1
|
Núñez-Ríos DL, Nagamatsu ST, Martínez-Magaña JJ, Hurd Y, Rompala G, Krystal JH, Montalvo-Ortiz JL. Mapping the epigenomic landscape of post-traumatic stress disorder in human cortical neurons. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.10.11.24315258. [PMID: 39484232 PMCID: PMC11527063 DOI: 10.1101/2024.10.11.24315258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
The study conducted a comprehensive genome-wide analysis of differential 5mC and 5hmC modifications at both CpG and non-CpG sites in postmortem orbitofrontal neurons from 25 PTSD cases and 13 healthy controls. It was observed that PTSD patients exhibit a greater number of differential 5hmC sites compared to 5mC sites. Specifically, individuals with PTSD tend to show hyper-5mC/5hmC at CpG sites, particularly within CpG islands and promoter regions, and hypo-5mC/5hmC at non-CpG sites, especially within intragenic regions. Functional enrichment analysis indicated distinct yet interconnected roles for 5mC and 5hmC in PTSD. The 5mC marks primarily regulate cell-cell adhesion processes, whereas 5hmC marks are involved in embryonic morphogenesis and cell fate commitment. By integrating published PTSD findings from central and peripheral tissues through multi-omics approaches, several biological mechanisms were prioritized, including developmental processes, HPA axis regulation, and immune responses. Based on the consistent enrichment in developmental processes, we hypothesize that if epigenetic changes occur during early developmental stages, they may increase the risk of developing PTSD following trauma exposure. Conversely, if these epigenetic changes occur in adulthood, they may influence neuronal apoptosis and survival mechanisms.
Collapse
Affiliation(s)
- Diana L. Núñez-Ríos
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- National Center of Post-Traumatic Stress Disorder, VA CT Healthcare System, West Haven, CT, USA
| | - Sheila T. Nagamatsu
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- National Center of Post-Traumatic Stress Disorder, VA CT Healthcare System, West Haven, CT, USA
| | - Jose Jaime Martínez-Magaña
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- National Center of Post-Traumatic Stress Disorder, VA CT Healthcare System, West Haven, CT, USA
| | - Yasmin Hurd
- Icahn School of Medicine at Mount Sinai, New York City, NY, USA
| | - Gregory Rompala
- Icahn School of Medicine at Mount Sinai, New York City, NY, USA
| | - John H. Krystal
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- National Center of Post-Traumatic Stress Disorder, VA CT Healthcare System, West Haven, CT, USA
| | | | - Janitza L. Montalvo-Ortiz
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- National Center of Post-Traumatic Stress Disorder, VA CT Healthcare System, West Haven, CT, USA
| |
Collapse
|
2
|
Milaneschi Y, Montanari S, Jansen R, Schranner D, Kastenmüller G, Arnold M, Janiri D, Sani G, Bhattacharyya S, Dehkordi SM, Dunlop B, Rush A, Penninx B, Kaddurah-Daouk R. Acylcarnitines metabolism in depression: association with diagnostic status, depression severity and symptom profile in the NESDA cohort. RESEARCH SQUARE 2024:rs.3.rs-4638158. [PMID: 39149483 PMCID: PMC11326352 DOI: 10.21203/rs.3.rs-4638158/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
Acylcarnitines (ACs) are involved in bioenergetics processes that may play a role in the pathophysiology of depression. Previous genomic evidence identified four ACs potentially linked to depression risk. We carried forward these ACs and tested the association of their circulating levels with Major Depressive Disorder (MDD) diagnosis, overall depression severity and specific symptom profiles. The sample from the Netherlands Study of Depression and Anxiety included participants with current (n = 1035) or remitted (n = 739) MDD and healthy controls (n = 800). Plasma levels of four ACs (short-chain: acetylcarnitine C2 and propionylcarnitine C3; medium-chain: octanoylcarnitine C8 and decanoylcarnitine C10) were measured. Overall depression severity as well as atypical/energy-related (AES), anhedonic and melancholic symptom profiles were derived from the Inventory of Depressive Symptomatology. As compared to healthy controls, subjects with current or remitted MDD presented similarly lower mean C2 levels (Cohen's d = 0.2, p ≤ 1e-4). Higher overall depression severity was significantly associated with higher C3 levels (ß=0.06, SE = 0.02, p = 1.21e-3). No associations were found for C8 and C10. Focusing on symptom profiles, only higher AES scores were linked to lower C2 (ß=-0.05, SE = 0.02, p = 1.85e-2) and higher C3 (ß=0.08, SE = 0.02, p = 3.41e-5) levels. Results were confirmed in analyses pooling data with an additional internal replication sample from the same subjects measured at 6-year follow-up (totaling 4141 observations). Small alterations in levels of short-chain acylcarnitine levels were related to the presence and severity of depression, especially for symptoms reflecting altered energy homeostasis. Cellular metabolic dysfunctions may represent a key pathway in depression pathophysiology potentially accessible through AC metabolism.
Collapse
Affiliation(s)
| | | | - Rick Jansen
- Amsterdam UMC location Vrije Universiteit Amsterdam
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
3
|
Tusconi M, Kätzel D, Sánchez-Gutiérrez T. Editorial: Reviews in psychiatry 2023: schizophrenia. Front Psychiatry 2024; 15:1444818. [PMID: 39026521 PMCID: PMC11255906 DOI: 10.3389/fpsyt.2024.1444818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 06/18/2024] [Indexed: 07/20/2024] Open
Affiliation(s)
| | - Dennis Kätzel
- Institute of Applied Physiology, Faculty of Medicine, University of Ulm, Ulm, Germany
| | | |
Collapse
|
4
|
Abbasi AF, Asim MN, Ahmed S, Vollmer S, Dengel A. Survival prediction landscape: an in-depth systematic literature review on activities, methods, tools, diseases, and databases. Front Artif Intell 2024; 7:1428501. [PMID: 39021434 PMCID: PMC11252047 DOI: 10.3389/frai.2024.1428501] [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: 05/07/2024] [Accepted: 06/12/2024] [Indexed: 07/20/2024] Open
Abstract
Survival prediction integrates patient-specific molecular information and clinical signatures to forecast the anticipated time of an event, such as recurrence, death, or disease progression. Survival prediction proves valuable in guiding treatment decisions, optimizing resource allocation, and interventions of precision medicine. The wide range of diseases, the existence of various variants within the same disease, and the reliance on available data necessitate disease-specific computational survival predictors. The widespread adoption of artificial intelligence (AI) methods in crafting survival predictors has undoubtedly revolutionized this field. However, the ever-increasing demand for more sophisticated and effective prediction models necessitates the continued creation of innovative advancements. To catalyze these advancements, it is crucial to bring existing survival predictors knowledge and insights into a centralized platform. The paper in hand thoroughly examines 23 existing review studies and provides a concise overview of their scope and limitations. Focusing on a comprehensive set of 90 most recent survival predictors across 44 diverse diseases, it delves into insights of diverse types of methods that are used in the development of disease-specific predictors. This exhaustive analysis encompasses the utilized data modalities along with a detailed analysis of subsets of clinical features, feature engineering methods, and the specific statistical, machine or deep learning approaches that have been employed. It also provides insights about survival prediction data sources, open-source predictors, and survival prediction frameworks.
