: 07/24/2024] [Indexed: 08/24/2024]
Abstract
Over the last decade, biology has begun utilizing 'big data' approaches, resulting in large, comprehensive atlases in modalities ranging from transcriptomics to neural connectomics. However, these approaches must be complemented and integrated with 'small data' approaches to efficiently utilize data from individual labs. Integration of smaller datasets with major reference atlases is critical to provide context to individual experiments, and approaches toward integration of large and small data have been a major focus in many fields in recent years. Here we discuss progress in integration of small data with consortium-sized atlases across multiple modalities, and its potential applications. We then examine promising future directions for utilizing the power of small data to maximize the information garnered from small-scale experiments. We envision that, in the near future, international consortia comprising many laboratories will work together to collaboratively build reference atlases and foundation models using small data methods.
Collapse
Affiliation(s)
- Kevin G Johnston
- Department of Mathematics, University of California, Irvine, Irvine, CA, USA
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, Irvine, CA, USA
| | - Steven F Grieco
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, Irvine, CA, USA
- Center for Neural Circuit Mapping, University of California, Irvine, Irvine, CA, USA
| | - Qing Nie
- Department of Mathematics, University of California, Irvine, Irvine, CA, USA.
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, USA.
| | - Fabian J Theis
- Helmholtz Center Munich-German Research Center for Environmental Health, Institute of Computational Biology, Neuherberg, Germany.
- School of Life Sciences Weihenstephan, Technical University of Munich, Munich, Germany.
- Department of Mathematics, Technical University of Munich, Munich, Germany.
| | - Xiangmin Xu
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, Irvine, CA, USA.
- Center for Neural Circuit Mapping, University of California, Irvine, Irvine, CA, USA.
| |
Collapse
|
5
|
Escamilla S, Salas-Lucia F. Thyroid Hormone and Alzheimer Disease: Bridging Epidemiology to Mechanism. Endocrinology 2024; 165:bqae124. [PMID: 39276028 DOI: 10.1210/endocr/bqae124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 08/12/2024] [Accepted: 09/12/2024] [Indexed: 09/16/2024]
Abstract
The identification of critical factors that can worsen the mechanisms contributing to the pathophysiology of Alzheimer disease is of paramount importance. Thyroid hormones (TH) fit this criterion. Epidemiological studies have identified an association between altered circulating TH levels and Alzheimer disease. The study of human and animal models indicates that TH can affect all the main cellular, molecular, and genetic mechanisms known as hallmarks of Alzheimer disease. This is true not only for the excessive production in the brain of protein aggregates leading to amyloid plaques and neurofibrillary tangles but also for the clearance of these molecules from the brain parenchyma via the blood-brain barrier and for the escalated process of neuroinflammation-and even for the effects of carrying Alzheimer-associated genetic variants. Suboptimal TH levels result in a greater accumulation of protein aggregates in the brain. The direct TH regulation of critical genes involved in amyloid beta production and clearance is remarkable, affecting the expression of multiple genes, including APP (related to amyloid beta production), APOE, LRP1, TREM2, AQP4, and ABCB1 (related to amyloid beta clearance). TH also affects microglia by increasing their migration and function and directly regulating the immunosuppressor gene CD73, impacting the immune response of these cells. Studies aiming to understand the mechanisms that could explain how changes in TH levels can contribute to the brain alterations seen in patients with Alzheimer disease are ongoing. These studies have potential implications for the management of patients with Alzheimer disease and ultimately can contribute to devising new interventions for these conditions.
Collapse
Affiliation(s)
- Sergio Escamilla
- Instituto de Neurociencias, CSIC-Universidad Miguel Hernández, Alicante 03550, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Alicante 03550, Spain
- Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), Alicante 03010, Spain
| | | |
Collapse
|
6
|
Filomena E, Picardi E, Tullo A, Pesole G, D’Erchia AM. Identification of deregulated lncRNAs in Alzheimer's disease: an integrated gene co-expression network analysis of hippocampus and fusiform gyrus RNA-seq datasets. Front Aging Neurosci 2024; 16:1437278. [PMID: 39086756 PMCID: PMC11288953 DOI: 10.3389/fnagi.2024.1437278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 07/08/2024] [Indexed: 08/02/2024] Open
Abstract
Introduction The deregulation of lncRNAs expression has been associated with neuronal damage in Alzheimer's disease (AD), but how or whether they can influence its onset is still unknown. We investigated 2 RNA-seq datasets consisting, respectively, of the hippocampal and fusiform gyrus transcriptomic profile of AD patients, matched with non-demented controls. Methods We performed a differential expression analysis, a gene correlation network analysis (WGCNA) and a pathway enrichment analysis of two RNA-seq datasets. Results We found deregulated lncRNAs in common between hippocampus and fusiform gyrus and deregulated gene groups associated to functional pathways related to neurotransmission and memory consolidation. lncRNAs, co-expressed with known AD-related coding genes, were identified from the prioritized modules of both brain regions. Discussion We found common deregulated lncRNAs in the AD hippocampus and fusiform gyrus, that could be considered common signatures of AD pathogenesis, providing an important source of information for understanding the molecular changes of AD.
Collapse
Affiliation(s)
- Ermes Filomena
- Department of Biosciences, Biotechnology and Environment, University of Bari Aldo Moro, Bari, Italy
| | - Ernesto Picardi
- Department of Biosciences, Biotechnology and Environment, University of Bari Aldo Moro, Bari, Italy
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, National Research Council, Bari, Italy
| | - Apollonia Tullo
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, National Research Council, Bari, Italy
| | - Graziano Pesole
- Department of Biosciences, Biotechnology and Environment, University of Bari Aldo Moro, Bari, Italy
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, National Research Council, Bari, Italy
| | - Anna Maria D’Erchia
- Department of Biosciences, Biotechnology and Environment, University of Bari Aldo Moro, Bari, Italy
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, National Research Council, Bari, Italy
| |
Collapse
|
7
|
Juvenal G, Meinerz C, Ayupe AC, Campos HC, Reis EM, Longo BM, Pillat MM, Ulrich H. Bradykinin promotes immune responses in differentiated embryonic neurospheres carrying APP swe and PS1 dE9 mutations. Cell Biosci 2024; 14:82. [PMID: 38890712 PMCID: PMC11184896 DOI: 10.1186/s13578-024-01251-3] [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: 02/07/2024] [Accepted: 05/24/2024] [Indexed: 06/20/2024] Open
Abstract
BACKGROUND Neural progenitor cells (NPCs) can be cultivated from developing brains, reproducing many of the processes that occur during neural development. They can be isolated from a variety of animal models, such as transgenic mice carrying mutations in amyloid precursor protein (APP) and presenilin 1 and 2 (PSEN 1 and 2), characteristic of familial Alzheimer's disease (fAD). Modulating the development of these cells with inflammation-related peptides, such as bradykinin (BK) and its antagonist HOE-140, enables the understanding of the impact of such molecules in a relevant AD model. RESULTS We performed a global gene expression analysis on transgenic neurospheres treated with BK and HOE-140. To validate the microarray data, quantitative real-time reverse-transcription polymerase chain reaction (RT-PCR) was performed on 8 important genes related to the immune response in AD such as CCL12, CCL5, CCL3, C3, CX3CR1, TLR2 and TNF alpha and Iba-1. Furthermore, comparative analysis of the transcriptional profiles was performed between treatments, including gene ontology and reactome enrichment, construction and analysis of protein-protein interaction networks and, finally, comparison of our data with human dataset from AD patients. The treatments affected the expression levels of genes mainly related to microglia-mediated neuroinflammatory responses, with BK promoting an increase in the expression of genes that enrich processes, biological pathways, and cellular components related to immune dysfunction, neurodegeneration and cell cycle. B2 receptor inhibition by HOE-140 resulted in the reduction of AD-related anomalies caused in this system. CONCLUSIONS BK is an important immunomodulatory agent and enhances the immunological changes identified in transgenic neurospheres carrying the genetic load of AD. Bradykinin treatments modulate the expression rates of genes related to microglia-mediated neuroinflammation. Inhibiting bradykinin activity in Alzheimer's disease may slow disease progression.
Collapse
Affiliation(s)
- Guilherme Juvenal
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, Av. Prof. Lineu Prestes 748, São Paulo, 05508-900, Brazil
| | - Carine Meinerz
- Department of Microbiology and Parasitology, Health Sciences Center, Federal University of Santa Maria, Santa Maria-RS, Brazil
| | - Ana Carolina Ayupe
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, Av. Prof. Lineu Prestes 748, São Paulo, 05508-900, Brazil
| | | | - Eduardo Moraes Reis
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, Av. Prof. Lineu Prestes 748, São Paulo, 05508-900, Brazil
| | | | - Micheli Mainardi Pillat
- Department of Microbiology and Parasitology, Health Sciences Center, Federal University of Santa Maria, Santa Maria-RS, Brazil.
| | - Henning Ulrich
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, Av. Prof. Lineu Prestes 748, São Paulo, 05508-900, Brazil.
| |
Collapse
|
8
|
Gujjala VA, Klimek I, Abyadeh M, Tyshkovskiy A, Oz N, Castro JP, Gladyshev VN, Newton J, Kaya A. A disease similarity approach identifies short-lived Niemann-Pick type C disease mice with accelerated brain aging as a novel mouse model for Alzheimer's disease and aging research. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.19.590328. [PMID: 38712089 PMCID: PMC11071364 DOI: 10.1101/2024.04.19.590328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
Since its first description in 1906 by Dr. Alois Alzheimer, Alzheimer's disease (AD) has been the most common type of dementia. Initially thought to be caused by age-associated accumulation of plaques, in recent years, research has increasingly associated AD with lysosomal storage and metabolic disorders, and the explanation of its pathogenesis has shifted from amyloid and tau accumulation to oxidative stress and impaired lipid and glucose metabolism aggravated by hypoxic conditions. However, the underlying mechanisms linking those cellular processes and conditions to disease progression have yet to be defined. Here, we applied a disease similarity approach to identify unknown molecular targets of AD by using transcriptomic data from congenital diseases known to increase AD risk, namely Down Syndrome, Niemann Pick Disease Type C (NPC), and Mucopolysaccharidoses I. We uncovered common pathways, hub genes, and miRNAs across in vitro and in vivo models of these diseases as potential molecular targets for neuroprotection and amelioration of AD pathology, many of which have never been associated with AD. We then investigated common molecular alterations in brain samples from an NPC disease mouse model by juxtaposing them with brain samples of both human and mouse models of AD. Detailed phenotypic and molecular analyses revealed that the NPC mut mouse model can serve as a potential short-lived in vivo model for AD research and for understanding molecular factors affecting brain aging. This research represents the first comprehensive approach to congenital disease association with neurodegeneration and a new perspective on AD research while highlighting shortcomings and lack of correlation in diverse in vitro models. Considering the lack of an AD mouse model that recapitulates the physiological hallmarks of brain aging, the characterization of a short-lived NPC mouse model will further accelerate the research in these fields and offer a unique model for understanding the molecular mechanisms of AD from a perspective of accelerated brain aging.
