1
|
Gupta R, Bhandari M, Grover A, Al-Shehari T, Kadrie M, Alfakih T, Alsalman H. Predictive modeling of ALS progression: an XGBoost approach using clinical features. BioData Min 2024; 17:54. [PMID: 39623504 PMCID: PMC11610297 DOI: 10.1186/s13040-024-00399-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 10/11/2024] [Indexed: 12/06/2024] Open
Abstract
This research presents a predictive model aimed at estimating the progression of Amyotrophic Lateral Sclerosis (ALS) based on clinical features collected from a dataset of 50 patients. Important features included evaluations of speech, mobility, and respiratory function. We utilized an XGBoost regression model to forecast scores on the ALS Functional Rating Scale (ALSFRS-R), achieving a training mean squared error (MSE) of 0.1651 and a testing MSE of 0.0073, with R² values of 0.9800 for training and 0.9993 for testing. The model demonstrates high accuracy, providing a useful tool for clinicians to track disease progression and enhance patient management and treatment strategies.
Collapse
Affiliation(s)
- Richa Gupta
- Department of Computer Science and Engineering, School of Engineering Sciences and Technology, Jamia Hamdard, Delhi, India.
| | - Mansi Bhandari
- Department of Computer Science and Engineering, School of Engineering Sciences and Technology, Jamia Hamdard, Delhi, India
| | - Anhad Grover
- Department of Computer Science and Engineering, School of Engineering Sciences and Technology, Jamia Hamdard, Delhi, India
| | - Taher Al-Shehari
- Computer Skills, Department of Self-Development Skill, Common First Year Deanship, King Saud University, Riyadhi, 11362, Saudi Arabia
| | - Mohammed Kadrie
- Computer Skills, Department of Self-Development Skill, Common First Year Deanship, King Saud University, Riyadhi, 11362, Saudi Arabia
| | - Taha Alfakih
- Department of Information Systems, College of Computer and Information Sciences, King Saud University, Riyadh, 11543, Saudi Arabia
| | - Hussain Alsalman
- Department of Computer Science, College of Computer and Information Sciences, King Saud University, Riyadh, 11543, Saudi Arabia
| |
Collapse
|
2
|
Kew SYN, Mok SY, Goh CH. Machine learning and brain-computer interface approaches in prognosis and individualized care strategies for individuals with amyotrophic lateral sclerosis: A systematic review. MethodsX 2024; 13:102765. [PMID: 39286440 PMCID: PMC11403252 DOI: 10.1016/j.mex.2024.102765] [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: 04/16/2024] [Accepted: 05/15/2024] [Indexed: 09/19/2024] Open
Abstract
Amyotrophic lateral sclerosis (ALS) characterized by progressive degeneration of motor neurons is a debilitating disease, posing substantial challenges in both prognosis and daily life assistance. However, with the advancement of machine learning (ML) which is renowned for tackling many real-world settings, it can offer unprecedented opportunities in prognostic studies and facilitate individuals with ALS in motor-imagery tasks. ML models, such as random forests (RF), have emerged as the most common and effective algorithms for predicting disease progression and survival time in ALS. The findings revealed that RF models had an excellent predictive performance for ALS, with a testing R2 of 0.524 and minimal treatment effects of 0.0717 for patient survival time. Despite significant limitations in sample size, with a maximum of 18 participants, which may not adequately reflect the population diversity being studied, ML approaches have been effectively applied to ALS datasets, and numerous prognostic models have been tested using neuroimaging data, longitudinal datasets, and core clinical variables. In many literatures, the constraints of ML models are seldom explicitly enunciated. Therefore, the main objective of this research is to provide a review of the most significant studies on the usage of ML models for analyzing ALS. This review covers a variation of ML algorithms involved in applications in ALS prognosis besides, leveraging ML to improve the efficacy of brain-computer interfaces (BCIs) for ALS individuals in later stages with restricted voluntary muscular control. The key future advances in individualized care and ALS prognosis may include the advancement of more personalized care aids that enable real-time input and ongoing validation of ML in diverse healthcare contexts.
Collapse
Affiliation(s)
- Stephanie Yen Nee Kew
- Department of Mechatronics and Biomedical Engineering, Lee Kong Chian Faculty of Engineering and Science, Universiti Tunku Abdul Rahman, 43000 Kajang, Selangor Darul Ehsan, Malaysia
| | - Siew-Ying Mok
- Department of Mechatronics and Biomedical Engineering, Lee Kong Chian Faculty of Engineering and Science, Universiti Tunku Abdul Rahman, 43000 Kajang, Selangor Darul Ehsan, Malaysia
| | - Choon-Hian Goh
- Department of Mechatronics and Biomedical Engineering, Lee Kong Chian Faculty of Engineering and Science, Universiti Tunku Abdul Rahman, 43000 Kajang, Selangor Darul Ehsan, Malaysia
| |
Collapse
|
3
|
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
|
4
|
Silva-Sousa T, Usuda JN, Al-Arawe N, Frias F, Hinterseher I, Catar R, Luecht C, Riesner K, Hackel A, Schimke LF, Dias HD, Filgueiras IS, Nakaya HI, Camara NOS, Fischer S, Riemekasten G, Ringdén O, Penack O, Winkler T, Duda G, Fonseca DLM, Cabral-Marques O, Moll G. The global evolution and impact of systems biology and artificial intelligence in stem cell research and therapeutics development: a scoping review. Stem Cells 2024; 42:929-944. [PMID: 39230167 DOI: 10.1093/stmcls/sxae054] [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: 06/13/2024] [Accepted: 08/07/2024] [Indexed: 09/05/2024]
Abstract
Advanced bioinformatics analysis, such as systems biology (SysBio) and artificial intelligence (AI) approaches, including machine learning (ML) and deep learning (DL), is increasingly present in stem cell (SC) research. An approximate timeline on these developments and their global impact is still lacking. We conducted a scoping review on the contribution of SysBio and AI analysis to SC research and therapy development based on literature published in PubMed between 2000 and 2024. We identified an 8 to 10-fold increase in research output related to all 3 search terms between 2000 and 2021, with a 10-fold increase in AI-related production since 2010. Use of SysBio and AI still predominates in preclinical basic research with increasing use in clinically oriented translational medicine since 2010. SysBio- and AI-related research was found all over the globe, with SysBio output led by the (US, n = 1487), (UK, n = 1094), Germany (n = 355), The Netherlands (n = 339), Russia (n = 215), and France (n = 149), while for AI-related research the US (n = 853) and UK (n = 258) take a strong lead, followed by Switzerland (n = 69), The Netherlands (n = 37), and Germany (n = 19). The US and UK are most active in SCs publications related to AI/ML and AI/DL. The prominent use of SysBio in ESC research was recently overtaken by prominent use of AI in iPSC and MSC research. This study reveals the global evolution and growing intersection among AI, SysBio, and SC research over the past 2 decades, with substantial growth in all 3 fields and exponential increases in AI-related research in the past decade.