Collapse
Affiliation(s)
- Ahtisham Fazeel Abbasi
- Department of Computer Science, Rhineland-Palatinate Technical University of Kaiserslautern-Landau, Kaiserslautern, Germany
- Smart Data & Knowledge Services, Deutsches Forschungszentrum für Künstliche Intelligenz (DFKI), Kaiserslautern, Germany
| | - Muhammad Nabeel Asim
- Smart Data & Knowledge Services, Deutsches Forschungszentrum für Künstliche Intelligenz (DFKI), Kaiserslautern, Germany
| | - Sheraz Ahmed
- Smart Data & Knowledge Services, Deutsches Forschungszentrum für Künstliche Intelligenz (DFKI), Kaiserslautern, Germany
| | - Sebastian Vollmer
- Department of Computer Science, Rhineland-Palatinate Technical University of Kaiserslautern-Landau, Kaiserslautern, Germany
- Smart Data & Knowledge Services, Deutsches Forschungszentrum für Künstliche Intelligenz (DFKI), Kaiserslautern, Germany
| | - Andreas Dengel
- Department of Computer Science, Rhineland-Palatinate Technical University of Kaiserslautern-Landau, Kaiserslautern, Germany
- Smart Data & Knowledge Services, Deutsches Forschungszentrum für Künstliche Intelligenz (DFKI), Kaiserslautern, Germany
| |
Collapse
|
5
|
Chang L, Wang T, Qu Y, Fan X, Zhou X, Wei Y, Hashimoto K. Identification of novel endoplasmic reticulum-related genes and their association with immune cell infiltration in major depressive disorder. J Affect Disord 2024; 356:190-203. [PMID: 38604455 DOI: 10.1016/j.jad.2024.04.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 03/25/2024] [Accepted: 04/08/2024] [Indexed: 04/13/2024]
Abstract
BACKGROUND Several lines of evidence point to an interaction between genetic predisposition and environmental factors in the onset of major depressive disorder (MDD). This study is aimed to investigate the pathogenesis of MDD by identifying key biomarkers, associated immune infiltration using bioinformatic analysis and human postmortem sample. METHODS The Gene Expression Omnibus (GEO) database of GSE98793 was adopted to identify hub genes linked to endoplasmic reticulum (ER) stress-related genes (ERGs) in MDD. Another GEO database of GSE76826 was employed to validate the novel target associated with ERGs and immune infiltration in MDD. Moreover, human postmortem sample from MDD patients was utilized to confirm the differential expression analysis of hub genes. RESULTS We discovered 12 ER stress-related differentially expressed genes (ERDEGs). A LASSO Cox regression analysis helped construct a diagnostic model for these ERDEGs, incorporating immune infiltration analysis revealed that three hub genes (ERLIN1, SEC61B, and USP13) show the significant and consistent expression differences between the two groups. Western blot analysis of postmortem brain samples indicated notably higher expression levels of ERLIN1 and SEC61B in the MDD group, with USP13 also tending to increase compared to control group. LIMITATIONS The utilization of the MDD gene chip in this analysis was sourced from the GEO database, which possesses a restricted number of pertinent gene chip samples. CONCLUSIONS These findings indicate that ERDEGs especially including ERLIN1, SEC61B, and USP13 associated the infiltration of immune cells may be potential diagnostic indicators for MDD.
Collapse
Affiliation(s)
- Lijia Chang
- Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba 260-8670, Japan
| | - Tong Wang
- Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Southwest Medical University, Luzhou 646000, Sichuan, China
| | - Youge Qu
- Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba 260-8670, Japan
| | - Xinrong Fan
- Department of Cardiology, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Xiangyu Zhou
- Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Ministry of Education, Southwest Medical University, Luzhou 646000, China; Department of Thyroid and Vascular Surgery, The Affiliated Hospital, Southwest Medical University, Luzhou 646000, China
| | - Yan Wei
- Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Southwest Medical University, Luzhou 646000, Sichuan, China.
| | - Kenji Hashimoto
- Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba 260-8670, Japan.
| |
Collapse
|
6
|
Welton T, Chew G, Mai AS, Ng JH, Chan LL, Tan EK. Association of Gene Expression and Tremor Network Structure. Mov Disord 2024; 39:1119-1130. [PMID: 38769620 DOI: 10.1002/mds.29831] [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: 09/17/2023] [Revised: 04/18/2024] [Accepted: 04/24/2024] [Indexed: 05/22/2024] Open
Abstract
BACKGROUND Transcriptomic changes in the essential tremor (ET)-associated cerebello-thalamo-cortical "tremor network" and their association to brain structure have not been investigated. OBJECTIVE The aim was to characterize molecular changes associated with network-level imaging-derived phenotypes (IDP) found in ET. METHODS We performed an imaging-transcriptomic study in British adults using imaging-genome-wide association study summary statistics (UK Biobank "BIG40" cohort; n = 33,224, aged 40-69 years). We imputed imaging-transcriptomic associations for 184 IDPs and analyzed functional enrichment of gene modules and aggregate network-level phenotypes. Validation was performed in cerebellar-tissue RNA-sequencing data from ET patients and controls (n = 55). RESULTS Among 237,896 individual predicted gene expression levels for 6063 unique genes/transcripts, we detected 2269 genome-wide significant associations (Bonferroni P < 2.102e-7, 0.95%). These were concentrated in intracellular volume fraction measures of white matter pathways and in genes with putative links to tremor (MAPT, ARL17A, KANSL1, SPPL2C, LRRC37A4P, PLEKHM1, and FMNL1). Whole-tremor-network cortical thickness was associated with a gene module linked to mitochondrial organization and protein quality control (r = 0.91, P = 2e-70), whereas white-gray T1-weighted magnetic resonance imaging (MRI) contrast in the tremor network was associated with a gene module linked to sphingolipid synthesis and ethanolamine metabolism (r = -0.90, P = 2e-68). Imputed association effect sizes and RNA-sequencing log-fold change in the validation dataset were significantly correlated for cerebellar peduncular diffusion MRI phenotypes, and there was a close overlap of significant associations between both datasets for gray matter phenotypes (χ2 = 6.40, P = 0.006). CONCLUSIONS The identified genes and processes are potential treatment targets for ET, and our results help characterize molecular changes that could in future be used for patient treatment selection or prognosis prediction. © 2024 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Thomas Welton
- Department of Research, National Neuroscience Institute, Singapore, Singapore
- Neuroscience and Behavioural Disorders, Duke-NUS Medical School, Singapore, Singapore
| | - Gabriel Chew
- Neuroscience and Behavioural Disorders, Duke-NUS Medical School, Singapore, Singapore
| | - Aaron Shengting Mai
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Jing Han Ng
- Department of Neurology, Singapore General Hospital, Singapore, Singapore
| | - Ling Ling Chan
- Department of Research, National Neuroscience Institute, Singapore, Singapore
- Neuroscience and Behavioural Disorders, Duke-NUS Medical School, Singapore, Singapore
- Department of Diagnostic Radiology, Singapore General Hospital, Singapore, Singapore
| | - Eng-King Tan
- Department of Research, National Neuroscience Institute, Singapore, Singapore
- Neuroscience and Behavioural Disorders, Duke-NUS Medical School, Singapore, Singapore
- Department of Neurology, Singapore General Hospital, Singapore, Singapore
| |
Collapse
|
7
|
Chen Z, Ge R, Wang C, Elazab A, Fu X, Min W, Qin F, Jia G, Fan X. Identification of important gene signatures in schizophrenia through feature fusion and genetic algorithm. Mamm Genome 2024; 35:241-255. [PMID: 38512459 DOI: 10.1007/s00335-024-10034-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 02/07/2024] [Indexed: 03/23/2024]
Abstract
Schizophrenia is a debilitating psychiatric disorder that can significantly affect a patient's quality of life and lead to permanent brain damage. Although medical research has identified certain genetic risk factors, the specific pathogenesis of the disorder remains unclear. Despite the prevalence of research employing magnetic resonance imaging, few studies have focused on the gene level and gene expression profile involving a large number of screened genes. However, the high dimensionality of genetic data presents a great challenge to accurately modeling the data. To tackle the current challenges, this study presents a novel feature selection strategy that utilizes heuristic feature fusion and a multi-objective optimization genetic algorithm. The goal is to improve classification performance and identify the key gene subset for schizophrenia diagnostics. Traditional gene screening techniques are inadequate for accurately determining the precise number of key genes associated with schizophrenia. Our innovative approach integrates a filter-based feature selection method to reduce data dimensionality and a multi-objective optimization genetic algorithm for improved classification tasks. By combining the filtering and wrapper methods, our strategy leverages their respective strengths in a deliberate manner, leading to superior classification accuracy and a more efficient selection of relevant genes. This approach has demonstrated significant improvements in classification results across 11 out of 14 relevant datasets. The performance on the remaining three datasets is comparable to the existing methods. Furthermore, visual and enrichment analyses have confirmed the practicality of our proposed method as a promising tool for the early detection of schizophrenia.
Collapse
Affiliation(s)
| | - Ruiquan Ge
- Hangzhou Dianzi University, Hangzhou, China.
- Hangzhou Institute of Advanced Technology, Hangzhou, China.