Collapse
|
9
|
Zhang W, Mou M, Hu W, Lu M, Zhang H, Zhang H, Luo Y, Xu H, Tao L, Dai H, Gao J, Zhu F. MOINER: A Novel Multiomics Early Integration Framework for Biomedical Classification and Biomarker Discovery. J Chem Inf Model 2024; 64:2720-2732. [PMID: 38373720 DOI: 10.1021/acs.jcim.4c00013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/21/2024]
Abstract
In the context of precision medicine, multiomics data integration provides a comprehensive understanding of underlying biological processes and is critical for disease diagnosis and biomarker discovery. One commonly used integration method is early integration through concatenation of multiple dimensionally reduced omics matrices due to its simplicity and ease of implementation. However, this approach is seriously limited by information loss and lack of latent feature interaction. Herein, a novel multiomics early integration framework (MOINER) based on information enhancement and image representation learning is thus presented to address the challenges. MOINER employs the self-attention mechanism to capture the intrinsic correlations of omics-features, which make it significantly outperform the existing state-of-the-art methods for multiomics data integration. Moreover, visualizing the attention embedding and identifying potential biomarkers offer interpretable insights into the prediction results. All source codes and model for MOINER are freely available https://github.com/idrblab/MOINER.
Collapse
Affiliation(s)
- Wei Zhang
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Alibaba-Zhejiang University Joint Research Center of Future Digital Healthcare, Hangzhou 330110, China
| | - Minjie Mou
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Wei Hu
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Mingkun Lu
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Hanyu Zhang
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Hongning Zhang
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Yongchao Luo
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Hongquan Xu
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China
| | - Lin Tao
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China
| | - Haibin Dai
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Jianqing Gao
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Feng Zhu
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Alibaba-Zhejiang University Joint Research Center of Future Digital Healthcare, Hangzhou 330110, China
| |
Collapse
|
10
|
Zhang Z, Liu X, Zhang S, Song Z, Lu K, Yang W. A review and analysis of key biomarkers in Alzheimer's disease. Front Neurosci 2024; 18:1358998. [PMID: 38445255 PMCID: PMC10912539 DOI: 10.3389/fnins.2024.1358998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 02/02/2024] [Indexed: 03/07/2024] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder that affects over 50 million elderly individuals worldwide. Although the pathogenesis of AD is not fully understood, based on current research, researchers are able to identify potential biomarker genes and proteins that may serve as effective targets against AD. This article aims to present a comprehensive overview of recent advances in AD biomarker identification, with highlights on the use of various algorithms, the exploration of relevant biological processes, and the investigation of shared biomarkers with co-occurring diseases. Additionally, this article includes a statistical analysis of key genes reported in the research literature, and identifies the intersection with AD-related gene sets from databases such as AlzGen, GeneCard, and DisGeNet. For these gene sets, besides enrichment analysis, protein-protein interaction (PPI) networks utilized to identify central genes among the overlapping genes. Enrichment analysis, protein interaction network analysis, and tissue-specific connectedness analysis based on GTEx database performed on multiple groups of overlapping genes. Our work has laid the foundation for a better understanding of the molecular mechanisms of AD and more accurate identification of key AD markers.
Collapse
Affiliation(s)
- Zhihao Zhang
- School of Computer Science and Technology, Xinjiang University, Ürümqi, China
- College of Medical Engineering and Technology, Xinjiang Medical University, Ürümqi, China
| | - Xiangtao Liu
- College of Medical Engineering and Technology, Xinjiang Medical University, Ürümqi, China
| | - Suixia Zhang
- College of Medical Engineering and Technology, Xinjiang Medical University, Ürümqi, China
- College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, China
- State Key Laboratory of Pathogenesis, Prevention, Treatment of Central Asian High Incidence Diseases, First Affiliated Hospital of Xinjiang Medical University, Ürümqi, China
| | - Zhixin Song
- College of Medical Engineering and Technology, Xinjiang Medical University, Ürümqi, China
| | - Ke Lu
- School of Computer Science and Technology, Xinjiang University, Ürümqi, China
| | - Wenzhong Yang
- School of Computer Science and Technology, Xinjiang University, Ürümqi, China
| |
Collapse
|
11
|
Luo H, Liang H, Liu H, Fan Z, Wei Y, Yao X, Cong S. TEMINET: A Co-Informative and Trustworthy Multi-Omics Integration Network for Diagnostic Prediction. Int J Mol Sci 2024; 25:1655. [PMID: 38338932 PMCID: PMC10855161 DOI: 10.3390/ijms25031655] [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: 12/29/2023] [Revised: 01/20/2024] [Accepted: 01/26/2024] [Indexed: 02/12/2024] Open
Abstract
Advancing the domain of biomedical investigation, integrated multi-omics data have shown exceptional performance in elucidating complex human diseases. However, as the variety of omics information expands, precisely perceiving the informativeness of intra- and inter-omics becomes challenging due to the intricate interrelations, thus presenting significant challenges in the integration of multi-omics data. To address this, we introduce a novel multi-omics integration approach, referred to as TEMINET. This approach enhances diagnostic prediction by leveraging an intra-omics co-informative representation module and a trustworthy learning strategy used to address inter-omics fusion. Considering the multifactorial nature of complex diseases, TEMINET utilizes intra-omics features to construct disease-specific networks; then, it applies graph attention networks and a multi-level framework to capture more collective informativeness than pairwise relations. To perceive the contribution of co-informative representations within intra-omics, we designed a trustworthy learning strategy to identify the reliability of each omics in integration. To integrate inter-omics information, a combined-beliefs fusion approach is deployed to harmonize the trustworthy representations of different omics types effectively. Our experiments across four different diseases using mRNA, methylation, and miRNA data demonstrate that TEMINET achieves advanced performance and robustness in classification tasks.
Collapse
Affiliation(s)
- Haoran Luo
- Qingdao Innovation and Development Center, Harbin Engineering University, Qingdao 266000, China; (H.L.); (Z.F.)
- College of Intelligent Systems Science and Engineering, Harbin Engineering University, Harbin 150001, China; (H.L.); (H.L.); (Y.W.)
| | - Hong Liang
- College of Intelligent Systems Science and Engineering, Harbin Engineering University, Harbin 150001, China; (H.L.); (H.L.); (Y.W.)
| | - Hongwei Liu
- College of Intelligent Systems Science and Engineering, Harbin Engineering University, Harbin 150001, China; (H.L.); (H.L.); (Y.W.)
| | - Zhoujie Fan
- Qingdao Innovation and Development Center, Harbin Engineering University, Qingdao 266000, China; (H.L.); (Z.F.)
| | - Yanhui Wei
- College of Intelligent Systems Science and Engineering, Harbin Engineering University, Harbin 150001, China; (H.L.); (H.L.); (Y.W.)
| | - Xiaohui Yao
- Qingdao Innovation and Development Center, Harbin Engineering University, Qingdao 266000, China; (H.L.); (Z.F.)
- College of Intelligent Systems Science and Engineering, Harbin Engineering University, Harbin 150001, China; (H.L.); (H.L.); (Y.W.)
| | - Shan Cong
- Qingdao Innovation and Development Center, Harbin Engineering University, Qingdao 266000, China; (H.L.); (Z.F.)
- College of Intelligent Systems Science and Engineering, Harbin Engineering University, Harbin 150001, China; (H.L.); (H.L.); (Y.W.)
| |
Collapse
|
12
|
Dai Y, Hsu YC, Fernandes BS, Zhang K, Li X, Enduru N, Liu A, Manuel AM, Jiang X, Zhao Z. Disentangling Accelerated Cognitive Decline from the Normal Aging Process and Unraveling Its Genetic Components: A Neuroimaging-Based Deep Learning Approach. J Alzheimers Dis 2024; 97:1807-1827. [PMID: 38306043 PMCID: PMC11649026 DOI: 10.3233/jad-231020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2024]
Abstract
Background The progressive cognitive decline, an integral component of Alzheimer's disease (AD), unfolds in tandem with the natural aging process. Neuroimaging features have demonstrated the capacity to distinguish cognitive decline changes stemming from typical brain aging and AD between different chronological points. Objective To disentangle the normal aging effect from the AD-related accelerated cognitive decline and unravel its genetic components using a neuroimaging-based deep learning approach. Methods We developed a deep-learning framework based on a dual-loss Siamese ResNet network to extract fine-grained information from the longitudinal structural magnetic resonance imaging (MRI) data from the Alzheimer's Disease Neuroimaging Initiative (ADNI) study. We then conducted genome-wide association studies (GWAS) and post-GWAS analyses to reveal the genetic basis of AD-related accelerated cognitive decline. Results We used our model to process data from 1,313 individuals, training it on 414 cognitively normal people and predicting cognitive assessment for all participants. In our analysis of accelerated cognitive decline GWAS, we identified two genome-wide significant loci: APOE locus (chromosome 19 p13.32) and rs144614292 (chromosome 11 p15.1). Variant rs144614292 (G > T) has not been reported in previous AD GWA studies. It is within the intronic region of NELL1, which is expressed in neurons and plays a role in controlling cell growth and differentiation. The cell-type-specific enrichment analysis and functional enrichment of GWAS signals highlighted the microglia and immune-response pathways. Conclusions Our deep learning model effectively extracted relevant neuroimaging features and predicted individual cognitive decline. We reported a novel variant (rs144614292) within the NELL1 gene.
