1
|
Huang K, Tian J, Sun L, Hu H, Huang X, Zhou S, Deng A, Zhou Z, Jiang M, Li G, Xie P, Wang Y, Jiang X. TransGeneSelector: using a transformer approach to mine key genes from small transcriptomic datasets in plant responses to various environments. BMC Genomics 2025; 26:259. [PMID: 40098114 PMCID: PMC11912617 DOI: 10.1186/s12864-025-11434-y] [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: 06/18/2024] [Accepted: 03/04/2025] [Indexed: 03/19/2025] Open
Abstract
Gene mining is crucial for understanding the regulatory mechanisms underlying complex biological processes, particularly in plants responding to environmental conditions. Traditional machine learning methods, while useful, often overlook important gene relationships due to their reliance on manual feature selection and limited ability to capture complex inter-gene regulatory dynamics. Deep learning approaches, while powerful, are often unsuitable for small sample sizes. This study introduces TransGeneSelector, the first deep learning framework specifically designed for mining key genes from small transcriptomic datasets. By integrating a Wasserstein Generative Adversarial Network with Gradient Penalty (WGAN-GP) for sample generation and a Transformer-based network for classification, TransGeneSelector efficiently addresses the challenges of small-sample transcriptomic data, capturing both global gene regulatory interactions and specific biological processes. Evaluated in Arabidopsis thaliana, the model achieved high classification accuracy in predicting seed germination and heat stress conditions, outperforming traditional methods like Random Forest and Support Vector Machines (SVM). Moreover, Shapley Additive Explanations (SHAP) analysis and gene regulatory network construction revealed that TransGeneSelector effectively identified genes that appear to have upstream regulatory functions based on our analyses, enriching them in multiple key pathways which are critical for seed germination and heat stress response. RT-qPCR validation further confirmed the model's gene selection accuracy, demonstrating consistent expression patterns across varying germination conditions. The findings underscore the potential of TransGeneSelector as a robust tool for gene mining, offering deeper insights into gene regulation and organism adaptation under diverse environmental conditions. This work provides a framework that leverages deep learning for key gene identification in small transcriptomic datasets.
Collapse
Affiliation(s)
- Kerui Huang
- Key Laboratory of Agricultural Products Processing and Food Safety in Hunan Higher Education, Hunan University of Arts and Science, Changde, 415000, China
| | - Jianhong Tian
- College of Life Sciences, Hunan Normal University, Changsha, 410081, China
| | - Lei Sun
- Key Laboratory of Research and Utilization of Ethnomedicinal Plant Resources of Hunan Province, College of Biological and Food Engineering, Huaihua University, Huaihua, 418000, China
| | - Haoliang Hu
- Key Laboratory of Agricultural Products Processing and Food Safety in Hunan Higher Education, Hunan University of Arts and Science, Changde, 415000, China
| | - Xuebin Huang
- Key Laboratory of Agricultural Products Processing and Food Safety in Hunan Higher Education, Hunan University of Arts and Science, Changde, 415000, China
| | - Shiqi Zhou
- Rice Research Institute of Jiangxi Academy of Agricultural Sciences, Nanchang, 330000, China
| | - Aihua Deng
- Key Laboratory of Agricultural Products Processing and Food Safety in Hunan Higher Education, Hunan University of Arts and Science, Changde, 415000, China
| | - Zhibo Zhou
- Key Laboratory of Agricultural Products Processing and Food Safety in Hunan Higher Education, Hunan University of Arts and Science, Changde, 415000, China
| | - Ming Jiang
- Key Laboratory of Agricultural Products Processing and Food Safety in Hunan Higher Education, Hunan University of Arts and Science, Changde, 415000, China
| | - Guiwu Li
- College of Life Sciences, Hunan Normal University, Changsha, 410081, China
| | - Peng Xie
- Key Laboratory of Agricultural Products Processing and Food Safety in Hunan Higher Education, Hunan University of Arts and Science, Changde, 415000, China.
| | - Yun Wang
- Key Laboratory of Agricultural Products Processing and Food Safety in Hunan Higher Education, Hunan University of Arts and Science, Changde, 415000, China.
| | - Xiaocheng Jiang
- College of Life Sciences, Hunan Normal University, Changsha, 410081, China.
| |
Collapse
|
2
|
Nehmeh B, Rebehmed J, Nehmeh R, Taleb R, Akoury E. Unlocking therapeutic frontiers: harnessing artificial intelligence in drug discovery for neurodegenerative diseases. Drug Discov Today 2024; 29:104216. [PMID: 39428082 DOI: 10.1016/j.drudis.2024.104216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 10/05/2024] [Accepted: 10/15/2024] [Indexed: 10/22/2024]
Abstract
Neurodegenerative diseases (NDs) pose serious healthcare challenges with limited therapeutic treatments and high social burdens. The integration of artificial intelligence (AI) into drug discovery has emerged as a promising approach to address these challenges. This review explores the application of AI techniques to unravel therapeutic frontiers for NDs. We examine the current landscape of AI-driven drug discovery and discuss the potentials of AI in accelerating the identification of novel therapeutic targets on ND research and drug development, optimization of drug candidates, and expediating personalized medicine approaches. Finally, we outline future directions and challenges in harnessing AI for the advancement of therapeutics in this critical area by emphasizing the importance of interdisciplinary collaboration and ethical considerations.
Collapse
Affiliation(s)
- Bilal Nehmeh
- Department of Physical Sciences, Lebanese American University, Beirut 1102-2801, Lebanon
| | - Joseph Rebehmed
- Department of Computer Science and Mathematics, Lebanese American University, Beirut 1102-2801, Lebanon
| | - Riham Nehmeh
- INSA Rennes, Institut d'électronique et de Télécommunications de Rennes IETR, UMR 6164, 35708 Rennes, France
| | - Robin Taleb
- Department of Physical Sciences, Lebanese American University, Byblos Campus, Blat, 4M8F+6QF, Lebanon
| | - Elias Akoury
- Department of Physical Sciences, Lebanese American University, Beirut 1102-2801, Lebanon.
| |
Collapse
|
3
|
Gonzalez-Latapi P, Bustos B, Dong S, Lubbe S, Simuni T, Krainc D. Alterations in Blood Methylome as Potential Epigenetic Biomarker in Sporadic Parkinson's Disease. Ann Neurol 2024; 95:1162-1172. [PMID: 38563317 DOI: 10.1002/ana.26923] [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: 09/29/2023] [Revised: 02/03/2024] [Accepted: 02/19/2024] [Indexed: 04/04/2024]
Abstract
OBJECTIVE To characterize DNA methylation (DNAm) differences between sporadic Parkinson's disease (PD) and healthy control (HC) individuals enrolled in the Parkinson's Progression Markers Initiative (PPMI). METHODS Using whole blood, we characterized longitudinal differences in DNAm between sporadic PD patients (n = 196) and HCs (n = 86) enrolled in PPMI. RNA sequencing (RNAseq) was used to conduct gene expression analyses for genes mapped to differentially methylated cytosine-guanine sites (CpGs). RESULTS At the time of patient enrollment, 5,178 CpGs were differentially methylated (2,683 hypermethylated and 2,495 hypomethylated) in PD compared to HC. Of these, 579 CpGs underwent significant methylation changes over 3 years. Several differentially methylated CpGs were found near the cytochrome P450 family 2 subfamily E member 1 (CYP2E1) gene. Additionally, multiple hypermethylated CpGs were associated with the N-myc downregulated gene family member 4 (NDRG4) gene. RNA-Seq analyses showed 75 differentially expressed genes in PD patients compared to controls. An integrative analysis of both differentially methylated sites and differentially expressed genes revealed 20 genes that exhibited hypomethylation concomitant with overexpression. Additionally, 1 gene, cathepsin H (CTSH), displayed hypermethylation that was associated with its decreased expression. INTERPRETATION We provide initial evidence of alterations in DNAm in blood of PD patients that may serve as potential epigenetic biomarker of disease. To evaluate the significance of these changes throughout the progression of PD, additional profiling at longer intervals and during the prodromal stages of disease will be necessary. ANN NEUROL 2024;95:1162-1172.
