1
|
Serrano E, Chandrasekaran SN, Bunten D, Brewer KI, Tomkinson J, Kern R, Bornholdt M, Fleming S, Pei R, Arevalo J, Tsang H, Rubinetti V, Tromans-Coia C, Becker T, Weisbart E, Bunne C, Kalinin AA, Senft R, Taylor SJ, Jamali N, Adeboye A, Abbasi HS, Goodman A, Caicedo JC, Carpenter AE, Cimini BA, Singh S, Way GP. Reproducible image-based profiling with Pycytominer. ARXIV 2024:arXiv:2311.13417v2. [PMID: 38045474 PMCID: PMC10690292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Subscribe] [Scholar Register] [Indexed: 12/05/2023]
Abstract
Advances in high-throughput microscopy have enabled the rapid acquisition of large numbers of high-content microscopy images. Whether by deep learning or classical algorithms, image analysis pipelines then produce single-cell features. To process these single-cells for downstream applications, we present Pycytominer, a user-friendly, open-source python package that implements the bioinformatics steps, known as "image-based profiling". We demonstrate Pycytominer's usefulness in a machine learning project to predict nuisance compounds that cause undesirable cell injuries.
Collapse
Affiliation(s)
- Erik Serrano
- Department of Biomedical Informatics, University of Colorado School of Medicine
| | | | - Dave Bunten
- Department of Biomedical Informatics, University of Colorado School of Medicine
| | | | - Jenna Tomkinson
- Department of Biomedical Informatics, University of Colorado School of Medicine
| | - Roshan Kern
- Department of Biomedical Informatics, University of Colorado School of Medicine
- Case Western Reserve University
| | | | | | - Ruifan Pei
- Imaging Platform, Broad Institute of MIT and Harvard
| | - John Arevalo
- Imaging Platform, Broad Institute of MIT and Harvard
| | - Hillary Tsang
- Imaging Platform, Broad Institute of MIT and Harvard
| | - Vincent Rubinetti
- Department of Biomedical Informatics, University of Colorado School of Medicine
| | | | - Tim Becker
- Imaging Platform, Broad Institute of MIT and Harvard
| | - Erin Weisbart
- Imaging Platform, Broad Institute of MIT and Harvard
| | | | | | - Rebecca Senft
- Imaging Platform, Broad Institute of MIT and Harvard
| | - Stephen J. Taylor
- Department of Biomedical Informatics, University of Colorado School of Medicine
| | - Nasim Jamali
- Imaging Platform, Broad Institute of MIT and Harvard
| | | | | | - Allen Goodman
- Imaging Platform, Broad Institute of MIT and Harvard
- Genentech gRED
| | - Juan C. Caicedo
- Imaging Platform, Broad Institute of MIT and Harvard
- Morgridge Institute for Research, University of Wisconsin-Madison
| | | | | | | | - Gregory P. Way
- Department of Biomedical Informatics, University of Colorado School of Medicine
| |
Collapse
|
2
|
Xu C, Alameri A, Leong W, Johnson E, Chen Z, Xu B, Leong KW. Multiscale engineering of brain organoids for disease modeling. Adv Drug Deliv Rev 2024; 210:115344. [PMID: 38810702 PMCID: PMC11265575 DOI: 10.1016/j.addr.2024.115344] [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: 02/13/2024] [Revised: 04/23/2024] [Accepted: 05/25/2024] [Indexed: 05/31/2024]
Abstract
Brain organoids hold great potential for modeling human brain development and pathogenesis. They recapitulate certain aspects of the transcriptional trajectory, cellular diversity, tissue architecture and functions of the developing brain. In this review, we explore the engineering strategies to control the molecular-, cellular- and tissue-level inputs to achieve high-fidelity brain organoids. We review the application of brain organoids in neural disorder modeling and emerging bioengineering methods to improve data collection and feature extraction at multiscale. The integration of multiscale engineering strategies and analytical methods has significant potential to advance insight into neurological disorders and accelerate drug development.
Collapse
Affiliation(s)
- Cong Xu
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - Alia Alameri
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - Wei Leong
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - Emily Johnson
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - Zaozao Chen
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - Bin Xu
- Department of Psychiatry, Columbia University, New York, NY 10032, USA.
| | - Kam W Leong
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA.
| |
Collapse
|
3
|
Tang Q, Ratnayake R, Seabra G, Jiang Z, Fang R, Cui L, Ding Y, Kahveci T, Bian J, Li C, Luesch H, Li Y. Morphological profiling for drug discovery in the era of deep learning. Brief Bioinform 2024; 25:bbae284. [PMID: 38886164 PMCID: PMC11182685 DOI: 10.1093/bib/bbae284] [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: 03/09/2024] [Revised: 05/13/2024] [Accepted: 06/03/2024] [Indexed: 06/20/2024] Open
Abstract
Morphological profiling is a valuable tool in phenotypic drug discovery. The advent of high-throughput automated imaging has enabled the capturing of a wide range of morphological features of cells or organisms in response to perturbations at the single-cell resolution. Concurrently, significant advances in machine learning and deep learning, especially in computer vision, have led to substantial improvements in analyzing large-scale high-content images at high throughput. These efforts have facilitated understanding of compound mechanism of action, drug repurposing, characterization of cell morphodynamics under perturbation, and ultimately contributing to the development of novel therapeutics. In this review, we provide a comprehensive overview of the recent advances in the field of morphological profiling. We summarize the image profiling analysis workflow, survey a broad spectrum of analysis strategies encompassing feature engineering- and deep learning-based approaches, and introduce publicly available benchmark datasets. We place a particular emphasis on the application of deep learning in this pipeline, covering cell segmentation, image representation learning, and multimodal learning. Additionally, we illuminate the application of morphological profiling in phenotypic drug discovery and highlight potential challenges and opportunities in this field.
Collapse
Affiliation(s)
- Qiaosi Tang
- Calico Life Sciences, South San Francisco, CA 94080, United States
| | - Ranjala Ratnayake
- Department of Medicinal Chemistry, Center for Natural Products, Drug Discovery and Development, University of Florida, Gainesville, FL 32610, United States
| | - Gustavo Seabra
- Department of Medicinal Chemistry, Center for Natural Products, Drug Discovery and Development, University of Florida, Gainesville, FL 32610, United States
| | - Zhe Jiang
- Department of Computer & Information Science & Engineering, University of Florida, Gainesville, FL 32611, United States
| | - Ruogu Fang
- Department of Computer & Information Science & Engineering, University of Florida, Gainesville, FL 32611, United States
- J. Crayton Pruitt Family Department of Biomedical Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, FL 32611, United States
| | - Lina Cui
- Department of Medicinal Chemistry, Center for Natural Products, Drug Discovery and Development, University of Florida, Gainesville, FL 32610, United States
| | - Yousong Ding
- Department of Medicinal Chemistry, Center for Natural Products, Drug Discovery and Development, University of Florida, Gainesville, FL 32610, United States
| | - Tamer Kahveci
- Department of Computer & Information Science & Engineering, University of Florida, Gainesville, FL 32611, United States
| | - Jiang Bian
- Department of Health Outcomes and Biomedical Informatics, College of Medicine, University of Florida, Gainesville, FL 32611, United States
| | - Chenglong Li
- Department of Medicinal Chemistry, Center for Natural Products, Drug Discovery and Development, University of Florida, Gainesville, FL 32610, United States
| | - Hendrik Luesch
- Department of Medicinal Chemistry, Center for Natural Products, Drug Discovery and Development, University of Florida, Gainesville, FL 32610, United States
| | - Yanjun Li
- Department of Medicinal Chemistry, Center for Natural Products, Drug Discovery and Development, University of Florida, Gainesville, FL 32610, United States
- Department of Computer & Information Science & Engineering, University of Florida, Gainesville, FL 32611, United States
| |
Collapse
|
4
|
Mikheeva AM, Bogomolov MA, Gasca VA, Sementsov MV, Spirin PV, Prassolov VS, Lebedev TD. Improving the power of drug toxicity measurements by quantitative nuclei imaging. Cell Death Discov 2024; 10:181. [PMID: 38637526 PMCID: PMC11026393 DOI: 10.1038/s41420-024-01950-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 03/29/2024] [Accepted: 04/08/2024] [Indexed: 04/20/2024] Open
Abstract
Imaging-based anticancer drug screens are becoming more prevalent due to development of automated fluorescent microscopes and imaging stations, as well as rapid advancements in image processing software. Automated cell imaging provides many benefits such as their ability to provide high-content data, modularity, dynamics recording and the fact that imaging is the most direct way to access cell viability and cell proliferation. However, currently most publicly available large-scale anticancer drugs screens, such as GDSC, CTRP and NCI-60, provide cell viability data measured by assays based on colorimetric or luminometric measurements of NADH or ATP levels. Although such datasets provide valuable data, it is unclear how well drug toxicity measurements can be integrated with imaging data. Here we explored the relations between drug toxicity data obtained by XTT assay, two quantitative nuclei imaging methods and trypan blue dye exclusion assay using a set of four cancer cell lines with different morphologies and 30 drugs with different mechanisms of action. We show that imaging-based approaches provide high accuracy and the differences between results obtained by different methods highly depend on drug mechanism of action. Selecting AUC metrics over IC50 or comparing data where significantly drugs reduced cell numbers noticeably improves consistency between methods. Using automated cell segmentation protocols we analyzed mitochondria activity in more than 11 thousand drug-treated cells and showed that XTT assay produces unreliable data for CDK4/6, Aurora A, VEGFR and PARP inhibitors due induced cell size growth and increase in individual mitochondria activity. We also explored several benefits of image-based analysis such as ability to monitor cell number dynamics, dissect changes in total and individual mitochondria activity from cell proliferation, and ability to identify chromatin remodeling drugs. Finally, we provide a web tool that allows comparing results obtained by different methods.
Collapse
Affiliation(s)
- Alesya M Mikheeva
- Department of Cancer Cell Biology, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 32 Vavilova str., Moscow, 119991, Russia
- Moscow Institute of Physics and Technology (National Research University), 9 Institutskiy per., Dolgoprudny, Moscow Region, 141701, Russia
| | - Mikhail A Bogomolov
- Department of Cancer Cell Biology, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 32 Vavilova str., Moscow, 119991, Russia
- Moscow Institute of Physics and Technology (National Research University), 9 Institutskiy per., Dolgoprudny, Moscow Region, 141701, Russia
| | - Valentina A Gasca
- Department of Cancer Cell Biology, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 32 Vavilova str., Moscow, 119991, Russia
- Moscow Institute of Physics and Technology (National Research University), 9 Institutskiy per., Dolgoprudny, Moscow Region, 141701, Russia
| | - Mikhail V Sementsov
- Department of Cancer Cell Biology, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 32 Vavilova str., Moscow, 119991, Russia
- Moscow Institute of Physics and Technology (National Research University), 9 Institutskiy per., Dolgoprudny, Moscow Region, 141701, Russia
| | - Pavel V Spirin
- Department of Cancer Cell Biology, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 32 Vavilova str., Moscow, 119991, Russia
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 32 Vavilova str., Moscow, 119991, Russia
| | - Vladimir S Prassolov
- Department of Cancer Cell Biology, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 32 Vavilova str., Moscow, 119991, Russia
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 32 Vavilova str., Moscow, 119991, Russia
| | - Timofey D Lebedev
- Department of Cancer Cell Biology, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 32 Vavilova str., Moscow, 119991, Russia.
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 32 Vavilova str., Moscow, 119991, Russia.
| |
Collapse
|
5
|
Maramraju S, Kowalczewski A, Kaza A, Liu X, Singaraju JP, Albert MV, Ma Z, Yang H. AI-organoid integrated systems for biomedical studies and applications. Bioeng Transl Med 2024; 9:e10641. [PMID: 38435826 PMCID: PMC10905559 DOI: 10.1002/btm2.10641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 12/07/2023] [Accepted: 12/13/2023] [Indexed: 03/05/2024] Open
Abstract
In this review, we explore the growing role of artificial intelligence (AI) in advancing the biomedical applications of human pluripotent stem cell (hPSC)-derived organoids. Stem cell-derived organoids, these miniature organ replicas, have become essential tools for disease modeling, drug discovery, and regenerative medicine. However, analyzing the vast and intricate datasets generated from these organoids can be inefficient and error-prone. AI techniques offer a promising solution to efficiently extract insights and make predictions from diverse data types generated from microscopy images, transcriptomics, metabolomics, and proteomics. This review offers a brief overview of organoid characterization and fundamental concepts in AI while focusing on a comprehensive exploration of AI applications in organoid-based disease modeling and drug evaluation. It provides insights into the future possibilities of AI in enhancing the quality control of organoid fabrication, label-free organoid recognition, and three-dimensional image reconstruction of complex organoid structures. This review presents the challenges and potential solutions in AI-organoid integration, focusing on the establishment of reliable AI model decision-making processes and the standardization of organoid research.