Collapse
Affiliation(s)
- Yulin Dai
- Center for Precision Health, McWilliams School of
Biomedical Informatics, The University of Texas Health Science Center at Houston,
Houston, TX, USA
| | - Yu-Chun Hsu
- Center for Secure Artificial Intelligence for Healthcare,
School of Biomedical Informatics, The University of Texas Health Science Center at
Houston, Houston, TX, USA
| | - Brisa S. Fernandes
- Center for Precision Health, McWilliams School of
Biomedical Informatics, The University of Texas Health Science Center at Houston,
Houston, TX, USA
| | - Kai Zhang
- Center for Secure Artificial Intelligence for Healthcare,
School of Biomedical Informatics, The University of Texas Health Science Center at
Houston, Houston, TX, USA
| | - Xiaoyang Li
- Center for Precision Health, McWilliams School of
Biomedical Informatics, The University of Texas Health Science Center at Houston,
Houston, TX, USA
- Department of Biostatistics and Data Science, School of
Public Health, The University of Texas Health Science Center at Houston, Houston,
TX, USA
| | - Nitesh Enduru
- Center for Precision Health, McWilliams School of
Biomedical Informatics, The University of Texas Health Science Center at Houston,
Houston, TX, USA
- Department of Epidemiology, Human Genetics and
Environmental Sciences, School of Public Health, The University of Texas Health
Science Center at Houston, Houston, TX, USA
| | - Andi Liu
- Center for Precision Health, McWilliams School of
Biomedical Informatics, The University of Texas Health Science Center at Houston,
Houston, TX, USA
- Department of Epidemiology, Human Genetics and
Environmental Sciences, School of Public Health, The University of Texas Health
Science Center at Houston, Houston, TX, USA
| | - Astrid M. Manuel
- Center for Precision Health, McWilliams School of
Biomedical Informatics, The University of Texas Health Science Center at Houston,
Houston, TX, USA
| | - Xiaoqian Jiang
- Center for Secure Artificial Intelligence for Healthcare,
School of Biomedical Informatics, The University of Texas Health Science Center at
Houston, Houston, TX, USA
| | - Zhongming Zhao
- Center for Precision Health, McWilliams School of
Biomedical Informatics, The University of Texas Health Science Center at Houston,
Houston, TX, USA
- Department of Epidemiology, Human Genetics and
Environmental Sciences, School of Public Health, The University of Texas Health
Science Center at Houston, Houston, TX, USA
- Department of Biomedical Informatics, Vanderbilt University
Medical enter, Nashville, TN, USA
| |
Collapse
|
13
|
Rojo D, Dal Cengio L, Badner A, Kim S, Sakai N, Greene J, Dierckx T, Mehl LC, Eisinger E, Ransom J, Arellano-Garcia C, Gumma ME, Soyk RL, Lewis CM, Lam M, Weigel MK, Damonte VM, Yalçın B, Jones SE, Ollila HM, Nishino S, Gibson EM. BMAL1 loss in oligodendroglia contributes to abnormal myelination and sleep. Neuron 2023; 111:3604-3618.e11. [PMID: 37657440 PMCID: PMC10873033 DOI: 10.1016/j.neuron.2023.08.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 06/28/2023] [Accepted: 08/03/2023] [Indexed: 09/03/2023]
Abstract
Myelination depends on the maintenance of oligodendrocytes that arise from oligodendrocyte precursor cells (OPCs). We show that OPC-specific proliferation, morphology, and BMAL1 are time-of-day dependent. Knockout of Bmal1 in mouse OPCs during development disrupts the expression of genes associated with circadian rhythms, proliferation, density, morphology, and migration, leading to changes in OPC dynamics in a spatiotemporal manner. Furthermore, these deficits translate into thinner myelin, dysregulated cognitive and motor functions, and sleep fragmentation. OPC-specific Bmal1 loss in adulthood does not alter OPC density at baseline but impairs the remyelination of a demyelinated lesion driven by changes in OPC morphology and migration. Lastly, we show that sleep fragmentation is associated with increased prevalence of the demyelinating disorder multiple sclerosis (MS), suggesting a link between MS and sleep that requires further investigation. These findings have broad mechanistic and therapeutic implications for brain disorders that include both myelin and sleep phenotypes.
Collapse
Affiliation(s)
- Daniela Rojo
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Palo Alto, CA 94305, USA
| | - Louisa Dal Cengio
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Palo Alto, CA 94305, USA
| | - Anna Badner
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Palo Alto, CA 94305, USA
| | - Samuel Kim
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Palo Alto, CA 94305, USA
| | - Noriaki Sakai
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Palo Alto, CA 94305, USA
| | - Jacob Greene
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Palo Alto, CA 94305, USA
| | - Tess Dierckx
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Palo Alto, CA 94305, USA
| | - Lindsey C Mehl
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Palo Alto, CA 94305, USA; Cancer Biology Graduate Program, Stanford University School of Medicine, Palo Alto, CA 94305, USA
| | - Ella Eisinger
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Palo Alto, CA 94305, USA
| | - Julia Ransom
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Palo Alto, CA 94305, USA
| | - Caroline Arellano-Garcia
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Palo Alto, CA 94305, USA; Biology Graduate Program, Stanford University, Palo Alto, CA 94305, USA
| | - Mohammad E Gumma
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Palo Alto, CA 94305, USA
| | - Rebecca L Soyk
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Palo Alto, CA 94305, USA
| | - Cheyanne M Lewis
- Neuroscience Graduate Program, Stanford University, Palo Alto, CA 94305, USA
| | - Mable Lam
- Department of Neurosurgery, Stanford University School of Medicine, Palo Alto, CA 94305, USA
| | - Maya K Weigel
- Department of Neurosurgery, Stanford University School of Medicine, Palo Alto, CA 94305, USA; Stem Cell Biology and Regenerative Medicine Program, Stanford University School of Medicine, Palo Alto, CA 94305, USA
| | - Valentina Martinez Damonte
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Palo Alto, CA 94305, USA
| | - Belgin Yalçın
- Department of Neurology & Neurological Sciences, Stanford University School of Medicine, Palo Alto, CA 94305, USA
| | - Samuel E Jones
- Institute for Molecular Medicine, HiLIFE, University of Helsinki, Helsinki 00014, Finland
| | - Hanna M Ollila
- Institute for Molecular Medicine, HiLIFE, University of Helsinki, Helsinki 00014, Finland; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, and Harvard Medical School, Boston, MA 02114, USA
| | - Seiji Nishino
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Palo Alto, CA 94305, USA
| | - Erin M Gibson
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Palo Alto, CA 94305, USA.
| |
Collapse
|
14
|
Rosewood TJ, Nho K, Risacher SL, Gao S, Shen L, Foroud T, Saykin AJ. Genome-Wide Association Analysis across Endophenotypes in Alzheimer's Disease: Main Effects and Disease Stage-Specific Interactions. Genes (Basel) 2023; 14:2010. [PMID: 38002954 PMCID: PMC10671827 DOI: 10.3390/genes14112010] [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: 09/11/2023] [Revised: 10/23/2023] [Accepted: 10/23/2023] [Indexed: 11/26/2023] Open
Abstract
The underlying genetic susceptibility for Alzheimer's disease (AD) is not yet fully understood. The heterogeneous nature of the disease challenges genetic association studies. Endophenotype approaches can help to address this challenge by more direct interrogation of biological traits related to the disease. AD endophenotypes based on amyloid-β, tau, and neurodegeneration (A/T/N) biomarkers and cognitive performance were selected from the Alzheimer's Disease Neuroimaging Initiative (ADNI) cohort (N = 1565). A genome-wide association study (GWAS) of quantitative phenotypes was performed using an SNP main effect and an SNP by Diagnosis interaction (SNP × DX) model to identify disease stage-specific genetic effects. Nine loci were identified as study-wide significant with one or more A/T/N endophenotypes in the main effect model, as well as additional findings significantly associated with cognitive measures. These nine loci include SNPs in or near the genes APOE, SRSF10, HLA-DQB1, XKR3, and KIAA1671. The SNP × DX model identified three study-wide significant genetic loci (BACH2, EP300, and PACRG-AS1) with a neuroprotective effect in later AD stage endophenotypes. An endophenotype approach identified novel genetic associations and provided insight into the molecular mechanisms underlying the genetic associations that may otherwise be missed using conventional case-control study designs.
Collapse
Affiliation(s)
- Thea J. Rosewood
- Indiana Alzheimer’s Disease Research Center, Indianapolis, IN 46202, USA; (T.J.R.); (S.L.R.); (S.G.); (T.F.)
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Kwangsik Nho
- Indiana Alzheimer’s Disease Research Center, Indianapolis, IN 46202, USA; (T.J.R.); (S.L.R.); (S.G.); (T.F.)
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- School of Informatics and Computing, Indiana University, Indianapolis, IN 46202, USA
| | - Shannon L. Risacher
- Indiana Alzheimer’s Disease Research Center, Indianapolis, IN 46202, USA; (T.J.R.); (S.L.R.); (S.G.); (T.F.)