Collapse
Affiliation(s)
- Paulina Gonzalez-Latapi
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Bernabe Bustos
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Siyuan Dong
- Biostatistics Collaboration Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Steven Lubbe
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Tanya Simuni
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Dimitri Krainc
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| |
Collapse
|
4
|
Li W, Wu H, Li J, Wang Z, Cai M, Liu X, Liu G. Transcriptomic analysis reveals associations of blood-based A-to-I editing with Parkinson's disease. J Neurol 2024; 271:976-985. [PMID: 37902879 DOI: 10.1007/s00415-023-12053-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 10/07/2023] [Accepted: 10/09/2023] [Indexed: 11/01/2023]
Abstract
BACKGROUND Adenosine-to-inosine (A-to-I) editing is the most common type of RNA editing in humans and the role of A-to-I RNA editing remains unclear in Parkinson's disease (PD). OBJECTIVE We aimed to explore the potential causal association between A-to-I editing and PD, and to assess whether changes in A-to-I editing were associated with cognitive progression in PD. METHODS The RNA-seq data from 380 PD patients and 178 healthy controls in the Parkinson's Progression Marker Initiative cohort was used to quantify A-to-I editing sites. We performed cis-RNA editing quantitative trait loci analysis and a two-sample Mendelian Randomization (MR) study by integrating genome-wide association studies to infer the potential causality between A-to-I editing and PD pathogenesis. The potential causal A-to-I editing sites were further confirmed by Summary-data-based MR analysis. Spearman's correlation analysis was performed to characterize the association between longitudinal A-to-I editing and cognitive progression in patients with PD. RESULTS We identified 17 potential causal A-to-I editing sites for PD and indicated that genetic risk variants may contribute to the risk of PD through A-to-I editing. These A-to-I editing sites were located in genes NCOR1, KANSL1 and BST1. Moreover, we observed 57 sites whose longitudinal A-to-I editing levels correlated with cognitive progression in PD. CONCLUSIONS We found potential causal A-to-I editing sites for PD onset and longitudinal changes of A-to-I editing were associated with cognitive progression in PD. We anticipate this study will provide new biological insights and drive the discovery of the epitranscriptomic role underlying Parkinson's disease.
Collapse
Affiliation(s)
- Weimin Li
- Shenzhen Key Laboratory of Systems Medicine in Inflammatory Diseases, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, No.66, Gongchang Road, Guangming District, Shenzhen, 518107, Guangdong, People's Republic of China
- Neurobiology Research Center, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, No.66, Gongchang Road, Guangming District, Shenzhen, 518107, Guangdong, People's Republic of China
| | - Hao Wu
- Shenzhen Key Laboratory of Systems Medicine in Inflammatory Diseases, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, No.66, Gongchang Road, Guangming District, Shenzhen, 518107, Guangdong, People's Republic of China
- Neurobiology Research Center, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, No.66, Gongchang Road, Guangming District, Shenzhen, 518107, Guangdong, People's Republic of China
| | - Jinxia Li
- Shenzhen Key Laboratory of Systems Medicine in Inflammatory Diseases, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, No.66, Gongchang Road, Guangming District, Shenzhen, 518107, Guangdong, People's Republic of China
- Neurobiology Research Center, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, No.66, Gongchang Road, Guangming District, Shenzhen, 518107, Guangdong, People's Republic of China
| | - Zhuo Wang
- Warshel Institute for Computational Biology, School of Medicine, The Chinese University of Hong Kong, Shenzhen, 518172, Guangdong, People's Republic of China
| | - Miao Cai
- Neurology Department, Zhejiang Hospital, Hangzhou, 310013, People's Republic of China
| | - Xiaoli Liu
- Neurology Department, Zhejiang Hospital, Hangzhou, 310013, People's Republic of China
| | - Ganqiang Liu
- Shenzhen Key Laboratory of Systems Medicine in Inflammatory Diseases, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, No.66, Gongchang Road, Guangming District, Shenzhen, 518107, Guangdong, People's Republic of China.
- Neurobiology Research Center, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, No.66, Gongchang Road, Guangming District, Shenzhen, 518107, Guangdong, People's Republic of China.
| |
Collapse
|
5
|
Larson KC, Martens LH, Marconi M, Dejesus C, Bruhn S, Miller TA, Tate B, Levenson JM. Preclinical translational platform of neuroinflammatory disease biology relevant to neurodegenerative disease. J Neuroinflammation 2024; 21:37. [PMID: 38297405 PMCID: PMC10832185 DOI: 10.1186/s12974-024-03029-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 01/23/2024] [Indexed: 02/02/2024] Open
Abstract
Neuroinflammation is a key driver of neurodegenerative disease, however the tools available to model this disease biology at the systems level are lacking. We describe a translational drug discovery platform based on organotypic culture of murine cortical brain slices that recapitulate disease-relevant neuroinflammatory biology. After an acute injury response, the brain slices assume a chronic neuroinflammatory state marked by transcriptomic profiles indicative of activation of microglia and astrocytes and loss of neuronal function. Microglia are necessary for manifestation of this neuroinflammation, as depletion of microglia prior to isolation of the brain slices prevents both activation of astrocytes and robust loss of synaptic function genes. The transcriptomic pattern of neuroinflammation in the mouse platform is present in published datasets derived from patients with amyotrophic lateral sclerosis, Huntington's disease, and frontotemporal dementia. Pharmacological utility of the platform was validated by demonstrating reversal of microglial activation and the overall transcriptomic signature with transforming growth factor-β. Additional anti-inflammatory targets were screened and inhibitors of glucocorticoid receptors, COX-2, dihydrofolate reductase, and NLRP3 inflammasome all failed to reverse the neuroinflammatory signature. Bioinformatics analysis of the neuroinflammatory signature identified protein tyrosine phosphatase non-receptor type 11 (PTPN11/SHP2) as a potential target. Three structurally distinct inhibitors of PTPN11 (RMC-4550, TN0155, IACS-13909) reversed the neuroinflammatory disease signature. Collectively, these results highlight the utility of this novel neuroinflammatory platform for facilitating identification and validation of targets for neuroinflammatory neurodegenerative disease drug discovery.
Collapse
Affiliation(s)
- Kelley C Larson
- Vigil Neuroscience, Watertown, USA
- Tiaki Therapeutics, Inc., c/o Dementia Discovery Fund, 201 Washington Street, 39th Floor, Boston, MA, 02108, USA
| | - Lauren H Martens
- , Neumora Therapeutics, Watertown, USA
- Tiaki Therapeutics, Inc., c/o Dementia Discovery Fund, 201 Washington Street, 39th Floor, Boston, MA, 02108, USA
| | - Michael Marconi
- Department of Molecular Pathology, Massachusetts General Hospital, Boston, USA
- Tiaki Therapeutics, Inc., c/o Dementia Discovery Fund, 201 Washington Street, 39th Floor, Boston, MA, 02108, USA
| | - Christopher Dejesus
- Atalanta Therapeutics, Boston, USA
- Tiaki Therapeutics, Inc., c/o Dementia Discovery Fund, 201 Washington Street, 39th Floor, Boston, MA, 02108, USA
| | - Suzanne Bruhn
- Charcot-Marie-Tooth Association, Glenolden, USA
- Tiaki Therapeutics, Inc., c/o Dementia Discovery Fund, 201 Washington Street, 39th Floor, Boston, MA, 02108, USA
| | - Thomas A Miller
- Walden Biosciences, Cambridge, USA
- Tiaki Therapeutics, Inc., c/o Dementia Discovery Fund, 201 Washington Street, 39th Floor, Boston, MA, 02108, USA
| | - Barbara Tate
- FARA, Homestead, USA
- Tiaki Therapeutics, Inc., c/o Dementia Discovery Fund, 201 Washington Street, 39th Floor, Boston, MA, 02108, USA
| | - Jonathan M Levenson
- FireCyte Therapeutics, Beverly, USA.