Collapse
Affiliation(s)
- Sudhiksha Maramraju
- Department of Biomedical EngineeringUniversity of North TexasDentonTexasUSA
- Texas Academy of Mathematics and ScienceUniversity of North TexasDentonTexasUSA
| | - Andrew Kowalczewski
- Department of Biomedical & Chemical EngineeringSyracuse UniversitySyracuseNew YorkUSA
- BioInspired Institute for Material and Living SystemsSyracuse UniversitySyracuseNew YorkUSA
| | - Anirudh Kaza
- Department of Biomedical EngineeringUniversity of North TexasDentonTexasUSA
- Texas Academy of Mathematics and ScienceUniversity of North TexasDentonTexasUSA
| | - Xiyuan Liu
- Department of Mechanical & Aerospace EngineeringSyracuse UniversitySyracuseNew YorkUSA
| | - Jathin Pranav Singaraju
- Department of Biomedical EngineeringUniversity of North TexasDentonTexasUSA
- Texas Academy of Mathematics and ScienceUniversity of North TexasDentonTexasUSA
| | - Mark V. Albert
- Department of Biomedical EngineeringUniversity of North TexasDentonTexasUSA
- Department of Computer Science and EngineeringUniversity of North TexasDentonTexasUSA
| | - Zhen Ma
- Department of Biomedical & Chemical EngineeringSyracuse UniversitySyracuseNew YorkUSA
- BioInspired Institute for Material and Living SystemsSyracuse UniversitySyracuseNew YorkUSA
| | - Huaxiao Yang
- Department of Biomedical EngineeringUniversity of North TexasDentonTexasUSA
| |
Collapse
|
6
|
Lau TA, Mair E, Rabbitts BM, Lohith A, Lokey RS. High-Content Image-Based Screening and Deep Learning for the Detection of Anti-Inflammatory Drug Leads. Chembiochem 2024; 25:e202300136. [PMID: 37815526 PMCID: PMC11126213 DOI: 10.1002/cbic.202300136] [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: 02/20/2023] [Revised: 10/02/2023] [Accepted: 10/10/2023] [Indexed: 10/11/2023]
Abstract
We developed a high-content image-based screen that utilizes the pro-inflammatory stimulus lipopolysaccharide (LPS) and murine macrophages (RAW264.7) with the goal of enabling the identification of novel anti-inflammatory lead compounds. We screened 2,259 bioactive compounds with annotated mechanisms of action (MOA) to identify compounds that block the LPS-induced phenotype in macrophages. We utilized a set of seven fluorescence microscopy probes to generate images that were used to train and optimize a deep neural network classifier to distinguish between unstimulated and LPS-stimulated macrophages. The top hits from the deep learning classifier were validated using a linear classifier trained on individual cells and subsequently investigated in a multiplexed cytokine secretion assay. All 12 hits significantly modulated the expression of at least one cytokine upon LPS stimulation. Seven of these were allosteric inhibitors of the mitogen-activated protein kinase kinase (MEK1/2) and showed similar effects on cytokine expression. This deep learning morphological assay identified compounds that modulate the innate immune response to LPS and may aid in identifying new anti-inflammatory drug leads.
Collapse
Affiliation(s)
- Tannia A Lau
- Department of Chemistry and Biochemistry, University of California Santa Cruz, Santa Cruz, CA 95064, USA
| | - Elmar Mair
- No affiliation, Santa Cruz, CA 95060, USA
| | - Beverley M Rabbitts
- Department of Chemistry and Biochemistry, University of California Santa Cruz, Santa Cruz, CA 95064, USA
| | - Akshar Lohith
- Department of Chemistry and Biochemistry, University of California Santa Cruz, Santa Cruz, CA 95064, USA
| | - R Scott Lokey
- Department of Chemistry and Biochemistry, University of California Santa Cruz, Santa Cruz, CA 95064, USA
| |
Collapse
|
7
|
Chai B, Efstathiou C, Yue H, Draviam VM. Opportunities and challenges for deep learning in cell dynamics research. Trends Cell Biol 2023:S0962-8924(23)00228-3. [PMID: 38030542 DOI: 10.1016/j.tcb.2023.10.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 09/30/2023] [Accepted: 10/13/2023] [Indexed: 12/01/2023]
Abstract
The growth of artificial intelligence (AI) has led to an increase in the adoption of computer vision and deep learning (DL) techniques for the evaluation of microscopy images and movies. This adoption has not only addressed hurdles in quantitative analysis of dynamic cell biological processes but has also started to support advances in drug development, precision medicine, and genome-phenome mapping. We survey existing AI-based techniques and tools, as well as open-source datasets, with a specific focus on the computational tasks of segmentation, classification, and tracking of cellular and subcellular structures and dynamics. We summarise long-standing challenges in microscopy video analysis from a computational perspective and review emerging research frontiers and innovative applications for DL-guided automation in cell dynamics research.
Collapse
Affiliation(s)
- Binghao Chai
- School of Biological and Behavioural Sciences, Queen Mary University of London (QMUL), London E1 4NS, UK
| | - Christoforos Efstathiou
- School of Biological and Behavioural Sciences, Queen Mary University of London (QMUL), London E1 4NS, UK
| | - Haoran Yue
- School of Biological and Behavioural Sciences, Queen Mary University of London (QMUL), London E1 4NS, UK
| | - Viji M Draviam
- School of Biological and Behavioural Sciences, Queen Mary University of London (QMUL), London E1 4NS, UK; The Alan Turing Institute, London NW1 2DB, UK.
| |
Collapse
|
8
|
Lamiable A, Champetier T, Leonardi F, Cohen E, Sommer P, Hardy D, Argy N, Massougbodji A, Del Nery E, Cottrell G, Kwon YJ, Genovesio A. Revealing invisible cell phenotypes with conditional generative modeling. Nat Commun 2023; 14:6386. [PMID: 37821450 PMCID: PMC10567685 DOI: 10.1038/s41467-023-42124-6] [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: 07/01/2022] [Accepted: 09/28/2023] [Indexed: 10/13/2023] Open
Abstract
Biological sciences, drug discovery and medicine rely heavily on cell phenotype perturbation and microscope observation. However, most cellular phenotypic changes are subtle and thus hidden from us by natural cell variability: two cells in the same condition already look different. In this study, we show that conditional generative models can be used to transform an image of cells from any one condition to another, thus canceling cell variability. We visually and quantitatively validate that the principle of synthetic cell perturbation works on discernible cases. We then illustrate its effectiveness in displaying otherwise invisible cell phenotypes triggered by blood cells under parasite infection, or by the presence of a disease-causing pathological mutation in differentiated neurons derived from iPSCs, or by low concentration drug treatments. The proposed approach, easy to use and robust, opens the door to more accessible discovery of biological and disease biomarkers.
Collapse
Affiliation(s)
- Alexis Lamiable
- Computational Bioimaging and Bioinformatics, Institut de Biologie de l'Ecole Normale Supérieure, PSL University, 46, rue d'Ulm, 75005, Paris, France
| | - Tiphaine Champetier
- Computational Bioimaging and Bioinformatics, Institut de Biologie de l'Ecole Normale Supérieure, PSL University, 46, rue d'Ulm, 75005, Paris, France
- Ksilink, 16 rue d'Ankara, 67000, Strasbourg, France
| | - Francesco Leonardi
- Computational Bioimaging and Bioinformatics, Institut de Biologie de l'Ecole Normale Supérieure, PSL University, 46, rue d'Ulm, 75005, Paris, France
- Université Paris-Cité, MERIT, IRD, F-75006, Paris, France
| | - Ethan Cohen
- Computational Bioimaging and Bioinformatics, Institut de Biologie de l'Ecole Normale Supérieure, PSL University, 46, rue d'Ulm, 75005, Paris, France
| | - Peter Sommer
- Ksilink, 16 rue d'Ankara, 67000, Strasbourg, France
| | - David Hardy
- Histopathology Platform, Institut Pasteur, F-75015, Paris, France
| | - Nicolas Argy
- Université Paris-Cité, MERIT, IRD, F-75006, Paris, France
- Laboratoire de parasitologie-mycologie, Hôpital Bichat-Claude bernard, APHP, Paris, France
| | | | - Elaine Del Nery
- Biophenics, Institut Curie, PSL Research University, Department of Translational Research, Cell and Tissue Imaging Facility (PICT-IBiSA), 26 rue d'Ulm, 75005, Paris, France
| | | | - Yong-Jun Kwon
- Ksilink, 16 rue d'Ankara, 67000, Strasbourg, France.
- Personalized Therapy Discovery, Department of Oncology, Luxembourg Institute of Health, Dudelange, Luxembourg.
| | - Auguste Genovesio
- Computational Bioimaging and Bioinformatics, Institut de Biologie de l'Ecole Normale Supérieure, PSL University, 46, rue d'Ulm, 75005, Paris, France.
| |
Collapse
|
9
|
Stossi F, Singh PK, Safari K, Marini M, Labate D, Mancini MA. High throughput microscopy and single cell phenotypic image-based analysis in toxicology and drug discovery. Biochem Pharmacol 2023; 216:115770. [PMID: 37660829 DOI: 10.1016/j.bcp.2023.115770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 08/23/2023] [Accepted: 08/25/2023] [Indexed: 09/05/2023]
Abstract
Measuring single cell responses to the universe of chemicals (drugs, natural products, environmental toxicants etc.) is of paramount importance to human health as phenotypic variability in sensing stimuli is a hallmark of biology that is considered during high throughput screening. One of the ways to approach this problem is via high throughput, microscopy-based assays coupled with multi-dimensional single cell analysis methods. Here, we will summarize some of the efforts in this vast and growing field, focusing on phenotypic screens (e.g., Cell Painting), single cell analytics and quality control, with particular attention to environmental toxicology and drug screening. We will discuss advantages and limitations of high throughput assays with various end points and levels of complexity.
Collapse
Affiliation(s)
- Fabio Stossi
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA; GCC Center for Advanced Microscopy and Image Informatics, Houston, TX, USA.
| | - Pankaj K Singh
- GCC Center for Advanced Microscopy and Image Informatics, Houston, TX, USA; Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M University, Houston, TX, USA
| | - Kazem Safari
- GCC Center for Advanced Microscopy and Image Informatics, Houston, TX, USA; Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M University, Houston, TX, USA
| | - Michela Marini
- GCC Center for Advanced Microscopy and Image Informatics, Houston, TX, USA; Department of Mathematics, University of Houston, Houston, TX, USA
| | - Demetrio Labate
- GCC Center for Advanced Microscopy and Image Informatics, Houston, TX, USA; Department of Mathematics, University of Houston, Houston, TX, USA
| | - Michael A Mancini
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA; GCC Center for Advanced Microscopy and Image Informatics, Houston, TX, USA; Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M University, Houston, TX, USA
| |
Collapse
|
10
|
Lejal V, Cerisier N, Rouquié D, Taboureau O. Assessment of Drug-Induced Liver Injury through Cell Morphology and Gene Expression Analysis. Chem Res Toxicol 2023; 36:1456-1470. [PMID: 37652439 PMCID: PMC10523580 DOI: 10.1021/acs.chemrestox.2c00381] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Indexed: 09/02/2023]
Abstract
Drug-induced liver injury (DILI) is a significant concern in drug development, often leading to drug withdrawal. Although many studies aim to identify biomarkers and gene/pathway signatures related to liver toxicity and aim to predict DILI compounds, this remains a challenge in drug discovery. With a strong development of high-content screening/imaging (HCS/HCI) for phenotypic screening, we explored the morphological cell perturbations induced by DILI compounds. In the first step, cell morphological signatures were associated with two datasets of DILI chemicals (DILIRank and eTox). The mechanisms of action were then analyzed for chemicals having transcriptomics data and sharing similar morphological perturbations. Signaling pathways associated with liver toxicity (cell cycle, cell growth, apoptosis, ...) were then captured, and a hypothetical relation between cell morphological perturbations and gene deregulation was illustrated within our analysis. Finally, using the cell morphological signatures, machine learning approaches were developed to predict chemicals with a potential risk of DILI. Some models showed relevant performance with validation set balanced accuracies between 0.645 and 0.739. Overall, our findings demonstrate the utility of combining HCI with transcriptomics data to identify the morphological and gene expression signatures related to DILI chemicals. Moreover, our protocol could be extended to other toxicity end points, offering a promising avenue for comprehensive toxicity assessment in drug discovery.
Collapse
Affiliation(s)
- Vanille Lejal
- Université
Paris Cité, Inserm U1133, CNRS
UMR 8251, 75013, Paris, France
| | - Natacha Cerisier
- Université
Paris Cité, Inserm U1133, CNRS
UMR 8251, 75013, Paris, France
| | - David Rouquié
- Bayer
SAS, Bayer Crop Science, 355 rue Dostoïevski, CS 90153, 06906 Valbonne, Sophia-Antipolis, France
- Université
Côte d’Azur 3IA Interdisciplinary Institute in Artificial Intelligence, 06103 Nice Cedex, France
| | - Olivier Taboureau
- Université
Paris Cité, Inserm U1133, CNRS
UMR 8251, 75013, Paris, France
| |
Collapse
|
11
|
Gerardo‐Nava JL, Jansen J, Günther D, Klasen L, Thiebes AL, Niessing B, Bergerbit C, Meyer AA, Linkhorst J, Barth M, Akhyari P, Stingl J, Nagel S, Stiehl T, Lampert A, Leube R, Wessling M, Santoro F, Ingebrandt S, Jockenhoevel S, Herrmann A, Fischer H, Wagner W, Schmitt RH, Kiessling F, Kramann R, De Laporte L. Transformative Materials to Create 3D Functional Human Tissue Models In Vitro in a Reproducible Manner. Adv Healthc Mater 2023; 12:e2301030. [PMID: 37311209 PMCID: PMC11468549 DOI: 10.1002/adhm.202301030] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 05/21/2023] [Indexed: 06/15/2023]
Abstract
Recreating human tissues and organs in the petri dish to establish models as tools in biomedical sciences has gained momentum. These models can provide insight into mechanisms of human physiology, disease onset, and progression, and improve drug target validation, as well as the development of new medical therapeutics. Transformative materials play an important role in this evolution, as they can be programmed to direct cell behavior and fate by controlling the activity of bioactive molecules and material properties. Using nature as an inspiration, scientists are creating materials that incorporate specific biological processes observed during human organogenesis and tissue regeneration. This article presents the reader with state-of-the-art developments in the field of in vitro tissue engineering and the challenges related to the design, production, and translation of these transformative materials. Advances regarding (stem) cell sources, expansion, and differentiation, and how novel responsive materials, automated and large-scale fabrication processes, culture conditions, in situ monitoring systems, and computer simulations are required to create functional human tissue models that are relevant and efficient for drug discovery, are described. This paper illustrates how these different technologies need to converge to generate in vitro life-like human tissue models that provide a platform to answer health-based scientific questions.