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Sujuan Gao
- Indiana Alzheimer’s Disease Research Center, Indianapolis, IN 46202, USA; (T.J.R.); (S.L.R.); (S.G.); (T.F.)
- Department of Biostatistics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Li Shen
- Department of Biostatistics, Epidemiology and Informatics, The Perelman School of Medicine, Philadelphia, PA 19104, USA;
| | - Tatiana Foroud
- Indiana Alzheimer’s Disease Research Center, Indianapolis, IN 46202, USA; (T.J.R.); (S.L.R.); (S.G.); (T.F.)
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Andrew J. Saykin
- Indiana Alzheimer’s Disease Research Center, Indianapolis, IN 46202, USA; (T.J.R.); (S.L.R.); (S.G.); (T.F.)
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | | |
Collapse
|
15
|
Tang S, Buchman AS, Wang Y, Avey D, Xu J, Tasaki S, Bennett DA, Zheng Q, Yang J. Differential gene expression analysis based on linear mixed model corrects false positive inflation for studying quantitative traits. Sci Rep 2023; 13:16570. [PMID: 37789141 PMCID: PMC10547771 DOI: 10.1038/s41598-023-43686-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 09/27/2023] [Indexed: 10/05/2023] Open
Abstract
Differential gene expression (DGE) analysis has been widely employed to identify genes expressed differentially with respect to a trait of interest using RNA sequencing (RNA-Seq) data. Recent RNA-Seq data with large samples pose challenges to existing DGE methods, which were mainly developed for dichotomous traits and small sample sizes. Especially, existing DGE methods are likely to result in inflated false positive rates. To address this gap, we employed a linear mixed model (LMM) that has been widely used in genetic association studies for DGE analysis of quantitative traits. We first applied the LMM method to the discovery RNA-Seq data of dorsolateral prefrontal cortex (DLPFC) tissue (n = 632) with four continuous measures of Alzheimer's Disease (AD) cognitive and neuropathologic traits. The quantile-quantile plots of p-values showed that false positive rates were well calibrated by LMM, whereas other methods not accounting for sample-specific mixed effects led to serious inflation. LMM identified 37 potentially significant genes with differential expression in DLPFC for at least one of the AD traits, 17 of which were replicated in the additional RNA-Seq data of DLPFC, supplemental motor area, spinal cord, and muscle tissues. This application study showed not only well calibrated DGE results by LMM, but also possibly shared gene regulatory mechanisms of AD traits across different relevant tissues.
Collapse
Affiliation(s)
- Shizhen Tang
- Department of Human Genetics, Center for Computational and Quantitative Genetics, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Department of Biostatistics and Bioinformatics, Emory University School of Public Health, Atlanta, GA, 30322, USA
| | - Aron S Buchman
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, 60612, USA
| | - Yanling Wang
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, 60612, USA
| | - Denis Avey
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, 60612, USA
| | - Jishu Xu
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, 60612, USA
| | - Shinya Tasaki
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, 60612, USA
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, 60612, USA
| | - Qi Zheng
- Department of Bioinformatics and Biostatistics, University of Louisville, 485 E. Gray St, Louisville, KY, 40202, USA.
| | - Jingjing Yang
- Department of Human Genetics, Center for Computational and Quantitative Genetics, Emory University School of Medicine, Atlanta, GA, 30322, USA.
| |
Collapse
|
16
|
Dai Y, Yu-Chun H, Fernandes BS, Zhang K, Xiaoyang L, Enduru N, Liu A, Manuel AM, Jiang X, Zhao Z. Disentangling accelerated cognitive decline from the normal aging process and unraveling its genetic components: A neuroimaging-based deep learning approach. RESEARCH SQUARE 2023:rs.3.rs-3328861. [PMID: 37720047 PMCID: PMC10503860 DOI: 10.21203/rs.3.rs-3328861/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/19/2023]
Abstract
Background The progressive cognitive decline that is an integral component of AD unfolds in tandem with the natural aging process. Neuroimaging features have demonstrated the capacity to distinguish cognitive decline changes stemming from typical brain aging and Alzheimer's disease between different chronological points. Methods We developed a deep-learning framework based on dual-loss Siamese ResNet network to extract fine-grained information from the longitudinal structural magnetic resonance imaging (MRI) data from the Alzheimer's Disease Neuroimaging Initiative (ADNI) study. We then conducted genome-wide association studies (GWAS) and post-GWAS analyses to reveal the genetic basis of AD-related accelerated cognitive decline. Results We used our model to process data from 1,313 individuals, training it on 414 cognitively normal people and predicting cognitive assessment for all participants. In our analysis of accelerated cognitive decline GWAS, we identified two genome-wide significant loci: APOE locus (chromosome 19 p13.32) and rs144614292 (chromosome 11 p15.1). Variant rs144614292 (G>T) has not been reported in previous AD GWA studies. It is within the intronic region of NELL1, which is expressed in neuron and plays a role in controlling cell growth and differentiation. In addition, MUC7 and PROL1/OPRPNon chromosome 4 were significant at the gene level. The cell-type-specific enrichment analysis and functional enrichment of GWAS signals highlighted the microglia and immune-response pathways. Furthermore, we found that the cognitive decline slope GWAS was positively correlated with previous AD GWAS. Conclusion Our deep learning model was demonstrated effective on extracting relevant neuroimaging features and predicting individual cognitive decline. We reported a novel variant (rs144614292) within the NELL1 gene. Our approach has the potential to disentangle accelerated cognitive decline from the normal aging process and to determine its related genetic factors, leveraging opportunities for early intervention.
Collapse
Affiliation(s)
- Yulin Dai
- The University of Texas Health Science Center at Houston
| | - Hsu Yu-Chun
- The University of Texas Health Science Center at Houston
| | | | - Kai Zhang
- The University of Texas Health Science Center at Houston
| | - Li Xiaoyang
- The University of Texas Health Science Center at Houston
| | - Nitesh Enduru
- The University of Texas Health Science Center at Houston
| | - Andi Liu
- The University of Texas Health Science Center at Houston
| | | | - Xiaoqian Jiang
- The University of Texas Health Science Center at Houston
| | - Zhongming Zhao
- The University of Texas Health Science Center at Houston
| |
Collapse
|
17
|
Luckett ES, Zielonka M, Kordjani A, Schaeverbeke J, Adamczuk K, De Meyer S, Van Laere K, Dupont P, Cleynen I, Vandenberghe R. Longitudinal APOE4- and amyloid-dependent changes in the blood transcriptome in cognitively intact older adults. Alzheimers Res Ther 2023; 15:121. [PMID: 37438770 PMCID: PMC10337180 DOI: 10.1186/s13195-023-01242-5] [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: 03/09/2023] [Accepted: 05/06/2023] [Indexed: 07/14/2023]
Abstract
BACKGROUND Gene expression is dysregulated in Alzheimer's disease (AD) patients, both in peripheral blood and post mortem brain. We investigated peripheral whole-blood gene (co)expression to determine molecular changes prior to symptom onset. METHODS RNA was extracted and sequenced for 65 cognitively healthy F-PACK participants (65 (56-80) years, 34 APOE4 non-carriers, 31 APOE4 carriers), at baseline and follow-up (interval: 5.0 (3.4-8.6) years). Participants received amyloid PET at both time points and amyloid rate of change derived. Accumulators were defined with rate of change ≥ 2.19 Centiloids. We performed differential gene expression and weighted gene co-expression network analysis to identify differentially expressed genes and networks of co-expressed genes, respectively, with respect to traits of interest (APOE4 status, amyloid accumulation (binary/continuous)), and amyloid positivity status, followed by Gene Ontology annotation. RESULTS There were 166 significant differentially expressed genes at follow-up compared to baseline in APOE4 carriers only, whereas 12 significant differentially expressed genes were found only in APOE4 non-carriers, over time. Among the significant genes in APOE4 carriers, several had strong evidence for a pathogenic role in AD based on direct association scores generated from the DISQOVER platform: NGRN, IGF2, GMPR, CLDN5, SMIM24. Top enrichment terms showed upregulated mitochondrial and metabolic pathways, and an exacerbated upregulation of ribosomal pathways in APOE4 carriers compared to non-carriers. Similarly, there were 33 unique significant differentially expressed genes at follow-up compared to baseline in individuals classified as amyloid negative at baseline and positive at follow-up or amyloid positive at both time points and 32 unique significant differentially expressed genes over time in individuals amyloid negative at both time points. Among the significant genes in the first group, the top five with the highest direct association scores were as follows: RPL17-C18orf32, HSP90AA1, MBP, SIRPB1, and GRINA. Top enrichment terms included upregulated metabolism and focal adhesion pathways. Baseline and follow-up gene co-expression networks were separately built. Seventeen baseline co-expression modules were derived, with one significantly negatively associated with amyloid accumulator status (r2 = - 0.25, p = 0.046). This was enriched for proteasomal protein catabolic process and myeloid cell development. Thirty-two follow-up modules were derived, with two significantly associated with APOE4 status: one downregulated (r2 = - 0.27, p = 0.035) and one upregulated (r2 = 0.26, p = 0.039) module. Top enrichment processes for the downregulated module included proteasomal protein catabolic process and myeloid cell homeostasis. Top enrichment processes for the upregulated module included cytoplasmic translation and rRNA processing. CONCLUSIONS We show that there are longitudinal gene expression changes that implicate a disrupted immune system, protein removal, and metabolism in cognitively intact individuals who carry APOE4 or who accumulate in cortical amyloid. This provides insight into the pathophysiology of AD, whilst providing novel targets for drug and therapeutic development.