- Key Laboratory of Discrete Industrial Internet of Things of Zhejiang Province, Hangzhou, China.
| | - Changmiao Wang
- Shenzhen Research Institute of Big Data, Shenzhen, China
| | - Ahmed Elazab
- Computer Science Department, Misr Higher Institute for Commerce and Computers, Mansoura, Egypt
| | - Xianjun Fu
- School of Artificial Intelligence, Zhejiang College of Security Technology, Wenzhou, China
| | - Wenwen Min
- School of Information Science and Engineering, Yunnan University, Kunming, China
| | - Feiwei Qin
- Hangzhou Dianzi University, Hangzhou, China
| | | | - Xiaopeng Fan
- Hangzhou Institute of Advanced Technology, Hangzhou, China
| |
Collapse
|
8
|
Xu Y, Cao L, Chen Y, Zhang Z, Liu W, Li H, Ding C, Pu J, Qian K, Xu W. Integrating Machine Learning in Metabolomics: A Path to Enhanced Diagnostics and Data Interpretation. SMALL METHODS 2024:e2400305. [PMID: 38682615 DOI: 10.1002/smtd.202400305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Revised: 04/07/2024] [Indexed: 05/01/2024]
Abstract
Metabolomics, leveraging techniques like NMR and MS, is crucial for understanding biochemical processes in pathophysiological states. This field, however, faces challenges in metabolite sensitivity, data complexity, and omics data integration. Recent machine learning advancements have enhanced data analysis and disease classification in metabolomics. This study explores machine learning integration with metabolomics to improve metabolite identification, data efficiency, and diagnostic methods. Using deep learning and traditional machine learning, it presents advancements in metabolic data analysis, including novel algorithms for accurate peak identification, robust disease classification from metabolic profiles, and improved metabolite annotation. It also highlights multiomics integration, demonstrating machine learning's potential in elucidating biological phenomena and advancing disease diagnostics. This work contributes significantly to metabolomics by merging it with machine learning, offering innovative solutions to analytical challenges and setting new standards for omics data analysis.
Collapse
Affiliation(s)
- Yudian Xu
- Department of Traditional Chinese Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, P. R. China
| | - Linlin Cao
- State Key Laboratory for Oncogenes and Related Genes, Division of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, 200127, P. R. China
| | - Yifan Chen
- State Key Laboratory for Oncogenes and Related Genes, Division of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, 200127, P. R. China
| | - Ziyue Zhang
- School of Biomedical Engineering, Institute of Medical Robotics and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, 200030, P. R. China
| | - Wanshan Liu
- School of Biomedical Engineering, Institute of Medical Robotics and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, 200030, P. R. China
| | - He Li
- Department of Traditional Chinese Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, P. R. China
| | - Chenhuan Ding
- Department of Traditional Chinese Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, P. R. China
| | - Jun Pu
- State Key Laboratory for Oncogenes and Related Genes, Division of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, 200127, P. R. China
| | - Kun Qian
- State Key Laboratory for Oncogenes and Related Genes, Division of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, 200127, P. R. China
- School of Biomedical Engineering, Institute of Medical Robotics and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, 200030, P. R. China
| | - Wei Xu
- State Key Laboratory for Oncogenes and Related Genes, Division of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, 200127, P. R. China
| |
Collapse
|
9
|
J SG, P D, P E. Enhancing drug discovery in schizophrenia: a deep learning approach for accurate drug-target interaction prediction - DrugSchizoNet. Comput Methods Biomech Biomed Engin 2024:1-18. [PMID: 38375638 DOI: 10.1080/10255842.2023.2282951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 10/17/2023] [Indexed: 02/21/2024]
Abstract
Drug discovery relies on the precise prognosis of drug-target interactions (DTI). Due to their ability to learn from raw data, deep learning (DL) methods have displayed outstanding performance over traditional approaches. However, challenges such as imbalanced data, noise, poor generalization, high cost, and time-consuming processes hinder progress in this field. To overcome the above challenges, we propose a DL-based model termed DrugSchizoNet for drug interaction (DI) prediction of Schizophrenia. Our model leverages drug-related data from the DrugBank and repoDB databases, employing three key preprocessing techniques. First, data cleaning eliminates duplicate or incomplete entries to ensure data integrity. Next, normalization is performed to enhance security and reduce costs associated with data acquisition. Finally, feature extraction is applied to improve the quality of input data. The three layers of the DrugSchizoNet model are the input, hidden and output layers. In the hidden layer, we employ dropout regularization to mitigate overfitting and improve generalization. The fully connected (FC) layer extracts relevant features, while the LSTM layer captures the sequential nature of DIs. In the output layer, our model provides confidence scores for potential DIs. To optimize the prediction accuracy, we utilize hyperparameter tuning through OB-MOA optimization. Experimental results demonstrate that DrugSchizoNet achieves a superior accuracy of 98.70%. The existing models, including CNN-RNN, DANN, CKA-MKL, DGAN, and CNN, across various evaluation metrics such as accuracy, recall, specificity, precision, F1 score, AUPR, and AUROC are compared with the proposed model. By effectively addressing the challenges of imbalanced data, noise, poor generalization, high cost and time-consuming processes, DrugSchizoNet offers a promising approach for accurate DTI prediction in Schizophrenia. Its superior performance demonstrates the potential of DL in advancing drug discovery and development processes.
Collapse
Affiliation(s)
- Sherine Glory J
- Department of Computer Science and Engineering, SRM Institute of Science and Technology, Vadapalani Campus, Chennai, India
| | - Durgadevi P
- Department of Computer Science and Engineering, SRM Institute of Science and Technology, Vadapalani Campus, Chennai, India
| | - Ezhumalai P
- Department of Computer Science and Engineering, R.M.D. Engineering College, Kavaraipettai, India
| |
Collapse
|
10
|
Montanari S, Jansen R, Schranner D, Kastenmüller G, Arnold M, Janiri D, Sani G, Bhattacharyya S, Dehkordi SM, Dunlop BW, Rush AJ, Penninx BWHJ, Kaddurah-Daouk R, Milaneschi Y. Acylcarnitines metabolism in depression: association with diagnostic status, depression severity and symptom profile in the NESDA cohort. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.02.14.24302813. [PMID: 38405847 PMCID: PMC10889013 DOI: 10.1101/2024.02.14.24302813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
Background Acylcarnitines (ACs) are involved in bioenergetics processes that may play a role in the pathophysiology of depression. Studies linking AC levels to depression are few and provide mixed findings. We examined the association of circulating ACs levels with Major Depressive Disorder (MDD) diagnosis, overall depression severity and specific symptom profiles. Methods The sample from the Netherlands Study of Depression and Anxiety included participants with current (n=1035) or remitted (n=739) MDD and healthy controls (n=800). Plasma levels of four ACs (short-chain: acetylcarnitine C2 and propionylcarnitine C3; medium-chain: octanoylcarnitine C8 and decanoylcarnitine C10) were measured. Overall depression severity as well as atypical/energy-related (AES), anhedonic and melancholic symptom profiles were derived from the Inventory of Depressive Symptomatology. Results As compared to healthy controls, subjects with current or remitted MDD presented similarly lower mean C2 levels (Cohen's d=0.2, p≤1e-4). Higher overall depression severity was significantly associated with higher C3 levels (ß=0.06, SE=0.02, p=1.21e-3). No associations were found for C8 and C10. Focusing on symptom profiles, only higher AES scores were linked to lower C2 (ß=-0.05, SE=0.02, p=1.85e-2) and higher C3 (ß=0.08, SE=0.02, p=3.41e-5) levels. Results were confirmed in analyses pooling data with an additional internal replication sample from the same subjects measured at 6-year follow-up (totaling 4195 observations). Conclusions Small alterations in levels of short-chain acylcarnitine levels were related to the presence and severity of depression, especially for symptoms reflecting altered energy homeostasis. Cellular metabolic dysfunctions may represent a key pathway in depression pathophysiology potentially accessible through AC metabolism.