Collapse
Affiliation(s)
- Thayna Silva-Sousa
- BIH Center for Regenerative Therapies (BCRT), Charité Universitätzsmedizin, corporate member of Freie Universität Berlin, Humboldt Universität zu Berlin, and Berlin Institute of Health (BIH), 10117 Berlin, Germany
- Julius Wolff Institute (JWI), Charité Universitätzsmedizin, 10117 Berlin, Germany
- Department of Vascular Surgery, Universitätsklinikum Ruppin-Brandenburg, Medizinische Hochschule Branderburg Theodor Fontane, 16816 Neuruppin, Germany
- Fakultät für Gesundheitswissenschaften Brandenburg, Gemeinsame Fakultät der Universität Potsdam, der Medizinischen Hochschule Brandenburg Theodor Fontane, und der Brandenburgischen Technischen Universität Cottbus-Senftenberg, 14476 Potsdam, Germany
| | - Júlia Nakanishi Usuda
- BIH Center for Regenerative Therapies (BCRT), Charité Universitätzsmedizin, corporate member of Freie Universität Berlin, Humboldt Universität zu Berlin, and Berlin Institute of Health (BIH), 10117 Berlin, Germany
- Julius Wolff Institute (JWI), Charité Universitätzsmedizin, 10117 Berlin, Germany
- Department of Vascular Surgery, Universitätsklinikum Ruppin-Brandenburg, Medizinische Hochschule Branderburg Theodor Fontane, 16816 Neuruppin, Germany
- Fakultät für Gesundheitswissenschaften Brandenburg, Gemeinsame Fakultät der Universität Potsdam, der Medizinischen Hochschule Brandenburg Theodor Fontane, und der Brandenburgischen Technischen Universität Cottbus-Senftenberg, 14476 Potsdam, Germany
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo (USP), São Paulo (SP), Brazil
| | - Nada Al-Arawe
- BIH Center for Regenerative Therapies (BCRT), Charité Universitätzsmedizin, corporate member of Freie Universität Berlin, Humboldt Universität zu Berlin, and Berlin Institute of Health (BIH), 10117 Berlin, Germany
- Julius Wolff Institute (JWI), Charité Universitätzsmedizin, 10117 Berlin, Germany
- Department of Vascular Surgery, Universitätsklinikum Ruppin-Brandenburg, Medizinische Hochschule Branderburg Theodor Fontane, 16816 Neuruppin, Germany
- Fakultät für Gesundheitswissenschaften Brandenburg, Gemeinsame Fakultät der Universität Potsdam, der Medizinischen Hochschule Brandenburg Theodor Fontane, und der Brandenburgischen Technischen Universität Cottbus-Senftenberg, 14476 Potsdam, Germany
- Department of Nephrology and Internal Intensive Care Medicine, Charité Universitätzsmedizin, 10117 Berlin, Germany
- Department of Hematology, Oncology, and Tumorimmunology, Charité Universitätzsmedizin, 10117 Berlin, Germany
| | - Francisca Frias
- BIH Center for Regenerative Therapies (BCRT), Charité Universitätzsmedizin, corporate member of Freie Universität Berlin, Humboldt Universität zu Berlin, and Berlin Institute of Health (BIH), 10117 Berlin, Germany
- Julius Wolff Institute (JWI), Charité Universitätzsmedizin, 10117 Berlin, Germany
- Department of Vascular Surgery, Universitätsklinikum Ruppin-Brandenburg, Medizinische Hochschule Branderburg Theodor Fontane, 16816 Neuruppin, Germany
- Fakultät für Gesundheitswissenschaften Brandenburg, Gemeinsame Fakultät der Universität Potsdam, der Medizinischen Hochschule Brandenburg Theodor Fontane, und der Brandenburgischen Technischen Universität Cottbus-Senftenberg, 14476 Potsdam, Germany
| | - Irene Hinterseher
- Department of Vascular Surgery, Universitätsklinikum Ruppin-Brandenburg, Medizinische Hochschule Branderburg Theodor Fontane, 16816 Neuruppin, Germany
- Fakultät für Gesundheitswissenschaften Brandenburg, Gemeinsame Fakultät der Universität Potsdam, der Medizinischen Hochschule Brandenburg Theodor Fontane, und der Brandenburgischen Technischen Universität Cottbus-Senftenberg, 14476 Potsdam, Germany
- Vascular Surgery, Charité Universitätzsmedizin, 10117 Berlin, Germany
| | - Rusan Catar
- Department of Nephrology and Internal Intensive Care Medicine, Charité Universitätzsmedizin, 10117 Berlin, Germany
| | - Christian Luecht
- Department of Nephrology and Internal Intensive Care Medicine, Charité Universitätzsmedizin, 10117 Berlin, Germany
| | - Katarina Riesner
- Department of Hematology, Oncology, and Tumorimmunology, Charité Universitätzsmedizin, 10117 Berlin, Germany
| | - Alexander Hackel
- Clinic for Rheumatology and Clinical Immunology, University Medical Center Schleswig Holstein Campus Lübeck, 23538 Lübeck, Germany
| | - Lena F Schimke
- Department of Immunology, Institute of Biomedical Sciences, USP, SP, Brazil
| | - Haroldo Dutra Dias
- Interunit Postgraduate Program on Bioinformatics, Institute of Mathematics and Statistics (IME), USP, SP, Brazil
| | | | - Helder I Nakaya
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo (USP), São Paulo (SP), Brazil
- Department of Medicine, Division of Molecular Medicine, Laboratory of Medical Investigation 29, USP School of Medicine (USPM), São Paulo (SP), Brazil
| | - Niels Olsen Saraiva Camara
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo (USP), São Paulo (SP), Brazil
| | - Stefan Fischer
- Clinic for Rheumatology and Clinical Immunology, University Medical Center Schleswig Holstein Campus Lübeck, 23538 Lübeck, Germany
| | - Gabriela Riemekasten
- Clinic for Rheumatology and Clinical Immunology, University Medical Center Schleswig Holstein Campus Lübeck, 23538 Lübeck, Germany
| | - Olle Ringdén
- Division of Pediatrics, Department of CLINTEC, Karolinska Institutet, Stockholm, Sweden
| | - Olaf Penack
- Department of Hematology, Oncology, and Tumorimmunology, Charité Universitätzsmedizin, 10117 Berlin, Germany
| | - Tobias Winkler
- BIH Center for Regenerative Therapies (BCRT), Charité Universitätzsmedizin, corporate member of Freie Universität Berlin, Humboldt Universität zu Berlin, and Berlin Institute of Health (BIH), 10117 Berlin, Germany
- Julius Wolff Institute (JWI), Charité Universitätzsmedizin, 10117 Berlin, Germany
| | - Georg Duda
- BIH Center for Regenerative Therapies (BCRT), Charité Universitätzsmedizin, corporate member of Freie Universität Berlin, Humboldt Universität zu Berlin, and Berlin Institute of Health (BIH), 10117 Berlin, Germany
- Julius Wolff Institute (JWI), Charité Universitätzsmedizin, 10117 Berlin, Germany
| | - Dennyson Leandro M Fonseca
- BIH Center for Regenerative Therapies (BCRT), Charité Universitätzsmedizin, corporate member of Freie Universität Berlin, Humboldt Universität zu Berlin, and Berlin Institute of Health (BIH), 10117 Berlin, Germany
- Julius Wolff Institute (JWI), Charité Universitätzsmedizin, 10117 Berlin, Germany
- Interunit Postgraduate Program on Bioinformatics, Institute of Mathematics and Statistics (IME), USP, SP, Brazil
| | - Otávio Cabral-Marques
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo (USP), São Paulo (SP), Brazil
- Department of Immunology, Institute of Biomedical Sciences, USP, SP, Brazil
- Interunit Postgraduate Program on Bioinformatics, Institute of Mathematics and Statistics (IME), USP, SP, Brazil
- Department of Medicine, Division of Molecular Medicine, Laboratory of Medical Investigation 29, USP School of Medicine (USPM), São Paulo (SP), Brazil
- D'OR Institute Research and Education, SP, Brazil
| | - Guido Moll
- BIH Center for Regenerative Therapies (BCRT), Charité Universitätzsmedizin, corporate member of Freie Universität Berlin, Humboldt Universität zu Berlin, and Berlin Institute of Health (BIH), 10117 Berlin, Germany
- Julius Wolff Institute (JWI), Charité Universitätzsmedizin, 10117 Berlin, Germany
- Department of Nephrology and Internal Intensive Care Medicine, Charité Universitätzsmedizin, 10117 Berlin, Germany
| |
Collapse
|
5
|
Umar TP, Jain N, Papageorgakopoulou M, Shaheen RS, Alsamhori JF, Muzzamil M, Kostiks A. Artificial intelligence for screening and diagnosis of amyotrophic lateral sclerosis: a systematic review and meta-analysis. Amyotroph Lateral Scler Frontotemporal Degener 2024; 25:425-436. [PMID: 38563056 DOI: 10.1080/21678421.2024.2334836] [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: 09/07/2023] [Revised: 03/04/2024] [Accepted: 03/18/2024] [Indexed: 04/04/2024]
Abstract
INTRODUCTION Amyotrophic lateral sclerosis (ALS) is a rare and fatal neurological disease that leads to progressive motor function degeneration. Diagnosing ALS is challenging due to the absence of a specific detection test. The use of artificial intelligence (AI) can assist in the investigation and treatment of ALS. METHODS We searched seven databases for literature on the application of AI in the early diagnosis and screening of ALS in humans. The findings were summarized using random-effects summary receiver operating characteristic curve. The risk of bias (RoB) analysis was carried out using QUADAS-2 or QUADAS-C tools. RESULTS In the 34 analyzed studies, a meta-prevalence of 47% for ALS was noted. For ALS detection, the pooled sensitivity of AI models was 94.3% (95% CI - 63.2% to 99.4%) with a pooled specificity of 98.9% (95% CI - 92.4% to 99.9%). For ALS classification, the pooled sensitivity of AI models was 90.9% (95% CI - 86.5% to 93.9%) with a pooled specificity of 92.3% (95% CI - 84.8% to 96.3%). Based on type of input for classification, the pooled sensitivity of AI models for gait, electromyography, and magnetic resonance signals was 91.2%, 92.6%, and 82.2%, respectively. The pooled specificity for gait, electromyography, and magnetic resonance signals was 94.1%, 96.5%, and 77.3%, respectively. CONCLUSIONS Although AI can play a significant role in the screening and diagnosis of ALS due to its high sensitivities and specificities, concerns remain regarding quality of evidence reported in the literature.
