1
|
Wang Y, Zhou S, Wang X, Lu D, Yang J, Lu Y, Fan X, Li C, Wang Y. Electroactive membranes enhance in-situ alveolar ridge preservation via spatiotemporal electrical modulation of cell motility. Biomaterials 2024; 317:123077. [PMID: 39756273 DOI: 10.1016/j.biomaterials.2024.123077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 11/27/2024] [Accepted: 12/30/2024] [Indexed: 01/07/2025]
Abstract
Post-extraction alveolar bone resorption invariably compromises implant placement and aesthetic restoration outcomes. Current non-resorbable membranes exhibit limited efficacy in alveolar ridge preservation (ARP) due to insufficient cell recruitment and osteoinductive capabilities. Herein, we introduce a multifunctional electroactive membrane (PPy-BTO/P(VDF-TrFE), PB/PT) designed to spatiotemporally regulate cell migration and osteogenesis, harmonizing with the socket healing process. Initially, the membrane's endogenous-level surface potential recruits stem cells from the socket. Subsequently, adherent cell-migration-triggered forces generate on-demand piezopotential, stimulating intracellular calcium ion fluctuations and activating the Ca2+/calcineurin/NFAT1 signaling pathway via Cav3.2 channels. This enhances cell motility and osteogenic differentiation predominantly in the coronal socket region, counteracting the natural healing trajectory. The membrane's self-powered energy supply, proportional to cell migration velocity and manifested as nanoparticle deformation, mitigates ridge shrinkage, both independently and in conjunction with bone grafts. This energy-autonomous membrane, based on the spatiotemporal modulation of cell motility, presents a novel approach for in-situ ARP treatment and the development of 4D bionic scaffolds.
Collapse
Affiliation(s)
- Yanlan Wang
- Hospital of Stomatology, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, 510055, China
| | - Shiqi Zhou
- Hospital of Stomatology, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, 510055, China
| | - Xiaoshuang Wang
- Hospital of Stomatology, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, 510055, China
| | - Dongheng Lu
- Hospital of Stomatology, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, 510055, China
| | - Jinghong Yang
- Hospital of Stomatology, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, 510055, China
| | - Yu Lu
- Hospital of Stomatology, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, 510055, China
| | - Xiaolei Fan
- Hospital of Stomatology, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, 510055, China
| | - Changhao Li
- State Key Laboratory of Digital Medical Engineering, Key Laboratory of Biomedical Engineering of Hainan Province, School of Biomedical Engineering, Hainan University, Sanya, 572025, China.
| | - Yan Wang
- Hospital of Stomatology, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, 510055, China.
| |
Collapse
|
2
|
Ghanegolmohammadi F, Eslami M, Ohya Y. Systematic data analysis pipeline for quantitative morphological cell phenotyping. Comput Struct Biotechnol J 2024; 23:2949-2962. [PMID: 39104709 PMCID: PMC11298594 DOI: 10.1016/j.csbj.2024.07.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 07/09/2024] [Accepted: 07/10/2024] [Indexed: 08/07/2024] Open
Abstract
Quantitative morphological phenotyping (QMP) is an image-based method used to capture morphological features at both the cellular and population level. Its interdisciplinary nature, spanning from data collection to result analysis and interpretation, can lead to uncertainties, particularly among those new to this actively growing field. High analytical specificity for a typical QMP is achieved through sophisticated approaches that can leverage subtle cellular morphological changes. Here, we outline a systematic workflow to refine the QMP methodology. For a practical review, we describe the main steps of a typical QMP; in each step, we discuss the available methods, their applications, advantages, and disadvantages, along with the R functions and packages for easy implementation. This review does not cover theoretical backgrounds, but provides several references for interested researchers. It aims to broaden the horizons for future phenome studies and demonstrate how to exploit years of endeavors to achieve more with less.