Collapse
Affiliation(s)
- Emma S Luckett
- Laboratory for Cognitive Neurology, Leuven Brain Institute, KU Leuven, Leuven, 3000, Belgium
- Alzheimer Research Centre KU Leuven, Leuven Brain Institute, Leuven, 3000, Belgium
- Laboratory for Complex Genetics, KU Leuven, Leuven, 3000, Belgium
| | - Magdalena Zielonka
- Alzheimer Research Centre KU Leuven, Leuven Brain Institute, Leuven, 3000, Belgium
- Laboratory for the Research of Neurodegenerative Diseases, VIB-KU Leuven, KU Leuven, Leuven, 3000, Belgium
| | - Amine Kordjani
- Laboratory for Complex Genetics, KU Leuven, Leuven, 3000, Belgium
| | - Jolien Schaeverbeke
- Laboratory for Cognitive Neurology, Leuven Brain Institute, KU Leuven, Leuven, 3000, Belgium
- Alzheimer Research Centre KU Leuven, Leuven Brain Institute, Leuven, 3000, Belgium
- Laboratory of Neuropathology, Leuven Brain Institute, KU Leuven, Leuven, 3000, Belgium
| | | | - Steffi De Meyer
- Laboratory for Cognitive Neurology, Leuven Brain Institute, KU Leuven, Leuven, 3000, Belgium
- Alzheimer Research Centre KU Leuven, Leuven Brain Institute, Leuven, 3000, Belgium
- Laboratory of Molecular Neurobiomarker Research, KU Leuven, Leuven, 3000, Belgium
| | - Koen Van Laere
- Division of Nuclear Medicine, UZ Leuven, Leuven, 3000, Belgium
- Nuclear Medicine and Molecular Imaging, Department of Imaging and Pathology, KU Leuven, Leuven, 3000, Belgium
| | - Patrick Dupont
- Laboratory for Cognitive Neurology, Leuven Brain Institute, KU Leuven, Leuven, 3000, Belgium
- Alzheimer Research Centre KU Leuven, Leuven Brain Institute, Leuven, 3000, Belgium
| | - Isabelle Cleynen
- Laboratory for Complex Genetics, KU Leuven, Leuven, 3000, Belgium
| | - Rik Vandenberghe
- Laboratory for Cognitive Neurology, Leuven Brain Institute, KU Leuven, Leuven, 3000, Belgium.
- Alzheimer Research Centre KU Leuven, Leuven Brain Institute, Leuven, 3000, Belgium.
- Neurology Department, University Hospitals Leuven, Herestraat 49, Leuven, 3000, Belgium.
| |
Collapse
|
18
|
Lake J, Warly Solsberg C, Kim JJ, Acosta-Uribe J, Makarious MB, Li Z, Levine K, Heutink P, Alvarado CX, Vitale D, Kang S, Gim J, Lee KH, Pina-Escudero SD, Ferrucci L, Singleton AB, Blauwendraat C, Nalls MA, Yokoyama JS, Leonard HL. Multi-ancestry meta-analysis and fine-mapping in Alzheimer's disease. Mol Psychiatry 2023; 28:3121-3132. [PMID: 37198259 PMCID: PMC10615750 DOI: 10.1038/s41380-023-02089-w] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 03/27/2023] [Accepted: 03/31/2023] [Indexed: 05/19/2023]
Abstract
Genome-wide association studies (GWAS) of Alzheimer's disease are predominantly carried out in European ancestry individuals despite the known variation in genetic architecture and disease prevalence across global populations. We leveraged published GWAS summary statistics from European, East Asian, and African American populations, and an additional GWAS from a Caribbean Hispanic population using previously reported genotype data to perform the largest multi-ancestry GWAS meta-analysis of Alzheimer's disease and related dementias to date. This method allowed us to identify two independent novel disease-associated loci on chromosome 3. We also leveraged diverse haplotype structures to fine-map nine loci with a posterior probability >0.8 and globally assessed the heterogeneity of known risk factors across populations. Additionally, we compared the generalizability of multi-ancestry- and single-ancestry-derived polygenic risk scores in a three-way admixed Colombian population. Our findings highlight the importance of multi-ancestry representation in uncovering and understanding putative factors that contribute to risk of Alzheimer's disease and related dementias.
Collapse
Affiliation(s)
- Julie Lake
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - Caroline Warly Solsberg
- Pharmaceutical Sciences and Pharmacogenomics, University of California, San Francisco, San Francisco, CA, USA
- Department of Neurology and Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
- Memory and Aging Center, University of California, San Francisco, San Francisco, CA, USA
| | - Jonggeol Jeffrey Kim
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
- Preventive Neurology Unit, Centre for Prevention Diagnosis and Detection, Wolfson Institute of Population Health, Queen Mary University of London, London, UK
| | - Juliana Acosta-Uribe
- Neuroscience Research Institute and the department of Molecular, Cellular and Developmental Biology, University of California, Santa Barbara, Santa Barbara, CA, USA
- Neuroscience Group of Antioquia, University of Antioquia, Medellín, Colombia
| | - Mary B Makarious
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, UK
- UCL Movement Disorders Centre, University College London, London, UK
| | - Zizheng Li
- Pharmaceutical Sciences and Pharmacogenomics, University of California, San Francisco, San Francisco, CA, USA
- Department of Neurology and Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Kristin Levine
- Data Tecnica International LLC, Washington, DC, USA
- Center for Alzheimer's and Related Dementias, National Institutes of Health, Bethesda, MD, USA
| | - Peter Heutink
- Alector, Inc. 131 Oyster Point Blvd, Suite 600, South San Francisco, CA, 94080, USA
| | - Chelsea X Alvarado
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
- Data Tecnica International LLC, Washington, DC, USA
- Center for Alzheimer's and Related Dementias, National Institutes of Health, Bethesda, MD, USA
| | - Dan Vitale
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
- Data Tecnica International LLC, Washington, DC, USA
- Center for Alzheimer's and Related Dementias, National Institutes of Health, Bethesda, MD, USA
| | - Sarang Kang
- Gwangju Alzheimer's disease and Related Dementia Cohort Research Center, Chosun University, Gwangju, 61452, Korea
- BK FOUR Department of Integrative Biological Sciences, Chosun University, Gwangju, 61452, Korea
| | - Jungsoo Gim
- Gwangju Alzheimer's disease and Related Dementia Cohort Research Center, Chosun University, Gwangju, 61452, Korea
- BK FOUR Department of Integrative Biological Sciences, Chosun University, Gwangju, 61452, Korea
- Department of Biomedical Science, Chosun University, Gwangju, 61452, Korea
| | - Kun Ho Lee
- Gwangju Alzheimer's disease and Related Dementia Cohort Research Center, Chosun University, Gwangju, 61452, Korea
- BK FOUR Department of Integrative Biological Sciences, Chosun University, Gwangju, 61452, Korea
- Department of Biomedical Science, Chosun University, Gwangju, 61452, Korea
- Korea Brain Research Institute, Daegu, 41062, Korea
| | - Stefanie D Pina-Escudero
- Pharmaceutical Sciences and Pharmacogenomics, University of California, San Francisco, San Francisco, CA, USA
- Department of Neurology and Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
- Memory and Aging Center, University of California, San Francisco, San Francisco, CA, USA
| | - Luigi Ferrucci
- Longitudinal Studies Section, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Andrew B Singleton
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
- Center for Alzheimer's and Related Dementias, National Institutes of Health, Bethesda, MD, USA
| | - Cornelis Blauwendraat
- Center for Alzheimer's and Related Dementias, National Institutes of Health, Bethesda, MD, USA
- Integrative Neurogenomics Unit, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - Mike A Nalls
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
- Data Tecnica International LLC, Washington, DC, USA
- Center for Alzheimer's and Related Dementias, National Institutes of Health, Bethesda, MD, USA
| | - Jennifer S Yokoyama
- Pharmaceutical Sciences and Pharmacogenomics, University of California, San Francisco, San Francisco, CA, USA
- Department of Neurology and Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
- Memory and Aging Center, University of California, San Francisco, San Francisco, CA, USA
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, USA
| | - Hampton L Leonard
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA.
- Data Tecnica International LLC, Washington, DC, USA.
- Center for Alzheimer's and Related Dementias, National Institutes of Health, Bethesda, MD, USA.
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany.
| |
Collapse
|
19
|
Zhao K, Wu Y, Zhao D, Zhang H, Lin J, Wang Y. Six mitophagy-related hub genes as peripheral blood biomarkers of Alzheimer's disease and their immune cell infiltration correlation. Front Neurosci 2023; 17:1125281. [PMID: 37274215 PMCID: PMC10232817 DOI: 10.3389/fnins.2023.1125281] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 03/30/2023] [Indexed: 06/06/2023] Open
Abstract
Background Alzheimer's disease (AD), a neurodegenerative disorder with progressive symptoms, seriously endangers human health worldwide. AD diagnosis and treatment are challenging, but molecular biomarkers show diagnostic potential. This study aimed to investigate AD biomarkers in the peripheral blood. Method Utilizing three microarray datasets, we systematically analyzed the differences in expression and predictive value of mitophagy-related hub genes (MRHGs) in the peripheral blood mononuclear cells of patients with AD to identify potential diagnostic biomarkers. Subsequently, a protein-protein interaction network was constructed to identify hub genes, and functional enrichment analyses were performed. Using consistent clustering analysis, AD subtypes with significant differences were determined. Finally, infiltration patterns of immune cells in AD subtypes and the relationship between MRHGs and immune cells were investigated by two algorithms, CIBERSORT and single-sample gene set enrichment analysis (ssGSEA). Results Our study identified 53 AD- and mitophagy-related differentially expressed genes and six MRHGs, which may be potential biomarkers for diagnosing AD. Functional analysis revealed that six MRHGs significantly affected biologically relevant functions and signaling pathways such as IL-4 Signaling Pathway, RUNX3 Regulates Notch Signaling Pathway, IL-1 and Megakaryocytes in Obesity Pathway, and Overview of Leukocyteintrinsic Hippo Pathway. Furthermore, CIBERSORT and ssGSEA algorithms were used for all AD samples to analyze the abundance of infiltrating immune cells in the two disease subtypes. The results showed that these subtypes were significantly related to immune cell types such as activated mast cells, regulatory T cells, M0 macrophages, and neutrophils. Moreover, specific MRHGs were significantly correlated with immune cell levels. Conclusion Our findings suggest that MRHGs may contribute to the development and prognosis of AD. The six identified MRHGs could be used as valuable diagnostic biomarkers for further research on AD. This study may provide new promising diagnostic and therapeutic targets in the peripheral blood of patients with AD.