Collapse
|
12
|
Laber S, Strobel S, Mercader JM, Dashti H, dos Santos FR, Kubitz P, Jackson M, Ainbinder A, Honecker J, Agrawal S, Garborcauskas G, Stirling DR, Leong A, Figueroa K, Sinnott-Armstrong N, Kost-Alimova M, Deodato G, Harney A, Way GP, Saadat A, Harken S, Reibe-Pal S, Ebert H, Zhang Y, Calabuig-Navarro V, McGonagle E, Stefek A, Dupuis J, Cimini BA, Hauner H, Udler MS, Carpenter AE, Florez JC, Lindgren C, Jacobs SB, Claussnitzer M. Discovering cellular programs of intrinsic and extrinsic drivers of metabolic traits using LipocyteProfiler. CELL GENOMICS 2023; 3:100346. [PMID: 37492099 PMCID: PMC10363917 DOI: 10.1016/j.xgen.2023.100346] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 08/22/2022] [Accepted: 05/26/2023] [Indexed: 07/27/2023]
Abstract
A primary obstacle in translating genetic associations with disease into therapeutic strategies is elucidating the cellular programs affected by genetic risk variants and effector genes. Here, we introduce LipocyteProfiler, a cardiometabolic-disease-oriented high-content image-based profiling tool that enables evaluation of thousands of morphological and cellular profiles that can be systematically linked to genes and genetic variants relevant to cardiometabolic disease. We show that LipocyteProfiler allows surveillance of diverse cellular programs by generating rich context- and process-specific cellular profiles across hepatocyte and adipocyte cell-state transitions. We use LipocyteProfiler to identify known and novel cellular mechanisms altered by polygenic risk of metabolic disease, including insulin resistance, fat distribution, and the polygenic contribution to lipodystrophy. LipocyteProfiler paves the way for large-scale forward and reverse deep phenotypic profiling in lipocytes and provides a framework for the unbiased identification of causal relationships between genetic variants and cellular programs relevant to human disease.
Collapse
Affiliation(s)
- Samantha Laber
- Programs in Metabolism and Medical and Population Genetics, Type 2 Diabetes Systems Genomics Initiative, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford OX3 7FZ, UK
- Wellcome Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
| | - Sophie Strobel
- Programs in Metabolism and Medical and Population Genetics, Type 2 Diabetes Systems Genomics Initiative, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Institute of Nutritional Medicine, School of Medicine, Technical University of Munich, 85354 Freising-Weihenstephan, Germany
| | - Josep M. Mercader
- Programs in Metabolism and Medical and Population Genetics, Type 2 Diabetes Systems Genomics Initiative, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Diabetes Unit and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
- Department of Medicine, Harvard Medical School, Boston, MA 02114, USA
| | - Hesam Dashti
- Programs in Metabolism and Medical and Population Genetics, Type 2 Diabetes Systems Genomics Initiative, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Department of Medicine, Harvard Medical School, Boston, MA 02114, USA
- The Novo Nordisk Foundation Center for Genomic Mechanisms of Disease, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Felipe R.C. dos Santos
- Programs in Metabolism and Medical and Population Genetics, Type 2 Diabetes Systems Genomics Initiative, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- The Novo Nordisk Foundation Center for Genomic Mechanisms of Disease, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Phil Kubitz
- Programs in Metabolism and Medical and Population Genetics, Type 2 Diabetes Systems Genomics Initiative, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Else Kröner-Fresenius-Centre for Nutritional Medicine, School of Life Sciences, Technical University of Munich, 85354 Freising-Weihenstephan, Germany
- The Novo Nordisk Foundation Center for Genomic Mechanisms of Disease, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Maya Jackson
- Programs in Metabolism and Medical and Population Genetics, Type 2 Diabetes Systems Genomics Initiative, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- The Novo Nordisk Foundation Center for Genomic Mechanisms of Disease, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Alina Ainbinder
- Programs in Metabolism and Medical and Population Genetics, Type 2 Diabetes Systems Genomics Initiative, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Julius Honecker
- Else Kröner-Fresenius-Centre for Nutritional Medicine, School of Life Sciences, Technical University of Munich, 85354 Freising-Weihenstephan, Germany
| | - Saaket Agrawal
- Programs in Metabolism and Medical and Population Genetics, Type 2 Diabetes Systems Genomics Initiative, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Garrett Garborcauskas
- Programs in Metabolism and Medical and Population Genetics, Type 2 Diabetes Systems Genomics Initiative, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - David R. Stirling
- Imaging Platform, Center for the Development of Therapeutics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Aaron Leong
- Programs in Metabolism and Medical and Population Genetics, Type 2 Diabetes Systems Genomics Initiative, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Diabetes Unit and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
- Department of Medicine, Harvard Medical School, Boston, MA 02114, USA
| | - Katherine Figueroa
- Programs in Metabolism and Medical and Population Genetics, Type 2 Diabetes Systems Genomics Initiative, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Diabetes Unit and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Nasa Sinnott-Armstrong
- Programs in Metabolism and Medical and Population Genetics, Type 2 Diabetes Systems Genomics Initiative, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Department of Genetics, Stanford University, San Francisco, CA, USA
| | - Maria Kost-Alimova
- Imaging Platform, Center for the Development of Therapeutics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Giacomo Deodato
- Programs in Metabolism and Medical and Population Genetics, Type 2 Diabetes Systems Genomics Initiative, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Alycen Harney
- Programs in Metabolism and Medical and Population Genetics, Type 2 Diabetes Systems Genomics Initiative, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Gregory P. Way
- Imaging Platform, Center for the Development of Therapeutics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Alham Saadat
- Programs in Metabolism and Medical and Population Genetics, Type 2 Diabetes Systems Genomics Initiative, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Sierra Harken
- Programs in Metabolism and Medical and Population Genetics, Type 2 Diabetes Systems Genomics Initiative, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Saskia Reibe-Pal
- Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford OX3 7FZ, UK
| | - Hannah Ebert
- Institute of Nutritional Science, University Hohenheim, 70599 Stuttgart, Germany
| | - Yixin Zhang
- Department of Biostatistics, Boston University School of Public Health, Boston, MA 02118, USA
| | - Virtu Calabuig-Navarro
- Programs in Metabolism and Medical and Population Genetics, Type 2 Diabetes Systems Genomics Initiative, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Institute of Nutritional Science, University Hohenheim, 70599 Stuttgart, Germany
| | - Elizabeth McGonagle
- Programs in Metabolism and Medical and Population Genetics, Type 2 Diabetes Systems Genomics Initiative, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Adam Stefek
- Programs in Metabolism and Medical and Population Genetics, Type 2 Diabetes Systems Genomics Initiative, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Josée Dupuis
- Department of Biostatistics, Boston University School of Public Health, Boston, MA 02118, USA
- Department of Epidemiology, Biostatistics and Occupational Health, McGill University, Montreal, QC H3A 1G1, Canada
| | - Beth A. Cimini
- Programs in Metabolism and Medical and Population Genetics, Type 2 Diabetes Systems Genomics Initiative, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Hans Hauner
- Institute of Nutritional Medicine, School of Medicine, Technical University of Munich, 85354 Freising-Weihenstephan, Germany
- Else Kröner-Fresenius-Centre for Nutritional Medicine, School of Life Sciences, Technical University of Munich, 85354 Freising-Weihenstephan, Germany
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany
| | - Miriam S. Udler
- Programs in Metabolism and Medical and Population Genetics, Type 2 Diabetes Systems Genomics Initiative, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Diabetes Unit and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
- Department of Medicine, Harvard Medical School, Boston, MA 02114, USA
| | - Anne E. Carpenter
- Imaging Platform, Center for the Development of Therapeutics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Jose C. Florez
- Programs in Metabolism and Medical and Population Genetics, Type 2 Diabetes Systems Genomics Initiative, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Diabetes Unit and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
- Department of Medicine, Harvard Medical School, Boston, MA 02114, USA
| | - Cecilia Lindgren
- Programs in Metabolism and Medical and Population Genetics, Type 2 Diabetes Systems Genomics Initiative, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford OX3 7FZ, UK
- Wellcome Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
| | - Suzanne B.R. Jacobs
- Programs in Metabolism and Medical and Population Genetics, Type 2 Diabetes Systems Genomics Initiative, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Diabetes Unit and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Melina Claussnitzer
- Programs in Metabolism and Medical and Population Genetics, Type 2 Diabetes Systems Genomics Initiative, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
- Diabetes Unit and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
- Department of Medicine, Harvard Medical School, Boston, MA 02114, USA
- The Novo Nordisk Foundation Center for Genomic Mechanisms of Disease, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| |
Collapse
|
13
|
Rathore D, Marino MJ, Nita-Lazar A. Omics and systems view of innate immune pathways. Proteomics 2023; 23:e2200407. [PMID: 37269203 DOI: 10.1002/pmic.202200407] [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: 02/14/2023] [Revised: 04/16/2023] [Accepted: 05/23/2023] [Indexed: 06/04/2023]
Abstract
Multiomics approaches to studying systems biology are very powerful techniques that can elucidate changes in the genomic, transcriptomic, proteomic, and metabolomic levels within a cell type in response to an infection. These approaches are valuable for understanding the mechanisms behind disease pathogenesis and how the immune system responds to being challenged. With the emergence of the COVID-19 pandemic, the importance and utility of these tools have become evident in garnering a better understanding of the systems biology within the innate and adaptive immune response and for developing treatments and preventative measures for new and emerging pathogens that pose a threat to human health. In this review, we focus on state-of-the-art omics technologies within the scope of innate immunity.
Collapse
Affiliation(s)
- Deepali Rathore
- Functional Cellular Networks Section, Laboratory of Immune Systems Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Matthew J Marino
- Functional Cellular Networks Section, Laboratory of Immune Systems Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Aleksandra Nita-Lazar
- Functional Cellular Networks Section, Laboratory of Immune Systems Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
14
|
Joos-Vandewalle J, Steenkamp V, Prinsloo E. A simplified workflow with end-point validation of real-time electrical cell-substrate impedance sensing of retinoic acid stimulated neurogenesis in human SH-SY5Y cells in vitro. BMC Res Notes 2023; 16:93. [PMID: 37264464 DOI: 10.1186/s13104-023-06369-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 05/25/2023] [Indexed: 06/03/2023] Open
Abstract
OBJECTIVE Retinoic acid (RA) is known to transition proliferating SH-SY5Y neuroblastoma cells towards functional neurons. However, the activity of RA is restricted due to its photolability where any findings from prolonged time course observations using microscopy may alter outcomes. The aim of the study was to establish a real-time, long-term (9-day) protocol for the screening of differentiation events using Electrical cell-substrate impedance sensing (ECIS). RESULTS AND DISCUSSION A differentiation baseline for SH-SY5Y cells was established. Cells were seeded and exposed to repeated spikes of RA using the xCELLigence real-time cell analyser single plate (RTCA-SP) for real-time monitoring and identification of differentiation activity over a 9 day period in order to be more representative of differentiation over a prolonged timeline. Specific features associated with differentiation (growth inhibition, neurite outgrowths) were confirmed by end-point analysis. RA-induced growth inhibition and assumed phenotypic changes (i.e. neurite outgrowth) were identified by the xCELLigence analysis and further confirmed by end-point metabolic and phenotypic assays. Change in cellular morphology and neurite outgrowth length was identified by end-point fluorescence detection followed by computational analysis. Based on this it was possible to identify SH-SY5Y phenotypic differentiation with distinct phases observed over 9 days using Electric cell-substrate impedance sensing (ECIS) cell index traces providing a path to application in larger scale neurotrophic factor screening using this scalable technology.
Collapse
Affiliation(s)
- Julia Joos-Vandewalle
- Biotechnology Innovation Centre, Rhodes University, P.O. Box 94, Makhanda, 6140, South Africa
| | - Vanessa Steenkamp
- Department of Pharmacology, Faculty of Health Sciences, University of Pretoria, Private Bag X323, Arcadia, 0007, South Africa
| | - Earl Prinsloo
- Biotechnology Innovation Centre, Rhodes University, P.O. Box 94, Makhanda, 6140, South Africa.
| |
Collapse
|
15
|
Heinrich L, Kumbier K, Li L, Altschuler SJ, Wu LF. Selection of Optimal Cell Lines for High-Content Phenotypic Screening. ACS Chem Biol 2023; 18:679-685. [PMID: 36920184 PMCID: PMC10127200 DOI: 10.1021/acschembio.2c00878] [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: 11/28/2022] [Accepted: 03/07/2023] [Indexed: 03/16/2023]
Abstract
High-content microscopy offers a scalable approach to screen against multiple targets in a single pass. Prior work has focused on methods to select "optimal" cellular readouts in microscopy screens. However, methods to select optimal cell line models have garnered much less attention. Here, we provide a roadmap for how to select the cell line or lines that are best suited to identify bioactive compounds and their mechanism of action (MOA). We test our approach on compounds targeting cancer-relevant pathways, ranking cell lines in two tasks: detecting compound activity ("phenoactivity") and grouping compounds with similar MOA by similar phenotype ("phenosimilarity"). Evaluating six cell lines across 3214 well-annotated compounds, we show that optimal cell line selection depends on both the task of interest (e.g., detecting phenoactivity vs inferring phenosimilarity) and distribution of MOAs within the compound library. Given a task of interest and a set of compounds, we provide a systematic framework for choosing optimal cell line(s). Our framework can be used to reduce the number of cell lines required to identify hits within a compound library and help accelerate the pace of early drug discovery.