Collapse
Affiliation(s)
- Farzan Ghanegolmohammadi
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Department of Integrated Biosciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Chiba, Japan
| | - Mohammad Eslami
- Harvard Ophthalmology AI Lab, Schepen’s Eye Research Institute of Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, USA
| | - Yoshikazu Ohya
- Department of Integrated Biosciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Chiba, Japan
| |
Collapse
|
3
|
Huang Y, Zhou Z, Liu T, Tang S, Xin X. Exploring heterogeneous cell population dynamics in different microenvironments by novel analytical strategy based on images. NPJ Syst Biol Appl 2024; 10:129. [PMID: 39505883 PMCID: PMC11542073 DOI: 10.1038/s41540-024-00459-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 10/21/2024] [Indexed: 11/08/2024] Open
Abstract
Understanding the dynamic states and transitions of heterogeneous cell populations is crucial for addressing fundamental biological questions. High-content imaging provides rich datasets, but it remains increasingly difficult to integrate and annotate high-dimensional and time-resolved datasets to profile heterogeneous cell population dynamics in different microenvironments. Using hepatic stellate cells (HSCs) LX-2 as model, we proposed a novel analytical strategy for image-based integration and annotation to profile dynamics of heterogeneous cell populations in 2D/3D microenvironments. High-dimensional features were extracted from extensive image datasets, and cellular states were identified based on feature profiles. Time-series clustering revealed distinct temporal patterns of cell shape and actin cytoskeleton reorganization. We found LX-2 showed more complex membrane dynamics and contractile systems with an M-shaped actin compactness trend in 3D culture, while they displayed rapid spreading in early 2D culture. This image-based integration and annotation strategy enhances our understanding of HSCs heterogeneity and dynamics in complex extracellular microenvironments.
Collapse
Affiliation(s)
- Yihong Huang
- Laboratory of Biophysics, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Zidong Zhou
- Laboratory of Biophysics, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Tianqi Liu
- Laboratory of Biophysics, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Shengnan Tang
- Laboratory of Biophysics, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Xuegang Xin
- Laboratory of Biophysics, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China.
| |
Collapse
|
4
|
Liu J, Du H, Huang L, Xie W, Liu K, Zhang X, Chen S, Zhang Y, Li D, Pan H. AI-Powered Microfluidics: Shaping the Future of Phenotypic Drug Discovery. ACS APPLIED MATERIALS & INTERFACES 2024; 16:38832-38851. [PMID: 39016521 DOI: 10.1021/acsami.4c07665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/18/2024]
Abstract
Phenotypic drug discovery (PDD), which involves harnessing biological systems directly to uncover effective drugs, has undergone a resurgence in recent years. The rapid advancement of artificial intelligence (AI) over the past few years presents numerous opportunities for augmenting phenotypic drug screening on microfluidic platforms, leveraging its predictive capabilities, data analysis, efficient data processing, etc. Microfluidics coupled with AI is poised to revolutionize the landscape of phenotypic drug discovery. By integrating advanced microfluidic platforms with AI algorithms, researchers can rapidly screen large libraries of compounds, identify novel drug candidates, and elucidate complex biological pathways with unprecedented speed and efficiency. This review provides an overview of recent advances and challenges in AI-based microfluidics and their applications in drug discovery. We discuss the synergistic combination of microfluidic systems for high-throughput screening and AI-driven analysis for phenotype characterization, drug-target interactions, and predictive modeling. In addition, we highlight the potential of AI-powered microfluidics to achieve an automated drug screening system. Overall, AI-powered microfluidics represents a promising approach to shaping the future of phenotypic drug discovery by enabling rapid, cost-effective, and accurate identification of therapeutically relevant compounds.