Collapse
Affiliation(s)
- Kun Zhao
- Department of Neurology, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Yinyan Wu
- Department of Neurology, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Dongliang Zhao
- Department of Neurology, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Hui Zhang
- Fujian Center for Safety Evaluation of New Drug, Fujian Medical University, Fuzhou, Fujian, China
| | - Jianyang Lin
- Department of General Surgery, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Yuanwei Wang
- Department of Neurology, Shuyang Hospital Affiliated to Xuzhou Medical University, Shuyang, Jiangsu, China
| |
Collapse
|
20
|
Yang M, Matan-Lithwick S, Wang Y, De Jager PL, Bennett DA, Felsky D. Multi-omic integration via similarity network fusion to detect molecular subtypes of ageing. Brain Commun 2023; 5:fcad110. [PMID: 37082508 PMCID: PMC10110975 DOI: 10.1093/braincomms/fcad110] [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: 01/23/2023] [Revised: 02/17/2023] [Accepted: 04/03/2023] [Indexed: 04/07/2023] Open
Abstract
Molecular subtyping of brain tissue provides insights into the heterogeneity of common neurodegenerative conditions, such as Alzheimer's disease. However, existing subtyping studies have mostly focused on single data modalities and only those individuals with severe cognitive impairment. To address these gaps, we applied similarity network fusion, a method capable of integrating multiple high-dimensional multi-omic data modalities simultaneously, to an elderly sample spanning the full spectrum of cognitive ageing trajectories. We analyzed human frontal cortex brain samples characterized by five omic modalities: bulk RNA sequencing (18 629 genes), DNA methylation (53 932 CpG sites), histone acetylation (26 384 peaks), proteomics (7737 proteins) and metabolomics (654 metabolites). Similarity network fusion followed by spectral clustering was used for subtype detection, and subtype numbers were determined by Eigen-gap and rotation cost statistics. Normalized mutual information determined the relative contribution of each modality to the fused network. Subtypes were characterized by associations with 13 age-related neuropathologies and cognitive decline. Fusion of all five data modalities (n = 111) yielded two subtypes (n S1 = 53, n S2 = 58), which were nominally associated with diffuse amyloid plaques; however, this effect was not significant after correction for multiple testing. Histone acetylation (normalized mutual information = 0.38), DNA methylation (normalized mutual information = 0.18) and RNA abundance (normalized mutual information = 0.15) contributed most strongly to this network. Secondary analysis integrating only these three modalities in a larger subsample (n = 513) indicated support for both three- and five-subtype solutions, which had significant overlap, but showed varying degrees of internal stability and external validity. One subtype showed marked cognitive decline, which remained significant even after correcting for tests across both three- and five-subtype solutions (p Bonf = 5.9 × 10-3). Comparison to single-modality subtypes demonstrated that the three-modal subtypes were able to uniquely capture cognitive variability. Comprehensive sensitivity analyses explored influences of sample size and cluster number parameters. We identified highly integrative molecular subtypes of ageing derived from multiple high dimensional, multi-omic data modalities simultaneously. Fusing RNA abundance, DNA methylation, and histone acetylation measures generated subtypes that were associated with cognitive decline. This work highlights the potential value and challenges of multi-omic integration in unsupervised subtyping of post-mortem brain.
Collapse
Affiliation(s)
- Mu Yang
- Division of Biostatistics, Dalla Lana School of Public Health, University of Toronto, Toronto, ON M5T 3M7, Canada
- The Krembil Centre for Neuroinformatics, Centre for Addiction and Mental Health, Toronto, ON M5T 1R8, Canada
| | - Stuart Matan-Lithwick
- The Krembil Centre for Neuroinformatics, Centre for Addiction and Mental Health, Toronto, ON M5T 1R8, Canada
| | - Yanling Wang
- Rush Alzheimer’s Disease Center, Rush University, Chicago, IL 60612, USA
| | - Philip L De Jager
- The Center for Translational and Computational Neuroimmunology, Columbia University Medical Center, New York, NY 10033, USA
| | - David A Bennett
- Rush Alzheimer’s Disease Center, Rush University, Chicago, IL 60612, USA
| | - Daniel Felsky
- Division of Biostatistics, Dalla Lana School of Public Health, University of Toronto, Toronto, ON M5T 3M7, Canada
- The Krembil Centre for Neuroinformatics, Centre for Addiction and Mental Health, Toronto, ON M5T 1R8, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON M5T 1R8, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON M5S 1A8, Canada
| |
Collapse
|
21
|
Kodam P, Sai Swaroop R, Pradhan SS, Sivaramakrishnan V, Vadrevu R. Integrated multi-omics analysis of Alzheimer's disease shows molecular signatures associated with disease progression and potential therapeutic targets. Sci Rep 2023; 13:3695. [PMID: 36879094 PMCID: PMC9986671 DOI: 10.1038/s41598-023-30892-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 03/02/2023] [Indexed: 03/08/2023] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease characterized by the formation of amyloid plaques implicated in neuronal death. Genetics, age, and sex are the risk factors attributed to AD. Though omics studies have helped to identify pathways associated with AD, an integrated systems analysis with the available data could help to understand mechanisms, potential biomarkers, and therapeutic targets. Analysis of transcriptomic data sets from the GEO database, and proteomic and metabolomic data sets from literature was performed to identify deregulated pathways and commonality analysis identified overlapping pathways among the data sets. The deregulated pathways included those of neurotransmitter synapses, oxidative stress, inflammation, vitamins, complement, and coagulation pathways. Cell type analysis of GEO data sets showed microglia, endothelial, myeloid, and lymphoid cells are affected. Microglia are associated with inflammation and pruning of synapses with implications for memory and cognition. Analysis of the protein-cofactor network of B2, B6, and pantothenate shows metabolic pathways modulated by these vitamins which overlap with the deregulated pathways from the multi-omics analysis. Overall, the integrated analysis identified the molecular signature associated with AD. Treatment with anti-oxidants, B2, B6, and pantothenate in genetically susceptible individuals in the pre-symptomatic stage might help in better management of the disease.
Collapse
Affiliation(s)
- Pradeep Kodam
- Department of Biological Sciences, Birla Institute of Technology and Science Pilani, Hyderabad Campus, Jawahar Nagar, Hyderabad, Telangana, 500078, India
| | - R Sai Swaroop
- Disease Biology Lab, Department of Biosciences, Sri Sathya Sai Institute of Higher Learning, Prasanthi Nilayam, Anantapur, Andhra Pradesh, 515134, India
| | - Sai Sanwid Pradhan
- Disease Biology Lab, Department of Biosciences, Sri Sathya Sai Institute of Higher Learning, Prasanthi Nilayam, Anantapur, Andhra Pradesh, 515134, India
| | - Venketesh Sivaramakrishnan
- Disease Biology Lab, Department of Biosciences, Sri Sathya Sai Institute of Higher Learning, Prasanthi Nilayam, Anantapur, Andhra Pradesh, 515134, India.
| | - Ramakrishna Vadrevu
- Department of Biological Sciences, Birla Institute of Technology and Science Pilani, Hyderabad Campus, Jawahar Nagar, Hyderabad, Telangana, 500078, India.
| |
Collapse
|
22
|
Picón-Pagès P, Bosch-Morató M, Subirana L, Rubio-Moscardó F, Guivernau B, Fanlo-Ucar H, Zeylan ME, Senyuz S, Herrera-Fernández V, Vicente R, Fernández-Fernández JM, García-Ojalvo J, Gursoy A, Keskin O, Oliva B, Posas F, de Nadal E, Muñoz FJ. A Genome-Wide Functional Screen Identifies Enhancer and Protective Genes for Amyloid Beta-Peptide Toxicity. Int J Mol Sci 2023; 24:ijms24021278. [PMID: 36674792 PMCID: PMC9865122 DOI: 10.3390/ijms24021278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 12/20/2022] [Accepted: 01/04/2023] [Indexed: 01/11/2023] Open
Abstract
Alzheimer's disease (AD) is known to be caused by amyloid β-peptide (Aβ) misfolded into β-sheets, but this knowledge has not yet led to treatments to prevent AD. To identify novel molecular players in Aβ toxicity, we carried out a genome-wide screen in Saccharomyces cerevisiae, using a library of 5154 gene knock-out strains expressing Aβ1-42. We identified 81 mammalian orthologue genes that enhance Aβ1-42 toxicity, while 157 were protective. Next, we performed interactome and text-mining studies to increase the number of genes and to identify the main cellular functions affected by Aβ oligomers (oAβ). We found that the most affected cellular functions were calcium regulation, protein translation and mitochondrial activity. We focused on SURF4, a protein that regulates the store-operated calcium channel (SOCE). An in vitro analysis using human neuroblastoma cells showed that SURF4 silencing induced higher intracellular calcium levels, while its overexpression decreased calcium entry. Furthermore, SURF4 silencing produced a significant reduction in cell death when cells were challenged with oAβ1-42, whereas SURF4 overexpression induced Aβ1-42 cytotoxicity. In summary, we identified new enhancer and protective activities for Aβ toxicity and showed that SURF4 contributes to oAβ1-42 neurotoxicity by decreasing SOCE activity.