Collapse
Affiliation(s)
- Louise Heinrich
- Department
of Pharmaceutical Chemistry, University
of California San Francisco, San Fancisco, California 94158, United States
| | - Karl Kumbier
- Department
of Pharmaceutical Chemistry, University
of California San Francisco, San Fancisco, California 94158, United States
| | - Li Li
- Department
of Pharmaceutical Chemistry, University
of California San Francisco, San Fancisco, California 94158, United States
| | - Steven J. Altschuler
- Department
of Pharmaceutical Chemistry, University
of California San Francisco, San Fancisco, California 94158, United States
| | - Lani F. Wu
- Department
of Pharmaceutical Chemistry, University
of California San Francisco, San Fancisco, California 94158, United States
| |
Collapse
|
16
|
Rao SPS, Manjunatha UH, Mikolajczak S, Ashigbie PG, Diagana TT. Drug discovery for parasitic diseases: powered by technology, enabled by pharmacology, informed by clinical science. Trends Parasitol 2023; 39:260-271. [PMID: 36803572 DOI: 10.1016/j.pt.2023.01.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/19/2023] [Accepted: 01/25/2023] [Indexed: 02/22/2023]
Abstract
While prevention is a bedrock of public health, innovative therapeutics are needed to complement the armamentarium of interventions required to achieve disease control and elimination targets for neglected diseases. Extraordinary advances in drug discovery technologies have occurred over the past decades, along with accumulation of scientific knowledge and experience in pharmacological and clinical sciences that are transforming many aspects of drug R&D across disciplines. We reflect on how these advances have propelled drug discovery for parasitic infections, focusing on malaria, kinetoplastid diseases, and cryptosporidiosis. We also discuss challenges and research priorities to accelerate discovery and development of urgently needed novel antiparasitic drugs.
Collapse
Affiliation(s)
| | | | | | - Paul G Ashigbie
- Novartis Institute for Tropical Diseases, Emeryville, CA, USA.
| | | |
Collapse
|
17
|
Papandreou A, Luft C, Barral S, Kriston-Vizi J, Kurian MA, Ketteler R. Automated high-content imaging in iPSC-derived neuronal progenitors. SLAS DISCOVERY : ADVANCING LIFE SCIENCES R & D 2023; 28:42-51. [PMID: 36610640 PMCID: PMC10602900 DOI: 10.1016/j.slasd.2022.12.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 12/18/2022] [Accepted: 12/31/2022] [Indexed: 01/07/2023]
Abstract
Induced pluripotent stem cells (iPSCs) have great potential as physiological disease models for human disorders where access to primary cells is difficult, such as neurons. In recent years, many protocols have been developed for the generation of iPSCs and the differentiation into specialised cell subtypes of interest. More recently, these models have been modified to allow large-scale phenotyping and high-content screening of small molecule compounds in iPSC-derived neuronal cells. Here, we describe the automated seeding of day 11 ventral midbrain progenitor cells into 96-well plates, administration of compounds, automated staining for immunofluorescence, the acquisition of images on a high-content screening platform and workflows for image analysis.
Collapse
Affiliation(s)
- Apostolos Papandreou
- University College London MRC Laboratory for Molecular Cell Biology, London, UK; Developmental Neurosciences, Zayed Centre for Research into Rare Disease in Children, University College London Great Ormond Street Institute of Child Health, London, UK
| | - Christin Luft
- University College London MRC Laboratory for Molecular Cell Biology, London, UK
| | - Serena Barral
- Developmental Neurosciences, Zayed Centre for Research into Rare Disease in Children, University College London Great Ormond Street Institute of Child Health, London, UK
| | - Janos Kriston-Vizi
- University College London MRC Laboratory for Molecular Cell Biology, London, UK
| | - Manju A Kurian
- Developmental Neurosciences, Zayed Centre for Research into Rare Disease in Children, University College London Great Ormond Street Institute of Child Health, London, UK
| | - Robin Ketteler
- University College London MRC Laboratory for Molecular Cell Biology, London, UK
| |
Collapse
|
18
|
Samernate T, Htoo HH, Sugie J, Chavasiri W, Pogliano J, Chaikeeratisak V, Nonejuie P. High-Resolution Bacterial Cytological Profiling Reveals Intrapopulation Morphological Variations upon Antibiotic Exposure. Antimicrob Agents Chemother 2023; 67:e0130722. [PMID: 36625642 PMCID: PMC9933734 DOI: 10.1128/aac.01307-22] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 12/05/2022] [Indexed: 01/11/2023] Open
Abstract
Phenotypic heterogeneity is crucial to bacterial survival and could provide insights into the mechanism of action (MOA) of antibiotics, especially those with polypharmacological actions. Although phenotypic changes among individual cells could be detected by existing profiling methods, due to the data complexity, only population average data were commonly used, thereby overlooking the heterogeneity. In this study, we developed a high-resolution bacterial cytological profiling method that can capture morphological variations of bacteria upon antibiotic treatment. With an unprecedented single-cell resolution, this method classifies morphological changes of individual cells into known MOAs with an overall accuracy above 90%. We next showed that combinations of two antibiotics induce altered cell morphologies that are either unique or similar to that of an antibiotic in the combinations. With these combinatorial profiles, this method successfully revealed multiple cytological changes caused by a natural product-derived compound that, by itself, is inactive against Acinetobacter baumannii but synergistically exerts its multiple antibacterial activities in the presence of colistin. The findings have paved the way for future single-cell profiling in bacteria and have highlighted previously underappreciated intrapopulation variations caused by antibiotic perturbation.
Collapse
Affiliation(s)
- Thanadon Samernate
- Institute of Molecular Biosciences, Mahidol University, Nakhon Pathom, Thailand
| | - Htut Htut Htoo
- Institute of Molecular Biosciences, Mahidol University, Nakhon Pathom, Thailand
| | - Joseph Sugie
- Division of Biological Sciences, University of California, San Diego, La Jolla, California, USA
| | - Warinthorn Chavasiri
- Center of Excellence in Natural Products Chemistry, Department of Chemistry, Chulalongkorn University, Bangkok, Thailand
| | - Joe Pogliano
- Division of Biological Sciences, University of California, San Diego, La Jolla, California, USA
| | | | - Poochit Nonejuie
- Institute of Molecular Biosciences, Mahidol University, Nakhon Pathom, Thailand
| |
Collapse
|
19
|
Pierozan P, Kosnik M, Karlsson O. High-content analysis shows synergistic effects of low perfluorooctanoic acid (PFOS) and perfluorooctane sulfonic acid (PFOA) mixture concentrations on human breast epithelial cell carcinogenesis. ENVIRONMENT INTERNATIONAL 2023; 172:107746. [PMID: 36731186 DOI: 10.1016/j.envint.2023.107746] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 01/04/2023] [Accepted: 01/09/2023] [Indexed: 06/18/2023]
Abstract
Perfluoroalkyl substances (PFAS) have been associated with cancer, but the potential underlying mechanisms need to be further elucidated and include studies of PFAS mixtures. This mechanistic study revealed that very low concentrations (500 pM) of the binary PFOS and PFOA mixture induced synergistic effects on human epithelial breast cell (MCF-10A) proliferation. The cell proliferation was mediated by pregnane X receptor (PXR) activation, an increase in cyclin D1 and CDK6/4 levels, decrease in p21 and p53 levels, and by regulation of phosphor-Akt and β-catenin. The PFAS mixture also altered histone modifications, epigenetic mechanisms implicated in tumorigenesis, and promoted cell migration and invasion by reducing the levels of occludin. High-content screening using the cell painting assay, revealed that hundreds of cell features were affected by the PFAS mixture even at the lowest concentration tested (100 pM). The detailed phenotype profiling further demonstrated that the PFAS mixture altered cell morphology, mostly in parameters related to intensity and texture associated with mitochondria, endoplasmic reticulum, and nucleoli. Exposure to higher concentrations (≥50 µM) of the PFOS and PFOA mixture caused cell death through synergistic interactions that induced oxidative stress, DNA/RNA damage, and lipid peroxidation, illustrating the complexity of mixture toxicology. Increased knowledge about mixture-induced effects is important for better understanding of PFAS' possible role in cancer etiology, and may impact the risk assessment of these and other compounds. This study shows the potential of image-based multiplexed fluorescence assays and high-content screening for development of new approach methodologies in toxicology.
Collapse
Affiliation(s)
- Paula Pierozan
- Science for Life Laboratory, Department of Environmental Science, Stockholm University, Stockholm 114 18, Sweden
| | - Marissa Kosnik
- Science for Life Laboratory, Department of Environmental Science, Stockholm University, Stockholm 114 18, Sweden
| | - Oskar Karlsson
- Science for Life Laboratory, Department of Environmental Science, Stockholm University, Stockholm 114 18, Sweden.
| |
Collapse
|
20
|
Heinrich L, Kumbier K, Li L, Altschuler SP, Wu LF. Selection of optimal cell lines for high-content phenotypic screening. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.11.523662. [PMID: 36711978 PMCID: PMC9882115 DOI: 10.1101/2023.01.11.523662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
High-content microscopy offers a scalable approach to screen against multiple targets in a single pass. Prior work has focused on methods to select "optimal" cellular readouts in microscopy screens. However, methods to select optimal cell line models have garnered much less attention. Here, we provide a roadmap for how to select the cell line or lines that are best suited to identify bioactive compounds and their mechanism of action (MOA). We test our approach on compounds targeting cancer-relevant pathways, ranking cell lines in two tasks: detecting compound activity ("phenoactivity") and grouping compounds with similar MOA by similar phenotype ("phenosimilarity"). Evaluating six cell lines across 3214 well-annotated compounds, we show that optimal cell line selection depends on both the task of interest (e.g. detecting phenoactivity vs. inferring phenosimilarity) and distribution of MOAs within the compound library. Given a task of interest and set of compounds, we provide a systematic framework for choosing optimal cell line(s). Our framework can be used to reduce the number of cell lines required to identify hits within a compound library and help accelerate the pace of early drug discovery.
Collapse
Affiliation(s)
- Louise Heinrich
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Fancisco, California, 94158, United States
| | - Karl Kumbier
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Fancisco, California, 94158, United States
| | - Li Li
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Fancisco, California, 94158, United States
| | - Steven P Altschuler
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Fancisco, California, 94158, United States
| | - Lani F Wu
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Fancisco, California, 94158, United States
| |
Collapse
|
21
|
Qi R, Zou Q. Trends and Potential of Machine Learning and Deep Learning in Drug Study at Single-Cell Level. RESEARCH (WASHINGTON, D.C.) 2023; 6:0050. [PMID: 36930772 PMCID: PMC10013796 DOI: 10.34133/research.0050] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Accepted: 12/27/2022] [Indexed: 01/12/2023]
Abstract
Cancer treatments always face challenging problems, particularly drug resistance due to tumor cell heterogeneity. The existing datasets include the relationship between gene expression and drug sensitivities; however, the majority are based on tissue-level studies. Study drugs at the single-cell level are perspective to overcome minimal residual disease caused by subclonal resistant cancer cells retained after initial curative therapy. Fortunately, machine learning techniques can help us understand how different types of cells respond to different cancer drugs from the perspective of single-cell gene expression. Good modeling using single-cell data and drug response information will not only improve machine learning for cell-drug outcome prediction but also facilitate the discovery of drugs for specific cancer subgroups and specific cancer treatments. In this paper, we review machine learning and deep learning approaches in drug research. By analyzing the application of these methods on cancer cell lines and single-cell data and comparing the technical gap between single-cell sequencing data analysis and single-cell drug sensitivity analysis, we hope to explore the trends and potential of drug research at the single-cell data level and provide more inspiration for drug research at the single-cell level. We anticipate that this review will stimulate the innovative use of machine learning methods to address new challenges in precision medicine more broadly.
Collapse
Affiliation(s)
- Ren Qi
- Yangtze Delta Region Institute (Quzhou), University of Electronic Science and Technology of China, Quzhou 324000, China.,School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, China
| | - Quan Zou
- Yangtze Delta Region Institute (Quzhou), University of Electronic Science and Technology of China, Quzhou 324000, China.,Institute of Fundamental and Frontier Sciences, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
22
|
Toth T, Bauer D, Sukosd F, Horvath P. Fisheye transformation enhances deep-learning-based single-cell phenotyping by including cellular microenvironment. CELL REPORTS METHODS 2022; 2:100339. [PMID: 36590690 PMCID: PMC9795324 DOI: 10.1016/j.crmeth.2022.100339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 08/22/2022] [Accepted: 10/21/2022] [Indexed: 11/23/2022]
Abstract
Incorporating information about the surroundings can have a significant impact on successfully determining the class of an object. This is of particular interest when determining the phenotypes of cells, for example, in the context of high-throughput screens. We hypothesized that an ideal approach would consider the fully featured view of the cell of interest, include its neighboring microenvironment, and give lesser weight to cells that are far from the cell of interest. To satisfy these criteria, we present an approach with a transformation similar to those characteristic of fisheye cameras. Using this transformation with proper settings, we could significantly increase the accuracy of single-cell phenotyping, both in the case of cell culture and tissue-based microscopy images, and we present improved results on a dataset containing images of wild animals.