- Tiaki Therapeutics, Inc., c/o Dementia Discovery Fund, 201 Washington Street, 39th Floor, Boston, MA, 02108, USA.
| |
Collapse
|
6
|
Sopić M, Karaduzovic-Hadziabdic K, Kardassis D, Maegdefessel L, Martelli F, Meerson A, Munjas J, Niculescu LS, Stoll M, Magni P, Devaux Y, CardioRNA COST Action CA17129 and AtheroNET COST Action CA21153. Transcriptomic research in atherosclerosis: Unravelling plaque phenotype and overcoming methodological challenges. JOURNAL OF MOLECULAR AND CELLULAR CARDIOLOGY PLUS 2023; 6:100048. [PMID: 39802625 PMCID: PMC11708385 DOI: 10.1016/j.jmccpl.2023.100048] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 09/11/2023] [Indexed: 01/16/2025]
Abstract
Atherosclerotic disease is a major cause of acute cardiovascular events. A deeper understanding of its underlying mechanisms will allow advancing personalized and patient-centered healthcare. Transcriptomic research has proven to be a powerful tool for unravelling the complex molecular pathways that drive atherosclerosis. However, low reproducibility of research findings and lack of standardization of procedures pose significant challenges in this field. In this review, we discuss how transcriptomic research can help in understanding the different phenotypes of the atherosclerotic plaque that contribute to the development and progression of atherosclerosis. We highlight the methodological challenges that need to be addressed to improve research outputs, and emphasize the importance of research protocols harmonization. We also discuss recent advances in transcriptomic research, including bulk or single-cell sequencing, and their added value in plaque phenotyping. Finally, we explore how integrated multiomics data and machine learning improve understanding of atherosclerosis and provide directions for future research.
Collapse
Affiliation(s)
- Miron Sopić
- Department of Medical Biochemistry, Faculty of Pharmacy, University of Belgrade, Luxembourg Institute of Health, L-1445 Strassen, Luxembourg
- Cardiovascular Research Unit, Department of Population Health, Luxembourg Institute of Health, L-1445 Strassen, Luxembourg
| | - Kanita Karaduzovic-Hadziabdic
- Faculty of Engineering and Natural Science, Computer Science, International University of Sarajevo, Bosnia and Herzegovina
| | - Dimitris Kardassis
- Laboratory of Biochemistry, University of Crete Medical School and Gene Regulation and Epigenetics Group, Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology of Hellas, Heraklion 71003, Greece
| | - Lars Maegdefessel
- Institute of Molecular Vascular Medicine, Klinikum rechts der Isar, Technical University Munich, Germany
- German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Department of Medicine, Karolinska Institute, Stockholm, Sweden
| | - Fabio Martelli
- Molecular Cardiology Laboratory, IRCCS-Policlinico San Donato, via Morandi 30, 20097, San Donato Milanese, Milan, Italy
| | - Ari Meerson
- Molecular Biology of Chronic Diseases Laboratory, Genomic Center, Galilee Research Institute (MIGAL), Kiryat Shmona, Israel
- Faculty of Sciences, Tel Hai Academic College, Israel
| | - Jelena Munjas
- Department of Medical Biochemistry, Faculty of Pharmacy, University of Belgrade, Serbia
| | - Loredan S. Niculescu
- Lipidomics Department, Institute of Cellular Biology and Pathology “Nicolae Simionescu” of the Romanian Academy, 8, B.P. Hasdeu Street, Bucharest 050568, Romania
| | - Monika Stoll
- University of Münster, Institute of Hunan Genetics, Division of Genetic Epidemiology, Münster, Germany
- Maastricht University, Dept. of Biochemistry, Genetic Epidemiology and Statistical Genetics, Maastricht, NL
| | - Paolo Magni
- Department of Pharmacological and Biomolecular Sciences “Rodolfo Paoletti”, Università degli Studi di Milano, via Balzaretti 9, 20133, Milan, Italy
- IRCCS MultiMedica, via Milanese 300, 20099 Sesto S. Giovanni, Milan, Italy
| | - Yvan Devaux
- Cardiovascular Research Unit, Department of Population Health, Luxembourg Institute of Health, L-1445 Strassen, Luxembourg
| | | |
Collapse
|
7
|
Nazari E, Khalili-Tanha G, Asadnia A, Pourali G, Maftooh M, Khazaei M, Nasiri M, Hassanian SM, Ghayour-Mobarhan M, Ferns GA, Kiani MA, Avan A. Bioinformatics analysis and machine learning approach applied to the identification of novel key genes involved in non-alcoholic fatty liver disease. Sci Rep 2023; 13:20489. [PMID: 37993474 PMCID: PMC10665370 DOI: 10.1038/s41598-023-46711-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 11/03/2023] [Indexed: 11/24/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) comprises a range of chronic liver diseases that result from the accumulation of excess triglycerides in the liver, and which, in its early phases, is categorized NAFLD, or hepato-steatosis with pure fatty liver. The mortality rate of non-alcoholic steatohepatitis (NASH) is more than NAFLD; therefore, diagnosing the disease in its early stages may decrease liver damage and increase the survival rate. In the current study, we screened the gene expression data of NAFLD patients and control samples from the public dataset GEO to detect DEGs. Then, the correlation betweenbetween the top selected DEGs and clinical data was evaluated. In the present study, two GEO datasets (GSE48452, GSE126848) were downloaded. The dysregulated expressed genes (DEGs) were identified by machine learning methods (Penalize regression models). Then, the shared DEGs between the two training datasets were validated using validation datasets. ROC-curve analysis was used to identify diagnostic markers. R software analyzed the interactions between DEGs, clinical data, and fatty liver. Ten novel genes, including ABCF1, SART3, APC5, NONO, KAT7, ZPR1, RABGAP1, SLC7A8, SPAG9, and KAT6A were found to have a differential expression between NAFLD and healthy individuals. Based on validation results and ROC analysis, NR4A2 and IGFBP1b were identified as diagnostic markers. These key genes may be predictive markers for the development of fatty liver. It is recommended that these key genes are assessed further as possible predictive markers during the development of fatty liver.
Collapse
Affiliation(s)
- Elham Nazari
- Department of Health Information Technology and Management, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ghazaleh Khalili-Tanha
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Alireza Asadnia
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ghazaleh Pourali
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mina Maftooh
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Majid Khazaei
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammadreza Nasiri
- Recombinant Proteins Research Group, The Research Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Seyed Mahdi Hassanian
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Basic Sciences Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Majid Ghayour-Mobarhan
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Gordon A Ferns
- Division of Medical Education, Brighton & Sussex Medical School, Falmer, Brighton, BN1 9PH, Sussex, UK
| | - Mohammad Ali Kiani
- Department of Pediatrics, Ghaem Hospital, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir Avan
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
- College of Medicine, University of Warith Al-Anbiyaa, Karbala, Iraq.