Collapse
Affiliation(s)
- Tungki Pratama Umar
- Department of Medical Profession, Faculty of Medicine, Universitas Sriwijaya, Palembang, Indonesia
| | - Nityanand Jain
- Faculty of Medicine, Riga Stradinš University, Riga, Latvia
| | | | | | | | - Muhammad Muzzamil
- Department of Public Health, Health Services Academy, Islamabad, Pakistan, and
| | - Andrejs Kostiks
- Department of Neurology, Riga East University Clinical Hospital, Riga, Latvia
| |
Collapse
|
6
|
Ara J, Khatun T. A literature review: machine learning-based stem cell investigation. ANNALS OF TRANSLATIONAL MEDICINE 2024; 12:52. [PMID: 38911568 PMCID: PMC11193562 DOI: 10.21037/atm-23-1937] [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: 11/27/2023] [Accepted: 01/08/2024] [Indexed: 06/25/2024]
Abstract
Background and Objective Stem cell (SC) is a crucial factor of the human organ that is significantly important for clinical solutions. However, consideration of SC in the therapeutic or disease classification process is complex in terms of accurate classification and prediction. To overcome this issue, Machine learning (ML) is the most effective technique that is frequently used in cell-based clinical applications for diagnosis, treatment, and disease identification. Recently it has been implemented for SC observation which is a crucial factor for clinical solutions. Thus, the objective of this review work is to represent the effectiveness of ML techniques for SC observation from clinical perspectives with current challenges and future direction for further improvement. Methods In this study, we conducted a short review of ML-based applications in SCs investigation and classification for the improvement of clinical solutions. We explored studies from five scientific databases (Web of Science, Google Scholar, Scopus, ScienceDirect, and PubMed) with several keywords related to the objective of our research study. After primary and secondary screening, 15 articles were utilized for this research study and summarized the observation results in terms of ten aspects (year of publication, focused area, objective, experimented datasets, selected ML classifiers, experimental procedure, classification parameter, overall performance in terms of accuracy, advancements, and limitations) with their current limitations and future improvement directions. Key Content and Findings The majority of the existing literature review works are limited to focusing on specific SC-based investigation, limited evaluation attributes, and lack of challenges and future improvement suggestions. Also, most of the review work didn't consider the investigation of the effectiveness of the ML technique in SC biology. Therefore, in this paper, we investigate existing literature related to the development of clinical solutions considering ML techniques, in the area of SC and cell culture processes and highlight current challenges and future directions. Conclusions The majority of studies focused on the disease identification process and implemented the convolutional neural network and support vector machine techniques. The prime limitations of the investigated studies are related to the focused area, investigated SCs, the small number of experimental datasets, and validation techniques. None of the studies provided complete evidence to determine an optimal ML technique for SC to build classification or predictive models. Therefore, further concern is required to develop and improve the developed solutions including other ML techniques, large datasets, and advanced evaluation processes.
Collapse
Affiliation(s)
- Jinat Ara
- Department of Electrical Engineering and Information Systems, University of Pannonia, Veszprem, Hungary
| | - Tanzila Khatun
- Department of Biochemistry and Biotechnology, Independent University of Bangladesh (IUB), Dhaka, Bangladesh
| |
Collapse
|
7
|
Shi Q, Song F, Zhou X, Chen X, Cao J, Na J, Fan Y, Zhang G, Zheng L. Early Predicting Osteogenic Differentiation of Mesenchymal Stem Cells Based on Deep Learning Within One Day. Ann Biomed Eng 2024; 52:1706-1718. [PMID: 38488988 DOI: 10.1007/s10439-024-03483-3] [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: 11/03/2023] [Accepted: 02/24/2024] [Indexed: 03/17/2024]
Abstract
Osteogenic differentiation of mesenchymal stem cells (MSCs) is proposed to be critical for bone tissue engineering and regenerative medicine. However, the current approach for evaluating osteogenic differentiation mainly involves immunohistochemical staining of specific markers which often can be detected at day 5-7 of osteogenic inducing. Deep learning (DL) is a significant technology for realizing artificial intelligence (AI). Computer vision, a branch of AI, has been proved to achieve high-precision image recognition using convolutional neural networks (CNNs). Our goal was to train CNNs to quantitatively measure the osteogenic differentiation of MSCs. To this end, bright-field images of MSCs during early osteogenic differentiation (day 0, 1, 3, 5, and 7) were captured using a simple optical phase contrast microscope to train CNNs. The results showed that the CNNs could be trained to recognize undifferentiated cells and differentiating cells with an accuracy of 0.961 on the independent test set. In addition, we found that CNNs successfully distinguished differentiated cells at a very early stage (only 1 day). Further analysis showed that overall morphological features of MSCs were the main basis for the CNN classification. In conclusion, MSCs differentiation detection can be achieved early and accurately through simple bright-field images and DL networks, which may also provide a potential and novel method for the field of cell detection in the near future.
Collapse
Affiliation(s)
- Qiusheng Shi
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, No. 37 Xueyuan Road, Haidian District, Beijing, 100191, China
| | - Fan Song
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, No. 37 Xueyuan Road, Haidian District, Beijing, 100191, China
| | - Xiaocheng Zhou
- Department of Statistics, The Chinese University of Hong Kong, Sha Tin, Hong Kong SAR, China
| | - Xinyuan Chen
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, No. 37 Xueyuan Road, Haidian District, Beijing, 100191, China
| | - Jingqi Cao
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, No. 37 Xueyuan Road, Haidian District, Beijing, 100191, China
| | - Jing Na
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, No. 37 Xueyuan Road, Haidian District, Beijing, 100191, China
| | - Yubo Fan
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, No. 37 Xueyuan Road, Haidian District, Beijing, 100191, China.
| | - Guanglei Zhang
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, No. 37 Xueyuan Road, Haidian District, Beijing, 100191, China.
| | - Lisha Zheng
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, No. 37 Xueyuan Road, Haidian District, Beijing, 100191, China.
| |
Collapse
|
8
|
Vo QD, Saito Y, Ida T, Nakamura K, Yuasa S. The use of artificial intelligence in induced pluripotent stem cell-based technology over 10-year period: A systematic scoping review. PLoS One 2024; 19:e0302537. [PMID: 38771829 PMCID: PMC11108174 DOI: 10.1371/journal.pone.0302537] [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: 02/02/2024] [Accepted: 04/09/2024] [Indexed: 05/23/2024] Open
Abstract
BACKGROUND Stem cell research, particularly in the domain of induced pluripotent stem cell (iPSC) technology, has shown significant progress. The integration of artificial intelligence (AI), especially machine learning (ML) and deep learning (DL), has played a pivotal role in refining iPSC classification, monitoring cell functionality, and conducting genetic analysis. These enhancements are broadening the applications of iPSC technology in disease modelling, drug screening, and regenerative medicine. This review aims to explore the role of AI in the advancement of iPSC research. METHODS In December 2023, data were collected from three electronic databases (PubMed, Web of Science, and Science Direct) to investigate the application of AI technology in iPSC processing. RESULTS This systematic scoping review encompassed 79 studies that met the inclusion criteria. The number of research studies in this area has increased over time, with the United States emerging as a leading contributor in this field. AI technologies have been diversely applied in iPSC technology, encompassing the classification of cell types, assessment of disease-specific phenotypes in iPSC-derived cells, and the facilitation of drug screening using iPSC. The precision of AI methodologies has improved significantly in recent years, creating a foundation for future advancements in iPSC-based technologies. CONCLUSIONS Our review offers insights into the role of AI in regenerative and personalized medicine, highlighting both challenges and opportunities. Although still in its early stages, AI technologies show significant promise in advancing our understanding of disease progression and development, paving the way for future clinical applications.
Collapse
Affiliation(s)
- Quan Duy Vo
- Faculty of Medicine, Department of Cardiovascular Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
- Faculty of Medicine, Nguyen Tat Thanh University, Ho Chi Minh City, Viet Nam
| | - Yukihiro Saito
- Department of Cardiovascular Medicine, Okayama University Hospital, Okayama, Japan
| | - Toshihiro Ida
- Faculty of Medicine, Department of Cardiovascular Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Kazufumi Nakamura
- Faculty of Medicine, Department of Cardiovascular Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Shinsuke Yuasa
- Faculty of Medicine, Department of Cardiovascular Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| |
Collapse
|
9
|
Otsuka Y, Imamura K, Oishi A, Asakawa K, Kondo T, Nakai R, Suga M, Inoue I, Sagara Y, Tsukita K, Teranaka K, Nishimura Y, Watanabe A, Umeyama K, Okushima N, Mitani K, Nagashima H, Kawakami K, Muguruma K, Tsujikawa A, Inoue H. Phototoxicity avoidance is a potential therapeutic approach for retinal dystrophy caused by EYS dysfunction. JCI Insight 2024; 9:e174179. [PMID: 38646933 PMCID: PMC11141876 DOI: 10.1172/jci.insight.174179] [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: 07/24/2023] [Accepted: 03/06/2024] [Indexed: 04/25/2024] Open
Abstract
Inherited retinal dystrophies (IRDs) are progressive diseases leading to vision loss. Mutation in the eyes shut homolog (EYS) gene is one of the most frequent causes of IRD. However, the mechanism of photoreceptor cell degeneration by mutant EYS has not been fully elucidated. Here, we generated retinal organoids from induced pluripotent stem cells (iPSCs) derived from patients with EYS-associated retinal dystrophy (EYS-RD). In photoreceptor cells of RD organoids, both EYS and G protein-coupled receptor kinase 7 (GRK7), one of the proteins handling phototoxicity, were not in the outer segment, where they are physiologically present. Furthermore, photoreceptor cells in RD organoids were vulnerable to light stimuli, and especially to blue light. Mislocalization of GRK7, which was also observed in eys-knockout zebrafish, was reversed by delivering control EYS into photoreceptor cells of RD organoids. These findings suggest that avoiding phototoxicity would be a potential therapeutic approach for EYS-RD.