Collapse
Affiliation(s)
- Timea Toth
- Synthetic and Systems Biology Unit, Biological Research Centre, Eötvös Loránd Research Network, Szeged, Hungary
- Doctoral School of Biology, University of Szeged, Szeged, Hungary
| | - David Bauer
- Synthetic and Systems Biology Unit, Biological Research Centre, Eötvös Loránd Research Network, Szeged, Hungary
| | - Farkas Sukosd
- Department of Pathology, University of Szeged, Szeged, Hungary
| | - Peter Horvath
- Synthetic and Systems Biology Unit, Biological Research Centre, Eötvös Loránd Research Network, Szeged, Hungary
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
- Single-Cell Technologies, Inc., Szeged, Hungary
| |
Collapse
|
23
|
Baručić D, Kaushik S, Kybic J, Stanková J, Džubák P, Hajdúch M. Characterization of drug effects on cell cultures from phase-contrast microscopy images. Comput Biol Med 2022; 151:106171. [PMID: 36306582 DOI: 10.1016/j.compbiomed.2022.106171] [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: 05/26/2022] [Revised: 08/30/2022] [Accepted: 10/01/2022] [Indexed: 12/27/2022]
Abstract
In this work, we classify chemotherapeutic agents (topoisomerase inhibitors) based on their effect on U-2 OS cells. We use phase-contrast microscopy images, which are faster and easier to obtain than fluorescence images and support live cell imaging. We use a convolutional neural network (CNN) trained end-to-end directly on the input images without requiring for manual segmentations or any other auxiliary data. Our method can distinguish between tested cytotoxic drugs with an accuracy of 98%, provided that their mechanism of action differs, outperforming previous work. The results are even better when substance-specific concentrations are used. We show the benefit of sharing the extracted features over all classes (drugs). Finally, a 2D visualization of these features reveals clusters, which correspond well to known class labels, suggesting the possible use of our methodology for drug discovery application in analyzing new, unseen drugs.
Collapse
Affiliation(s)
- Denis Baručić
- Faculty of Electrical Engineering, Czech Technical University in Prague, Prague, Czech Republic.
| | - Sumit Kaushik
- Faculty of Electrical Engineering, Czech Technical University in Prague, Prague, Czech Republic.
| | - Jan Kybic
- Faculty of Electrical Engineering, Czech Technical University in Prague, Prague, Czech Republic.
| | - Jarmila Stanková
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| | - Petr Džubák
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| | - Marián Hajdúch
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| |
Collapse
|
24
|
Berker Y, ElHarouni D, Peterziel H, Fiesel P, Witt O, Oehme I, Schlesner M, Oppermann S. Patient-by-Patient Deep Transfer Learning for Drug-Response Profiling Using Confocal Fluorescence Microscopy of Pediatric Patient-Derived Tumor-Cell Spheroids. IEEE TRANSACTIONS ON MEDICAL IMAGING 2022; 41:3981-3999. [PMID: 36099221 DOI: 10.1109/tmi.2022.3205554] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Image-based phenotypic drug profiling is receiving increasing attention in drug discovery and precision medicine. Compared to classical end-point measurements quantifying drug response, image-based profiling enables both the quantification of drug response and characterization of disease entities and drug-induced cell-death phenotypes. Here, we aim to quantify image-based drug responses in patient-derived 3D spheroid tumor cell cultures, tackling the challenges of a lack of single-cell-segmentation methods and limited patient-derived material. Therefore, we investigate deep transfer learning with patient-by-patient fine-tuning for cell-viability quantification. We fine-tune a convolutional neural network (pre-trained on ImageNet) with 210 control images specific to a single training cell line and 54 additional screen -specific assay control images. This method of image-based drug profiling is validated on 6 cell lines with known drug sensitivities, and further tested with primary patient-derived samples in a medium-throughput setting. Network outputs at different drug concentrations are used for drug-sensitivity scoring, and dense-layer activations are used in t-distributed stochastic neighbor embeddings of drugs to visualize groups of drugs with similar cell-death phenotypes. Image-based cell-line experiments show strong correlation to metabolic results ( R ≈ 0.7 ) and confirm expected hits, indicating the predictive power of deep learning to identify drug-hit candidates for individual patients. In patient-derived samples, combining drug sensitivity scoring with phenotypic analysis may provide opportunities for complementary combination treatments. Deep transfer learning with patient-by-patient fine-tuning is a promising, segmentation-free image-analysis approach for precision medicine and drug discovery.
Collapse
|
25
|
Nevarez AJ, Hao N. Quantitative cell imaging approaches to metastatic state profiling. Front Cell Dev Biol 2022; 10:1048630. [PMID: 36393865 PMCID: PMC9640958 DOI: 10.3389/fcell.2022.1048630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 10/13/2022] [Indexed: 11/13/2022] Open
Abstract
Genetic heterogeneity of metastatic dissemination has proven challenging to identify exploitable markers of metastasis; this bottom-up approach has caused a stalemate between advances in metastasis and the late stage of the disease. Advancements in quantitative cellular imaging have allowed the detection of morphological phenotype changes specific to metastasis, the morphological changes connected to the underlying complex signaling pathways, and a robust readout of metastatic cell state. This review focuses on the recent machine and deep learning developments to gain detailed information about the metastatic cell state using light microscopy. We describe the latest studies using quantitative cell imaging approaches to identify cell appearance-based metastatic patterns. We discuss how quantitative cancer biologists can use these frameworks to work backward toward exploitable hidden drivers in the metastatic cascade and pioneering new Frontier drug discoveries specific for metastasis.
Collapse
Affiliation(s)
| | - Nan Hao
- *Correspondence: Andres J. Nevarez, ; Nan Hao,
| |
Collapse
|
26
|
Ceran Y, Ergüder H, Ladner K, Korenfeld S, Deniz K, Padmanabhan S, Wong P, Baday M, Pengo T, Lou E, Patel CB. TNTdetect.AI: A Deep Learning Model for Automated Detection and Counting of Tunneling Nanotubes in Microscopy Images. Cancers (Basel) 2022; 14:4958. [PMID: 36230881 PMCID: PMC9562025 DOI: 10.3390/cancers14194958] [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: 06/05/2022] [Revised: 09/22/2022] [Accepted: 09/30/2022] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Tunneling nanotubes (TNTs) are cellular structures connecting cell membranes and mediating intercellular communication. TNTs are manually identified and counted by a trained investigator; however, this process is time-intensive. We therefore sought to develop an automated approach for quantitative analysis of TNTs. METHODS We used a convolutional neural network (U-Net) deep learning model to segment phase contrast microscopy images of both cancer and non-cancer cells. Our method was composed of preprocessing and model development. We developed a new preprocessing method to label TNTs on a pixel-wise basis. Two sequential models were employed to detect TNTs. First, we identified the regions of images with TNTs by implementing a classification algorithm. Second, we fed parts of the image classified as TNT-containing into a modified U-Net model to estimate TNTs on a pixel-wise basis. RESULTS The algorithm detected 49.9% of human expert-identified TNTs, counted TNTs, and calculated the number of TNTs per cell, or TNT-to-cell ratio (TCR); it detected TNTs that were not originally detected by the experts. The model had 0.41 precision, 0.26 recall, and 0.32 f-1 score on a test dataset. The predicted and true TCRs were not significantly different across the training and test datasets (p = 0.78). CONCLUSIONS Our automated approach labeled and detected TNTs and cells imaged in culture, resulting in comparable TCRs to those determined by human experts. Future studies will aim to improve on the accuracy, precision, and recall of the algorithm.
Collapse
Affiliation(s)
- Yasin Ceran
- School of Information Systems and Technology, San José State University, San José, CA 95192, USA
- Department of Management Information Systems, Korea Advanced Institute of Science and Technology, Daejeon 34141, Korea
| | - Hamza Ergüder
- Department of Electronics and Communication Engineering, Yildiz Technical University, 34349 Istanbul, Turkey
| | - Katherine Ladner
- Department of Medicine Division of Hematology, Oncology and Transplantation, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Sophie Korenfeld
- Department of Medicine Division of Hematology, Oncology and Transplantation, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Karina Deniz
- Department of Medicine Division of Hematology, Oncology and Transplantation, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Sanyukta Padmanabhan
- Department of Medicine Division of Hematology, Oncology and Transplantation, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Phillip Wong
- Department of Medicine Division of Hematology, Oncology and Transplantation, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Murat Baday
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA
- Precision Health and Integrated Diagnostics Center, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Thomas Pengo
- Informatics Institute, University of Minnesota, Minneapolis, MN 55455, USA
| | - Emil Lou
- Department of Medicine Division of Hematology, Oncology and Transplantation, University of Minnesota Medical School, Minneapolis, MN 55455, USA
- Masonic Cancer Center, Minneapolis, MN 55455, USA
| | - Chirag B. Patel
- Department of Neuro-Oncology, MD Anderson Cancer Center, The University of Texas System, Houston, TX 77030, USA
- Neuroscience Graduate Program, MD Anderson UTHealth Graduate School of Biomedical Sciences, Houston, TX 77030, USA
- Cancer Biology Graduate Program, MD Anderson UTHealth Graduate School of Biomedical Sciences, Houston, TX 77030, USA
| |
Collapse
|
27
|
Zhang X, Kupczyk E, Schmitt-Kopplin P, Mueller C. Current and future approaches for in vitro hit discovery in diabetes mellitus. Drug Discov Today 2022; 27:103331. [PMID: 35926826 DOI: 10.1016/j.drudis.2022.07.016] [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: 02/04/2022] [Revised: 06/10/2022] [Accepted: 07/26/2022] [Indexed: 12/15/2022]
Abstract
Type 2 diabetes mellitus (T2DM) is a serious public health problem. In this review, we discuss current and promising future drugs, targets, in vitro assays and emerging omics technologies in T2DM. Importantly, we open the perspective to image-based high-content screening (HCS), with the focus of combining it with metabolomics or lipidomics. HCS has become a strong technology in phenotypic screens because it allows comprehensive screening for the cell-modulatory activity of small molecules. Metabolomics and lipidomics screen for perturbations at the molecular level. The combination of these data-intensive comprehensive technologies is enabled by the rapid development of artificial intelligence. It promises a deep cellular and molecular phenotyping directly linked to chemical information about the applied drug candidates or complex mixtures.
Collapse
Affiliation(s)
- Xin Zhang
- Research Unit Analytical BioGeoChemistry, Helmholtz Zentrum München, Ingolstaedter Landstr. 1, 85764 Neuherberg, Germany
| | - Erwin Kupczyk
- Research Unit Analytical BioGeoChemistry, Helmholtz Zentrum München, Ingolstaedter Landstr. 1, 85764 Neuherberg, Germany; Comprehensive Foodomics Platform, Chair of Analytical Food Chemistry, TUM School of Life Sciences, Technical University of Munich, Maximus-von-Imhof-Forum 2, 85354 Freising, Germany
| | - Philippe Schmitt-Kopplin
- Research Unit Analytical BioGeoChemistry, Helmholtz Zentrum München, Ingolstaedter Landstr. 1, 85764 Neuherberg, Germany; Comprehensive Foodomics Platform, Chair of Analytical Food Chemistry, TUM School of Life Sciences, Technical University of Munich, Maximus-von-Imhof-Forum 2, 85354 Freising, Germany.
| | - Constanze Mueller
- Research Unit Analytical BioGeoChemistry, Helmholtz Zentrum München, Ingolstaedter Landstr. 1, 85764 Neuherberg, Germany.
| |
Collapse
|
28
|
Self-supervised deep learning encodes high-resolution features of protein subcellular localization. Nat Methods 2022; 19:995-1003. [PMID: 35879608 PMCID: PMC9349041 DOI: 10.1038/s41592-022-01541-z] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 05/26/2022] [Indexed: 12/16/2022]
Abstract
Explaining the diversity and complexity of protein localization is essential to fully understand cellular architecture. Here we present cytoself, a deep-learning approach for fully self-supervised protein localization profiling and clustering. Cytoself leverages a self-supervised training scheme that does not require preexisting knowledge, categories or annotations. Training cytoself on images of 1,311 endogenously labeled proteins from the OpenCell database reveals a highly resolved protein localization atlas that recapitulates major scales of cellular organization, from coarse classes, such as nuclear and cytoplasmic, to the subtle localization signatures of individual protein complexes. We quantitatively validate cytoself’s ability to cluster proteins into organelles and protein complexes, showing that cytoself outperforms previous self-supervised approaches. Moreover, to better understand the inner workings of our model, we dissect the emergent features from which our clustering is derived, interpret them in the context of the fluorescence images, and analyze the performance contributions of each component of our approach. Cytoself is a self-supervised deep learning-based approach for profiling and clustering protein localization from fluorescence images. Cytoself outperforms established approaches and can accurately predict protein subcellular localization.
Collapse
|
29
|
Artificial intelligence and machine-learning approaches in structure and ligand-based discovery of drugs affecting central nervous system. Mol Divers 2022; 27:959-985. [PMID: 35819579 DOI: 10.1007/s11030-022-10489-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 06/21/2022] [Indexed: 12/11/2022]
Abstract
CNS disorders are indications with a very high unmet medical needs, relatively smaller number of available drugs, and a subpar satisfaction level among patients and caregiver. Discovery of CNS drugs is extremely expensive affair with its own unique challenges leading to extremely high attrition rates and low efficiency. With explosion of data in information age, there is hardly any aspect of life that has not been touched by data driven technologies such as artificial intelligence (AI) and machine learning (ML). Drug discovery is no exception, emergence of big data via genomic, proteomic, biological, and chemical technologies has driven pharmaceutical giants to collaborate with AI oriented companies to revolutionise drug discovery, with the goal of increasing the efficiency of the process. In recent years many examples of innovative applications of AI and ML techniques in CNS drug discovery has been reported. Research on therapeutics for diseases such as schizophrenia, Alzheimer's and Parkinsonism has been provided with a new direction and thrust from these developments. AI and ML has been applied to both ligand-based and structure-based drug discovery and design of CNS therapeutics. In this review, we have summarised the general aspects of AI and ML from the perspective of drug discovery followed by a comprehensive coverage of the recent developments in the applications of AI/ML techniques in CNS drug discovery.