Collapse
Affiliation(s)
- Junchi Liu
- Department of Anesthesiology, The First Hospital of Jilin University, 71 Xinmin Street, Changchun 130012, China
| | - Hanze Du
- Department of Endocrinology, Key Laboratory of Endocrinology of National Health Commission, Translation Medicine Centre, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, China
- Key Laboratory of Endocrinology of National Health Commission, Department of Endocrinology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100730, China
| | - Lei Huang
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, School and Hospital of Stomatology, Jilin University, 1500 Qinghua Road, Changchun 130012, China
| | - Wangni Xie
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, School and Hospital of Stomatology, Jilin University, 1500 Qinghua Road, Changchun 130012, China
| | - Kexuan Liu
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, School and Hospital of Stomatology, Jilin University, 1500 Qinghua Road, Changchun 130012, China
| | - Xue Zhang
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, School and Hospital of Stomatology, Jilin University, 1500 Qinghua Road, Changchun 130012, China
| | - Shi Chen
- Department of Endocrinology, Key Laboratory of Endocrinology of National Health Commission, Translation Medicine Centre, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, China
- Key Laboratory of Endocrinology of National Health Commission, Department of Endocrinology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100730, China
| | - Yuan Zhang
- Department of Anesthesiology, The First Hospital of Jilin University, 71 Xinmin Street, Changchun 130012, China
| | - Daowei Li
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, School and Hospital of Stomatology, Jilin University, 1500 Qinghua Road, Changchun 130012, China
| | - Hui Pan
- Department of Endocrinology, Key Laboratory of Endocrinology of National Health Commission, Translation Medicine Centre, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, China
- Key Laboratory of Endocrinology of National Health Commission, Department of Endocrinology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100730, China
| |
Collapse
|
5
|
Grosicki M, Wojnar-Lason K, Mosiolek S, Mateuszuk L, Stojak M, Chlopicki S. Distinct profile of antiviral drugs effects in aortic and pulmonary endothelial cells revealed by high-content microscopy and cell painting assays. Toxicol Appl Pharmacol 2024; 490:117030. [PMID: 38981531 DOI: 10.1016/j.taap.2024.117030] [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: 04/03/2024] [Revised: 06/28/2024] [Accepted: 07/05/2024] [Indexed: 07/11/2024]
Abstract
Antiretroviral therapy have significantly improved the treatment of viral infections and reduced the associated mortality and morbidity rates. However, highly effective antiretroviral therapy (HAART) may lead to an increased risk of cardiovascular diseases, which could be related to endothelial toxicity. Here, seven antiviral drugs (remdesivir, PF-00835231, ritonavir, lopinavir, efavirenz, zidovudine and abacavir) were characterized against aortic (HAEC) and pulmonary (hLMVEC) endothelial cells, using high-content microscopy. The colourimetric study (MTS test) revealed similar toxicity profiles of all antiviral drugs tested in the concentration range of 1 nM-50 μM in aortic and pulmonary endothelial cells. Conversely, the drugs' effects on morphological parameters were more pronounced in HAECs as compared with hLMVECs. Based on the antiviral drugs' effects on the cytoplasmic and nuclei architecture (analyzed by multiple pre-defined parameters including SER texture and STAR morphology), the studied compounds were classified into five distinct morphological subgroups, each linked to a specific cellular response profile. In relation to morphological subgroup classification, antiviral drugs induced a loss of mitochondrial membrane potential, elevated ROS, changed lipid droplets/lysosomal content, decreased von Willebrand factor expression and micronuclei formation or dysregulated cellular autophagy. In conclusion, based on specific changes in endothelial cytoplasm, nuclei and subcellular morphology, the distinct endothelial response was identified for remdesivir, ritonavir, lopinavir, efavirenz, zidovudine and abacavir treatments. The effects detected in aortic endothelial cells were not detected in pulmonary endothelial cells. Taken together, high-content microscopy has proven to be a robust and informative method for endothelial drug profiling that may prove useful in predicting the organ-specific endothelial toxicity of various drugs.
Collapse
Affiliation(s)
- Marek Grosicki
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Krakow, Poland.
| | - Kamila Wojnar-Lason
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Krakow, Poland; Department of Pharmacology, Jagiellonian University Medical College, Krakow, Poland
| | - Sylwester Mosiolek
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Krakow, Poland; Jagiellonian University, Doctoral School of Exact and Natural Sciences, Krakow, Poland
| | - Lukasz Mateuszuk
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Krakow, Poland
| | - Marta Stojak
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Krakow, Poland
| | - Stefan Chlopicki
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Krakow, Poland; Department of Pharmacology, Jagiellonian University Medical College, Krakow, Poland.
| |
Collapse
|
6
|
Green RA, Khaliullin RN, Zhao Z, Ochoa SD, Hendel JM, Chow TL, Moon H, Biggs RJ, Desai A, Oegema K. Automated profiling of gene function during embryonic development. Cell 2024; 187:3141-3160.e23. [PMID: 38759650 PMCID: PMC11166207 DOI: 10.1016/j.cell.2024.04.012] [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: 05/27/2023] [Revised: 02/10/2024] [Accepted: 04/12/2024] [Indexed: 05/19/2024]
Abstract
Systematic functional profiling of the gene set that directs embryonic development is an important challenge. To tackle this challenge, we used 4D imaging of C. elegans embryogenesis to capture the effects of 500 gene knockdowns and developed an automated approach to compare developmental phenotypes. The automated approach quantifies features-including germ layer cell numbers, tissue position, and tissue shape-to generate temporal curves whose parameterization yields numerical phenotypic signatures. In conjunction with a new similarity metric that operates across phenotypic space, these signatures enabled the generation of ranked lists of genes predicted to have similar functions, accessible in the PhenoBank web portal, for ∼25% of essential development genes. The approach identified new gene and pathway relationships in cell fate specification and morphogenesis and highlighted the utilization of specialized energy generation pathways during embryogenesis. Collectively, the effort establishes the foundation for comprehensive analysis of the gene set that builds a multicellular organism.