Collapse
Affiliation(s)
- Yuki Otsuka
- iPSC-based Drug discovery and Development Team, RIKEN BioResource Research Center, Kyoto, Japan
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
- Department of Ophthalmology and Visual Sciences, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Keiko Imamura
- iPSC-based Drug discovery and Development Team, RIKEN BioResource Research Center, Kyoto, Japan
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
- RIKEN Center for Advanced Intelligence Project (AIP), Kyoto, Japan
| | - Akio Oishi
- Department of Ophthalmology and Visual Sciences, Nagasaki University, Nagasaki, Japan
| | - Kazuhide Asakawa
- Division of Molecular and Developmental Biology, National Institute of Genetics, Mishima, Japan
| | - Takayuki Kondo
- iPSC-based Drug discovery and Development Team, RIKEN BioResource Research Center, Kyoto, Japan
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
- RIKEN Center for Advanced Intelligence Project (AIP), Kyoto, Japan
| | - Risako Nakai
- iPSC-based Drug discovery and Development Team, RIKEN BioResource Research Center, Kyoto, Japan
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Mika Suga
- iPSC-based Drug discovery and Development Team, RIKEN BioResource Research Center, Kyoto, Japan
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Ikuyo Inoue
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
- RIKEN Center for Advanced Intelligence Project (AIP), Kyoto, Japan
| | - Yukako Sagara
- iPSC-based Drug discovery and Development Team, RIKEN BioResource Research Center, Kyoto, Japan
| | - Kayoko Tsukita
- iPSC-based Drug discovery and Development Team, RIKEN BioResource Research Center, Kyoto, Japan
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Kaori Teranaka
- Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yu Nishimura
- Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Akira Watanabe
- Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Kazuhiro Umeyama
- Meiji University International Institute for Bio-Resource Research, Kawasaki, Japan
| | - Nanako Okushima
- Division of Systems Medicine and Gene Therapy, Faculty of Medicine, Saitama Medical University, Saitama, Japan
| | - Kohnosuke Mitani
- Division of Systems Medicine and Gene Therapy, Faculty of Medicine, Saitama Medical University, Saitama, Japan
| | - Hiroshi Nagashima
- Meiji University International Institute for Bio-Resource Research, Kawasaki, Japan
| | - Koichi Kawakami
- Division of Molecular and Developmental Biology, National Institute of Genetics, Mishima, Japan
| | - Keiko Muguruma
- Department of iPS Cell Applied Medicine, Graduate School of Medicine, Kansai Medical University, Hirakata, Osaka, Japan
| | - Akitaka Tsujikawa
- Department of Ophthalmology and Visual Sciences, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Haruhisa Inoue
- iPSC-based Drug discovery and Development Team, RIKEN BioResource Research Center, Kyoto, Japan
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
- RIKEN Center for Advanced Intelligence Project (AIP), Kyoto, Japan
| |
Collapse
|
10
|
Nakamura R, Tohnai G, Nakatochi M, Atsuta N, Watanabe H, Ito D, Katsuno M, Hirakawa A, Izumi Y, Morita M, Hirayama T, Kano O, Kanai K, Hattori N, Taniguchi A, Suzuki N, Aoki M, Iwata I, Yabe I, Shibuya K, Kuwabara S, Oda M, Hashimoto R, Aiba I, Ishihara T, Onodera O, Yamashita T, Abe K, Mizoguchi K, Shimizu T, Ikeda Y, Yokota T, Hasegawa K, Tanaka F, Nakashima K, Kaji R, Niwa JI, Doyu M, Terao C, Ikegawa S, Fujimori K, Nakamura S, Ozawa F, Morimoto S, Onodera K, Ito T, Okada Y, Okano H, Sobue G. Genetic factors affecting survival in Japanese patients with sporadic amyotrophic lateral sclerosis: a genome-wide association study and verification in iPSC-derived motor neurons from patients. J Neurol Neurosurg Psychiatry 2023; 94:816-824. [PMID: 37142397 DOI: 10.1136/jnnp-2022-330851] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 04/18/2023] [Indexed: 05/06/2023]
Abstract
BACKGROUND Several genetic factors are associated with the pathogenesis of sporadic amyotrophic lateral sclerosis (ALS) and its phenotypes, such as disease progression. Here, in this study, we aimed to identify the genes that affect the survival of patients with sporadic ALS. METHODS We enrolled 1076 Japanese patients with sporadic ALS with imputed genotype data of 7 908 526 variants. We used Cox proportional hazards regression analysis with an additive model adjusted for sex, age at onset and the first two principal components calculated from genotyped data to conduct a genome-wide association study. We further analysed messenger RNA (mRNA) and phenotype expression in motor neurons derived from induced pluripotent stem cells (iPSC-MNs) of patients with ALS. RESULTS Three novel loci were significantly associated with the survival of patients with sporadic ALS-FGF1 at 5q31.3 (rs11738209, HR=2.36 (95% CI, 1.77 to 3.15), p=4.85×10-9), THSD7A at 7p21.3 (rs2354952, 1.38 (95% CI, 1.24 to 1.55), p=1.61×10-8) and LRP1 at 12q13.3 (rs60565245, 2.18 (95% CI, 1.66 to 2.86), p=2.35×10-8). FGF1 and THSD7A variants were associated with decreased mRNA expression of each gene in iPSC-MNs and reduced in vitro survival of iPSC-MNs obtained from patients with ALS. The iPSC-MN in vitro survival was reduced when the expression of FGF1 and THSD7A was partially disrupted. The rs60565245 was not associated with LRP1 mRNA expression. CONCLUSIONS We identified three loci associated with the survival of patients with sporadic ALS, decreased mRNA expression of FGF1 and THSD7A and the viability of iPSC-MNs from patients. The iPSC-MN model reflects the association between patient prognosis and genotype and can contribute to target screening and validation for therapeutic intervention.