Collapse
Affiliation(s)
- Silvia Montanari
- Department of Neuroscience, Section of Psychiatry, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Rick Jansen
- Department of Psychiatry, Amsterdam UMC,Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Amsterdam Public Health, Mental Health program, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Mood, Anxiety, Psychosis, Sleep & Stress program, Amsterdam, The Netherlands
| | - Daniela Schranner
- Institute of Computational Biology, Helmholtz Zentrum München, Neuherberg, Germany
| | - Gabi Kastenmüller
- Institute of Computational Biology, Helmholtz Zentrum München, Neuherberg, Germany
| | - Matthias Arnold
- Institute of Computational Biology, Helmholtz Zentrum München, Neuherberg, Germany
- Department of Psychiatry and Behavioral Sciences, Duke University, Durham, NC, USA
| | - Delfina Janiri
- Department of Neuroscience, Section of Psychiatry, Università Cattolica del Sacro Cuore, Rome, Italy
- Department of Psychiatry, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Gabriele Sani
- Department of Neuroscience, Section of Psychiatry, Università Cattolica del Sacro Cuore, Rome, Italy
- Department of Psychiatry, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Sudeepa Bhattacharyya
- Arkansas Biosciences Institute, Department of Biological Sciences, Arkansas State University, AR, USA
| | | | - Boadie W Dunlop
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, USA
| | - A. John Rush
- Department of Psychiatry and Behavioral Sciences, Duke University, Durham, NC, USA
- Duke-National University of Singapore, Singapore
| | - Brenda W. H. J. Penninx
- Department of Psychiatry, Amsterdam UMC,Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Amsterdam Public Health, Mental Health program, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Mood, Anxiety, Psychosis, Sleep & Stress program, Amsterdam, The Netherlands
| | - Rima Kaddurah-Daouk
- Department of Psychiatry and Behavioral Sciences, Duke University, Durham, NC, USA
- Duke Institute of Brain Sciences, Duke University, Durham, NC, USA
- Department of Medicine, Duke University, Durham, NC, USA
| | - Yuri Milaneschi
- Department of Psychiatry, Amsterdam UMC,Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Amsterdam Public Health, Mental Health program, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Mood, Anxiety, Psychosis, Sleep & Stress program, Amsterdam, The Netherlands
| |
Collapse
|
11
|
Bhattacharyya U, John J, Lam M, Fisher J, Sun B, Baird D, Chen CY, Lencz T. Large-Scale Mendelian Randomization Study Reveals Circulating Blood-based Proteomic Biomarkers for Psychopathology and Cognitive Task Performance. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.01.18.24301455. [PMID: 38293198 PMCID: PMC10827252 DOI: 10.1101/2024.01.18.24301455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2024]
Abstract
Background Research on peripheral (e.g., blood-based) biomarkers for psychiatric illness has typically been low-throughput in terms of both the number of subjects and the range of assays performed. Moreover, traditional case-control studies examining blood-based biomarkers are subject to potential confounds of treatment and other exposures common to patients with psychiatric illnesses. Our research addresses these challenges by leveraging large-scale, high-throughput proteomics data and Mendelian Randomization (MR) to examine the causal impact of circulating proteins on psychiatric phenotypes and cognitive task performance. Methods We utilized plasma proteomics data from the UK Biobank (3,072 proteins assayed in 34,557 European-ancestry individuals) and deCODE Genetics (4,719 proteins measured across 35,559 Icelandic individuals). Significant proteomic quantitative trait loci (both cis-pQTLs and trans-pQTLs) served as MR instruments, with the most recent GWAS for schizophrenia, bipolar disorder, major depressive disorder, and cognitive task performance (all excluding overlapping UK Biobank participants) as phenotypic outcomes. Results MR revealed 109 Bonferroni-corrected causal associations (44 novel) involving 88 proteins across the four phenotypes. Several immune-related proteins, including interleukins and complement factors, stood out as pleiotropic across multiple outcome phenotypes. Drug target enrichment analysis identified several novel potential pharmacologic repurposing opportunities, including anti-inflammatory agents for schizophrenia and bipolar disorder and duloxetine for cognitive performance. Conclusions Identification of causal effects for these circulating proteins suggests potential biomarkers for these conditions and offers insights for developing innovative therapeutic strategies. The findings also indicate substantial evidence for the pleiotropic effects of many proteins across different phenotypes, shedding light on the shared etiology among psychiatric conditions and cognitive ability.
Collapse
Affiliation(s)
- Upasana Bhattacharyya
- Institute of Behavioral Science, Feinstein Institutes for Medical Research, Manhasset, NY
- Division of Psychiatry Research, Zucker Hillside Hospital, Glen Oaks, NY
| | - Jibin John
- Institute of Behavioral Science, Feinstein Institutes for Medical Research, Manhasset, NY
- Division of Psychiatry Research, Zucker Hillside Hospital, Glen Oaks, NY
| | - Max Lam
- Institute of Behavioral Science, Feinstein Institutes for Medical Research, Manhasset, NY
- Division of Psychiatry Research, Zucker Hillside Hospital, Glen Oaks, NY
| | - Jonah Fisher
- Biogen Inc., Cambridge, MA
- Harvard T.H. Chan School of Public Health, Cambridge, MA
| | | | | | | | - Todd Lencz
- Institute of Behavioral Science, Feinstein Institutes for Medical Research, Manhasset, NY
- Division of Psychiatry Research, Zucker Hillside Hospital, Glen Oaks, NY
- Departments of Psychiatry and Molecular Medicine, Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY
| |
Collapse
|
12
|
Gu YW, Fan JW, Zhào H, Zhao SW, Liu XF, Yu R, Ren L, Wang X, Yin H, Cui LB. Imaging Transcriptomics of the Brain for Schizophrenia. ALPHA PSYCHIATRY 2024; 25:9-14. [PMID: 38799487 PMCID: PMC11114239 DOI: 10.5152/alphapsychiatry.2024.231369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 12/04/2023] [Indexed: 05/29/2024]
Abstract
Schizophrenia is a severe mental disorder with a neurodevelopmental origin. Although schizophrenia results from changes in the brain, the underlying biological mechanisms are unknown. Transcriptomics studies quantitative expression changes or qualitative changes of all genes and isoforms, providing a more meaningful biological insight. Magnetic resonance imaging (MRI) techniques play roles in revealing brain structure and function. We give a narrative focused review on the current transcriptome combined with MRI studies related to schizophrenia and summarize the research methodology and content of these studies to identify the research commonalities as well as the implications for future research, in an attempt to provide new insights into the mechanism, clinical diagnosis, and treatments of schizophrenia.
Collapse
Affiliation(s)
- Yue-Wen Gu
- Department of Clinical Psychology, Air Force Medical University, Xi’an, China
- Department of Radiology, Xijing Hospital, Air Force Medical University, Xi’an, China
| | - Jing-Wen Fan
- Department of Clinical Psychology, Air Force Medical University, Xi’an, China
- Department of Radiology, Xijing Hospital, Air Force Medical University, Xi’an, China
| | - Hóngyi Zhào
- Department of Neurology, NO. 984 Hospital of PLA, Beijing, China
| | - Shu-Wan Zhao
- Department of Clinical Psychology, Air Force Medical University, Xi’an, China
- Department of Radiology, Xijing Hospital, Air Force Medical University, Xi’an, China
| | - Xiao-Fan Liu
- Department of Clinical Psychology, Air Force Medical University, Xi’an, China
| | - Renqiang Yu
- Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Lei Ren
- Department of Clinical Psychology, Air Force Medical University, Xi’an, China
| | - Xinjiang Wang
- Department of Radiology, The Second Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Hong Yin
- Department of Radiology, Xijing Hospital, Air Force Medical University, Xi’an, China
- Department of Radiology, Xi’an People’s Hospital (Xi’an Fourth Hospital), Xi’an, China
| | - Long-Biao Cui
- Department of Clinical Psychology, Air Force Medical University, Xi’an, China
- Department of Radiology, The Second Medical Center, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
13
|
Hai Y, Ma J, Yang K, Wen Y. Bayesian linear mixed model with multiple random effects for prediction analysis on high-dimensional multi-omics data. Bioinformatics 2023; 39:btad647. [PMID: 37882747 PMCID: PMC10627352 DOI: 10.1093/bioinformatics/btad647] [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: 06/27/2023] [Revised: 09/24/2023] [Accepted: 10/24/2023] [Indexed: 10/27/2023] Open
Abstract
MOTIVATION Accurate disease risk prediction is an essential step in the modern quest for precision medicine. While high-dimensional multi-omics data have provided unprecedented data resources for prediction studies, their high-dimensionality and complex inter/intra-relationships have posed significant analytical challenges. RESULTS We proposed a two-step Bayesian linear mixed model framework (TBLMM) for risk prediction analysis on multi-omics data. TBLMM models the predictive effects from multi-omics data using a hybrid of the sparsity regression and linear mixed model with multiple random effects. It can resemble the shape of the true effect size distributions and accounts for non-linear, including interaction effects, among multi-omics data via kernel fusion. It infers its parameters via a computationally efficient variational Bayes algorithm. Through extensive simulation studies and the prediction analyses on the positron emission tomography imaging outcomes using data obtained from the Alzheimer's Disease Neuroimaging Initiative, we have demonstrated that TBLMM can consistently outperform the existing method in predicting the risk of complex traits. AVAILABILITY AND IMPLEMENTATION The corresponding R package is available on GitHub (https://github.com/YaluWen/TBLMM).