Collapse
|
30
|
Priya S, Tripathi G, Singh DB, Jain P, Kumar A. Machine learning approaches and their applications in drug discovery and design. Chem Biol Drug Des 2022; 100:136-153. [PMID: 35426249 DOI: 10.1111/cbdd.14057] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 03/30/2022] [Accepted: 04/10/2022] [Indexed: 01/04/2023]
Abstract
This review is focused on several machine learning approaches used in chemoinformatics. Machine learning approaches provide tools and algorithms to improve drug discovery. Many physicochemical properties of drugs like toxicity, absorption, drug-drug interaction, carcinogenesis, and distribution have been effectively modeled by QSAR techniques. Machine learning is a subset of artificial intelligence, and this technique has shown tremendous potential in the field of drug discovery. Techniques discussed in this review are capable of modeling non-linear datasets, as well as big data of increasing depth and complexity. Various machine learning-based approaches are being used for drug target prediction, modeling the structure of drug target, binding site prediction, ligand-based similarity searching, de novo designing of ligands with desired properties, developing scoring functions for molecular docking, building QSAR model for biological activity prediction, and prediction of pharmacokinetic and pharmacodynamic properties of ligands. In recent years, these predictive tools and models have achieved good accuracy. By the use of more related input data, relevant parameters, and appropriate algorithms, the accuracy of these predictions can be further improved.
Collapse
Affiliation(s)
- Sonal Priya
- Department of Chemistry, T. N. B. College, TMBU, Bhagalpur, India
| | - Garima Tripathi
- Department of Chemistry, T. N. B. College, TMBU, Bhagalpur, India
| | - Dev Bukhsh Singh
- Department of Biotechnology, Siddharth University, Siddharth Nagar, India
| | - Priyanka Jain
- National Institute of Plant Genome Research, New Delhi, India
| | - Abhijeet Kumar
- Department of Chemistry, Mahatma Gandhi Central University, Motihari, India
| |
Collapse
|
31
|
Wang Y, Jeon H. 3D cell cultures toward quantitative high-throughput drug screening. Trends Pharmacol Sci 2022; 43:569-581. [DOI: 10.1016/j.tips.2022.03.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 03/29/2022] [Accepted: 03/30/2022] [Indexed: 01/16/2023]
|
32
|
Vulliard L, Hancock J, Kamnev A, Fell CW, Ferreira da Silva J, Loizou JI, Nagy V, Dupré L, Menche J. BioProfiling.jl: profiling biological perturbations with high-content imaging in single cells and heterogeneous populations. Bioinformatics 2022; 38:1692-1699. [PMID: 34935929 PMCID: PMC8896612 DOI: 10.1093/bioinformatics/btab853] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 12/15/2021] [Accepted: 12/16/2021] [Indexed: 02/03/2023] Open
Abstract
MOTIVATION High-content imaging screens provide a cost-effective and scalable way to assess cell states across diverse experimental conditions. The analysis of the acquired microscopy images involves assembling and curating raw cellular measurements into morphological profiles suitable for testing biological hypotheses. Despite being a critical step, general-purpose and adaptable tools for morphological profiling are lacking and no solution is available for the high-performance Julia programming language. RESULTS Here, we introduce BioProfiling.jl, an efficient end-to-end solution for compiling and filtering informative morphological profiles in Julia. The package contains all the necessary data structures to curate morphological measurements and helper functions to transform, normalize and visualize profiles. Robust statistical distances and permutation tests enable quantification of the significance of the observed changes despite the high fraction of outliers inherent to high-content screens. This package also simplifies visual artifact diagnostics, thus streamlining a bottleneck of morphological analyses. We showcase the features of the package by analyzing a chemical imaging screen, in which the morphological profiles prove to be informative about the compounds' mechanisms of action and can be conveniently integrated with the network localization of molecular targets. AVAILABILITY AND IMPLEMENTATION The Julia package is available on GitHub: https://github.com/menchelab/BioProfiling.jl. We also provide Jupyter notebooks reproducing our analyses: https://github.com/menchelab/BioProfilingNotebooks. The data underlying this article are available from FigShare, at https://doi.org/10.6084/m9.figshare.14784678.v2. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Loan Vulliard
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna 1090, Austria
- Department of Structural and Computational Biology, Max Perutz Labs, University of Vienna, Vienna 1030, Austria
| | - Joel Hancock
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna 1090, Austria
- Department of Structural and Computational Biology, Max Perutz Labs, University of Vienna, Vienna 1030, Austria
| | - Anton Kamnev
- Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases, Vienna 1090, Austria
- Department of Dermatology, Medical University of Vienna, Vienna 1090, Austria
| | - Christopher W Fell
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna 1090, Austria
- Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases, Vienna 1090, Austria
- Department of Neurology, Medical University of Vienna, Vienna 1090, Austria
| | - Joana Ferreira da Silva
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna 1090, Austria
- Institute of Cancer Research, Department of Medicine I, Medical University of Vienna and Comprehensive Cancer Center, Vienna 1090, Austria
| | - Joanna I Loizou
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna 1090, Austria
- Institute of Cancer Research, Department of Medicine I, Medical University of Vienna and Comprehensive Cancer Center, Vienna 1090, Austria
| | - Vanja Nagy
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna 1090, Austria
- Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases, Vienna 1090, Austria
- Department of Neurology, Medical University of Vienna, Vienna 1090, Austria
| | - Loïc Dupré
- Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases, Vienna 1090, Austria
- Department of Dermatology, Medical University of Vienna, Vienna 1090, Austria
- Toulouse Institute for Infectious and Inflammatory Diseases (INFINITy), INSERM UMR1291, CNRS UMR5051, Toulouse III Paul Sabatier University, Toulouse 31024, France
| | | |
Collapse
|
33
|
Trapotsi MA, Hosseini-Gerami L, Bender A. Computational analyses of mechanism of action (MoA): data, methods and integration. RSC Chem Biol 2022; 3:170-200. [PMID: 35360890 PMCID: PMC8827085 DOI: 10.1039/d1cb00069a] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 12/09/2021] [Indexed: 12/15/2022] Open
Abstract
The elucidation of a compound's Mechanism of Action (MoA) is a challenging task in the drug discovery process, but it is important in order to rationalise phenotypic findings and to anticipate potential side-effects. Bioinformatic approaches, advances in machine learning techniques and the increasing deposition of high-throughput data in public databases have significantly contributed to recent advances in the field, but it is not straightforward to decide which data and methods are most suitable to use in a given case. In this review, we focus on these methods and data and their applications in generating MoA hypotheses for subsequent experimental validation. We discuss compound-specific data such as -omics, cell morphology and bioactivity data, as well as commonly used supplementary prior knowledge such as network and pathway data, and provide information on databases where this data can be accessed. In terms of methodologies, we discuss both well-established methods (connectivity mapping, pathway enrichment) as well as more developing methods (neural networks and multi-omics integration). Finally, we review case studies where the MoA of a compound was successfully suggested from computational analysis by incorporating multiple data modalities and/or methodologies. Our aim for this review is to provide researchers with insights into the benefits and drawbacks of both the data and methods in terms of level of understanding, biases and interpretation - and to highlight future avenues of investigation which we foresee will improve the field of MoA elucidation, including greater public access to -omics data and methodologies which are capable of data integration.
Collapse
Affiliation(s)
- Maria-Anna Trapotsi
- Centre for Molecular Informatics, Yusuf Hamied Department of Chemistry, University of Cambridge UK
| | - Layla Hosseini-Gerami
- Centre for Molecular Informatics, Yusuf Hamied Department of Chemistry, University of Cambridge UK
| | - Andreas Bender
- Centre for Molecular Informatics, Yusuf Hamied Department of Chemistry, University of Cambridge UK
| |
Collapse
|
34
|
Gerckens M, Schorpp K, Pelizza F, Wögrath M, Reichau K, Ma H, Dworsky AM, Sengupta A, Stoleriu MG, Heinzelmann K, Merl-Pham J, Irmler M, Alsafadi HN, Trenkenschuh E, Sarnova L, Jirouskova M, Frieß W, Hauck SM, Beckers J, Kneidinger N, Behr J, Hilgendorff A, Hadian K, Lindner M, Königshoff M, Eickelberg O, Gregor M, Plettenburg O, Yildirim AÖ, Burgstaller G. Phenotypic drug screening in a human fibrosis model identified a novel class of antifibrotic therapeutics. SCIENCE ADVANCES 2021; 7:eabb3673. [PMID: 34936468 PMCID: PMC8694600 DOI: 10.1126/sciadv.abb3673] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/09/2023]
Abstract
Fibrogenic processes instigate fatal chronic diseases leading to organ failure and death. Underlying biological processes involve induced massive deposition of extracellular matrix (ECM) by aberrant fibroblasts. We subjected diseased primary human lung fibroblasts to an advanced three-dimensional phenotypic high-content assay and screened a repurposing drug library of small molecules for inhibiting ECM deposition. Fibrotic Pattern Detection by Artificial Intelligence identified tranilast as an effective inhibitor. Structure-activity relationship studies confirmed N-(2-butoxyphenyl)-3-(phenyl)acrylamides (N23Ps) as a novel and highly potent compound class. N23Ps suppressed myofibroblast transdifferentiation, ECM deposition, cellular contractility, and altered cell shapes, thus advocating a unique mode of action. Mechanistically, transcriptomics identified SMURF2 as a potential therapeutic target network. Antifibrotic activity of N23Ps was verified by proteomics in a human ex vivo tissue fibrosis disease model, suppressing profibrotic markers SERPINE1 and CXCL8. Conclusively, N23Ps are a novel class of highly potent compounds inhibiting organ fibrosis in patients.
Collapse
Affiliation(s)
- Michael Gerckens
- Institute of Lung Biology and Disease (ILBD) and Comprehensive Pneumology Center (CPC), Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Kenji Schorpp
- Assay Development and Screening Platform, Institute of Molecular Toxicology and Pharmacology, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
| | - Francesco Pelizza
- Chemical and Process Engineering, Strathclyde University, Glasgow, Scotland, UK
| | - Melanie Wögrath
- Institute of Lung Biology and Disease (ILBD) and Comprehensive Pneumology Center (CPC), Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
- CPC-M bioArchive, Helmholtz Zentrum München, Comprehensive Pneumology Center Munich DZL/CPC-M, Munich, Germany
| | - Kora Reichau
- Institute of Medicinal Chemistry, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
- Leibniz Universität Hannover, Institute of Organic Chemistry and Center for Biomolecular Drug Research (BMWZ), Hannover, Germany
| | - Huilong Ma
- Institute of Medicinal Chemistry, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
- Leibniz Universität Hannover, Institute of Organic Chemistry and Center for Biomolecular Drug Research (BMWZ), Hannover, Germany
| | - Armando-Marco Dworsky
- Institute of Lung Biology and Disease (ILBD) and Comprehensive Pneumology Center (CPC), Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
- CPC-M bioArchive, Helmholtz Zentrum München, Comprehensive Pneumology Center Munich DZL/CPC-M, Munich, Germany
| | - Arunima Sengupta
- Institute of Lung Biology and Disease (ILBD) and Comprehensive Pneumology Center (CPC), Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Mircea Gabriel Stoleriu
- Institute of Lung Biology and Disease (ILBD) and Comprehensive Pneumology Center (CPC), Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
- CPC-M bioArchive, Helmholtz Zentrum München, Comprehensive Pneumology Center Munich DZL/CPC-M, Munich, Germany
- Asklepios Fachkliniken Munich-Gauting, Munich, Germany
| | - Katharina Heinzelmann
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Comprehensive Pneumology Center (CPC), Research Unit Lung Repair and Regeneration, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Juliane Merl-Pham
- Research Unit Protein Science, Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, Neuherberg, Germany
| | - Martin Irmler
- Institute of Experimental Genetics, Helmholtz Zentrum München, 85764 Neuherberg, Germany
| | - Hani N. Alsafadi
- CPC-M bioArchive, Helmholtz Zentrum München, Comprehensive Pneumology Center Munich DZL/CPC-M, Munich, Germany
- Comprehensive Pneumology Center (CPC), Research Unit Lung Repair and Regeneration, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
- Wallenberg Center for Molecular Medicine (WCMM), Department of Experimental Medical Sciences, Lund University, Lund, Sweden
| | - Eduard Trenkenschuh
- Department of Pharmacy–Center for Drug Research, Pharmaceutical Technology and Biopharmaceutics, Ludwig-Maximillians University of Munich, Munich, Germany
| | - Lenka Sarnova
- Laboratory of Integrative Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Marketa Jirouskova
- Laboratory of Integrative Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Wolfgang Frieß
- Department of Pharmacy–Center for Drug Research, Pharmaceutical Technology and Biopharmaceutics, Ludwig-Maximillians University of Munich, Munich, Germany
| | - Stefanie M. Hauck
- Research Unit Protein Science, Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, Neuherberg, Germany
| | - Johannes Beckers
- Institute of Experimental Genetics, Helmholtz Zentrum München, 85764 Neuherberg, Germany
- German Center for Diabetes Research (DZD e.V.), 85764 Neuherberg, Germany
- Chair of Experimental Genetics, Technische Universität München, 85354 Freising, Germany
| | - Nikolaus Kneidinger
- CPC-M bioArchive, Helmholtz Zentrum München, Comprehensive Pneumology Center Munich DZL/CPC-M, Munich, Germany
- Department of Internal Medicine V, Ludwig-Maximillians University of Munich, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Jürgen Behr
- CPC-M bioArchive, Helmholtz Zentrum München, Comprehensive Pneumology Center Munich DZL/CPC-M, Munich, Germany
- Asklepios Fachkliniken Munich-Gauting, Munich, Germany
- Department of Internal Medicine V, Ludwig-Maximillians University of Munich, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Anne Hilgendorff
- Institute of Lung Biology and Disease (ILBD) and Comprehensive Pneumology Center (CPC), Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
- CPC-M bioArchive, Helmholtz Zentrum München, Comprehensive Pneumology Center Munich DZL/CPC-M, Munich, Germany
| | - Kamyar Hadian
- Assay Development and Screening Platform, Institute of Molecular Toxicology and Pharmacology, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
| | - Michael Lindner
- CPC-M bioArchive, Helmholtz Zentrum München, Comprehensive Pneumology Center Munich DZL/CPC-M, Munich, Germany
- Asklepios Fachkliniken Munich-Gauting, Munich, Germany
- Paracelsus Medical Private University, Salzburg, Austria
| | - Melanie Königshoff
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Comprehensive Pneumology Center (CPC), Research Unit Lung Repair and Regeneration, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Oliver Eickelberg
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Martin Gregor
- Laboratory of Integrative Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Oliver Plettenburg
- Institute of Medicinal Chemistry, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
- Leibniz Universität Hannover, Institute of Organic Chemistry and Center for Biomolecular Drug Research (BMWZ), Hannover, Germany
- Institute for Lung Health (ILH), Justus Liebig University, Giessen, Germany
| | - Ali Önder Yildirim
- Institute of Lung Biology and Disease (ILBD) and Comprehensive Pneumology Center (CPC), Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Gerald Burgstaller
- Institute of Lung Biology and Disease (ILBD) and Comprehensive Pneumology Center (CPC), Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
- CPC-M bioArchive, Helmholtz Zentrum München, Comprehensive Pneumology Center Munich DZL/CPC-M, Munich, Germany
- Corresponding author.