Collapse
Affiliation(s)
- Rebecca A Green
- Ludwig Institute for Cancer Research, La Jolla, CA 92093, USA; Department of Cell and Developmental Biology, School of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA.
| | | | - Zhiling Zhao
- Ludwig Institute for Cancer Research, La Jolla, CA 92093, USA
| | - Stacy D Ochoa
- Ludwig Institute for Cancer Research, La Jolla, CA 92093, USA
| | | | | | - HongKee Moon
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstr. 108, 01307 Dresden, Germany
| | - Ronald J Biggs
- Ludwig Institute for Cancer Research, La Jolla, CA 92093, USA
| | - Arshad Desai
- Ludwig Institute for Cancer Research, La Jolla, CA 92093, USA; Department of Cell and Developmental Biology, School of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA; Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Karen Oegema
- Ludwig Institute for Cancer Research, La Jolla, CA 92093, USA; Department of Cell and Developmental Biology, School of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA; Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
7
|
Ryoo H, Kimmel H, Rondo E, Underhill GH. Advances in high throughput cell culture technologies for therapeutic screening and biological discovery applications. Bioeng Transl Med 2024; 9:e10627. [PMID: 38818120 PMCID: PMC11135158 DOI: 10.1002/btm2.10627] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 11/13/2023] [Accepted: 11/14/2023] [Indexed: 06/01/2024] Open
Abstract
Cellular phenotypes and functional responses are modulated by the signals present in their microenvironment, including extracellular matrix (ECM) proteins, tissue mechanical properties, soluble signals and nutrients, and cell-cell interactions. To better recapitulate and analyze these complex signals within the framework of more physiologically relevant culture models, high throughput culture platforms can be transformative. High throughput methodologies enable scientists to extract increasingly robust and broad datasets from individual experiments, screen large numbers of conditions for potential hits, better qualify and predict responses for preclinical applications, and reduce reliance on animal studies. High throughput cell culture systems require uniformity, assay miniaturization, specific target identification, and process simplification. In this review, we detail the various techniques that researchers have used to face these challenges and explore cellular responses in a high throughput manner. We highlight several common approaches including two-dimensional multiwell microplates, microarrays, and microfluidic cell culture systems as well as unencapsulated and encapsulated three-dimensional high throughput cell culture systems, featuring multiwell microplates, micromolds, microwells, microarrays, granular hydrogels, and cell-encapsulated microgels. We also discuss current applications of these high throughput technologies, namely stem cell sourcing, drug discovery and predictive toxicology, and personalized medicine, along with emerging opportunities and future impact areas.