Collapse
Affiliation(s)
- Yang Hai
- Department of Health Statistics, Shanxi Medical University, Taiyuan, Shanxi Province 030000, China
- Department of Statistics, University of Auckland, Auckland 1010, New Zealand
| | - Jixiang Ma
- Department of Health Statistics, Shanxi Medical University, Taiyuan, Shanxi Province 030000, China
| | - Kaixin Yang
- Department of Health Statistics, Shanxi Medical University, Taiyuan, Shanxi Province 030000, China
| | - Yalu Wen
- Department of Health Statistics, Shanxi Medical University, Taiyuan, Shanxi Province 030000, China
- Department of Statistics, University of Auckland, Auckland 1010, New Zealand
| |
Collapse
|
14
|
Wu Y, Zhang CY, Wang L, Li Y, Xiao X. Genetic Insights of Schizophrenia via Single Cell RNA-Sequencing Analyses. Schizophr Bull 2023; 49:914-922. [PMID: 36805283 PMCID: PMC10318862 DOI: 10.1093/schbul/sbad002] [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] [Indexed: 02/22/2023]
Abstract
BACKGROUND Schizophrenia is a complex and heterogeneous disorder involving multiple regions and types of cells in the brain. Despite rapid progress made by genome-wide association studies (GWAS) of schizophrenia, the mechanisms of the illness underlying the GWAS significant loci remain less clear. STUDY DESIGN We investigated schizophrenia risk genes using summary-data-based Mendelian randomization based on single-cell sequencing data, and explored the types of brain cells involved in schizophrenia through the expression weighted cell-type enrichment analysis. RESULTS We identified 54 schizophrenia risk genes (two-thirds of these genes were not identified using sequencing data of bulk tissues) using single-cell RNA-sequencing data. Further cell type enrichment analysis showed that schizophrenia risk genes were highly expressed in excitatory neurons and caudal ganglionic eminence interneurons, suggesting putative roles of these cells in the pathogenesis of schizophrenia. We also found that these risk genes identified using single-cell sequencing results could form a large protein-protein interaction network with genes affected by disease-causing rare variants. CONCLUSIONS Through integrative analyses using expression data at single-cell levels, we identified 54 risk genes associated with schizophrenia. Notably, many of these genes were only identified using single-cell RNA-sequencing data, and their altered expression levels in particular types of cells, rather than in the bulk tissues, were related to the increased risk of schizophrenia. Our results provide novel insight into the biological mechanisms of schizophrenia, and future single-cell studies are necessary to further facilitate the understanding of the disorder.
Collapse
Affiliation(s)
- Yong Wu
- Research Center for Mental Health and Neuroscience, Wuhan Mental Health Center, Wuhan, Hubei, China
- Affiliated Wuhan Mental Health Center, Jianghan University, Wuhan, Hubei, China
| | - Chu-Yi Zhang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Lu Wang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Yi Li
- Research Center for Mental Health and Neuroscience, Wuhan Mental Health Center, Wuhan, Hubei, China
- Affiliated Wuhan Mental Health Center, Jianghan University, Wuhan, Hubei, China
- Research Center for Psychological and Health Sciences, China University of Geosciences, Wuhan, Hubei, China
| | - Xiao Xiao
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, China
| |
Collapse
|
15
|
Abstract
The field of psychiatry is facing an important paradigm shift in the provision of clinical care and mental health service organization toward personalization and integration of multimodal data science. This approach, termed precision psychiatry, aims at identifying subgroups of patients more prone to the development of a certain phenotype, such as symptoms or severe mental disorders (risk detection), and/or to guide treatment selection. Pharmacogenomics and computational psychiatry are two fundamental tools of precision psychiatry, which have seen increasing levels of integration in clinical settings. Here we present a brief overview of these two applications of precision psychiatry in clinical settings.
Collapse
Affiliation(s)
- Mirko Manchia
- Section of Psychiatry, Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, 09127, Italy
- Unit of Clinical Psychiatry, University Hospital Agency of Cagliari, Cagliari, 09127,Italy
- Department of Pharmacology, Dalhousie University, Halifax, NS, B3H 4R2, Canada
| | - Martino Belvederi Murri
- Institute of Psychiatry, Department of Neuroscience and Rehabilitation, University of Ferrara, Ferrara, 44121, Italy
| |
Collapse
|
16
|
Fan JW, Gu YW, Wang DB, Liu XF, Zhao SW, Li X, Li B, Yin H, Wu WJ, Cui LB. Transcriptomics and magnetic resonance imaging in major psychiatric disorders. Front Psychiatry 2023; 14:1185471. [PMID: 37383618 PMCID: PMC10296768 DOI: 10.3389/fpsyt.2023.1185471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 05/16/2023] [Indexed: 06/30/2023] Open
Abstract
Major psychiatric disorders create a significant public health burden, and mental disorders such as major depressive disorder, bipolar disorder, and schizophrenia are major contributors to the national disease burden. The search for biomarkers has been a leading endeavor in the field of biological psychiatry in recent decades. And the application of cross-scale and multi-omics approaches combining genes and imaging in major psychiatric studies has facilitated the elucidation of gene-related pathogenesis and the exploration of potential biomarkers. In this article, we summarize the results of using combined transcriptomics and magnetic resonance imaging to understand structural and functional brain changes associated with major psychiatric disorders in the last decade, demonstrating the neurobiological mechanisms of genetically related structural and functional brain alterations in multiple directions, and providing new avenues for the development of quantifiable objective biomarkers, as well as clinical diagnostic and prognostic indicators.
Collapse
Affiliation(s)
- Jing-Wen Fan
- Department of Clinical Psychology, Fourth Military Medical University, Xi'an, China
- Department of Radiology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Yue-Wen Gu
- Department of Clinical Psychology, Fourth Military Medical University, Xi'an, China
- Department of Radiology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Dong-Bao Wang
- Schizophrenia Imaging Lab, Fourth Military Medical University, Xi’an, China
| | - Xiao-Fan Liu
- Department of Clinical Psychology, Fourth Military Medical University, Xi'an, China
| | - Shu-Wan Zhao
- Department of Clinical Psychology, Fourth Military Medical University, Xi'an, China
- Department of Radiology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Xiao Li
- Department of Psychiatry, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Baojuan Li
- School of Biomedical Engineering, Fourth Military Medical University, Xi'an, China
| | - Hong Yin
- Department of Radiology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
- Department of Radiology, Xi'an People's Hospital (Xi'an Fourth Hospital), Xi'an, China
| | - Wen-Jun Wu
- Department of Psychiatry, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Long-Biao Cui
- Department of Clinical Psychology, Fourth Military Medical University, Xi'an, China
- Schizophrenia Imaging Lab, Fourth Military Medical University, Xi’an, China
- Department of Radiology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| |
Collapse
|
17
|
Goel T, Varaprasad SA, Tanveer M, Pilli R. Investigating White Matter Abnormalities Associated with Schizophrenia Using Deep Learning Model and Voxel-Based Morphometry. Brain Sci 2023; 13:brainsci13020267. [PMID: 36831810 PMCID: PMC9954172 DOI: 10.3390/brainsci13020267] [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: 12/30/2022] [Revised: 01/19/2023] [Accepted: 01/27/2023] [Indexed: 02/09/2023] Open
Abstract
Schizophrenia (SCZ) is a devastating mental condition with significant negative consequences for patients, making correct and prompt diagnosis crucial. The purpose of this study is to use structural magnetic resonance image (MRI) to better classify individuals with SCZ from control normals (CN) and to locate a region of the brain that represents abnormalities associated with SCZ. Deep learning (DL), which is based on the nervous system, could be a very useful tool for doctors to accurately predict, diagnose, and treat SCZ. Gray Matter (GM), Cerebrospinal Fluid (CSF), and White Matter (WM) brain regions are extracted from 99 MRI images obtained from the open-source OpenNeuro database to demonstrate SCZ's regional relationship. In this paper, we use a pretrained ResNet-50 deep network to extract features from MRI images and an ensemble deep random vector functional link (edRVFL) network to classify those features. By examining the results obtained, the edRVFL deep model provides the highest classification accuracy of 96.5% with WM and is identified as the best-performing algorithm compared to the traditional algorithms. Furthermore, we examined the GM, WM, and CSF tissue volumes in CN subjects and SCZ patients using voxel-based morphometry (VBM), and the results show 1363 significant voxels, 6.90 T-value, and 6.21 Z-value in the WM region of SCZ patients. In SCZ patients, WM is most closely linked to structural alterations, as evidenced by VBM analysis and the DL model.