Collapse
Affiliation(s)
- Ryoichi Nakamura
- Department of Neurology, Aichi Medical University School of Medicine, Nagakute, Aichi, Japan
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Genki Tohnai
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
- Division of ALS Research, Aichi Medical University School of Medicine, Nagakute, Aichi, Japan
| | - Masahiro Nakatochi
- Public Health Informatics Unit, Department of Integrated Health Sciences, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Naoki Atsuta
- Department of Neurology, Aichi Medical University School of Medicine, Nagakute, Aichi, Japan
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Hirohisa Watanabe
- Department of Neurology, Fujita Health University, Toyoake, Aichi, Japan
- Brain and Mind Research Center, Nagoya University, Nagoya, Aichi, Japan
| | - Daisuke Ito
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Masahisa Katsuno
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
- Department of Clinical Research Education, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Akihiro Hirakawa
- Department of Clinical Biostatistics, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo, Japan
| | - Yuishin Izumi
- Department of Neurology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Mitsuya Morita
- Division of Neurology, Department of Internal Medicine, Jichi Medical University, Shimotsuke, Tochigi, Japan
| | - Takehisa Hirayama
- Department of Neurology, Toho University Faculty of Medicine, Ota-ku, Tokyo, Japan
| | - Osamu Kano
- Department of Neurology, Toho University Faculty of Medicine, Ota-ku, Tokyo, Japan
| | - Kazuaki Kanai
- Department of Neurology, Fukushima Medical University School of Medicine, Fukushima, Japan
- Department of Neurology, Juntendo University School of Medicine, Bunkyo-ku, Tokyo, Japan
| | - Nobutaka Hattori
- Department of Neurology, Juntendo University School of Medicine, Bunkyo-ku, Tokyo, Japan
| | - Akira Taniguchi
- Department of Neurology, Mie University Graduate School of Medicine, Tsu, Mie, Japan
| | - Naoki Suzuki
- Department of Neurology, Tohoku University School of Medicine, Sendai, Miyagi, Japan
| | - Masashi Aoki
- Department of Neurology, Tohoku University School of Medicine, Sendai, Miyagi, Japan
| | - Ikuko Iwata
- Department of Neurology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Ichiro Yabe
- Department of Neurology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Kazumoto Shibuya
- Department of Neurology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Satoshi Kuwabara
- Department of Neurology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Masaya Oda
- Department of Neurology, Vihara Hananosato Hospital, Miyoshi, Hiroshima, Japan
| | - Rina Hashimoto
- Department of Neurology, National Hospital Organization Higashinagoya National Hospital, Nagoya, Aichi, Japan
| | - Ikuko Aiba
- Department of Neurology, National Hospital Organization Higashinagoya National Hospital, Nagoya, Aichi, Japan
| | - Tomohiko Ishihara
- Department of Neurology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Osamu Onodera
- Department of Neurology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Toru Yamashita
- Department of Neurology, Okayama University Graduate School of Medicine, Okayama, Japan
| | - Koji Abe
- Department of Neurology, Okayama University Graduate School of Medicine, Okayama, Japan
| | - Kouichi Mizoguchi
- Department of Neurology, National Hospital Organization Shizuoka Medical Center, Shizuoka, Japan
| | - Toshio Shimizu
- Department of Neurology, Tokyo Metropolitan Neurological Hospital, Fuchu, Tokyo, Japan
| | - Yoshio Ikeda
- Department of Neurology, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| | - Takanori Yokota
- Department of Neurology and Neurological Science, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo, Japan
| | - Kazuko Hasegawa
- Division of Neurology, National Hospital Organization, Sagamihara National Hospital, Sagamihara, Kanagawa, Japan
| | - Fumiaki Tanaka
- Department of Neurology and Stroke Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Kanagawa, Japan
| | - Kenji Nakashima
- Department of Neurology, National Hospital Organization, Matsue Medical Center, Matsue, Shimane, Japan
| | - Ryuji Kaji
- Department of Neurology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Jun-Ichi Niwa
- Department of Neurology, Aichi Medical University School of Medicine, Nagakute, Aichi, Japan
| | - Manabu Doyu
- Department of Neurology, Aichi Medical University School of Medicine, Nagakute, Aichi, Japan
| | - Chikashi Terao
- Laboratory for Statistical and Translational Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa, Japan
| | - Shiro Ikegawa
- Laboratory for Bone and Joint Diseases, RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa, Japan
| | - Koki Fujimori
- Department of Physiology, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Shiho Nakamura
- Department of Physiology, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Fumiko Ozawa
- Department of Physiology, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Satoru Morimoto
- Department of Physiology, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Kazunari Onodera
- Department of Neurology, Aichi Medical University School of Medicine, Nagakute, Aichi, Japan
| | - Takuji Ito
- Department of Neurology, Aichi Medical University School of Medicine, Nagakute, Aichi, Japan
| | - Yohei Okada
- Department of Neurology, Aichi Medical University School of Medicine, Nagakute, Aichi, Japan
| | - Hideyuki Okano
- Department of Physiology, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Gen Sobue
- Brain and Mind Research Center, Nagoya University, Nagoya, Aichi, Japan
- Aichi Medical University, Nagakute, Aichi, Japan
| |
Collapse
|
11
|
Menduti G, Boido M. Recent Advances in High-Content Imaging and Analysis in iPSC-Based Modelling of Neurodegenerative Diseases. Int J Mol Sci 2023; 24:14689. [PMID: 37834135 PMCID: PMC10572296 DOI: 10.3390/ijms241914689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Revised: 09/24/2023] [Accepted: 09/25/2023] [Indexed: 10/15/2023] Open
Abstract
In the field of neurodegenerative pathologies, the platforms for disease modelling based on patient-derived induced pluripotent stem cells (iPSCs) represent a valuable molecular diagnostic/prognostic tool. Indeed, they paved the way for the in vitro recapitulation of the pathological mechanisms underlying neurodegeneration and for characterizing the molecular heterogeneity of disease manifestations, also enabling drug screening approaches for new therapeutic candidates. A major challenge is related to the choice and optimization of the morpho-functional study designs in human iPSC-derived neurons to deeply detail the cell phenotypes as markers of neurodegeneration. In recent years, the specific combination of high-throughput screening with subcellular resolution microscopy for cell-based high-content imaging (HCI) screening allowed in-depth analyses of cell morphology and neurite trafficking in iPSC-derived neuronal cells by using specific cutting-edge microscopes and automated computational assays. The present work aims to describe the main recent protocols and advances achieved with the HCI analysis in iPSC-based modelling of neurodegenerative diseases, highlighting technical and bioinformatics tips and tricks for further uses and research. To this end, microscopy requirements and the latest computational pipelines to analyze imaging data will be explored, while also providing an overview of the available open-source high-throughput automated platforms.
Collapse
Affiliation(s)
- Giovanna Menduti
- Department of Neuroscience “Rita Levi Montalcini”, Neuroscience Institute Cavalieri Ottolenghi, University of Turin, Regione Gonzole 10, Orbassano, 10043 Turin, TO, Italy;
| | | |
Collapse
|
12
|
Marzec-Schmidt K, Ghosheh N, Stahlschmidt SR, Küppers-Munther B, Synnergren J, Ulfenborg B. Artificial Intelligence Supports Automated Characterization of Differentiated Human Pluripotent Stem Cells. Stem Cells 2023; 41:850-861. [PMID: 37357747 PMCID: PMC10502778 DOI: 10.1093/stmcls/sxad049] [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: 03/26/2023] [Accepted: 06/05/2023] [Indexed: 06/27/2023]
Abstract
Revolutionary advances in AI and deep learning in recent years have resulted in an upsurge of papers exploring applications within the biomedical field. Within stem cell research, promising results have been reported from analyses of microscopy images to, that is, distinguish between pluripotent stem cells and differentiated cell types derived from stem cells. In this work, we investigated the possibility of using a deep learning model to predict the differentiation stage of pluripotent stem cells undergoing differentiation toward hepatocytes, based on morphological features of cell cultures. We were able to achieve close to perfect classification of images from early and late time points during differentiation, and this aligned very well with the experimental validation of cell identity and function. Our results suggest that deep learning models can distinguish between different cell morphologies, and provide alternative means of semi-automated functional characterization of stem cell cultures.
Collapse
Affiliation(s)
- Katarzyna Marzec-Schmidt
- Department of Soil and Environment, Swedish University of Agricultural Sciences (SLU), Skara, Sweden
| | - Nidal Ghosheh
- Takara Bio Europe, Gothenburg, Sweden
- Department of Biology and Bioinformatics, School of Bioscience, University of Skövde, Skövde, Sweden
| | | | | | - Jane Synnergren
- Department of Biology and Bioinformatics, School of Bioscience, University of Skövde, Skövde, Sweden
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Benjamin Ulfenborg
- Department of Biology and Bioinformatics, School of Bioscience, University of Skövde, Skövde, Sweden
| |
Collapse
|
13
|
Lopez-Bernal D, Balderas D, Ponce P, Rojas M, Molina A. Implications of Artificial Intelligence Algorithms in the Diagnosis and Treatment of Motor Neuron Diseases-A Review. Life (Basel) 2023; 13:life13041031. [PMID: 37109560 PMCID: PMC10146231 DOI: 10.3390/life13041031] [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: 12/22/2022] [Revised: 02/17/2023] [Accepted: 03/29/2023] [Indexed: 04/29/2023] Open
Abstract
Motor neuron diseases (MNDs) are a group of chronic neurological disorders characterized by the progressive failure of the motor system. Currently, these disorders do not have a definitive treatment; therefore, it is of huge importance to propose new and more advanced diagnoses and treatment options for MNDs. Nowadays, artificial intelligence is being applied to solve several real-life problems in different areas, including healthcare. It has shown great potential to accelerate the understanding and management of many health disorders, including neurological ones. Therefore, the main objective of this work is to offer a review of the most important research that has been done on the application of artificial intelligence models for analyzing motor disorders. This review includes a general description of the most commonly used AI algorithms and their usage in MND diagnosis, prognosis, and treatment. Finally, we highlight the main issues that must be overcome to take full advantage of what AI can offer us when dealing with MNDs.
Collapse
Affiliation(s)
- Diego Lopez-Bernal
- Tecnologico de Monterrey, National Department of Research, Puente 222, Del. Tlalpan, Mexico City 14380, Mexico
| | - David Balderas
- Tecnologico de Monterrey, National Department of Research, Puente 222, Del. Tlalpan, Mexico City 14380, Mexico
| | - Pedro Ponce
- Tecnologico de Monterrey, National Department of Research, Puente 222, Del. Tlalpan, Mexico City 14380, Mexico
| | - Mario Rojas
- Tecnologico de Monterrey, National Department of Research, Puente 222, Del. Tlalpan, Mexico City 14380, Mexico
| | - Arturo Molina
- Tecnologico de Monterrey, National Department of Research, Puente 222, Del. Tlalpan, Mexico City 14380, Mexico
| |
Collapse
|
14
|
Du H, Huo Z, Chen Y, Zhao Z, Meng F, Wang X, Liu S, Zhang H, Zhou F, Liu J, Zhang L, Zhou S, Guan Y, Wang X. Induced Pluripotent Stem Cells and Their Applications in Amyotrophic Lateral Sclerosis. Cells 2023; 12:cells12060971. [PMID: 36980310 PMCID: PMC10047679 DOI: 10.3390/cells12060971] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 03/20/2023] [Accepted: 03/20/2023] [Indexed: 03/30/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a progressive neurodegenerative disease that results in the loss of motor function in the central nervous system (CNS) and ultimately death. The mechanisms underlying ALS pathogenesis have not yet been fully elucidated, and ALS cannot be treated effectively. Most studies have applied animal or single-gene intervention cell lines as ALS disease models, but they cannot accurately reflect the pathological characteristics of ALS. Induced pluripotent stem cells (iPSCs) can be reprogrammed from somatic cells, possessing the ability to self-renew and differentiate into a variety of cells. iPSCs can be obtained from ALS patients with different genotypes and phenotypes, and the genetic background of the donor cells remains unchanged during reprogramming. iPSCs can differentiate into neurons and glial cells related to ALS. Therefore, iPSCs provide an excellent method to evaluate the impact of diseases on ALS patients. Moreover, patient-derived iPSCs are obtained from their own somatic cells, avoiding ethical concerns and posing only a low risk of immune rejection. The iPSC technology creates new hope for ALS treatment. Here, we review recent studies on iPSCs and their applications in disease modeling, drug screening and cell therapy in ALS, with a particular focus on the potential for ALS treatment.