Collapse
Affiliation(s)
- Pol Picón-Pagès
- Laboratory of Molecular Physiology, Department of Medicine and Life Sciences, Faculty of Health and Life Sciences, Universitat Pompeu Fabra, 08003 Barcelona, Spain
| | - Mònica Bosch-Morató
- Laboratory of Molecular Physiology, Department of Medicine and Life Sciences, Faculty of Health and Life Sciences, Universitat Pompeu Fabra, 08003 Barcelona, Spain
| | - Laia Subirana
- Department of Medicine and Life Sciences, Faculty of Health and Life Sciences, Universitat Pompeu Fabra, 08003 Barcelona, Spain
| | - Francisca Rubio-Moscardó
- Laboratory of Molecular Physiology, Department of Medicine and Life Sciences, Faculty of Health and Life Sciences, Universitat Pompeu Fabra, 08003 Barcelona, Spain
| | - Biuse Guivernau
- Laboratory of Molecular Physiology, Department of Medicine and Life Sciences, Faculty of Health and Life Sciences, Universitat Pompeu Fabra, 08003 Barcelona, Spain
| | - Hugo Fanlo-Ucar
- Laboratory of Molecular Physiology, Department of Medicine and Life Sciences, Faculty of Health and Life Sciences, Universitat Pompeu Fabra, 08003 Barcelona, Spain
| | - Melisa Ece Zeylan
- Computational Sciences and Engineering, Koc University, Istanbul 34450, Turkey
| | - Simge Senyuz
- Computational Sciences and Engineering, Koc University, Istanbul 34450, Turkey
| | - Víctor Herrera-Fernández
- Laboratory of Molecular Physiology, Department of Medicine and Life Sciences, Faculty of Health and Life Sciences, Universitat Pompeu Fabra, 08003 Barcelona, Spain
| | - Rubén Vicente
- Laboratory of Molecular Physiology, Department of Medicine and Life Sciences, Faculty of Health and Life Sciences, Universitat Pompeu Fabra, 08003 Barcelona, Spain
| | - José M. Fernández-Fernández
- Laboratory of Molecular Physiology, Department of Medicine and Life Sciences, Faculty of Health and Life Sciences, Universitat Pompeu Fabra, 08003 Barcelona, Spain
| | - Jordi García-Ojalvo
- Laboratory of Dynamical Systems Biology, Department of Medicine and Life Sciences, Faculty of Health and Life Sciences, Universitat Pompeu Fabra, 08003 Barcelona, Spain
| | - Attila Gursoy
- College of Engineering, Koc University, Istanbul 34450, Turkey
| | - Ozlem Keskin
- College of Engineering, Koc University, Istanbul 34450, Turkey
| | - Baldomero Oliva
- Laboratory of Structural Bioinformatics (GRIB), Department of Medicine and Life Sciences, Faculty of Health and Life Sciences, Universitat Pompeu Fabra, 08003 Barcelona, Spain
| | - Francesc Posas
- Department of Medicine and Life Sciences, Faculty of Health and Life Sciences, Universitat Pompeu Fabra, 08003 Barcelona, Spain
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, 08028 Barcelona, Spain
| | - Eulàlia de Nadal
- Department of Medicine and Life Sciences, Faculty of Health and Life Sciences, Universitat Pompeu Fabra, 08003 Barcelona, Spain
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, 08028 Barcelona, Spain
| | - Francisco J. Muñoz
- Laboratory of Molecular Physiology, Department of Medicine and Life Sciences, Faculty of Health and Life Sciences, Universitat Pompeu Fabra, 08003 Barcelona, Spain
- Correspondence:
| |
Collapse
|
23
|
Zhang Y, Xue Y, Wang L, Han Z, Wang T, Zhang H, Liu G, Xiao X. rs56405341 Variant Associates with Expression of C4orf33 and C4orf33 Was Downregulated in Alzheimer's Disease and Progressive Supranuclear Palsy. J Alzheimers Dis 2023; 96:57-64. [PMID: 37742642 DOI: 10.3233/jad-230327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/26/2023]
Abstract
The first primary age-related tauopathy (PART) genome-wide association study confirmed significant associations of Alzheimer's disease (AD) and progressive supranuclear palsy (PSP) genetic variants with PART, and highlighted a novel genetic variant rs56405341. Here, we perform a comprehensive analysis of rs56405341. We found that rs56405341 was significantly associated with C4orf33 mRNA expression, but not JADE1 mRNA expression in multiple brain tissues. C4orf33 was mainly expressed in cerebellar hemisphere and cerebellum, and JADE1 was mainly expressed in thyroid, and coronary artery. Meanwhile, we found significantly downregulated C4orf33 expression both AD and PSP compared with normal controls, respectively.
Collapse
Affiliation(s)
- Yan Zhang
- Department of Pathology, The Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Yanli Xue
- School of Biomedical Engineering, Capital Medical University, Beijing, China
| | - Longcai Wang
- Department of Anesthesiology, The Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Zhifa Han
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Beijing, China
| | - Tao Wang
- Chinese Institute for Brain Research, Beijing, China
| | - Haihua Zhang
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, China
| | - Guiyou Liu
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, China
- Chinese Institute for Brain Research, Beijing, China
- Key Laboratory of Cerebral Microcirculation in Universities of Shandong; Department of Neurology, Second Affiliated Hospital; Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, Shandong, China
- Beijing Key Laboratory of Hypoxia Translational Medicine, National Engineering Laboratory of Internet Medical Diagnosis and Treatment Technology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Xingjun Xiao
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
24
|
Rodríguez-Giraldo M, González-Reyes RE, Ramírez-Guerrero S, Bonilla-Trilleras CE, Guardo-Maya S, Nava-Mesa MO. Astrocytes as a Therapeutic Target in Alzheimer's Disease-Comprehensive Review and Recent Developments. Int J Mol Sci 2022; 23:13630. [PMID: 36362415 PMCID: PMC9654484 DOI: 10.3390/ijms232113630] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 10/25/2022] [Accepted: 10/26/2022] [Indexed: 09/20/2023] Open
Abstract
Alzheimer's disease (AD) is a frequent and disabling neurodegenerative disorder, in which astrocytes participate in several pathophysiological processes including neuroinflammation, excitotoxicity, oxidative stress and lipid metabolism (along with a critical role in apolipoprotein E function). Current evidence shows that astrocytes have both neuroprotective and neurotoxic effects depending on the disease stage and microenvironmental factors. Furthermore, astrocytes appear to be affected by the presence of amyloid-beta (Aβ), with alterations in calcium levels, gliotransmission and proinflammatory activity via RAGE-NF-κB pathway. In addition, astrocytes play an important role in the metabolism of tau and clearance of Aβ through the glymphatic system. In this review, we will discuss novel pharmacological and non-pharmacological treatments focused on astrocytes as therapeutic targets for AD. These interventions include effects on anti-inflammatory/antioxidant systems, glutamate activity, lipid metabolism, neurovascular coupling and glymphatic system, calcium dysregulation, and in the release of peptides which affects glial and neuronal function. According to the AD stage, these therapies may be of benefit in either preventing or delaying the progression of the disease.
Collapse
Affiliation(s)
| | | | | | | | | | - Mauricio O. Nava-Mesa
- Grupo de Investigación en Neurociencias (NeURos), Centro de Neurociencias Neurovitae-UR, Instituto de Medicina Traslacional (IMT), Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Bogotá 111711, Colombia
| |
Collapse
|
25
|
Kellogg MK, Tikhonova EB, Karamyshev AL. Signal Recognition Particle in Human Diseases. Front Genet 2022; 13:898083. [PMID: 35754847 PMCID: PMC9214365 DOI: 10.3389/fgene.2022.898083] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 05/05/2022] [Indexed: 01/14/2023] Open
Abstract
The signal recognition particle (SRP) is a ribonucleoprotein complex with dual functions. It co-translationally targets proteins with a signal sequence to the endoplasmic reticulum (ER) and protects their mRNA from degradation. If SRP is depleted or cannot recognize the signal sequence, then the Regulation of Aberrant Protein Production (RAPP) is activated, which results in the loss of secretory protein mRNA. If SRP recognizes the substrates but is unable to target them to ER, they may mislocalize or degrade. All these events lead to dramatic consequence for protein biogenesis, activating protein quality control pathways, and creating pressure on cell physiology, and might lead to the pathogenesis of disease. Indeed, SRP dysfunction is involved in many different human diseases, including: congenital neutropenia; idiopathic inflammatory myopathy; viral, protozoal, and prion infections; and cancer. In this work, we analyze diseases caused by SRP failure and discuss their possible molecular mechanisms.
Collapse
Affiliation(s)
- Morgana K Kellogg
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - Elena B Tikhonova
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - Andrey L Karamyshev
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| |
Collapse
|
26
|
He B, Gorijala P, Xie L, Cao S, Yan J. Gene co-expression changes underlying the functional connectomic alterations in Alzheimer's disease. BMC Med Genomics 2022; 15:92. [PMID: 35461274 PMCID: PMC9035246 DOI: 10.1186/s12920-022-01244-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 04/19/2022] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND There is growing evidence indicating that a number of functional connectivity networks are disrupted at each stage of the full clinical Alzheimer's disease spectrum. Such differences are also detectable in cognitive normal (CN) carrying mutations of AD risk genes, suggesting a substantial relationship between genetics and AD-altered functional brain networks. However, direct genetic effect on functional connectivity networks has not been measured. METHODS Leveraging existing AD functional connectivity studies collected in NeuroSynth, we performed a meta-analysis to identify two sets of brain regions: ones with altered functional connectivity in resting state network and ones without. Then with the brain-wide gene expression data in the Allen Human Brain Atlas, we applied a new biclustering method to identify a set of genes with differential co-expression patterns between these two set of brain regions. RESULTS Differential co-expression analysis using biclustering method led to a subset of 38 genes which showed distinctive co-expression patterns between AD-related and non AD-related brain regions in default mode network. More specifically, we observed 4 sub-clusters with noticeable co-expression difference, where the difference in correlations is above 0.5 on average. CONCLUSIONS This work applies a new biclustering method to search for a subset of genes with altered co-expression patterns in AD-related default mode network regions. Compared with traditional differential expression analysis, differential co-expression analysis yielded many more significant hits with extra insights into the wiring mechanism between genes. Particularly, the differential co-expression pattern was observed between two sets of genes, suggesting potential upstream genetic regulators in AD development.