Collapse
Affiliation(s)
- Tripti Goel
- Biomedical Imaging Lab, Department of Electronics and Communication Engineering, National Institute of Technology Silchar, Silchar 788010, Assam, India
- Correspondence: (T.G.); (M.T.)
| | - Sirigineedi A. Varaprasad
- Biomedical Imaging Lab, Department of Electronics and Communication Engineering, National Institute of Technology Silchar, Silchar 788010, Assam, India
| | - M. Tanveer
- Department of Mathematics, Indian Institute of Technology Indore, Simrol 453552, Madhya Pradesh, India
- Correspondence: (T.G.); (M.T.)
| | - Raveendra Pilli
- Biomedical Imaging Lab, Department of Electronics and Communication Engineering, National Institute of Technology Silchar, Silchar 788010, Assam, India
| |
Collapse
|
18
|
Schizophrenia and psychedelic state: Dysconnection versus hyper-connection. A perspective on two different models of psychosis stemming from dysfunctional integration processes. Mol Psychiatry 2023; 28:59-67. [PMID: 35931756 DOI: 10.1038/s41380-022-01721-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 07/15/2022] [Accepted: 07/22/2022] [Indexed: 01/07/2023]
Abstract
Psychotic symptoms are a cross-sectional dimension affecting multiple diagnostic categories, despite schizophrenia represents the prototype of psychoses. Initially, dopamine was considered the most involved molecule in the neurobiology of schizophrenia. Over the next years, several biological factors were added to the discussion helping to constitute the concept of schizophrenia as a disease marked by a deficit of functional integration, contributing to the formulation of the Dysconnection Hypothesis in 1995. Nowadays the notion of dysconnection persists in the conceptualization of schizophrenia enriched by neuroimaging findings which corroborate the hypothesis. At the same time, in recent years, psychedelics received a lot of attention by the scientific community and astonishing findings emerged about the rearrangement of brain networks under the effect of these compounds. Specifically, a global decrease in functional connectivity was found, highlighting the disintegration of preserved and functional circuits and an increase of overall connectivity in the brain. The aim of this paper is to compare the biological bases of dysconnection in schizophrenia with the alterations of neuronal cyto-architecture induced by psychedelics and the consequent state of cerebral hyper-connection. These two models of psychosis, despite diametrically opposed, imply a substantial deficit of integration of neural signaling reached through two opposite paths.
Collapse
|
19
|
de Bartolomeis A, De Simone G, Ciccarelli M, Castiello A, Mazza B, Vellucci L, Barone A. Antipsychotics-Induced Changes in Synaptic Architecture and Functional Connectivity: Translational Implications for Treatment Response and Resistance. Biomedicines 2022; 10:3183. [PMID: 36551939 PMCID: PMC9776416 DOI: 10.3390/biomedicines10123183] [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: 10/27/2022] [Revised: 12/02/2022] [Accepted: 12/04/2022] [Indexed: 12/14/2022] Open
Abstract
Schizophrenia is a severe mental illness characterized by alterations in processes that regulate both synaptic plasticity and functional connectivity between brain regions. Antipsychotics are the cornerstone of schizophrenia pharmacological treatment and, beyond occupying dopamine D2 receptors, can affect multiple molecular targets, pre- and postsynaptic sites, as well as intracellular effectors. Multiple lines of evidence point to the involvement of antipsychotics in sculpting synaptic architecture and remodeling the neuronal functional unit. Furthermore, there is an increasing awareness that antipsychotics with different receptor profiles could yield different interregional patterns of co-activation. In the present systematic review, we explored the fundamental changes that occur under antipsychotics' administration, the molecular underpinning, and the consequences in both acute and chronic paradigms. In addition, we investigated the relationship between synaptic plasticity and functional connectivity and systematized evidence on different topographical patterns of activation induced by typical and atypical antipsychotics.
Collapse
Affiliation(s)
- Andrea de Bartolomeis
- Section of Psychiatry, Laboratory of Translational and Molecular Psychiatry and Unit of Treatment-Resistant Psychosis, Department of Neuroscience, Reproductive Sciences and Odontostomatology, University Medical School of Naples “Federico II”, Via Pansini 5, 80131 Naples, Italy
| | | | | | | | | | | | | |
Collapse
|
20
|
Genetic Modifiers of Sickle Cell Disease. Hematol Oncol Clin North Am 2022; 36:1097-1124. [DOI: 10.1016/j.hoc.2022.06.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
21
|
Wang Y, Tang S, Ma R, Zamit I, Wei Y, Pan Y. Multi-modal intermediate integrative methods in neuropsychiatric disorders: A review. Comput Struct Biotechnol J 2022; 20:6149-6162. [PMID: 36420153 PMCID: PMC9674886 DOI: 10.1016/j.csbj.2022.11.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 11/04/2022] [Accepted: 11/04/2022] [Indexed: 11/09/2022] Open
Abstract
The etiology of neuropsychiatric disorders involves complex biological processes at different omics layers, such as genomics, transcriptomics, epigenetics, proteomics, and metabolomics. The advent of high-throughput technology, as well as the availability of large open-source datasets, has ushered in a new era in system biology, necessitating the integration of various types of omics data. The complexity of biological mechanisms, the limitations of integrative strategies, and the heterogeneity of multi-omics data have all presented significant challenges to computational scientists. In comparison to early and late integration, intermediate integration may transform each data type into appropriate intermediate representations using various data transformation techniques, allowing it to capture more complementary information contained in each omics and highlight new interactions across omics layers. Here, we reviewed multi-modal intermediate integrative techniques based on component analysis, matrix factorization, similarity network, multiple kernel learning, Bayesian network, artificial neural networks, and graph transformation, as well as their applications in neuropsychiatric domains. We depicted advancements in these approaches and compared the strengths and weaknesses of each method examined. We believe that our findings will aid researchers in their understanding of the transformation and integration of multi-omics data in neuropsychiatric disorders.
Collapse
Affiliation(s)
- Yanlin Wang
- Center for High Performance Computing, Joint Engineering Research Center for Health Big Data Intelligent Analysis Technology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
| | - Shi Tang
- Li Chiu Kong Family Sleep Assessment Unit, Department of Psychiatry, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong Special Administrative Region
| | - Ruimin Ma
- Center for High Performance Computing, Joint Engineering Research Center for Health Big Data Intelligent Analysis Technology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
| | - Ibrahim Zamit
- Center for High Performance Computing, Joint Engineering Research Center for Health Big Data Intelligent Analysis Technology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yanjie Wei
- Center for High Performance Computing, Joint Engineering Research Center for Health Big Data Intelligent Analysis Technology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
| | - Yi Pan
- Center for High Performance Computing, Joint Engineering Research Center for Health Big Data Intelligent Analysis Technology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
| |
Collapse
|
22
|
Wang X, Xiao L, Wang Z, Zhi L, Li Q. Common variants in GNL3 gene contributed the susceptibility of hand osteoarthritis in Han Chinese population. Sci Rep 2022; 12:16110. [PMID: 36167888 PMCID: PMC9515075 DOI: 10.1038/s41598-022-20287-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 09/12/2022] [Indexed: 11/09/2022] Open
Abstract
Osteoarthritis (OA) is one of the most popular degenerative joint diseases. The nucleolar GTP binding protein 3 (GNL3) gene encodes guanine nucleotide binding protein-like 3, which is related in cell proliferation, differentiation, and cell cycle regulation. Our study aimed to examine the contribution of GNL3 gene polymorphisms to the risk of hand OA and its related clinical features. A total of 3387 study participants including 1160 patients with hand OA and 2227 controls were recruited in this study. Eleven SNPs in GNL3 gene were selected for genotyping. Genetic association signals were examined using Plink. Relationships between significant SNPs and clinical features of hand OA were also explored. SNP rs11177 was found to be strongly associated with susceptibility of hand OA (P = 4.32 × 10-5). The minor allele of rs11177 was associated with increased susceptibility of hand OA. In addition, significant associations were also identified between genotypes of rs11177 and clinical features of hand OA patients including K-L grade (P < 0.01) and categorized pain scores (P < 0.01). Significant eQTL signals for rs11177 on GNL3 in multiple types of human tissues were also identified in GTEx database. Our results have established the link between GNL3 gene and susceptibility of hand OA.
Collapse
Affiliation(s)
- Xi Wang
- Department of Knee Joint Surgery, Xi'an Honghui Hospital, Xi'an, Shaanxi, China
| | - Lin Xiao
- Department of Knee Joint Surgery, Xi'an Honghui Hospital, Xi'an, Shaanxi, China
| | - Zhiyuan Wang
- Department of Knee Joint Surgery, Xi'an Honghui Hospital, Xi'an, Shaanxi, China
| | - Liqiang Zhi
- Department of Knee Joint Surgery, Xi'an Honghui Hospital, Xi'an, Shaanxi, China
| | - Qiang Li
- Department of Hand Surgery, Xi'an Honghui Hospital, No. 555 Youyi East Road, Xi'an, 710054, Shaanxi, China.
| |
Collapse
|
23
|
Guo X, Han J, Song Y, Yin Z, Liu S, Shang X. Using expression quantitative trait loci data and graph-embedded neural networks to uncover genotype–phenotype interactions. Front Genet 2022; 13:921775. [PMID: 36046233 PMCID: PMC9421127 DOI: 10.3389/fgene.2022.921775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Accepted: 07/04/2022] [Indexed: 11/13/2022] Open
Abstract
Motivation: A central goal of current biology is to establish a complete functional link between the genotype and phenotype, known as the so-called genotype–phenotype map. With the continuous development of high-throughput technology and the decline in sequencing costs, multi-omics analysis has become more widely employed. While this gives us new opportunities to uncover the correlation mechanisms between single-nucleotide polymorphism (SNP), genes, and phenotypes, multi-omics still faces certain challenges, specifically: 1) When the sample size is large enough, the number of omics types is often not large enough to meet the requirements of multi-omics analysis; 2) each omics’ internal correlations are often unclear, such as the correlation between genes in genomics; 3) when analyzing a large number of traits (p), the sample size (n) is often smaller than p, n << p, hindering the application of machine learning methods in the classification of disease outcomes.Results: To solve these issues with multi-omics and build a robust classification model, we propose a graph-embedded deep neural network (G-EDNN) based on expression quantitative trait loci (eQTL) data, which achieves sparse connectivity between network layers to prevent overfitting. The correlation within each omics is also considered such that the model more closely resembles biological reality. To verify the capabilities of this method, we conducted experimental analysis using the GSE28127 and GSE95496 data sets from the Gene Expression Omnibus (GEO) database, tested various neural network architectures, and used prior data for feature selection and graph embedding. Results show that the proposed method could achieve a high classification accuracy and easy-to-interpret feature selection. This method represents an extended application of genotype–phenotype association analysis in deep learning networks.