Collapse
Affiliation(s)
- Hongmei Du
- Department of Histology and Embryology, School of Basic Medical Sciences, Weifang Medical University, Weifang 261053, China
- Neurologic Disorders and Regenerative Repair Laboratory, Weifang Medical University, Weifang 261053, China
| | - Zijun Huo
- Department of Histology and Embryology, School of Basic Medical Sciences, Weifang Medical University, Weifang 261053, China
| | - Yanchun Chen
- Department of Histology and Embryology, School of Basic Medical Sciences, Weifang Medical University, Weifang 261053, China
- Neurologic Disorders and Regenerative Repair Laboratory, Weifang Medical University, Weifang 261053, China
| | - Zhenhan Zhao
- Department of Histology and Embryology, School of Basic Medical Sciences, Weifang Medical University, Weifang 261053, China
| | - Fandi Meng
- Department of Histology and Embryology, School of Basic Medical Sciences, Weifang Medical University, Weifang 261053, China
| | - Xuemei Wang
- Department of Histology and Embryology, School of Basic Medical Sciences, Weifang Medical University, Weifang 261053, China
| | - Shiyue Liu
- Neurologic Disorders and Regenerative Repair Laboratory, Weifang Medical University, Weifang 261053, China
| | - Haoyun Zhang
- Neurologic Disorders and Regenerative Repair Laboratory, Weifang Medical University, Weifang 261053, China
| | - Fenghua Zhou
- Neurologic Disorders and Regenerative Repair Laboratory, Weifang Medical University, Weifang 261053, China
- Department of Pathology, School of Basic Medical Sciences, Weifang Medical University, Weifang 261053, China
| | - Jinmeng Liu
- Neurologic Disorders and Regenerative Repair Laboratory, Weifang Medical University, Weifang 261053, China
| | - Lingyun Zhang
- Neurologic Disorders and Regenerative Repair Laboratory, Weifang Medical University, Weifang 261053, China
| | - Shuanhu Zhou
- Harvard Medical School and Harvard Stem Cell Institute, Harvard University, Boston, MA 02115, USA
| | - Yingjun Guan
- Department of Histology and Embryology, School of Basic Medical Sciences, Weifang Medical University, Weifang 261053, China
- Neurologic Disorders and Regenerative Repair Laboratory, Weifang Medical University, Weifang 261053, China
| | - Xin Wang
- Harvard Medical School and Harvard Stem Cell Institute, Harvard University, Boston, MA 02115, USA
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
15
|
Vidovic M, Müschen LH, Brakemeier S, Machetanz G, Naumann M, Castro-Gomez S. Current State and Future Directions in the Diagnosis of Amyotrophic Lateral Sclerosis. Cells 2023; 12:736. [PMID: 36899872 PMCID: PMC10000757 DOI: 10.3390/cells12050736] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Revised: 02/22/2023] [Accepted: 02/23/2023] [Indexed: 03/02/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease characterized by loss of upper and lower motor neurons, resulting in progressive weakness of all voluntary muscles and eventual respiratory failure. Non-motor symptoms, such as cognitive and behavioral changes, frequently occur over the course of the disease. Considering its poor prognosis with a median survival time of 2 to 4 years and limited causal treatment options, an early diagnosis of ALS plays an essential role. In the past, diagnosis has primarily been determined by clinical findings supported by electrophysiological and laboratory measurements. To increase diagnostic accuracy, reduce diagnostic delay, optimize stratification in clinical trials and provide quantitative monitoring of disease progression and treatment responsivity, research on disease-specific and feasible fluid biomarkers, such as neurofilaments, has been intensely pursued. Advances in imaging techniques have additionally yielded diagnostic benefits. Growing perception and greater availability of genetic testing facilitate early identification of pathogenic ALS-related gene mutations, predictive testing and access to novel therapeutic agents in clinical trials addressing disease-modified therapies before the advent of the first clinical symptoms. Lately, personalized survival prediction models have been proposed to offer a more detailed disclosure of the prognosis for the patient. In this review, the established procedures and future directions in the diagnostics of ALS are summarized to serve as a practical guideline and to improve the diagnostic pathway of this burdensome disease.
Collapse
Affiliation(s)
- Maximilian Vidovic
- Department of Neurology, University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
| | | | - Svenja Brakemeier
- Department of Neurology and Center for Translational Neuro and Behavioral Sciences (C-TNBS), University Hospital Essen, 45147 Essen, Germany
| | - Gerrit Machetanz
- Department of Neurology, Klinikum Rechts der Isar, Technical University of Munich, 81675 Munich, Germany
| | - Marcel Naumann
- Translational Neurodegeneration Section “Albrecht Kossel”, Department of Neurology, University Medical Center, University of Rostock, 18147 Rostock, Germany
| | - Sergio Castro-Gomez
- Department of Neurodegenerative Disease and Geriatric Psychiatry/Neurology, University Hospital Bonn, 53127 Bonn, Germany
- Institute of Physiology II, University Hospital Bonn, 53115 Bonn, Germany
- Department of Neuroimmunology, Institute of Innate Immunity, University Hospital Bonn, 53127 Bonn, Germany
| |
Collapse
|
16
|
Das T, Kaur H, Gour P, Prasad K, Lynn AM, Prakash A, Kumar V. Intersection of network medicine and machine learning towards investigating the key biomarkers and pathways underlying amyotrophic lateral sclerosis: a systematic review. Brief Bioinform 2022; 23:6780269. [PMID: 36411673 DOI: 10.1093/bib/bbac442] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 08/12/2022] [Accepted: 09/13/2022] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND Network medicine is an emerging area of research that focuses on delving into the molecular complexity of the disease, leading to the discovery of network biomarkers and therapeutic target discovery. Amyotrophic lateral sclerosis (ALS) is a complicated rare disease with unknown pathogenesis and no available treatment. In ALS, network properties appear to be potential biomarkers that can be beneficial in disease-related applications when explored independently or in tandem with machine learning (ML) techniques. OBJECTIVE This systematic literature review explores recent trends in network medicine and implementations of network-based ML algorithms in ALS. We aim to provide an overview of the identified primary studies and gather details on identifying the potential biomarkers and delineated pathways. METHODS The current study consists of searching for and investigating primary studies from PubMed and Dimensions.ai, published between 2018 and 2022 that reported network medicine perspectives and the coupling of ML techniques. Each abstract and full-text study was individually evaluated, and the relevant studies were finally included in the review for discussion once they met the inclusion and exclusion criteria. RESULTS We identified 109 eligible publications from primary studies representing this systematic review. The data coalesced into two themes: application of network science to identify disease modules and promising biomarkers in ALS, along with network-based ML approaches. Conclusion This systematic review gives an overview of the network medicine approaches and implementations of network-based ML algorithms in ALS to determine new disease genes, and identify critical pathways and therapeutic target discovery for personalized treatment.