Collapse
Affiliation(s)
- Hyeon Ryoo
- Bioengineering DepartmentUniversity of Illinois Urbana‐ChampaignUrbanaIllinoisUSA
| | - Hannah Kimmel
- Bioengineering DepartmentUniversity of Illinois Urbana‐ChampaignUrbanaIllinoisUSA
| | - Evi Rondo
- Bioengineering DepartmentUniversity of Illinois Urbana‐ChampaignUrbanaIllinoisUSA
| | - Gregory H. Underhill
- Bioengineering DepartmentUniversity of Illinois Urbana‐ChampaignUrbanaIllinoisUSA
| |
Collapse
|
8
|
Razdaibiedina A, Brechalov A, Friesen H, Mattiazzi Usaj M, Masinas MPD, Garadi Suresh H, Wang K, Boone C, Ba J, Andrews B. PIFiA: self-supervised approach for protein functional annotation from single-cell imaging data. Mol Syst Biol 2024; 20:521-548. [PMID: 38472305 PMCID: PMC11066028 DOI: 10.1038/s44320-024-00029-6] [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: 08/15/2023] [Revised: 02/27/2024] [Accepted: 02/28/2024] [Indexed: 03/14/2024] Open
Abstract
Fluorescence microscopy data describe protein localization patterns at single-cell resolution and have the potential to reveal whole-proteome functional information with remarkable precision. Yet, extracting biologically meaningful representations from cell micrographs remains a major challenge. Existing approaches often fail to learn robust and noise-invariant features or rely on supervised labels for accurate annotations. We developed PIFiA (Protein Image-based Functional Annotation), a self-supervised approach for protein functional annotation from single-cell imaging data. We imaged the global yeast ORF-GFP collection and applied PIFiA to generate protein feature profiles from single-cell images of fluorescently tagged proteins. We show that PIFiA outperforms existing approaches for molecular representation learning and describe a range of downstream analysis tasks to explore the information content of the feature profiles. Specifically, we cluster extracted features into a hierarchy of functional organization, study cell population heterogeneity, and develop techniques to distinguish multi-localizing proteins and identify functional modules. Finally, we confirm new PIFiA predictions using a colocalization assay, suggesting previously unappreciated biological roles for several proteins. Paired with a fully interactive website ( https://thecellvision.org/pifia/ ), PIFiA is a resource for the quantitative analysis of protein organization within the cell.
Collapse
Affiliation(s)
- Anastasia Razdaibiedina
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
- The Donnelly Centre, University of Toronto, Toronto, ON, Canada
- Vector Institute for Artificial Intelligence, Toronto, ON, Canada
| | - Alexander Brechalov
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
- The Donnelly Centre, University of Toronto, Toronto, ON, Canada
| | - Helena Friesen
- The Donnelly Centre, University of Toronto, Toronto, ON, Canada
| | - Mojca Mattiazzi Usaj
- The Donnelly Centre, University of Toronto, Toronto, ON, Canada
- Department of Chemistry and Biology, Toronto Metropolitan University, Toronto, ON, Canada
| | | | | | - Kyle Wang
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
- The Donnelly Centre, University of Toronto, Toronto, ON, Canada
| | - Charles Boone
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada.
- The Donnelly Centre, University of Toronto, Toronto, ON, Canada.
- RIKEN Center for Sustainable Resource Science, 2-1 Hirosawa, Wako, Saitama, Japan.
| | - Jimmy Ba
- Vector Institute for Artificial Intelligence, Toronto, ON, Canada.
- Department of Computer Science, University of Toronto, Toronto, ON, Canada.
| | - Brenda Andrews
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada.
- The Donnelly Centre, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
9
|
Wu Y, Xi Z, Liu F, Hu W, Feng H, Zhang Q. A deep semantic network-based image segmentation of soybean rust pathogens. FRONTIERS IN PLANT SCIENCE 2024; 15:1340584. [PMID: 38601300 PMCID: PMC11004377 DOI: 10.3389/fpls.2024.1340584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Accepted: 03/11/2024] [Indexed: 04/12/2024]
Abstract
Introduction Asian soybean rust is a highly aggressive leaf-based disease triggered by the obligate biotrophic fungus Phakopsora pachyrhizi which can cause up to 80% yield loss in soybean. The precise image segmentation of fungus can characterize fungal phenotype transitions during growth and help to discover new medicines and agricultural biocides using large-scale phenotypic screens. Methods The improved Mask R-CNN method is proposed to accomplish the segmentation of densely distributed, overlapping and intersecting microimages. First, Res2net is utilized to layer the residual connections in a single residual block to replace the backbone of the original Mask R-CNN, which is then combined with FPG to enhance the feature extraction capability of the network model. Secondly, the loss function is optimized and the CIoU loss function is adopted as the loss function for boundary box regression prediction, which accelerates the convergence speed of the model and meets the accurate classification of high-density spore images. Results The experimental results show that the mAP for detection and segmentation, accuracy of the improved algorithm is improved by 6.4%, 12.3% and 2.2% respectively over the original Mask R-CNN algorithm. Discussion This method is more suitable for the segmentation of fungi images and provide an effective tool for large-scale phenotypic screens of plant fungal pathogens.