Collapse
Affiliation(s)
- Xinpeng Guo
- School of Computer Science and Engineering, Northwestern Polytechnical University, Xi’an, China
- School of Air and Missile Defense, Air Force Engineering University, Xi’an, China
| | - Jinyu Han
- School of Economics and Management, Chang ‘an University, Xi’an, China
| | - Yafei Song
- School of Air and Missile Defense, Air Force Engineering University, Xi’an, China
| | - Zhilei Yin
- School of Computer Science and Engineering, Northwestern Polytechnical University, Xi’an, China
| | - Shuaichen Liu
- School of Marine Science and Technology, Northwestern Polytechnical University, Xi’an, China
| | - Xuequn Shang
- School of Computer Science and Engineering, Northwestern Polytechnical University, Xi’an, China
- *Correspondence: Xuequn Shang,
| |
Collapse
|
24
|
Giordano GM, Caporusso E, Pezzella P, Galderisi S. Updated perspectives on the clinical significance of negative symptoms in patients with schizophrenia. Expert Rev Neurother 2022; 22:541-555. [PMID: 35758871 DOI: 10.1080/14737175.2022.2092402] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Negative symptoms in schizophrenia are associated with poor response to available treatments, poor quality of life, and functional outcome. Therefore, they represent a substantial burden for people with schizophrenia, their families, and health-care systems. AREAS COVERED In this manuscript, we will provide an update on the conceptualization, assessment, and treatment of this complex psychopathological dimension of schizophrenia. EXPERT OPINION Despite the progress in the conceptualization of negative symptoms and in the development of state-of-the-art assessment instruments made in the last decades, these symptoms are still poorly recognized, and not always assessed in line with current conceptualization. Every effort should be made to disseminate the current knowledge on negative symptoms, on their assessment instruments and available treatments whose efficacy is supported by research evidence. Longitudinal studies should be promoted to evaluate the natural course of negative symptoms, improve our ability to identify the different sources of secondary negative symptoms, provide effective interventions, and target primary and persistent negative symptoms with innovative treatment strategies. Further research is needed to identify pathophysiological mechanisms of primary negative symptoms and foster the development of new treatments.
Collapse
|
25
|
Song P, Wang Y, Yuan X, Wang S, Song X. Exploring Brain Structural and Functional Biomarkers in Schizophrenia via Brain-Network-Constrained Multi-View SCCA. Front Neurosci 2022; 16:879703. [PMID: 35794950 PMCID: PMC9252525 DOI: 10.3389/fnins.2022.879703] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Accepted: 04/04/2022] [Indexed: 11/17/2022] Open
Abstract
Recent studies have proved that dynamic regional measures extracted from the resting-state functional magnetic resonance imaging, such as the dynamic fractional amplitude of low-frequency fluctuation (d-fALFF), could provide a great insight into brain dynamic characteristics of the schizophrenia. However, the unimodal feature is limited for delineating the complex patterns of brain deficits. Thus, functional and structural imaging data are usually analyzed together for uncovering the neural mechanism of schizophrenia. Investigation of neural function-structure coupling enables to find the potential biomarkers and further helps to understand the biological basis of schizophrenia. Here, a brain-network-constrained multi-view sparse canonical correlation analysis (BN-MSCCA) was proposed to explore the intrinsic associations between brain structure and dynamic brain function. Specifically, the d-fALFF was first acquired based on the sliding window method, whereas the gray matter map was computed based on voxel-based morphometry analysis. Then, the region-of-interest (ROI)-based features were extracted and further selected by performing the multi-view sparse canonical correlation analysis jointly with the diagnosis information. Moreover, the brain-network-based structural constraint was introduced to prompt the detected biomarkers more interpretable. The experiments were conducted on 191 patients with schizophrenia and 191 matched healthy controls. Results showed that the BN-MSCCA could identify the critical ROIs with more sparse canonical weight patterns, which are corresponding to the specific brain networks. These are biologically meaningful findings and could be treated as the potential biomarkers. The proposed method also obtained a higher canonical correlation coefficient for the testing data, which is more consistent with the results on training data, demonstrating its promising capability for the association identification. To demonstrate the effectiveness of the potential clinical applications, the detected biomarkers were further analyzed on a schizophrenia-control classification task and a correlation analysis task. The experimental results showed that our method had a superior performance with a 5-8% increment in accuracy and 6-10% improvement in area under the curve. Furthermore, two of the top-ranked biomarkers were significantly negatively correlated with the positive symptom score of Positive and Negative Syndrome Scale (PANSS). Overall, the proposed method could find the association between brain structure and dynamic brain function, and also help to identify the biological meaningful biomarkers of schizophrenia. The findings enable our further understanding of this disease.
Collapse
Affiliation(s)
- Peilun Song
- School of Information Engineering, Zhengzhou University, Zhengzhou, China
| | - Yaping Wang
- School of Information Engineering, Zhengzhou University, Zhengzhou, China
| | - Xiuxia Yuan
- Department of Psychiatry, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Biological Psychiatry International Joint Laboratory of Henan/Zhengzhou University, Zhengzhou, China
| | - Shuying Wang
- Department of Psychiatry, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Biological Psychiatry International Joint Laboratory of Henan/Zhengzhou University, Zhengzhou, China
| | - Xueqin Song
- Department of Psychiatry, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Biological Psychiatry International Joint Laboratory of Henan/Zhengzhou University, Zhengzhou, China
| |
Collapse
|
26
|
Gupta C, Chandrashekar P, Jin T, He C, Khullar S, Chang Q, Wang D. Bringing machine learning to research on intellectual and developmental disabilities: taking inspiration from neurological diseases. J Neurodev Disord 2022; 14:28. [PMID: 35501679 PMCID: PMC9059371 DOI: 10.1186/s11689-022-09438-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 04/07/2022] [Indexed: 12/31/2022] Open
Abstract
Intellectual and Developmental Disabilities (IDDs), such as Down syndrome, Fragile X syndrome, Rett syndrome, and autism spectrum disorder, usually manifest at birth or early childhood. IDDs are characterized by significant impairment in intellectual and adaptive functioning, and both genetic and environmental factors underpin IDD biology. Molecular and genetic stratification of IDDs remain challenging mainly due to overlapping factors and comorbidity. Advances in high throughput sequencing, imaging, and tools to record behavioral data at scale have greatly enhanced our understanding of the molecular, cellular, structural, and environmental basis of some IDDs. Fueled by the "big data" revolution, artificial intelligence (AI) and machine learning (ML) technologies have brought a whole new paradigm shift in computational biology. Evidently, the ML-driven approach to clinical diagnoses has the potential to augment classical methods that use symptoms and external observations, hoping to push the personalized treatment plan forward. Therefore, integrative analyses and applications of ML technology have a direct bearing on discoveries in IDDs. The application of ML to IDDs can potentially improve screening and early diagnosis, advance our understanding of the complexity of comorbidity, and accelerate the identification of biomarkers for clinical research and drug development. For more than five decades, the IDDRC network has supported a nexus of investigators at centers across the USA, all striving to understand the interplay between various factors underlying IDDs. In this review, we introduced fast-increasing multi-modal data types, highlighted example studies that employed ML technologies to illuminate factors and biological mechanisms underlying IDDs, as well as recent advances in ML technologies and their applications to IDDs and other neurological diseases. We discussed various molecular, clinical, and environmental data collection modes, including genetic, imaging, phenotypical, and behavioral data types, along with multiple repositories that store and share such data. Furthermore, we outlined some fundamental concepts of machine learning algorithms and presented our opinion on specific gaps that will need to be filled to accomplish, for example, reliable implementation of ML-based diagnosis technology in IDD clinics. We anticipate that this review will guide researchers to formulate AI and ML-based approaches to investigate IDDs and related conditions.