Collapse
Affiliation(s)
- Trishala Das
- School of Computational & Integrative Sciences, Jawaharlal Nehru University, New Delhi-110067, India
| | - Harbinder Kaur
- School of Computational & Integrative Sciences, Jawaharlal Nehru University, New Delhi-110067, India
| | - Pratibha Gour
- Dept. of Plant Molecular Biology, University of Delhi, South Campus, New Delhi-110021, India
| | - Kartikay Prasad
- Amity Institute of Neuropsychology & Neurosciences (AINN), Amity University, Noida, UP-201303, India
| | - Andrew M Lynn
- School of Computational & Integrative Sciences, Jawaharlal Nehru University, New Delhi-110067, India
| | - Amresh Prakash
- Amity Institute of Integrative Sciences and Health, Amity University Haryana, Gurgaon-122413, India
| | - Vijay Kumar
- Amity Institute of Neuropsychology & Neurosciences (AINN), Amity University, Noida, UP-201303, India
| |
Collapse
|
17
|
Imamura K, Izumi Y, Nagai M, Nishiyama K, Watanabe Y, Hanajima R, Egawa N, Ayaki T, Oki R, Fujita K, Uozumi R, Morinaga A, Hirohashi T, Fujii Y, Yamamoto T, Tatebe H, Tokuda T, Takahashi N, Morita S, Takahashi R, Inoue H. Safety and tolerability of bosutinib in patients with amyotrophic lateral sclerosis (iDReAM study): A multicentre, open-label, dose-escalation phase 1 trial. EClinicalMedicine 2022; 53:101707. [PMID: 36467452 PMCID: PMC9716331 DOI: 10.1016/j.eclinm.2022.101707] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 09/29/2022] [Accepted: 09/30/2022] [Indexed: 11/06/2022] Open
Abstract
BACKGROUND Amyotrophic lateral sclerosis (ALS) is a progressive neurodegenerative disease caused by the loss of motor neurons, and development of effective medicines is urgently required. Induced pluripotent stem cell (iPSC)-based drug repurposing identified the Src/c-Abl inhibitor bosutinib, which is approved for the treatment of chronic myelogenous leukemia (CML), as a candidate for the molecular targeted therapy of ALS. METHODS An open-label, multicentre, dose-escalation phase 1 study using a 3 + 3 design was conducted in 4 hospitals in Japan to evaluate the safety and tolerability of bosutinib in patients with ALS. Furthermore, the exploratory efficacy was evaluated using Revised ALS Functional Rating Scale (ALSFRS-R), predictive biomarkers including plasma neurofilament light chain (NFL) were explored, and single-cell RNA sequencing of iPSC-derived motor neurons was conducted. Patients, whose total ALSFRS-R scores decreased by 1-3 points during the 12-week, received escalating doses starting from 100 mg quaque die (QD) up to 400 mg QD based on dose-limiting toxicity (DLT) occurrence, and all participants who received one dose of the study drug were included in the primary analysis. This trial is registered with ClinicalTrials.gov, NCT04744532, as Induced pluripotent stem cell-based Drug Repurposing for Amyotrophic Lateral Sclerosis Medicine (iDReAM) study. FINDINGS Between March 29, 2019 and May 7, 2021, 20 patients were enrolled, 13 of whom received bosutinib treatment and 12 were included in the safety and efficacy analyses. No DLTs were observed up to 300 mg QD, but DLTs were observed in 3/3 patients of the 400 mg QD cohort. In all patients receiving 100 mg-400 mg, the prevalent adverse events (AEs) were gastrointestinal AEs in 12 patients (92.3%), liver function related AEs in 7 patients (53.8%), and rash in 3 patients (23.1%). The safety profile was consistent with that known for CML treatment, and ALS-specific AEs were not observed. A subset of patients (5/9 patients) was found to respond well to bosutinib treatment over the 12-week treatment period. It was found that the treatment-responsive patients could be distinguished by their lower levels of plasma NFL. Furthermore, single-cell RNA sequencing of iPSC-derived motor neurons revealed the pathogenesis related molecular signature in patients with ALS showing responsiveness to bosutinib. INTERPRETATION This is the first trial of a Src/c-Abl inhibitor, bosutinib, for patients with ALS. The safety and tolerability of bosutinib up to 300 mg, not 400 mg, in ALS were described, and responsiveness of patients on motor function was observed. Since this was an open-label trial within a short period with a limited number of patients, further clinical trials will be required. FUNDING AMED and iPS Cell Research Fund.
Collapse
Affiliation(s)
- Keiko Imamura
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Yuishin Izumi
- Department of Neurology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Makiko Nagai
- Department of Neurology, Kitasato University School of Medicine, Sagamihara, Japan
| | - Kazutoshi Nishiyama
- Department of Neurology, Kitasato University School of Medicine, Sagamihara, Japan
| | - Yasuhiro Watanabe
- Division of Neurology, Department of Brain and Neurosciences, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Ritsuko Hanajima
- Division of Neurology, Department of Brain and Neurosciences, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Naohiro Egawa
- Department of Neurology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Takashi Ayaki
- Department of Neurology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Ryosuke Oki
- Department of Neurology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Koji Fujita
- Department of Neurology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Ryuji Uozumi
- Department of Biomedical Statistics and Bioinformatics, Kyoto University, Kyoto, Japan
| | | | | | | | - Takuya Yamamoto
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Harutsugu Tatebe
- Department of Functional Brain Imaging, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology, Chiba, Japan
| | - Takahiko Tokuda
- Department of Functional Brain Imaging, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology, Chiba, Japan
| | - Naoto Takahashi
- Department of Hematology, Nephrology, and Rheumatology, Akita University Graduate School of Medicine, Akita, Japan
| | - Satoshi Morita
- Department of Biomedical Statistics and Bioinformatics, Kyoto University, Kyoto, Japan
| | - Ryosuke Takahashi
- Department of Neurology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Haruhisa Inoue
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
- Corresponding author. 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, Kyoto Pref., 606-8507, Japan.
| |
Collapse
|
18
|
Issa J, Abou Chaar M, Kempisty B, Gasiorowski L, Olszewski R, Mozdziak P, Dyszkiewicz-Konwińska M. Artificial-Intelligence-Based Imaging Analysis of Stem Cells: A Systematic Scoping Review. BIOLOGY 2022; 11:1412. [PMID: 36290317 PMCID: PMC9598508 DOI: 10.3390/biology11101412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Revised: 09/22/2022] [Accepted: 09/24/2022] [Indexed: 11/20/2022]
Abstract
This systematic scoping review aims to map and identify the available artificial-intelligence-based techniques for imaging analysis, the characterization of stem cell differentiation, and trans-differentiation pathways. On the ninth of March 2022, data were collected from five electronic databases (PubMed, Medline, Web of Science, Cochrane, and Scopus) and manual citation searching; all data were gathered in Zotero 5.0. A total of 4422 articles were collected after deduplication; only twenty-seven studies were included in this systematic scoping review after a two-phase screening against inclusion criteria by two independent reviewers. The amount of research in this field is significantly increasing over the years. While the current state of artificial intelligence (AI) can tackle a multitude of medical problems, the consensus amongst researchers remains that AI still falls short in multiple ways that investigators should examine, ranging from the quality of images used in training sets and appropriate sample size, as well as the unexpected events that may occur which the algorithm cannot predict.
Collapse
Affiliation(s)
- Julien Issa
- Department of Diagnostics, Poznań University of Medical Sciences, Bukowska 70, 60-812 Poznań, Poland
- Doctoral School, Poznań University of Medical Sciences, Bukowska 70, 60-812 Poznań, Poland
| | - Mazen Abou Chaar
- Department of Anatomy, Poznan University of Medical Sciences, 60-701 Poznan, Poland
| | - Bartosz Kempisty
- Department of Anatomy, Poznan University of Medical Sciences, 60-701 Poznan, Poland
- Prestage Department of Poultry Sciences, North Carolina State University, Raleigh, NC 27695, USA
- Department of Histology and Embryology, Poznan University of Medical Sciences, 60-701 Poznan, Poland
- Department of Veterinary Surgery, Institute of Veterinary Medicine, Nicolaus Copernicus University in Torun, 87-100 Torun, Poland
| | - Lukasz Gasiorowski
- Department of Medical Simulation, Poznan University of Medical Sciences, 60-701 Poznan, Poland
| | - Raphael Olszewski
- Department of Oral and Maxillofacial Surgery, Cliniques Univeristaires Saint-Luc, UCLouvain, 1200 Brussels, Belgium
| | - Paul Mozdziak
- Prestage Department of Poultry Sciences, North Carolina State University, Raleigh, NC 27695, USA
- Physiology Graduate Program, North Carolina State University, Raleigh, NC 27695, USA
| | | |
Collapse
|
19
|
Kusumoto D, Yuasa S, Fukuda K. Induced Pluripotent Stem Cell-Based Drug Screening by Use of Artificial Intelligence. Pharmaceuticals (Basel) 2022; 15:562. [PMID: 35631387 PMCID: PMC9145330 DOI: 10.3390/ph15050562] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 04/25/2022] [Accepted: 04/27/2022] [Indexed: 12/10/2022] Open
Abstract
Induced pluripotent stem cells (iPSCs) are terminally differentiated somatic cells that differentiate into various cell types. iPSCs are expected to be used for disease modeling and for developing novel treatments because differentiated cells from iPSCs can recapitulate the cellular pathology of patients with genetic mutations. However, a barrier to using iPSCs for comprehensive drug screening is the difficulty of evaluating their pathophysiology. Recently, the accuracy of image analysis has dramatically improved with the development of artificial intelligence (AI) technology. In the field of cell biology, it has become possible to estimate cell types and states by examining cellular morphology obtained from simple microscopic images. AI can evaluate disease-specific phenotypes of iPS-derived cells from label-free microscopic images; thus, AI can be utilized for disease-specific drug screening using iPSCs. In addition to image analysis, various AI-based methods can be applied to drug development, including phenotype prediction by analyzing genomic data and virtual screening by analyzing structural formulas and protein-protein interactions of compounds. In the future, combining AI methods may rapidly accelerate drug discovery using iPSCs. In this review, we explain the details of AI technology and the application of AI for iPSC-based drug screening.