Collapse
Affiliation(s)
- Yalin Wu
- Lushan Botanical Garden, Jiangxi Province and Chinese Academy of Sciences, Jiujiang, China
| | - Zhuobin Xi
- Mechanical Electrical Engineering School, Beijing Information Science & Technology University, Beijing, China
| | - Fen Liu
- Lushan Botanical Garden, Jiangxi Province and Chinese Academy of Sciences, Jiujiang, China
| | - Weiming Hu
- Lushan Botanical Garden, Jiangxi Province and Chinese Academy of Sciences, Jiujiang, China
| | - Hongjuan Feng
- Zhongzhen Kejian (ShenZhen) Holdings Co., Ltd, Shenzhen, China
| | - Qinjian Zhang
- Mechanical Electrical Engineering School, Beijing Information Science & Technology University, Beijing, China
| |
Collapse
|
10
|
Jan M, Spangaro A, Lenartowicz M, Mattiazzi Usaj M. From pixels to insights: Machine learning and deep learning for bioimage analysis. Bioessays 2024; 46:e2300114. [PMID: 38058114 DOI: 10.1002/bies.202300114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2023] [Revised: 10/25/2023] [Accepted: 11/13/2023] [Indexed: 12/08/2023]
Abstract
Bioimage analysis plays a critical role in extracting information from biological images, enabling deeper insights into cellular structures and processes. The integration of machine learning and deep learning techniques has revolutionized the field, enabling the automated, reproducible, and accurate analysis of biological images. Here, we provide an overview of the history and principles of machine learning and deep learning in the context of bioimage analysis. We discuss the essential steps of the bioimage analysis workflow, emphasizing how machine learning and deep learning have improved preprocessing, segmentation, feature extraction, object tracking, and classification. We provide examples that showcase the application of machine learning and deep learning in bioimage analysis. We examine user-friendly software and tools that enable biologists to leverage these techniques without extensive computational expertise. This review is a resource for researchers seeking to incorporate machine learning and deep learning in their bioimage analysis workflows and enhance their research in this rapidly evolving field.
Collapse
Affiliation(s)
- Mahta Jan
- Department of Chemistry and Biology, Toronto Metropolitan University, Toronto, Canada
| | - Allie Spangaro
- Department of Chemistry and Biology, Toronto Metropolitan University, Toronto, Canada
| | - Michelle Lenartowicz
- Department of Chemistry and Biology, Toronto Metropolitan University, Toronto, Canada
| | - Mojca Mattiazzi Usaj
- Department of Chemistry and Biology, Toronto Metropolitan University, Toronto, Canada
| |
Collapse
|
11
|
Li T, Gachet Y, Tournier S. MAARS Software for Automatic and Quantitative Analysis of Mitotic Progression. Methods Mol Biol 2024; 2740:275-293. [PMID: 38393482 DOI: 10.1007/978-1-0716-3557-5_17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2024]
Abstract
In this chapter, we describe a software called MAARS (Mitotic Analysis And Recording System) that enables automatic and quantitative analysis of mitotic progression on an open-source platform. This computer-assisted analysis of cell division allows the unbiased acquisition of multiple parameters such as cell shape or size, metaphase or anaphase delays, as well as various mitotic abnormalities. This chapter describes the power of such an expert system to highlight the complexity of the mechanisms required to prevent mitotic chromosome segregation errors, leading to aneuploidy.
Collapse
Affiliation(s)
- Tong Li
- MCD, Centre de Biologie Intégrative, Université de Toulouse, CNRS, UPS, Toulouse Cedex, France
- Wellcome Sanger Institute, Cambridge, UK
| | - Yannick Gachet
- MCD, Centre de Biologie Intégrative, Université de Toulouse, CNRS, UPS, Toulouse Cedex, France.
| | - Sylvie Tournier
- MCD, Centre de Biologie Intégrative, Université de Toulouse, CNRS, UPS, Toulouse Cedex, France.
| |
Collapse
|
12
|
Ortiz-Perez A, Zhang M, Fitzpatrick LW, Izquierdo-Lozano C, Albertazzi L. Advanced optical imaging for the rational design of nanomedicines. Adv Drug Deliv Rev 2024; 204:115138. [PMID: 37980951 DOI: 10.1016/j.addr.2023.115138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 11/06/2023] [Accepted: 11/08/2023] [Indexed: 11/21/2023]
Abstract
Despite the enormous potential of nanomedicines to shape the future of medicine, their clinical translation remains suboptimal. Translational challenges are present in every step of the development pipeline, from a lack of understanding of patient heterogeneity to insufficient insights on nanoparticle properties and their impact on material-cell interactions. Here, we discuss how the adoption of advanced optical microscopy techniques, such as super-resolution optical microscopies, correlative techniques, and high-content modalities, could aid the rational design of nanocarriers, by characterizing the cell, the nanomaterial, and their interaction with unprecedented spatial and/or temporal detail. In this nanomedicine arena, we will discuss how the implementation of these techniques, with their versatility and specificity, can yield high volumes of multi-parametric data; and how machine learning can aid the rapid advances in microscopy: from image acquisition to data interpretation.