| |
Collapse
|
35
|
Yin Z, Wong STC. Artificial intelligence unifies knowledge and actions in drug repositioning. Emerg Top Life Sci 2021; 5:803-813. [PMID: 34881780 PMCID: PMC8923082 DOI: 10.1042/etls20210223] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 11/08/2021] [Accepted: 11/09/2021] [Indexed: 11/17/2022]
Abstract
Drug repositioning aims to reuse existing drugs, shelved drugs, or drug candidates that failed clinical trials for other medical indications. Its attraction is sprung from the reduction in risk associated with safety testing of new medications and the time to get a known drug into the clinics. Artificial Intelligence (AI) has been recently pursued to speed up drug repositioning and discovery. The essence of AI in drug repositioning is to unify the knowledge and actions, i.e. incorporating real-world and experimental data to map out the best way forward to identify effective therapeutics against a disease. In this review, we share positive expectations for the evolution of AI and drug repositioning and summarize the role of AI in several methods of drug repositioning.
Collapse
Affiliation(s)
- Zheng Yin
- Department of Systems Medicine and Bioengineering, Houston Methodist Cancer Center and Ting Tsung & Wei Fong Chao Center for BRAIN, Houston Methodist Research Institute, Weill Cornell Medicine, Houston, TX 77030, U.S.A
| | - Stephen T C Wong
- Department of Systems Medicine and Bioengineering, Houston Methodist Cancer Center and Ting Tsung & Wei Fong Chao Center for BRAIN, Houston Methodist Research Institute, Weill Cornell Medicine, Houston, TX 77030, U.S.A
| |
Collapse
|
36
|
Chu HY, Wong ASL. Facilitating Machine Learning-Guided Protein Engineering with Smart Library Design and Massively Parallel Assays. ADVANCED GENETICS (HOBOKEN, N.J.) 2021; 2:2100038. [PMID: 36619853 PMCID: PMC9744531 DOI: 10.1002/ggn2.202100038] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 11/08/2021] [Indexed: 01/11/2023]
Abstract
Protein design plays an important role in recent medical advances from antibody therapy to vaccine design. Typically, exhaustive mutational screens or directed evolution experiments are used for the identification of the best design or for improvements to the wild-type variant. Even with a high-throughput screening on pooled libraries and Next-Generation Sequencing to boost the scale of read-outs, surveying all the variants with combinatorial mutations for their empirical fitness scores is still of magnitudes beyond the capacity of existing experimental settings. To tackle this challenge, in-silico approaches using machine learning to predict the fitness of novel variants based on a subset of empirical measurements are now employed. These machine learning models turn out to be useful in many cases, with the premise that the experimentally determined fitness scores and the amino-acid descriptors of the models are informative. The machine learning models can guide the search for the highest fitness variants, resolve complex epistatic relationships, and highlight bio-physical rules for protein folding. Using machine learning-guided approaches, researchers can build more focused libraries, thus relieving themselves from labor-intensive screens and fast-tracking the optimization process. Here, we describe the current advances in massive-scale variant screens, and how machine learning and mutagenesis strategies can be integrated to accelerate protein engineering. More specifically, we examine strategies to make screens more economical, informative, and effective in discovery of useful variants.
Collapse
Affiliation(s)
- Hoi Yee Chu
- Laboratory of Combinatorial Genetics and Synthetic BiologySchool of Biomedical SciencesThe University of Hong KongHong Kong852China
| | - Alan S. L. Wong
- Laboratory of Combinatorial Genetics and Synthetic BiologySchool of Biomedical SciencesThe University of Hong KongHong Kong852China
- Electrical and Electronic EngineeringThe University of Hong KongPokfulamHong Kong852China
| |
Collapse
|
37
|
Maurya R, Pathak VK, Dutta MK. Deep learning based microscopic cell images classification framework using multi-level ensemble. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2021; 211:106445. [PMID: 34627021 DOI: 10.1016/j.cmpb.2021.106445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Accepted: 09/26/2021] [Indexed: 06/13/2023]
Abstract
BACKGROUND AND OBJECTIVES Advancement of the ultra-fast microscopic images acquisition and generation techniques give rise to the automated artificial intelligence (AI)-based microscopic images classification systems. The earlier cell classification systems classify the cell images of a specific type captured using a specific microscopy technique, therefore the motivation behind the present study is to develop a generic framework that can be used for the classification of cell images of multiple types captured using a variety of microscopic techniques. METHODS The proposed framework for microscopic cell images classification is based on the transfer learning-based multi-level ensemble approach. The ensemble is made by training the same base model with different optimisation methods and different learning rates. An important contribution of the proposed framework lies in its ability to capture different granularities of features extracted from multiple scales of an input microscopic cell image. The base learners used in the proposed ensemble encapsulates the aggregation of low-level coarse features and high-level semantic features, thus, represent the different granular microscopic cell image features present at different scales of input cell images. The batch normalisation layer has been added to the base models for the fast convergence in the proposed ensemble for microscopic cell images classification. RESULTS The general applicability of the proposed framework for microscopic cell image classification has been tested with five different public datasets. The proposed method has outperformed the experimental results obtained in several other similar works. CONCLUSIONS The proposed framework for microscopic cell classification outperforms the other state-of-the-art classification methods in the same domain with a comparatively lesser amount of training data.
Collapse
Affiliation(s)
- Ritesh Maurya
- Centre for Advanced Studies, Dr. A.P.J. Abdul Kalam Technical University, Lucknow, India.
| | | | - Malay Kishore Dutta
- Centre for Advanced Studies, Dr. A.P.J. Abdul Kalam Technical University, Lucknow, India.
| |
Collapse
|
38
|
Selvaraj C, Chandra I, Singh SK. Artificial intelligence and machine learning approaches for drug design: challenges and opportunities for the pharmaceutical industries. Mol Divers 2021; 26:1893-1913. [PMID: 34686947 PMCID: PMC8536481 DOI: 10.1007/s11030-021-10326-z] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Accepted: 09/24/2021] [Indexed: 12/27/2022]
Abstract
The global spread of COVID-19 has raised the importance of pharmaceutical drug development as intractable and hot research. Developing new drug molecules to overcome any disease is a costly and lengthy process, but the process continues uninterrupted. The critical point to consider the drug design is to use the available data resources and to find new and novel leads. Once the drug target is identified, several interdisciplinary areas work together with artificial intelligence (AI) and machine learning (ML) methods to get enriched drugs. These AI and ML methods are applied in every step of the computer-aided drug design, and integrating these AI and ML methods results in a high success rate of hit compounds. In addition, this AI and ML integration with high-dimension data and its powerful capacity have taken a step forward. Clinical trials output prediction through the AI/ML integrated models could further decrease the clinical trials cost by also improving the success rate. Through this review, we discuss the backend of AI and ML methods in supporting the computer-aided drug design, along with its challenge and opportunity for the pharmaceutical industry. From the available information or data, the AI and ML based prediction for the high throughput virtual screening. After this integration of AI and ML, the success rate of hit identification has gained a momentum with huge success by providing novel drugs.
Collapse
Affiliation(s)
- Chandrabose Selvaraj
- CADD and Molecular Modelling Lab, Department of Bioinformatics, Alagappa University, Science Block, Karaikudi, Tamil Nadu, 630004, India.
| | - Ishwar Chandra
- CADD and Molecular Modelling Lab, Department of Bioinformatics, Alagappa University, Science Block, Karaikudi, Tamil Nadu, 630004, India
| | - Sanjeev Kumar Singh
- CADD and Molecular Modelling Lab, Department of Bioinformatics, Alagappa University, Science Block, Karaikudi, Tamil Nadu, 630004, India.
| |
Collapse
|
39
|
Organoids in image-based phenotypic chemical screens. Exp Mol Med 2021; 53:1495-1502. [PMID: 34663938 PMCID: PMC8569209 DOI: 10.1038/s12276-021-00641-8] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 04/08/2021] [Accepted: 05/04/2021] [Indexed: 12/18/2022] Open
Abstract
Image-based phenotypic screening relies on the extraction of multivariate information from cells cultured under a large variety of conditions. Technical advances in high-throughput microscopy enable screening in increasingly complex and biologically relevant model systems. To this end, organoids hold great potential for high-content screening because they recapitulate many aspects of parent tissues and can be derived from patient material. However, screening is substantially more difficult in organoids than in classical cell lines from both technical and analytical standpoints. In this review, we present an overview of studies employing organoids for screening applications. We discuss the promises and challenges of small-molecule treatments in organoids and give practical advice on designing, running, and analyzing high-content organoid-based phenotypic screens.
Collapse
|
40
|
Mak KK, Balijepalli MK, Pichika MR. Success stories of AI in drug discovery - where do things stand? Expert Opin Drug Discov 2021; 17:79-92. [PMID: 34553659 DOI: 10.1080/17460441.2022.1985108] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
INTRODUCTION Artificial intelligence (AI) in drug discovery and development (DDD) has gained more traction in the past few years. Many scientific reviews have already been made available in this area. Thus, in this review, the authors have focused on the success stories of AI-driven drug candidates and the scientometric analysis of the literature in this field. AREA COVERED The authors explore the literature to compile the success stories of AI-driven drug candidates that are currently being assessed in clinical trials or have investigational new drug (IND) status. The authors also provide the reader with their expert perspectives for future developments and their opinions on the field. EXPERT OPINION Partnerships between AI companies and the pharma industry are booming. The early signs of the impact of AI on DDD are encouraging, and the pharma industry is hoping for breakthroughs. AI can be a promising technology to unveil the greatest successes, but it has yet to be proven as AI is still at the embryonic stage.
Collapse
Affiliation(s)
- Kit-Kay Mak
- School of Postgraduate Studies and Research, International Medical University, Bukit Jalil, Malaysia.,Department of Pharmaceutical Chemistry, School of Pharmacy, International Medical University, Bukit Jalil, Malaysia.,Centre for Bioactive Molecules and Drug Delivery, Institute for Research, Development, and Innovation (Irdi), International Medical University, Bukit Jalil, Malaysia
| | | | - Mallikarjuna Rao Pichika
- Department of Pharmaceutical Chemistry, School of Pharmacy, International Medical University, Bukit Jalil, Malaysia.,Centre for Bioactive Molecules and Drug Delivery, Institute for Research, Development, and Innovation (Irdi), International Medical University, Bukit Jalil, Malaysia
| |
Collapse
|
41
|
Marine dissolved organic matter: a vast and unexplored molecular space. Appl Microbiol Biotechnol 2021; 105:7225-7239. [PMID: 34536106 PMCID: PMC8494709 DOI: 10.1007/s00253-021-11489-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 07/29/2021] [Accepted: 07/31/2021] [Indexed: 01/02/2023]
Abstract
Abstract Marine dissolved organic matter (DOM) comprises a vast and unexplored molecular space. Most of it resided in the oceans for thousands of years. It is among the most diverse molecular mixtures known, consisting of millions of individual compounds. More than 1 Eg of this material exists on the planet. As such, it comprises a formidable source of natural products promising significant potential for new biotechnological purposes. Great emphasis has been placed on understanding the role of DOM in biogeochemical cycles and climate attenuation, its lifespan, interaction with microorganisms, as well as its molecular composition. Yet, probing DOM bioactivities is in its infancy, largely because it is technically challenging due to the chemical complexity of the material. It is of considerable interest to develop technologies capable to better discern DOM bioactivities. Modern screening technologies are opening new avenues allowing accelerated identification of bioactivities for small molecules from natural products. These methods diminish a priori the need for laborious chemical fractionation. We examine here the application of untargeted metabolomics and multiplexed high-throughput molecular-phenotypic screening techniques that are providing first insights on previously undetectable DOM bioactivities. Key points • Marine DOM is a vast, unexplored biotechnological resource. • Untargeted bioscreening approaches are emerging for natural product screening. • Perspectives for developing bioscreening platforms for marine DOM are discussed.