- Faculty of Health, School of Biomedical Sciences, Queensland University of Technology (QUT), Brisbane, 4000, Australia.
| |
Collapse
|
8
|
Lai H, Li XY, Xu F, Zhu J, Li X, Song Y, Wang X, Wang Z, Wang C. Applications of Machine Learning to Diagnosis of Parkinson's Disease. Brain Sci 2023; 13:1546. [PMID: 38002506 PMCID: PMC10670005 DOI: 10.3390/brainsci13111546] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 10/28/2023] [Accepted: 10/31/2023] [Indexed: 11/26/2023] Open
Abstract
BACKGROUND Accurate diagnosis of Parkinson's disease (PD) is challenging due to its diverse manifestations. Machine learning (ML) algorithms can improve diagnostic precision, but their generalizability across medical centers in China is underexplored. OBJECTIVE To assess the accuracy of an ML algorithm for PD diagnosis, trained and tested on data from different medical centers in China. METHODS A total of 1656 participants were included, with 1028 from Beijing (training set) and 628 from Fuzhou (external validation set). Models were trained using the least absolute shrinkage and selection operator-logistic regression (LASSO-LR), decision tree (DT), random forest (RF), eXtreme gradient boosting (XGboost), support vector machine (SVM), and k-nearest neighbor (KNN) techniques. Hyperparameters were optimized using five-fold cross-validation and grid search techniques. Model performance was evaluated using the area under the curve (AUC) of the receiver operating characteristic (ROC) curve, accuracy, sensitivity (recall), specificity, precision, and F1 score. Variable importance was assessed for all models. RESULTS SVM demonstrated the best differentiation between healthy controls (HCs) and PD patients (AUC: 0.928, 95% CI: 0.908-0.947; accuracy: 0.844, 95% CI: 0.814-0.871; sensitivity: 0.826, 95% CI: 0.786-0.866; specificity: 0.861, 95% CI: 0.820-0.898; precision: 0.849, 95% CI: 0.807-0.891; F1 score: 0.837, 95% CI: 0.803-0.868) in the validation set. Constipation, olfactory decline, and daytime somnolence significantly influenced predictability. CONCLUSION We identified multiple pivotal variables and SVM as a precise and clinician-friendly ML algorithm for prediction of PD in Chinese patients.
Collapse
Affiliation(s)
- Hong Lai
- Department of Neurology, Xuanwu Hospital of Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing 100053, China; (H.L.); (X.-Y.L.); (F.X.); (J.Z.); (X.L.); (Y.S.); (X.W.); (Z.W.)
- Department of Neurology, The First Affiliated Hospital of Gannan Medical University, Ganzhou 341000, China
| | - Xu-Ying Li
- Department of Neurology, Xuanwu Hospital of Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing 100053, China; (H.L.); (X.-Y.L.); (F.X.); (J.Z.); (X.L.); (Y.S.); (X.W.); (Z.W.)
| | - Fanxi Xu
- Department of Neurology, Xuanwu Hospital of Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing 100053, China; (H.L.); (X.-Y.L.); (F.X.); (J.Z.); (X.L.); (Y.S.); (X.W.); (Z.W.)
| | - Junge Zhu
- Department of Neurology, Xuanwu Hospital of Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing 100053, China; (H.L.); (X.-Y.L.); (F.X.); (J.Z.); (X.L.); (Y.S.); (X.W.); (Z.W.)
| | - Xian Li
- Department of Neurology, Xuanwu Hospital of Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing 100053, China; (H.L.); (X.-Y.L.); (F.X.); (J.Z.); (X.L.); (Y.S.); (X.W.); (Z.W.)
| | - Yang Song
- Department of Neurology, Xuanwu Hospital of Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing 100053, China; (H.L.); (X.-Y.L.); (F.X.); (J.Z.); (X.L.); (Y.S.); (X.W.); (Z.W.)
| | - Xianlin Wang
- Department of Neurology, Xuanwu Hospital of Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing 100053, China; (H.L.); (X.-Y.L.); (F.X.); (J.Z.); (X.L.); (Y.S.); (X.W.); (Z.W.)
| | - Zhanjun Wang
- Department of Neurology, Xuanwu Hospital of Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing 100053, China; (H.L.); (X.-Y.L.); (F.X.); (J.Z.); (X.L.); (Y.S.); (X.W.); (Z.W.)
| | - Chaodong Wang
- Department of Neurology, Xuanwu Hospital of Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing 100053, China; (H.L.); (X.-Y.L.); (F.X.); (J.Z.); (X.L.); (Y.S.); (X.W.); (Z.W.)
| |
Collapse
|
9
|
Shusharina N, Yukhnenko D, Botman S, Sapunov V, Savinov V, Kamyshov G, Sayapin D, Voznyuk I. Modern Methods of Diagnostics and Treatment of Neurodegenerative Diseases and Depression. Diagnostics (Basel) 2023; 13:573. [PMID: 36766678 PMCID: PMC9914271 DOI: 10.3390/diagnostics13030573] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 01/27/2023] [Accepted: 01/29/2023] [Indexed: 02/09/2023] Open
Abstract
This paper discusses the promising areas of research into machine learning applications for the prevention and correction of neurodegenerative and depressive disorders. These two groups of disorders are among the leading causes of decline in the quality of life in the world when estimated using disability-adjusted years. Despite decades of research, the development of new approaches for the assessment (especially pre-clinical) and correction of neurodegenerative diseases and depressive disorders remains among the priority areas of research in neurophysiology, psychology, genetics, and interdisciplinary medicine. Contemporary machine learning technologies and medical data infrastructure create new research opportunities. However, reaching a consensus on the application of new machine learning methods and their integration with the existing standards of care and assessment is still a challenge to overcome before the innovations could be widely introduced to clinics. The research on the development of clinical predictions and classification algorithms contributes towards creating a unified approach to the use of growing clinical data. This unified approach should integrate the requirements of medical professionals, researchers, and governmental regulators. In the current paper, the current state of research into neurodegenerative and depressive disorders is presented.
Collapse
Affiliation(s)
- Natalia Shusharina
- Baltic Center for Neurotechnologies and Artificial Intelligence, Immanuel Kant Baltic Federal University, 236041 Kaliningrad, Russia
| | - Denis Yukhnenko
- Department of Social Security and Humanitarian Technologies, N. I. Lobachevsky State University of Nizhny Novgorod, 603022 Nizhny Novgorod, Russia
| | - Stepan Botman
- Baltic Center for Neurotechnologies and Artificial Intelligence, Immanuel Kant Baltic Federal University, 236041 Kaliningrad, Russia
| | - Viktor Sapunov
- Baltic Center for Neurotechnologies and Artificial Intelligence, Immanuel Kant Baltic Federal University, 236041 Kaliningrad, Russia
| | - Vladimir Savinov
- Baltic Center for Neurotechnologies and Artificial Intelligence, Immanuel Kant Baltic Federal University, 236041 Kaliningrad, Russia
| | - Gleb Kamyshov
- Baltic Center for Neurotechnologies and Artificial Intelligence, Immanuel Kant Baltic Federal University, 236041 Kaliningrad, Russia
| | - Dmitry Sayapin
- Baltic Center for Neurotechnologies and Artificial Intelligence, Immanuel Kant Baltic Federal University, 236041 Kaliningrad, Russia
| | - Igor Voznyuk
- Baltic Center for Neurotechnologies and Artificial Intelligence, Immanuel Kant Baltic Federal University, 236041 Kaliningrad, Russia
- Department of Neurology, Pavlov First Saint Petersburg State Medical University, 197022 Saint Petersburg, Russia
| |
Collapse
|
10
|
Harvey J, Reijnders RA, Cavill R, Duits A, Köhler S, Eijssen L, Rutten BPF, Shireby G, Torkamani A, Creese B, Leentjens AFG, Lunnon K, Pishva E. Machine learning-based prediction of cognitive outcomes in de novo Parkinson's disease. NPJ Parkinsons Dis 2022; 8:150. [PMID: 36344548 PMCID: PMC9640625 DOI: 10.1038/s41531-022-00409-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 10/11/2022] [Indexed: 11/09/2022] Open
Abstract
Cognitive impairment is a debilitating symptom in Parkinson's disease (PD). We aimed to establish an accurate multivariate machine learning (ML) model to predict cognitive outcome in newly diagnosed PD cases from the Parkinson's Progression Markers Initiative (PPMI). Annual cognitive assessments over an 8-year time span were used to define two cognitive outcomes of (i) cognitive impairment, and (ii) dementia conversion. Selected baseline variables were organized into three subsets of clinical, biofluid and genetic/epigenetic measures and tested using four different ML algorithms. Irrespective of the ML algorithm used, the models consisting of the clinical variables performed best and showed better prediction of cognitive impairment outcome over dementia conversion. We observed a marginal improvement in the prediction performance when clinical, biofluid, and epigenetic/genetic variables were all included in one model. Several cerebrospinal fluid measures and an epigenetic marker showed high predictive weighting in multiple models when included alongside clinical variables.