Collapse
Affiliation(s)
- Dai Kusumoto
- Department of Cardiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan;
- Center for Preventive Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Shinsuke Yuasa
- Department of Cardiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan;
| | - Keiichi Fukuda
- Department of Cardiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan;
| |
Collapse
|
20
|
Johns AE, Maragakis NJ. Exploring Motor Neuron Diseases Using iPSC Platforms. Stem Cells 2022; 40:2-13. [PMID: 35511862 PMCID: PMC9199844 DOI: 10.1093/stmcls/sxab006] [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: 06/18/2021] [Accepted: 09/17/2021] [Indexed: 01/21/2023]
Abstract
The degeneration of motor neurons is a pathological hallmark of motor neuron diseases (MNDs), but emerging evidence suggests that neuronal vulnerability extends well beyond this cell subtype. The ability to assess motor function in the clinic is limited to physical examination, electrophysiological measures, and tissue-based or neuroimaging techniques which lack the resolution to accurately assess neuronal dysfunction as the disease progresses. Spinal muscular atrophy (SMA), spinal and bulbar muscular atrophy (SBMA), hereditary spastic paraplegia (HSP), and amyotrophic lateral sclerosis (ALS) are all MNDs with devastating clinical outcomes that contribute significantly to disease burden as patients are no longer able to carry out normal activities of daily living. The critical need to accurately assess the cause and progression of motor neuron dysfunction, especially in the early stages of those diseases, has motivated the use of human iPSC-derived motor neurons (hiPSC-MN) to study the neurobiological mechanisms underlying disease pathogenesis and to generate platforms for therapeutic discovery and testing. As our understanding of MNDs has grown, so too has our need to develop more complex in vitro models which include hiPSC-MN co-cultured with relevant non-neuronal cells in 2D as well as in 3D organoid and spheroid systems. These more complex hiPSC-derived culture systems have led to the implementation of new technologies, including microfluidics, multielectrode array, and machine learning which offer novel insights into the functional correlates of these emerging model systems.
Collapse
Affiliation(s)
- Alexandra E Johns
- Department of Neurology, Johns Hopkins University, Baltimore, MD, USA
| | | |
Collapse
|
21
|
Okano H, Morimoto S. iPSC-based disease modeling and drug discovery in cardinal neurodegenerative disorders. Cell Stem Cell 2022; 29:189-208. [PMID: 35120619 DOI: 10.1016/j.stem.2022.01.007] [Citation(s) in RCA: 80] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
It has been 15 years since the birth of human induced pluripotent stem cell (iPSC) technology in 2007, and the scope of its application has been expanding. In addition to the development of cell therapies using iPSC-derived cells, pathological analyses using disease-specific iPSCs and clinical trials to confirm the safety and efficacy of drugs developed using iPSCs are progressing. With the innovation of related technologies, iPSC applications are about to enter a new stage. This review outlines advances in iPSC modeling and therapeutic development for cardinal neurodegenerative diseases, such as amyotrophic lateral sclerosis, Parkinson's disease, and Alzheimer's disease.
Collapse
Affiliation(s)
- Hideyuki Okano
- Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, Japan; Laboratory for Marmoset Neural Architecture, RIKEN Center for Brain Science, Wako-shi, Saitama 351-0198, Japan.
| | - Satoru Morimoto
- Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, Japan
| |
Collapse
|
22
|
Bede P, Murad A, Lope J, Li Hi Shing S, Finegan E, Chipika RH, Hardiman O, Chang KM. Phenotypic categorisation of individual subjects with motor neuron disease based on radiological disease burden patterns: A machine-learning approach. J Neurol Sci 2022; 432:120079. [PMID: 34875472 DOI: 10.1016/j.jns.2021.120079] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 11/25/2021] [Accepted: 11/29/2021] [Indexed: 12/20/2022]
Abstract
Motor neuron disease is an umbrella term encompassing a multitude of clinically heterogeneous phenotypes. The early and accurate categorisation of patients is hugely important, as MND phenotypes are associated with markedly different prognoses, progression rates, care needs and benefit from divergent management strategies. The categorisation of patients shortly after symptom onset is challenging, and often lengthy clinical monitoring is needed to assign patients to the appropriate phenotypic subgroup. In this study, a multi-class machine-learning strategy was implemented to classify 300 patients based on their radiological profile into diagnostic labels along the UMN-LMN spectrum. A comprehensive panel of cortical thickness measures, subcortical grey matter variables, and white matter integrity metrics were evaluated in a multilayer perceptron (MLP) model. Additional exploratory analyses were also carried out using discriminant function analyses (DFA). Excellent classification accuracy was achieved for amyotrophic lateral sclerosis in the testing cohort (93.7%) using the MLP model, but poor diagnostic accuracy was detected for primary lateral sclerosis (43.8%) and poliomyelitis survivors (60%). Feature importance analyses highlighted the relevance of white matter diffusivity metrics and the evaluation of cerebellar indices, cingulate measures and thalamic radiation variables to discriminate MND phenotypes. Our data suggest that radiological data from single patients may be meaningfully interpreted if large training data sets are available and the provision of diagnostic probability outcomes may be clinically useful in patients with short symptom duration. The computational interpretation of multimodal radiology datasets herald viable diagnostic, prognostic and clinical trial applications.
Collapse
Affiliation(s)
- Peter Bede
- Computational Neuroimaging Group, Biomedical Sciences Institute, Trinity College Dublin, Ireland; Pitié-Salpêtrière University Hospital, Sorbonne University, Paris, France.
| | - Aizuri Murad
- Computational Neuroimaging Group, Biomedical Sciences Institute, Trinity College Dublin, Ireland
| | - Jasmin Lope
- Computational Neuroimaging Group, Biomedical Sciences Institute, Trinity College Dublin, Ireland
| | - Stacey Li Hi Shing
- Computational Neuroimaging Group, Biomedical Sciences Institute, Trinity College Dublin, Ireland
| | - Eoin Finegan
- Computational Neuroimaging Group, Biomedical Sciences Institute, Trinity College Dublin, Ireland
| | - Rangariroyashe H Chipika
- Computational Neuroimaging Group, Biomedical Sciences Institute, Trinity College Dublin, Ireland
| | - Orla Hardiman
- Computational Neuroimaging Group, Biomedical Sciences Institute, Trinity College Dublin, Ireland
| | - Kai Ming Chang
- Computational Neuroimaging Group, Biomedical Sciences Institute, Trinity College Dublin, Ireland; Department of Electronics and Computer Science, University of Southampton, UK
| |
Collapse
|
23
|
Ainsbury EA, Moquet J, Sun M, Barnard S, Ellender M, Lloyd D. The future of biological dosimetry in mass casualty radiation emergency response, personalized radiation risk estimation and space radiation protection. Int J Radiat Biol 2021; 98:421-427. [PMID: 34515621 DOI: 10.1080/09553002.2021.1980629] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
PURPOSE The aim of this brief personal, high level review is to consider the state of the art for biological dosimetry for radiation routine and emergency response, and the potential future progress in this fascinating and active field. Four areas in which biomarkers may contribute to scientific advancement through improved dose and exposure characterization, as well as potential contributions to personalized risk estimation, are considered: emergency dosimetry, molecular epidemiology, personalized medical dosimetry, and space travel. CONCLUSION Ionizing radiation biodosimetry is an exciting field which will continue to benefit from active networking and collaboration with the wider fields of radiation research and radiation emergency response to ensure effective, joined up approaches to triage; radiation epidemiology to assess long term, low dose, radiation risk; radiation protection of workers, optimization and justification of radiation for diagnosis or treatment of patients in clinical uses, and protection of individuals traveling to space.
Collapse
Affiliation(s)
- Elizabeth A Ainsbury
- Public Health England, Centre for Radiation, Chemical and Environmental Hazards, Chilton, UK.,Environmental Research Group within the School of Public Health, Faculty of Medicine at Imperial College of Science, Technology and Medicine, London, UK
| | - Jayne Moquet
- Public Health England, Centre for Radiation, Chemical and Environmental Hazards, Chilton, UK
| | - Mingzhu Sun
- Public Health England, Centre for Radiation, Chemical and Environmental Hazards, Chilton, UK
| | - Stephen Barnard
- Public Health England, Centre for Radiation, Chemical and Environmental Hazards, Chilton, UK
| | - Michele Ellender
- Public Health England, Centre for Radiation, Chemical and Environmental Hazards, Chilton, UK
| | - David Lloyd
- Public Health England, Centre for Radiation, Chemical and Environmental Hazards, Chilton, UK
| |
Collapse
|