Collapse
Affiliation(s)
- Bing He
- Department of BioHealth Informatics, Indiana University Purdue University Indianapolis, Indianapolis, IN, USA
| | - Priyanka Gorijala
- Department of BioHealth Informatics, Indiana University Purdue University Indianapolis, Indianapolis, IN, USA
| | - Linhui Xie
- Department of Electrical and Computer Engineering, Indiana University Purdue University Indianapolis, Indianapolis, IN, USA
| | - Sha Cao
- Department of Biostatistics and Health Data Sciences, School of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Jingwen Yan
- Department of BioHealth Informatics, Indiana University Purdue University Indianapolis, Indianapolis, IN, USA
| |
Collapse
|
27
|
Deprez M, Moreira J, Sermesant M, Lorenzi M. Decoding Genetic Markers of Multiple Phenotypic Layers Through Biologically Constrained Genome-To-Phenome Bayesian Sparse Regression. FRONTIERS IN MOLECULAR MEDICINE 2022; 2:830956. [PMID: 39086978 PMCID: PMC11285669 DOI: 10.3389/fmmed.2022.830956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 02/18/2022] [Indexed: 08/02/2024]
Abstract
The applicability of multivariate approaches for the joint analysis of genomics and phenomics information is currently limited by the lack of scalability, and by the difficulty of interpreting the related findings from a biological perspective. To tackle these limitations, we present Bayesian Genome-to-Phenome Sparse Regression (G2PSR), a novel multivariate regression method based on sparse SNP-gene constraints. The statistical framework of G2PSR is based on a Bayesian neural network, were constraints on SNPs-genes associations are integrated by incorporating a priori knowledge linking variants to their respective genes, to then reconstruct the phenotypic data in the output layer. Interpretability is promoted by inducing sparsity on the genes through variational dropout, allowing to estimate the uncertainty associated with each gene, and related SNPs, in the reconstruction task. Ultimately, G2PSR is conceived to prevent multiple testing correction and to assess the combined effect of SNPs, thus increasing the statistical power in detecting genome-to-phenome associations. The effectiveness of G2PSR was demonstrated on synthetic and real data, with respect to state-of-the-art methods based on group-wise sparsity constraints. The application on real data consisted in an imaging-genetics analysis on the Alzheimer's Disease Neuroimaging Initiative data, relating SNPs from more than 3,500 genes to clinical and multi-variate brain volumetric information. The experimental results show that our method can provide accurate selection of relevant genes in dataset with large SNPs-to-samples ratio, thus overcoming the main limitations of current genome-to-phenome association methods.
Collapse
Affiliation(s)
- Marie Deprez
- University of Côte d’Azur, Nice, France
- INRIA, Epione Project-Team, Valbonne, France
| | - Julien Moreira
- University of Côte d’Azur, Nice, France
- INRIA, Epione Project-Team, Valbonne, France
| | - Maxime Sermesant
- University of Côte d’Azur, Nice, France
- INRIA, Epione Project-Team, Valbonne, France
| | - Marco Lorenzi
- University of Côte d’Azur, Nice, France
- INRIA, Epione Project-Team, Valbonne, France
| |
Collapse
|
28
|
Liu X, Lin L, Lu L, Li X, Han Y, Qiu Z, Li X, Li Y, Song X, Cao W, Li T. Comparative Transcriptional Analysis Identified Characteristic Genes and Patterns in HIV-Infected Immunological Non-Responders. Front Immunol 2022; 13:807890. [PMID: 35154126 PMCID: PMC8832504 DOI: 10.3389/fimmu.2022.807890] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 01/04/2022] [Indexed: 12/12/2022] Open
Abstract
Purpose The incomplete immune reconstitution is a complex phenomenon among human immunodeficiency virus (HIV)-infected patients despite the fact that they have achieved persistent viral suppression under the combined antiretroviral therapy. This study aims to screen and verify the immunological characteristics and underlying mechanisms of immunological non-responders (INRs). Methods The RNA-seq and the differentially expressed genes (DEGs) analysis were used to explore potential characteristics among INRs. Gene Ontology (GO) enrichment, ingenuity pathway analysis (IPA) analysis, Gene set enrichment analysis (GSEA) analysis, and the weighted gene co-expression network analysis (WGCNA) were used to explore the potential mechanism. The transcriptional meta-analysis was used to analyze the external efficiency. Results The RNA-seq identified 316 DEGs among INRs. The interferon signaling pathway was enriched via GO and IPA analysis among DEGs. The combined GSEA and WGCNA analysis confirmed that the IFN response was more correlated with INR. Furthermore, IFI27 (IFN-α Inducible Protein 27, also known as ISG12) was chosen based on combined DEG analysis, WGCNA analysis, and the transcriptional meta-analysis conducted on other published datasets about INRs. The expression of IFI27 was significantly negatively correlated with the CD4+ T-cell counts of PLWH, and the predictive efficiency of IFI27 level in distinguishing PLWH with poor immune recovery was also with significant power (AUC = 0.848). Conclusion The enhanced expression of IFI27 and the IFN response pathway are among the important immunological characteristics of INRs and exhibited promising efficiency as biomarkers for CD4+ T-cell recovery.
Collapse
Affiliation(s)
- Xiaosheng Liu
- Tsinghua-Peking Center for Life Sciences, Beijing, China.,Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, China.,Department of Infectious Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Ling Lin
- Department of Infectious Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Lianfeng Lu
- Department of Infectious Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Xiaodi Li
- Department of Infectious Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Yang Han
- Department of Infectious Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Zhifeng Qiu
- Department of Infectious Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Xiaoxia Li
- Department of Infectious Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Yanling Li
- Department of Infectious Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Xiaojing Song
- Department of Infectious Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Wei Cao
- Department of Infectious Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Taisheng Li
- Tsinghua-Peking Center for Life Sciences, Beijing, China.,Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, China.,Department of Infectious Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
29
|
Vigneswaran J, Muthukumar SA, Shafras M, Pant G. An insight into Alzheimer’s disease and its on-setting novel genes. THE EGYPTIAN JOURNAL OF NEUROLOGY, PSYCHIATRY AND NEUROSURGERY 2021. [DOI: 10.1186/s41983-021-00420-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
AbstractAccording to the World Health Organisation, as of 2019, globally around 50 million people suffer from dementia, with approximately another 10 million getting added to the list every year, wherein Alzheimer’s disease (AD) stands responsible for almost a whopping 60–70% for the existing number of cases. Alzheimer’s disease is one of the progressive, cognitive-declining, age-dependent, neurodegenerative diseases which is distinguished by histopathological symptoms, such as formation of amyloid plaque, senile plaque, neurofibrillary tangles, etc. Majorly four vital transcripts are identified in the AD complications which include Amyloid precursor protein (APP), Apolipoprotein E (ApoE), and two multi-pass transmembrane domain proteins—Presenilin 1 and 2. In addition, the formation of the abnormal filaments such as amyloid beta (Aβ) and tau and their tangling with some necessary factors contributing to the formation of plaques, neuroinflammation, and apoptosis which in turn leads to the emergence of AD. Although multiple molecular mechanisms have been elucidated so far, they are still counted as hypotheses ending with neuronal death on the basal forebrain and hippocampal area which results in AD. This review article is aimed at addressing the overview of the molecular mechanisms surrounding AD and the functional forms of the genes associated with it.
Collapse
|
30
|
Li QS, Cai D. Integrated miRNA-Seq and mRNA-Seq Study to Identify miRNAs Associated With Alzheimer's Disease Using Post-mortem Brain Tissue Samples. Front Neurosci 2021; 15:620899. [PMID: 33833661 PMCID: PMC8021900 DOI: 10.3389/fnins.2021.620899] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Accepted: 02/23/2021] [Indexed: 01/06/2023] Open
Abstract
Alzheimer's disease (AD), the leading form of dementia, is associated with abnormal tau and β-amyloid accumulation in the brain. We conducted a miRNA-seq study to identify miRNAs associated with AD in the post-mortem brain from the inferior frontal gyrus (IFG, n = 69) and superior temporal gyrus (STG, n = 81). Four and 64 miRNAs were differentially expressed (adjusted p-value < 0.05) in AD compared to cognitively normal controls in the IFG and STG, respectively. We observed down-regulation of several miRNAs that have previously been implicated in AD, including hsa-miR-212-5p and hsa-miR-132-5p, in AD samples across both brain regions, and up-regulation of hsa-miR-146a-5p, hsa-miR-501-3p, hsa-miR-34a-5p, and hsa-miR-454-3p in the STG. The differentially expressed miRNAs were previously implicated in the formation of amyloid-β plaques, the dysregulation of tau, and inflammation. We have also observed differential expressions for dozens of other miRNAs in the STG, including hsa-miR-4446-3p, that have not been described previously. Putative targets of these miRNAs (adjusted p-value < 0.1) were found to be involved in Wnt signaling pathway, MAPK family signaling cascades, sphingosine 1-phosphate (S1P) pathway, adaptive immune system, innate immune system, and neurogenesis. Our results support the finding of dysregulated miRNAs previously implicated in AD and propose additional miRNAs that appear to be dysregulated in AD for experimental follow-up.
Collapse
Affiliation(s)
- Qingqin S. Li
- Neuroscience, Janssen Research & Development, LLC, Titusville, NJ, United States
| | | |
Collapse
|