Collapse
Affiliation(s)
- Joshua Harvey
- Medical School, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| | - Rick A Reijnders
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), Maastricht University, Maastricht, The Netherlands
| | - Rachel Cavill
- Department of Advanced Computing Sciences, FSE, Maastricht University, Maastricht, The Netherlands
| | - Annelien Duits
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), Maastricht University, Maastricht, The Netherlands
- Department of Medical Psychology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Sebastian Köhler
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), Maastricht University, Maastricht, The Netherlands
| | - Lars Eijssen
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), Maastricht University, Maastricht, The Netherlands
- Department of Bioinformatics-BiGCaT, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, Maastricht, The Netherlands
| | - Bart P F Rutten
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), Maastricht University, Maastricht, The Netherlands
| | - Gemma Shireby
- Medical School, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| | - Ali Torkamani
- Department of Integrative Structural and Computational Biology, Scripps Research, La Jolla, CA, 92037, USA
| | - Byron Creese
- Medical School, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| | - Albert F G Leentjens
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), Maastricht University, Maastricht, The Netherlands
| | - Katie Lunnon
- Medical School, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| | - Ehsan Pishva
- Medical School, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK.
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), Maastricht University, Maastricht, The Netherlands.
| |
Collapse
|
11
|
Wang DD, Li YF, Zhang C, He SM, Chen X. Predicting the effect of sirolimus on disease activity in patients with systemic lupus erythematosus using machine learning. J Clin Pharm Ther 2022; 47:1845-1850. [PMID: 36131617 DOI: 10.1111/jcpt.13778] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 08/03/2022] [Accepted: 09/04/2022] [Indexed: 11/30/2022]
Abstract
WHAT IS KNOWN AND OBJECTIVES The present study aimed to predict the effect of sirolimus on disease activity in patients with systemic lupus erythematosus (SLE) using machine learning and to recommend appropriate sirolimus dosage regimen for patients with SLE. METHODS The Emax model was selected for machine learning, where the evaluation indicator was the change rate of systemic lupus erythematosus disease activity index from baseline value. RESULTS A total 103 patients with SLE were included for modelling, where the Emax , ET50 were -53.9%, 1.53 months in the final model respectively, and the evaluation of the final model was good. Further simulation found that the follow-up time to achieve 25%, 50%, 75% and 80% (plateau) Emax of sirolimus effecting on disease activity in patients with SLE were 0.51, 1.53, 4.59 and 6.12 months, respectively. In addition, the sirolimus dosage was flexible and adjusted according to drug concentration, where the intersection of sirolimus concentration range included in this study was about 8-10 ng/ml. WHAT IS NEW AND CONCLUSIONS This study was the first time to predict the effect of sirolimus on disease activity in patients with SLE and in order to achieve better therapeutic effect maintaining a concentration of 8-10 ng/ml sirolimus for at least 6.12 months was necessary.
Collapse
Affiliation(s)
- Dong-Dong Wang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, School of Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Ya-Feng Li
- Department of Pharmacy, Feng Xian People's Hospital, Xuzhou, Jiangsu, China
| | - Cun Zhang
- Department of Pharmacy, Xuzhou Oriental Hospital Affiliated to Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Su-Mei He
- Department of Pharmacy, Suzhou Science & Technology Town Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu, China
| | - Xiao Chen
- School of Nursing, Xuzhou Medical University, Xuzhou, Jiangsu, China
| |
Collapse
|
12
|
Suri JS, Maindarkar MA, Paul S, Ahluwalia P, Bhagawati M, Saba L, Faa G, Saxena S, Singh IM, Chadha PS, Turk M, Johri A, Khanna NN, Viskovic K, Mavrogeni S, Laird JR, Miner M, Sobel DW, Balestrieri A, Sfikakis PP, Tsoulfas G, Protogerou AD, Misra DP, Agarwal V, Kitas GD, Kolluri R, Teji JS, Al-Maini M, Dhanjil SK, Sockalingam M, Saxena A, Sharma A, Rathore V, Fatemi M, Alizad A, Krishnan PR, Omerzu T, Naidu S, Nicolaides A, Paraskevas KI, Kalra M, Ruzsa Z, Fouda MM. Deep Learning Paradigm for Cardiovascular Disease/Stroke Risk Stratification in Parkinson's Disease Affected by COVID-19: A Narrative Review. Diagnostics (Basel) 2022; 12:1543. [PMID: 35885449 PMCID: PMC9324237 DOI: 10.3390/diagnostics12071543] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Revised: 06/14/2022] [Accepted: 06/16/2022] [Indexed: 11/16/2022] Open
Abstract
Background and Motivation: Parkinson's disease (PD) is one of the most serious, non-curable, and expensive to treat. Recently, machine learning (ML) has shown to be able to predict cardiovascular/stroke risk in PD patients. The presence of COVID-19 causes the ML systems to become severely non-linear and poses challenges in cardiovascular/stroke risk stratification. Further, due to comorbidity, sample size constraints, and poor scientific and clinical validation techniques, there have been no well-explained ML paradigms. Deep neural networks are powerful learning machines that generalize non-linear conditions. This study presents a novel investigation of deep learning (DL) solutions for CVD/stroke risk prediction in PD patients affected by the COVID-19 framework. Method: The PRISMA search strategy was used for the selection of 292 studies closely associated with the effect of PD on CVD risk in the COVID-19 framework. We study the hypothesis that PD in the presence of COVID-19 can cause more harm to the heart and brain than in non-COVID-19 conditions. COVID-19 lung damage severity can be used as a covariate during DL training model designs. We, therefore, propose a DL model for the estimation of, (i) COVID-19 lesions in computed tomography (CT) scans and (ii) combining the covariates of PD, COVID-19 lesions, office and laboratory arterial atherosclerotic image-based biomarkers, and medicine usage for the PD patients for the design of DL point-based models for CVD/stroke risk stratification. Results: We validated the feasibility of CVD/stroke risk stratification in PD patients in the presence of a COVID-19 environment and this was also verified. DL architectures like long short-term memory (LSTM), and recurrent neural network (RNN) were studied for CVD/stroke risk stratification showing powerful designs. Lastly, we examined the artificial intelligence bias and provided recommendations for early detection of CVD/stroke in PD patients in the presence of COVID-19. Conclusion: The DL is a very powerful tool for predicting CVD/stroke risk in PD patients affected by COVID-19.
Collapse
Affiliation(s)
- Jasjit S. Suri
- Stroke Monitoring and Diagnostic Division, AtheroPoint™, Roseville, CA 95661, USA; (M.A.M.); (I.M.S.); (P.S.C.); (S.K.D.)
| | - Mahesh A. Maindarkar
- Stroke Monitoring and Diagnostic Division, AtheroPoint™, Roseville, CA 95661, USA; (M.A.M.); (I.M.S.); (P.S.C.); (S.K.D.)