Collapse
Affiliation(s)
- Chirag Gupta
- Waisman Center, University of Wisconsin-Madison, Madison, WI, 53705, USA
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Pramod Chandrashekar
- Waisman Center, University of Wisconsin-Madison, Madison, WI, 53705, USA
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Ting Jin
- Waisman Center, University of Wisconsin-Madison, Madison, WI, 53705, USA
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Chenfeng He
- Waisman Center, University of Wisconsin-Madison, Madison, WI, 53705, USA
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Saniya Khullar
- Waisman Center, University of Wisconsin-Madison, Madison, WI, 53705, USA
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Qiang Chang
- Waisman Center, University of Wisconsin-Madison, Madison, WI, 53705, USA
- Department of Medical Genetics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, 53705, USA
- Department of Neurology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Daifeng Wang
- Waisman Center, University of Wisconsin-Madison, Madison, WI, 53705, USA.
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, WI, 53706, USA.
- Department of Computer Sciences, University of Wisconsin-Madison, Madison, WI, 53706, USA.
| |
Collapse
|
27
|
Shoubridge AP, Choo JM, Martin AM, Keating DJ, Wong ML, Licinio J, Rogers GB. The gut microbiome and mental health: advances in research and emerging priorities. Mol Psychiatry 2022; 27:1908-1919. [PMID: 35236957 DOI: 10.1038/s41380-022-01479-w] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 01/17/2022] [Accepted: 02/08/2022] [Indexed: 12/19/2022]
Abstract
The gut microbiome exerts a considerable influence on human neurophysiology and mental health. Interactions between intestinal microbiology and host regulatory systems have now been implicated both in the development of psychiatric conditions and in the efficacy of many common therapies. With the growing acceptance of the role played by the gut microbiome in mental health outcomes, the focus of research is now beginning to shift from identifying relationships between intestinal microbiology and pathophysiology, and towards using this newfound insight to improve clinical outcomes. Here, we review recent advances in our understanding of gut microbiome-brain interactions, the mechanistic underpinnings of these relationships, and the ongoing challenge of distinguishing association and causation. We set out an overarching model of the evolution of microbiome-CNS interaction and examine how a growing knowledge of these complex systems can be used to determine disease susceptibility and reduce risk in a targeted manner.
Collapse
Affiliation(s)
- Andrew P Shoubridge
- Microbiome and Host Health, South Australian Health and Medical Research Institute, Adelaide, SA, 5001, Australia.,Infection and Immunity, Flinders Health and Medical Research Institute, College of Medicine and Public Health, Flinders University, Bedford Park, SA, 5042, Australia
| | - Jocelyn M Choo
- Microbiome and Host Health, South Australian Health and Medical Research Institute, Adelaide, SA, 5001, Australia.,Infection and Immunity, Flinders Health and Medical Research Institute, College of Medicine and Public Health, Flinders University, Bedford Park, SA, 5042, Australia
| | - Alyce M Martin
- Neuroscience, Flinders Health and Medical Research Institute, College of Medicine and Public Health, Flinders University, Bedford Park, SA, 5042, Australia
| | - Damien J Keating
- Neuroscience, Flinders Health and Medical Research Institute, College of Medicine and Public Health, Flinders University, Bedford Park, SA, 5042, Australia
| | - Ma-Li Wong
- Department of Psychiatry and Behavioral Sciences and Department of Neuroscience and Physiology, State University of New York Upstate Medical University, Syracuse, NY, 13210, USA
| | - Julio Licinio
- Department of Psychiatry and Behavioral Sciences and Department of Neuroscience and Physiology, State University of New York Upstate Medical University, Syracuse, NY, 13210, USA.,Department of Psychiatry, Flinders University College of Medicine and Public Health, Flinders University, Bedford Park, SA, 5042, Australia
| | - Geraint B Rogers
- Microbiome and Host Health, South Australian Health and Medical Research Institute, Adelaide, SA, 5001, Australia. .,Infection and Immunity, Flinders Health and Medical Research Institute, College of Medicine and Public Health, Flinders University, Bedford Park, SA, 5042, Australia.
| |
Collapse
|
28
|
Risk of migraine contributed by genetic polymorphisms of ANKDD1B gene: a case-control study based on Chinese Han population. Neurol Sci 2021; 43:2735-2743. [PMID: 34669083 DOI: 10.1007/s10072-021-05645-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Accepted: 09/29/2021] [Indexed: 10/20/2022]
Abstract
Early studies have indicated that the risk of migraine is contributed by both genetic and environmental factors. We aimed to evaluate the association between the risk of migraine and genetic polymorphisms in the ANKDD1B gene in a large sample of Chinese Han populations. A total of 882 patients with MO and 1,784 age-matched controls were recruited. A list of 12 tag SNPs located within the ANKDD1B gene region was genotyped. Distributions of SNP genotypes and alleles between patients and controls were examined to investigate the associations between the risk of migraine and genetic polymorphisms. The GTEx database was used to examine the effects of the significant SNPs on gene expressions. A stop-gain SNP, rs34358, was discovered to be significantly related with the risk of migraine (χ2 = 25.02, P = 5.66 × 10-7). The A allele of this SNP was significantly associated with a decreased risk of migraine (OR [95% CI] = 0.73 [0.65-0.83]). A dose-dependent pattern was identified in the genotypic analyses. The OR with 95% confidence interval for genotype AA versus GG was 0.55 [0.42-0.72], while for AG versus GG it was 0.74 [0.62-0.88]. Further bioinformatics analysis showed multiple significant signals (20 out of 47) for the association between SNP rs34358 and gene expression levels of ANKDD1B. In conclusion, we have provided population-based evidence for the association between genetic polymorphisms of the ANKDD1B gene and the risk of migraine. A protein-truncating variant was significantly associated with a decreased risk of migraine in the samples recruited from the Chinese Han population.
Collapse
|
29
|
Zhao SW, Xu X, Wang XY, Yan TC, Cao Y, Yan QH, Chen K, Jin YC, Zhang YH, Yin H, Cui LB. Shaping the Trans-Scale Properties of Schizophrenia via Cerebral Alterations on Magnetic Resonance Imaging and Single-Nucleotide Polymorphisms of Coding and Non-Coding Regions. Front Hum Neurosci 2021; 15:720239. [PMID: 34566604 PMCID: PMC8458928 DOI: 10.3389/fnhum.2021.720239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 08/05/2021] [Indexed: 11/13/2022] Open
Abstract
Schizophrenia is a complex mental illness with genetic heterogeneity, which is often accompanied by alterations in brain structure and function. The neurobiological mechanism of schizophrenia associated with heredity remains unknown. Recently, the development of trans-scale and multi-omics methods that integrate gene and imaging information sheds new light on the nature of schizophrenia. In this article, we summarized the results of brain structural and functional changes related to the specific single-nucleotide polymorphisms (SNPs) in the past decade, and the SNPs were divided into non-coding regions and coding regions, respectively. It is hoped that the relationship between SNPs and cerebral alterations can be displayed more clearly and intuitively, so as to provide fresh approaches for the discovery of potential biomarkers and the development of clinical accurate individualized treatment decision-making.
Collapse
Affiliation(s)
- Shu-Wan Zhao
- Department of Clinical Psychology, School of Medical Psychology, Fourth Military Medical University, Xi'an, China.,Department of Radiology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Xian Xu
- Department of Radiology, The Second Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Xian-Yang Wang
- Department of Clinical Psychology, School of Medical Psychology, Fourth Military Medical University, Xi'an, China
| | - Tian-Cai Yan
- Department of Clinical Psychology, School of Medical Psychology, Fourth Military Medical University, Xi'an, China
| | - Yang Cao
- Department of Clinical Psychology, School of Medical Psychology, Fourth Military Medical University, Xi'an, China
| | - Qing-Hong Yan
- Department of Psychiatry, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Kun Chen
- Department of Anatomy and K. K. Leung Brain Research Centre, Fourth Military Medical University, Xi'an, China
| | - Yin-Chuan Jin
- Department of Clinical Psychology, School of Medical Psychology, Fourth Military Medical University, Xi'an, China
| | - Ya-Hong Zhang
- Department of Psychiatry, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Hong Yin
- Department of Radiology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Long-Biao Cui
- Department of Clinical Psychology, School of Medical Psychology, Fourth Military Medical University, Xi'an, China.,Department of Radiology, The Second Medical Center, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|