Collapse
|
42
|
Ashraf M, Khalilitousi M, Laksman Z. Applying Machine Learning to Stem Cell Culture and Differentiation. Curr Protoc 2021; 1:e261. [PMID: 34529356 DOI: 10.1002/cpz1.261] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Machine learning techniques are increasingly becoming incorporated into biological research workflows in a variety of disciplines, most notably cancer research and drug discovery. Efforts in stem cell research comparatively lag behind. We detail key paradigms in machine learning, with a focus on equipping stem cell biologists with the understanding necessary to begin conceptualizing and designing machine learning workflows within their own domain of expertise. Supervised approaches in both regression and classification as well as unsupervised clustering techniques are all covered, with examples from across the biological sciences. High-throughput, high-content, multiplex assays for data acquisition are also discussed in the form of single-cell RNA sequencing and image-based approaches. Lastly, potential applications in stem cell biology, including the development of novel cell types, and improving model maturation are also discussed. Machine learning approaches applied in stem cell biology show promise in accelerating progress in developmental biology, drug screening, disease modeling, and personalized medicine. © 2021 Wiley Periodicals LLC.
Collapse
Affiliation(s)
- Mishal Ashraf
- School of Biomedical Engineering, University of British Columbia, Vancouver, British Columbia, Canada.,Centre for Heart Lung Innovation, Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Mohammadali Khalilitousi
- School of Biomedical Engineering, University of British Columbia, Vancouver, British Columbia, Canada
| | - Zachary Laksman
- School of Biomedical Engineering, University of British Columbia, Vancouver, British Columbia, Canada.,Centre for Heart Lung Innovation, Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
43
|
Renner H, Schöler HR, Bruder JM. Combining Automated Organoid Workflows With Artificial Intelligence-Based Analyses: Opportunities to Build a New Generation of Interdisciplinary High-Throughput Screens for Parkinson's Disease and Beyond. Mov Disord 2021; 36:2745-2762. [PMID: 34498298 DOI: 10.1002/mds.28775] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 08/05/2021] [Accepted: 08/09/2021] [Indexed: 12/14/2022] Open
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disease and primarily characterized by the loss of dopaminergic neurons in the substantia nigra pars compacta of the midbrain. Despite decades of research and the development of various disease model systems, there is no curative treatment. This could be due to current model systems, including cell culture and animal models, not adequately recapitulating human PD etiology. More complex human disease models, including human midbrain organoids, are maturing technologies that increasingly enable the strategic incorporation of the missing components needed to model PD in vitro. The resulting organoid-based biological complexity provides new opportunities and challenges in data analysis of rich multimodal data sets. Emerging artificial intelligence (AI) capabilities can take advantage of large, broad data sets and even correlate results across disciplines. Current organoid technologies no longer lack the prerequisites for large-scale high-throughput screening (HTS) and can generate complex yet reproducible data suitable for AI-based data mining. We have recently developed a fully scalable and HTS-compatible workflow for the generation, maintenance, and analysis of three-dimensional (3D) microtissues mimicking key characteristics of the human midbrain (called "automated midbrain organoids," AMOs). AMOs build a reproducible, scalable foundation for creating next-generation 3D models of human neural disease that can fuel mechanism-agnostic phenotypic drug discovery in human in vitro PD models and beyond. Here, we explore the opportunities and challenges resulting from the convergence of organoid HTS and AI-driven data analytics and outline potential future avenues toward the discovery of novel mechanisms and drugs in PD research. © 2021 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Henrik Renner
- Department of Cell and Developmental Biology, Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | - Hans R Schöler
- Department of Cell and Developmental Biology, Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | - Jan M Bruder
- Department of Cell and Developmental Biology, Max Planck Institute for Molecular Biomedicine, Münster, Germany
| |
Collapse
|
44
|
Brockman AA, Mobley BC, Ihrie RA. Histological Studies of the Ventricular-Subventricular Zone as Neural Stem Cell and Glioma Stem Cell Niche. J Histochem Cytochem 2021; 69:819-834. [PMID: 34310246 DOI: 10.1369/00221554211032003] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The neural stem cell niche of the ventricular-subventricular zone supports the persistence of stem and progenitor cells in the mature brain. This niche has many notable cytoarchitectural features that affect the activity of stem cells and may also support the survival and growth of invading tumor cells. Histochemical studies of the niche have revealed many proteins that, in combination, can help to reveal stem-like cells in the normal or cancer context, although many caveats persist in the quest to consistently identify these cells in the human brain. Here, we explore the complex relationship between the persistent proliferative capacity of the neural stem cell niche and the malignant proliferation of brain tumors, with a special focus on histochemical identification of stem cells and stem-like tumor cells and an eye toward the potential application of high-dimensional imaging approaches to the field.
Collapse
Affiliation(s)
- Asa A Brockman
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Bret C Mobley
- Departments of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Rebecca A Ihrie
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, Tennessee.,Departments of Neurological Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
| |
Collapse
|
45
|
Wilson SL, Way GP, Bittremieux W, Armache JP, Haendel MA, Hoffman MM. Sharing biological data: why, when, and how. FEBS Lett 2021; 595:847-863. [PMID: 33843054 PMCID: PMC10390076 DOI: 10.1002/1873-3468.14067] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Samantha L Wilson
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Gregory P Way
- Imaging Platform, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Wout Bittremieux
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA.,Department of Computer Science, University of Antwerp, Antwerpen, Belgium
| | - Jean-Paul Armache
- Department of Biochemistry & Molecular Biology, The Huck Institutes of Life Sciences, Pennsylvania State University, University Park, PA, USA
| | | | - Michael M Hoffman
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada.,Department of Medical Biophysics, Department of Computer Science, University of Toronto, Toronto, ON, Canada.,Vector Institute, Toronto, ON, Canada
| |
Collapse
|
46
|
Zaritsky A, Jamieson AR, Welf ES, Nevarez A, Cillay J, Eskiocak U, Cantarel BL, Danuser G. Interpretable deep learning uncovers cellular properties in label-free live cell images that are predictive of highly metastatic melanoma. Cell Syst 2021; 12:733-747.e6. [PMID: 34077708 PMCID: PMC8353662 DOI: 10.1016/j.cels.2021.05.003] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Revised: 01/22/2021] [Accepted: 05/07/2021] [Indexed: 12/22/2022]
Abstract
Deep learning has emerged as the technique of choice for identifying hidden patterns in cell imaging data but is often criticized as "black box." Here, we employ a generative neural network in combination with supervised machine learning to classify patient-derived melanoma xenografts as "efficient" or "inefficient" metastatic, validate predictions regarding melanoma cell lines with unknown metastatic efficiency in mouse xenografts, and use the network to generate in silico cell images that amplify the critical predictive cell properties. These exaggerated images unveiled pseudopodial extensions and increased light scattering as hallmark properties of metastatic cells. We validated this interpretation using live cells spontaneously transitioning between states indicative of low and high metastatic efficiency. This study illustrates how the application of artificial intelligence can support the identification of cellular properties that are predictive of complex phenotypes and integrated cell functions but are too subtle to be identified in the raw imagery by a human expert. A record of this paper's transparent peer review process is included in the supplemental information. VIDEO ABSTRACT.
Collapse
Affiliation(s)
- Assaf Zaritsky
- Lyda Hill Department of Bioinformatics, UT Southwestern Medical Center, Dallas, TX 75390, USA; Department of Software and Information Systems Engineering, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel.
| | - Andrew R Jamieson
- Lyda Hill Department of Bioinformatics, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Erik S Welf
- Lyda Hill Department of Bioinformatics, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Andres Nevarez
- Lyda Hill Department of Bioinformatics, UT Southwestern Medical Center, Dallas, TX 75390, USA; Section of Molecular Biology, Division of Biological Sciences, University of California San Diego, 9500 Gilman Drive, San Diego, La Jolla, CA 92093, USA
| | - Justin Cillay
- Lyda Hill Department of Bioinformatics, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Ugur Eskiocak
- Children's Research Institute and Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Brandi L Cantarel
- Lyda Hill Department of Bioinformatics, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Gaudenz Danuser
- Lyda Hill Department of Bioinformatics, UT Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
47
|
Abstract
Induced pluripotent stem cell (iPSC) technology holds promise for modeling neurodegenerative diseases. Traditional approaches for disease modeling using animal and cellular models require knowledge of disease mutations. However, many patients with neurodegenerative diseases do not have a known genetic cause. iPSCs offer a way to generate patient-specific models and study pathways of dysfunction in an in vitro setting in order to understand the causes and subtypes of neurodegeneration. Furthermore, iPSC-based models can be used to search for candidate therapeutics using high-throughput screening. Here we review how iPSC-based models are currently being used to further our understanding of neurodegenerative diseases, as well as discuss their challenges and future directions.
Collapse
Affiliation(s)
- Jonathan Li
- Computational and Systems Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA;
| | - Ernest Fraenkel
- Computational and Systems Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA; .,Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| |
Collapse
|
48
|
Palano G, Foinquinos A, Müllers E. In vitro Assays and Imaging Methods for Drug Discovery for Cardiac Fibrosis. Front Physiol 2021; 12:697270. [PMID: 34305651 PMCID: PMC8298031 DOI: 10.3389/fphys.2021.697270] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 06/07/2021] [Indexed: 12/20/2022] Open
Abstract
As a result of stress, injury, or aging, cardiac fibrosis is characterized by excessive deposition of extracellular matrix (ECM) components resulting in pathological remodeling, tissue stiffening, ventricular dilatation, and cardiac dysfunction that contribute to heart failure (HF) and eventually death. Currently, there are no effective therapies specifically targeting cardiac fibrosis, partially due to limited understanding of the pathological mechanisms and the lack of predictive in vitro models for high-throughput screening of antifibrotic compounds. The use of more relevant cell models, three-dimensional (3D) models, and coculture systems, together with high-content imaging (HCI) and machine learning (ML)-based image analysis, is expected to improve predictivity and throughput of in vitro models for cardiac fibrosis. In this review, we present an overview of available in vitro assays for cardiac fibrosis. We highlight the potential of more physiological 3D cardiac organoids and coculture systems and discuss HCI and automated artificial intelligence (AI)-based image analysis as key methods able to capture the complexity of cardiac fibrosis in vitro. As 3D and coculture models will soon be sufficiently mature for application in large-scale preclinical drug discovery, we expect the combination of more relevant models and high-content analysis to greatly increase translation from in vitro to in vivo models and facilitate the discovery of novel targets and drugs against cardiac fibrosis.
Collapse
Affiliation(s)
- Giorgia Palano
- Division of Physiological Chemistry I, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Ariana Foinquinos
- Bioscience Cardiovascular, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Erik Müllers
- Bioscience Cardiovascular, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| |
Collapse
|
49
|
Schuhmacher A, Gatto A, Kuss M, Gassmann O, Hinder M. Big Techs and startups in pharmaceutical R&D - A 2020 perspective on artificial intelligence. Drug Discov Today 2021; 26:2226-2231. [PMID: 33965571 DOI: 10.1016/j.drudis.2021.04.028] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 03/05/2021] [Accepted: 04/30/2021] [Indexed: 11/29/2022]
Abstract
We investigated what kind of artificial intelligence (AI) technologies are utilized in pharmaceutical research and development (R&D) and which sources of AI-related competencies can be leveraged by pharmaceutical companies. First, we found that machine learning (ML) is the dominating AI technology currently used in pharmaceutical R&D. Second, both Big Techs and AI startups are competent knowledge bases for AI applications. Big Techs have long-lasting experience in the digital field and offer more general IT solutions to support pharmaceutical companies in cloud computing, health monitoring, diagnostics or clinical trial management, whereas startups can provide more specific AI services to address special issues in the drug-discovery space.
Collapse
Affiliation(s)
- Alexander Schuhmacher
- Reutlingen University, Alteburgstrasse 150, DE-72762 Reutlingen, Germany; Institute of Technology Management, University of St Gallen, Dufourstrasse 40a, CH-9000 St Gallen, Switzerland.
| | - Alexander Gatto
- Sony Europe B.V. - Stuttgart Technology Center, Hedelfinger Strasse 61, DE-70327 Stuttgart, Germany
| | - Michael Kuss
- PricewaterhouseCoopers AG, Birchstrasse 160, CH-8050 Zurich, Switzerland
| | - Oliver Gassmann
- Institute of Technology Management, University of St Gallen, Dufourstrasse 40a, CH-9000 St Gallen, Switzerland
| | - Markus Hinder
- Novartis Institute of BioMedical Research, Postfach, Forum 1, CH-4002 Basel, Switzerland
| |
Collapse
|
50
|
Machine learning-assisted high-content analysis of pluripotent stem cell-derived embryos in vitro. Stem Cell Reports 2021; 16:1331-1346. [PMID: 33891867 PMCID: PMC8185434 DOI: 10.1016/j.stemcr.2021.03.018] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 03/17/2021] [Accepted: 03/17/2021] [Indexed: 12/13/2022] Open
Abstract
Stem cell-based embryo models by cultured pluripotent and extra-embryonic lineage stem cells are novel platforms to model early postimplantation development. We showed that induced pluripotent stem cells (iPSCs) could form ITS (iPSCs and trophectoderm stem cells) and ITX (iPSCs, trophectoderm stem cells, and XEN cells) embryos, resembling the early gastrula embryo developed in vivo. To facilitate the efficient and unbiased analysis of the stem cell-based embryo model, we set up a machine learning workflow to extract multi-dimensional features and perform quantification of ITS embryos using 3D images collected from a high-content screening system. We found that different PSC lines differ in their ability to form embryo-like structures. Through high-content screening of small molecules and cytokines, we identified that BMP4 best promoted the morphogenesis of the ITS embryo. Our study established an innovative strategy to analyze stem cell-based embryo models and uncovered new roles of BMP4 in stem cell-based embryo models.
Collapse
|