- Department of Biomedical Engineering, North Eastern Hill University, Shillong 793022, India; (S.P.); (M.B.)
| | - Sudip Paul
- Department of Biomedical Engineering, North Eastern Hill University, Shillong 793022, India; (S.P.); (M.B.)
| | - Puneet Ahluwalia
- Max Institute of Cancer Care, Max Super Specialty Hospital, New Delhi 110017, India;
| | - Mrinalini Bhagawati
- Department of Biomedical Engineering, North Eastern Hill University, Shillong 793022, India; (S.P.); (M.B.)
| | - Luca Saba
- Department of Radiology, and Pathology, Azienda Ospedaliero Universitaria, 09123 Cagliari, Italy; (L.S.); (G.F.)
| | - Gavino Faa
- Department of Radiology, and Pathology, Azienda Ospedaliero Universitaria, 09123 Cagliari, Italy; (L.S.); (G.F.)
| | - Sanjay Saxena
- Department of CSE, International Institute of Information Technology, Bhuneshwar 751029, India;
| | - Inder M. Singh
- Stroke Monitoring and Diagnostic Division, AtheroPoint™, Roseville, CA 95661, USA; (M.A.M.); (I.M.S.); (P.S.C.); (S.K.D.)
| | - Paramjit S. Chadha
- Stroke Monitoring and Diagnostic Division, AtheroPoint™, Roseville, CA 95661, USA; (M.A.M.); (I.M.S.); (P.S.C.); (S.K.D.)
| | - Monika Turk
- Department of Neurology, University Medical Centre Maribor, 2000 Maribor, Slovenia; (M.T.); (T.O.)
| | - Amer Johri
- Department of Medicine, Division of Cardiology, Queen’s University, Kingston, ON K7L 3N6, Canada;
| | - Narendra N. Khanna
- Department of Cardiology, Indraprastha APOLLO Hospitals, New Delhi 110076, India; (N.N.K.); (A.S.)
| | - Klaudija Viskovic
- Department of Radiology and Ultrasound, University Hospital for Infectious Diseases, 10000 Zagreb, Croatia;
| | - Sofia Mavrogeni
- Cardiology Clinic, Onassis Cardiac Surgery Centre, 176 74 Athens, Greece;
| | - John R. Laird
- Heart and Vascular Institute, Adventist Health St. Helena, St. Helena, CA 94574, USA;
| | - Martin Miner
- Men’s Health Centre, Miriam Hospital, Providence, RI 02906, USA;
| | - David W. Sobel
- Rheumatology Unit, National Kapodistrian University of Athens, 157 72 Athens, Greece; (D.W.S.); (P.P.S.)
| | | | - Petros P. Sfikakis
- Rheumatology Unit, National Kapodistrian University of Athens, 157 72 Athens, Greece; (D.W.S.); (P.P.S.)
| | - George Tsoulfas
- Department of Surgery, Aristoteleion University of Thessaloniki, 541 24 Thessaloniki, Greece;
| | - Athanase D. Protogerou
- Cardiovascular Prevention and Research Unit, Department of Pathophysiology, National & Kapodistrian University of Athens, 157 72 Athens, Greece;
| | - Durga Prasanna Misra
- Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow 226014, India; (D.P.M.); (V.A.)
| | - Vikas Agarwal
- Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow 226014, India; (D.P.M.); (V.A.)
| | - George D. Kitas
- Academic Affairs, Dudley Group NHS Foundation Trust, Dudley DY1 2HQ, UK;
- Arthritis Research UK Epidemiology Unit, Manchester University, Manchester M13 9PL, UK
| | - Raghu Kolluri
- OhioHealth Heart and Vascular, Mansfield, OH 44905, USA;
| | - Jagjit S. Teji
- Ann and Robert H. Lurie Children’s Hospital of Chicago, Chicago, IL 60611, USA;
| | - Mustafa Al-Maini
- Allergy, Clinical Immunology, and Rheumatology Institute, Toronto, ON M5G 1N8, Canada;
| | - Surinder K. Dhanjil
- Stroke Monitoring and Diagnostic Division, AtheroPoint™, Roseville, CA 95661, USA; (M.A.M.); (I.M.S.); (P.S.C.); (S.K.D.)
| | | | - Ajit Saxena
- Department of Cardiology, Indraprastha APOLLO Hospitals, New Delhi 110076, India; (N.N.K.); (A.S.)
| | - Aditya Sharma
- Division of Cardiovascular Medicine, University of Virginia, Charlottesville, VA 22908, USA;
| | - Vijay Rathore
- Nephrology Department, Kaiser Permanente, Sacramento, CA 95823, USA;
| | - Mostafa Fatemi
- Department of Physiology & Biomedical Engineering, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA;
| | - Azra Alizad
- Department of Radiology, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA;
| | | | - Tomaz Omerzu
- Department of Neurology, University Medical Centre Maribor, 2000 Maribor, Slovenia; (M.T.); (T.O.)
| | - Subbaram Naidu
- Electrical Engineering Department, University of Minnesota, Duluth, MN 55812, USA;
| | - Andrew Nicolaides
- Vascular Screening and Diagnostic Centre, University of Nicosia Medical School, Engomi 2408, Cyprus;
| | - Kosmas I. Paraskevas
- Department of Vascular Surgery, Central Clinic of Athens, 106 80 Athens, Greece;
| | - Mannudeep Kalra
- Department of Radiology, Harvard Medical School, Boston, MA 02115, USA;
| | - Zoltán Ruzsa
- Invasive Cardiology Division, Faculty of Medicine, University of Szeged, 6720 Szeged, Hungary;
| | - Mostafa M. Fouda
- Department of Electrical and Computer Engineering, Idaho State University, Pocatello, ID 83209, USA;
| |
Collapse
|
13
|
Chiricosta L, D’Angiolini S, Gugliandolo A, Mazzon E. Artificial Intelligence Predictor for Alzheimer’s Disease Trained on Blood Transcriptome: The Role of Oxidative Stress. Int J Mol Sci 2022; 23:ijms23095237. [PMID: 35563628 PMCID: PMC9104709 DOI: 10.3390/ijms23095237] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 05/04/2022] [Accepted: 05/05/2022] [Indexed: 02/01/2023] Open
Abstract
Alzheimer’s disease (AD) is an incurable neurodegenerative disease diagnosed by clinicians through healthcare records and neuroimaging techniques. These methods lack sensitivity and specificity, so new antemortem non-invasive strategies to diagnose AD are needed. Herein, we designed a machine learning predictor based on transcriptomic data obtained from the blood of AD patients and individuals without dementia (non-AD) through an 8 × 60 K microarray. The dataset was used to train different models with different hyperparameters. The support vector machines method allowed us to reach a Receiver Operating Characteristic score of 93% and an accuracy of 89%. High score levels were also achieved by the neural network and logistic regression methods. Furthermore, the Gene Ontology enrichment analysis of the features selected to train the model along with the genes differentially expressed between the non-AD and AD transcriptomic profiles shows the “mitochondrial translation” biological process to be the most interesting. In addition, inspection of the KEGG pathways suggests that the accumulation of β-amyloid triggers electron transport chain impairment, enhancement of reactive oxygen species and endoplasmic reticulum stress. Taken together, all these elements suggest that the oxidative stress induced by β-amyloid is a key feature trained by the model for the prediction of AD with high accuracy.
Collapse
|
14
|
Pantaleo E, Monaco A, Amoroso N, Lombardi A, Bellantuono L, Urso D, Lo Giudice C, Picardi E, Tafuri B, Nigro S, Pesole G, Tangaro S, Logroscino G, Bellotti R. A Machine Learning Approach to Parkinson’s Disease Blood Transcriptomics. Genes (Basel) 2022; 13:genes13050727. [PMID: 35627112 PMCID: PMC9141063 DOI: 10.3390/genes13050727] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Revised: 04/16/2022] [Accepted: 04/18/2022] [Indexed: 12/23/2022] Open
Abstract
The increased incidence and the significant health burden associated with Parkinson’s disease (PD) have stimulated substantial research efforts towards the identification of effective treatments and diagnostic procedures. Despite technological advancements, a cure is still not available and PD is often diagnosed a long time after onset when irreversible damage has already occurred. Blood transcriptomics represents a potentially disruptive technology for the early diagnosis of PD. We used transcriptome data from the PPMI study, a large cohort study with early PD subjects and age matched controls (HC), to perform the classification of PD vs. HC in around 550 samples. Using a nested feature selection procedure based on Random Forests and XGBoost we reached an AUC of 72% and found 493 candidate genes. We further discussed the importance of the selected genes through a functional analysis based on GOs and KEGG pathways.
Collapse
Affiliation(s)
- Ester Pantaleo
- Istituto Nazionale di Fisica Nucleare (INFN), Sezione di Bari, Via A. Orabona 4, 70125 Bari, Italy; (E.P.); (A.M.); (N.A.); (L.B.); (S.T.); (R.B.)
- Dipartimento di Scienze Mediche di Base, Neuroscienze e Organi di Senso, Università degli Studi di Bari Aldo Moro, Piazza G. Cesare 11, 70124 Bari, Italy;
- Dipartimento Interateneo di Fisica M. Merlin, Università degli Studi di Bari Aldo Moro, Via G. Amendola 173, 70125 Bari, Italy
| | - Alfonso Monaco
- Istituto Nazionale di Fisica Nucleare (INFN), Sezione di Bari, Via A. Orabona 4, 70125 Bari, Italy; (E.P.); (A.M.); (N.A.); (L.B.); (S.T.); (R.B.)
| | - Nicola Amoroso
- Istituto Nazionale di Fisica Nucleare (INFN), Sezione di Bari, Via A. Orabona 4, 70125 Bari, Italy; (E.P.); (A.M.); (N.A.); (L.B.); (S.T.); (R.B.)
- Dipartimento di Farmacia-Scienze del Farmaco, Università degli Studi di Bari Aldo Moro, Via A. Orabona 4, 70125 Bari, Italy
| | - Angela Lombardi
- Istituto Nazionale di Fisica Nucleare (INFN), Sezione di Bari, Via A. Orabona 4, 70125 Bari, Italy; (E.P.); (A.M.); (N.A.); (L.B.); (S.T.); (R.B.)
- Dipartimento Interateneo di Fisica M. Merlin, Università degli Studi di Bari Aldo Moro, Via G. Amendola 173, 70125 Bari, Italy
- Correspondence:
| | - Loredana Bellantuono
- Istituto Nazionale di Fisica Nucleare (INFN), Sezione di Bari, Via A. Orabona 4, 70125 Bari, Italy; (E.P.); (A.M.); (N.A.); (L.B.); (S.T.); (R.B.)
- Dipartimento di Scienze Mediche di Base, Neuroscienze e Organi di Senso, Università degli Studi di Bari Aldo Moro, Piazza G. Cesare 11, 70124 Bari, Italy;
| | - Daniele Urso
- Centro per le Malattie Neurodegenerative e l’Invecchiamento Cerebrale, Dipartimento di Ricerca Clinica in Neurologia, Università degli Studi di Bari Aldo Moro, Pia Fondazione Cardinale G. Panico, 73039 Tricase, Italy; (D.U.); (B.T.); (S.N.)
- Institute of Psychiatry, Psychology and Neuroscience, King’s College London, De Crespigny Park, London SE5 8AF, UK
| | - Claudio Lo Giudice
- Dipartimento di Bioscienze, Biotecnologie e Biofarmaceutica, Università degli Studi di Bari Aldo Moro, Via A. Orabona 4, 70125 Bari, Italy; (C.L.G.); (E.P.); (G.P.)
| | - Ernesto Picardi
- Dipartimento di Bioscienze, Biotecnologie e Biofarmaceutica, Università degli Studi di Bari Aldo Moro, Via A. Orabona 4, 70125 Bari, Italy; (C.L.G.); (E.P.); (G.P.)
- Istituto di Biomembrane, Bioenergetica e Biotecnologie Molecolari, Consiglio Nazionale delle Ricerche, Via G. Amendola 122/O, 70126 Bari, Italy
| | - Benedetta Tafuri
- Centro per le Malattie Neurodegenerative e l’Invecchiamento Cerebrale, Dipartimento di Ricerca Clinica in Neurologia, Università degli Studi di Bari Aldo Moro, Pia Fondazione Cardinale G. Panico, 73039 Tricase, Italy; (D.U.); (B.T.); (S.N.)
| | - Salvatore Nigro
- Centro per le Malattie Neurodegenerative e l’Invecchiamento Cerebrale, Dipartimento di Ricerca Clinica in Neurologia, Università degli Studi di Bari Aldo Moro, Pia Fondazione Cardinale G. Panico, 73039 Tricase, Italy; (D.U.); (B.T.); (S.N.)
- Istituto di Nanotecnologia (NANOTEC), Consiglio Nazionale delle Ricerche, Via Monteroni, 73100 Lecce, Italy
| | - Graziano Pesole
- Dipartimento di Bioscienze, Biotecnologie e Biofarmaceutica, Università degli Studi di Bari Aldo Moro, Via A. Orabona 4, 70125 Bari, Italy; (C.L.G.); (E.P.); (G.P.)
- Istituto di Biomembrane, Bioenergetica e Biotecnologie Molecolari, Consiglio Nazionale delle Ricerche, Via G. Amendola 122/O, 70126 Bari, Italy
| | - Sabina Tangaro
- Istituto Nazionale di Fisica Nucleare (INFN), Sezione di Bari, Via A. Orabona 4, 70125 Bari, Italy; (E.P.); (A.M.); (N.A.); (L.B.); (S.T.); (R.B.)
- Dipartimento di Scienze del Suolo, della Pianta e degli Alimenti, Università degli Studi di Bari Aldo Moro, Via A. Orabona 4, 70125 Bari, Italy
| | - Giancarlo Logroscino
- Dipartimento di Scienze Mediche di Base, Neuroscienze e Organi di Senso, Università degli Studi di Bari Aldo Moro, Piazza G. Cesare 11, 70124 Bari, Italy;
- Centro per le Malattie Neurodegenerative e l’Invecchiamento Cerebrale, Dipartimento di Ricerca Clinica in Neurologia, Università degli Studi di Bari Aldo Moro, Pia Fondazione Cardinale G. Panico, 73039 Tricase, Italy; (D.U.); (B.T.); (S.N.)
| | - Roberto Bellotti
- Istituto Nazionale di Fisica Nucleare (INFN), Sezione di Bari, Via A. Orabona 4, 70125 Bari, Italy; (E.P.); (A.M.); (N.A.); (L.B.); (S.T.); (R.B.)
- Dipartimento Interateneo di Fisica M. Merlin, Università degli Studi di Bari Aldo Moro, Via G. Amendola 173, 70125 Bari, Italy
| |
Collapse
|
15
|
Paul S, Maindarkar M, Saxena S, Saba L, Turk M, Kalra M, Krishnan PR, Suri JS. Bias Investigation in Artificial Intelligence Systems for Early Detection of Parkinson's Disease: A Narrative Review. Diagnostics (Basel) 2022; 12:166. [PMID: 35054333 PMCID: PMC8774851 DOI: 10.3390/diagnostics12010166] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 12/27/2021] [Accepted: 01/01/2022] [Indexed: 12/13/2022] Open
Abstract
Background and Motivation: Diagnosis of Parkinson's disease (PD) is often based on medical attention and clinical signs. It is subjective and does not have a good prognosis. Artificial Intelligence (AI) has played a promising role in the diagnosis of PD. However, it introduces bias due to lack of sample size, poor validation, clinical evaluation, and lack of big data configuration. The purpose of this study is to compute the risk of bias (RoB) automatically. METHOD The PRISMA search strategy was adopted to select the best 39 AI studies out of 85 PD studies closely associated with early diagnosis PD. The studies were used to compute 30 AI attributes (based on 6 AI clusters), using AP(ai)Bias 1.0 (AtheroPointTM, Roseville, CA, USA), and the mean aggregate score was computed. The studies were ranked and two cutoffs (Moderate-Low (ML) and High-Moderate (MH)) were determined to segregate the studies into three bins: low-, moderate-, and high-bias. RESULT The ML and HM cutoffs were 3.50 and 2.33, respectively, which constituted 7, 13, and 6 for low-, moderate-, and high-bias studies. The best and worst architectures were "deep learning with sketches as outcomes" and "machine learning with Electroencephalography," respectively. We recommend (i) the usage of power analysis in big data framework, (ii) that it must undergo scientific validation using unseen AI models, and (iii) that it should be taken towards clinical evaluation for reliability and stability tests. CONCLUSION The AI is a vital component for the diagnosis of early PD and the recommendations must be followed to lower the RoB.
Collapse
Affiliation(s)
- Sudip Paul
- Department of Biomedical Engineering, North Eastern Hill University, Shillong 793022, India
| | - Maheshrao Maindarkar
- Department of Biomedical Engineering, North Eastern Hill University, Shillong 793022, India
| | - Sanjay Saxena
- Department of CSE, International Institute of Information Technology, Bhuneshwar 751003, India
| | - Luca Saba
- Department of Radiology, University of Cagliari, 09121 Cagliari, Italy
| | - Monika Turk
- Department of Neurology, University Medical Centre Maribor, 1262 Maribor, Slovenia
| | - Manudeep Kalra
- Department of Radiology, Harvard Medical School, Boston, MA 02115, USA
| | | | - Jasjit S Suri
- Stroke Monitoring and Diagnostic Division, AtheroPoint™, Roseville, CA 95661, USA
| |
Collapse
|
16
|
Machine Learning and Intracranial Aneurysms: From Detection to Outcome Prediction. ACTA NEUROCHIRURGICA. SUPPLEMENT 2021; 134:319-331. [PMID: 34862556 DOI: 10.1007/978-3-030-85292-4_36] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Machine learning (ML) is a rapidly rising research tool in biomedical sciences whose applications include segmentation, classification, disease detection, and outcome prediction. With respect to traditional statistical methods, ML algorithms have the potential to learn and improve their predictive performance when fed with large data sets without the need of being specifically programmed. In recent years, this technology has been increasingly applied for tackling clinical issues in intracranial aneurysm (IA) research. Several studies attempted to provide reliable models for enhanced aneurysm detection. Convolutional neural networks trained with variable degrees of human interaction on data from diverse imaging modalities showed high sensitivity in aneurysm detection tasks, also outperforming expert image analysis. Algorithms were also shown to differentiate ruptured from unruptured IAs, with however limited clinical relevance. For prediction of rupture and stability assessment, ML was preliminarily shown to achieve better performance compared to conventional statistical methods and existing risk scores. ML-based complication and functional outcome prediction in the event of SAH have been more extensively reported, in contrast with periprocedural outcome investigation in unruptured IA patients. ML has the potential to be a game changer in IA patient management. Currently clinical translation of experimental results is limited.
Collapse
|
17
|
Li R, Li L, Xu Y, Yang J. Machine learning meets omics: applications and perspectives. Brief Bioinform 2021; 23:6425809. [PMID: 34791021 DOI: 10.1093/bib/bbab460] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 09/29/2021] [Accepted: 10/07/2021] [Indexed: 02/07/2023] Open
Abstract
The innovation of biotechnologies has allowed the accumulation of omics data at an alarming rate, thus introducing the era of 'big data'. Extracting inherent valuable knowledge from various omics data remains a daunting problem in bioinformatics. Better solutions often need some kind of more innovative methods for efficient handlings and effective results. Recent advancements in integrated analysis and computational modeling of multi-omics data helped address such needs in an increasingly harmonious manner. The development and application of machine learning have largely advanced our insights into biology and biomedicine and greatly promoted the development of therapeutic strategies, especially for precision medicine. Here, we propose a comprehensive survey and discussion on what happened, is happening and will happen when machine learning meets omics. Specifically, we describe how artificial intelligence can be applied to omics studies and review recent advancements at the interface between machine learning and the ever-widest range of omics including genomics, transcriptomics, proteomics, metabolomics, radiomics, as well as those at the single-cell resolution. We also discuss and provide a synthesis of ideas, new insights, current challenges and perspectives of machine learning in omics.
Collapse
Affiliation(s)
- Rufeng Li
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an 710061, P. R. China
| | - Lixin Li
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an 710061, P. R. China
| | - Yungang Xu
- School of Electronics and Information, Northwestern Polytechnical University, Xi'an, 710129, China
| | - Juan Yang
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an 710061, P. R. China.,Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education of China, Xi'an 710061, P. R. China
| |
Collapse
|
18
|
Rodrigues J, Amin A, Raghushaker CR, Chandra S, Joshi MB, Prasad K, Rai S, Nayak SG, Ray S, Mahato KK. Exploring photoacoustic spectroscopy-based machine learning together with metabolomics to assess breast tumor progression in a xenograft model ex vivo. J Transl Med 2021; 101:952-965. [PMID: 33875792 PMCID: PMC8214996 DOI: 10.1038/s41374-021-00597-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 03/06/2021] [Accepted: 03/06/2021] [Indexed: 12/24/2022] Open
Abstract
In the current study, a breast tumor xenograft was established in athymic nude mice by subcutaneous injection of the MCF-7 cell line and assessed the tumor progression by photoacoustic spectroscopy combined with machine learning tools. The advancement of breast tumors in nude mice was validated by tumor volume kinetics and histopathology and corresponding image analysis by TissueQuant software compared to controls. The ex vivo tumors in progressive conditions belonging to time points, day 5th, 10th, 15th & 20th, were excited with 281 nm pulsed laser light and recorded the corresponding photoacoustic spectra in time domain. The spectra were then pre-processed, augmented for a 10-fold increase in the data strength, and subjected to wavelet packet transformation for feature extraction and selection using MATLAB software. In the present study, the top 10 features from all the time point groups under study were selected based on their prediction ranking values using the mRMR algorithm. The chosen features of all the time-point groups were then subjected to multi-class Support Vector Machine (SVM) algorithms for learning and classifying into respective time point groups under study. The analysis demonstrated accuracy values of 95.2%, 99.5%, and 80.3% with SVM- Radial Basis Function (SVM-RBF), SVM-Polynomial & SVM-Linear, respectively. The serum metabolomic levels during tumor progression complemented photoacoustic patterns of tumor progression, depicting breast cancer pathophysiology.
Collapse
Affiliation(s)
- Jackson Rodrigues
- Department of Biophysics, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Ashwini Amin
- Department of Electronics & Communication Engineering, Manipal Institute of Technology, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | | | - Subhash Chandra
- Department of Biophysics, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Manjunath B Joshi
- Department of Ageing Research, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Keerthana Prasad
- Manipal School of Information Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Sharada Rai
- Department of Pathology, Kasturba Medical College, Mangalore, Manipal Academy of Higher Education, Mangalore, Karnataka, India
| | - Subramanya G Nayak
- Department of Electronics & Communication Engineering, Manipal Institute of Technology, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Satadru Ray
- Department of Surgery, Kasturba Medical College, Mangalore, Manipal Academy of Higher Education, Mangalore, Karnataka, India
| | - Krishna Kishore Mahato
- Department of Biophysics, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India.
| |
Collapse
|
19
|
Lake J, Storm CS, Makarious MB, Bandres-Ciga S. Genetic and Transcriptomic Biomarkers in Neurodegenerative Diseases: Current Situation and the Road Ahead. Cells 2021; 10:1030. [PMID: 33925602 PMCID: PMC8170880 DOI: 10.3390/cells10051030] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 04/21/2021] [Accepted: 04/24/2021] [Indexed: 12/19/2022] Open
Abstract
Neurodegenerative diseases are etiologically and clinically heterogeneous conditions, often reflecting a spectrum of disease rather than well-defined disorders. The underlying molecular complexity of these diseases has made the discovery and validation of useful biomarkers challenging. The search of characteristic genetic and transcriptomic indicators for preclinical disease diagnosis, prognosis, or subtyping is an area of ongoing effort and interest. The next generation of biomarker studies holds promise by implementing meaningful longitudinal and multi-modal approaches in large scale biobank and healthcare system scale datasets. This work will only be possible in an open science framework. This review summarizes the current state of genetic and transcriptomic biomarkers in Parkinson's disease, Alzheimer's disease, and amyotrophic lateral sclerosis, providing a comprehensive landscape of recent literature and future directions.
Collapse
Affiliation(s)
- Julie Lake
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA; (J.L.); (M.B.M.)
| | - Catherine S. Storm
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London WC1N 3BG, UK;
- UCL Movement Disorders Centre, University College London, London WC1E 6BT, UK
| | - Mary B. Makarious
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA; (J.L.); (M.B.M.)
| | - Sara Bandres-Ciga
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA; (J.L.); (M.B.M.)
| |
Collapse
|