Collapse
Affiliation(s)
- Ana Ortiz-Perez
- Department of Biomedical Engineering, Institute of Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Miao Zhang
- Department of Biomedical Engineering, Institute of Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Laurence W Fitzpatrick
- Department of Biomedical Engineering, Institute of Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Cristina Izquierdo-Lozano
- Department of Biomedical Engineering, Institute of Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Lorenzo Albertazzi
- Department of Biomedical Engineering, Institute of Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands.
| |
Collapse
|
13
|
Pajanoja C, Kerosuo L. ShapeMetrics: A 3D Cell Segmentation Pipeline for Single-Cell Spatial Morphometric Analysis. Methods Mol Biol 2024; 2767:263-273. [PMID: 37219813 DOI: 10.1007/7651_2023_489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
There is a growing need for single-cell level data analysis in correlation with the advancements of microscopy techniques. Morphology-based statistics gathered from individual cells are essential for detection and quantification of even subtle changes within the complex tissues, yet the information available from high-resolution imaging is oftentimes sub-optimally utilized due to the lack of proper computational analysis software. Here we present ShapeMetrics, a 3D cell segmentation pipeline that we have developed to identify, analyze, and quantify single cells in an image. This MATLAB-based script enables users to extract morphological parameters, such as ellipticity, longest axis, cell elongation, or the ratio between cell volume and surface area. We have specifically invested in creating a user-friendly pipeline, aimed for biologists with a limited computational background. Our pipeline is presented with detailed stepwise instructions, starting from the establishment of machine learning-based prediction files of immuno-labeled cell membranes followed by the application of 3D cell segmentation and parameter extraction script, leading to the morphometric analysis and spatial visualization of cell clusters defined by their morphometric features.
Collapse
Affiliation(s)
- Ceren Pajanoja
- Neural Crest Development and Disease Unit, National Institute of Dental and Craniofacial Research, Intramural Research Program, Neural Crest Development and Disease Unit, National Institutes of Health, Bethesda, ML, USA
- Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Laura Kerosuo
- Neural Crest Development and Disease Unit, National Institute of Dental and Craniofacial Research, Intramural Research Program, Neural Crest Development and Disease Unit, National Institutes of Health, Bethesda, ML, USA
| |
Collapse
|
14
|
Wang R, Shi X, Li C. Insights into the Surface Binding and Structural Interference of Polyphenols with the Membrane Raft Domains in Relation to Their Distinctive Ability to Inhibit Preadipocyte Differentiation in 3T3-L1 Cells. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:19845-19855. [PMID: 38050784 DOI: 10.1021/acs.jafc.3c06747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/06/2023]
Abstract
Polyphenols with different structures have shown distinct variations in their ability to inhibit the differentiation of 3T3-L1 preadipocytes. However, the underlying mechanisms for these differences remain unclear. In the present study, the surface binding of polyphenols to different membrane domains was explored using coarse-grained molecular dynamics simulation (CG-MDs). Subsequently, this surface binding was confirmed in the liposome system by microscale thermophoresis. Additionally, the interference of polyphenols on the membrane raft's structure was studied through atomic force microscopy and high-content screening fluorescence microscopy. The results indicated that polyphenols with a differentiation-inhibitory ability, such as epicatechin-3-gallate (ECG) and epicatechin-3-gallate-(4β → 8, 2β → O → 7)-epicatechin-3-gallate (A-type ECG dimer), exhibited strong binding to ordered domains enriched in sphingolipids and cholesterol. This binding led to the structural disruption of membrane rafts by altering their size and shape, with the binding constant of 3.8 μM for ECG and 0.3 μM for A-type ECG dimer, respectively. In contrast, epicatechin (EC) with little differentiation-inhibitory ability had no effects on membrane rafts, and its binding constant with the ordered domain was 380.6 μM. Overall, the surface binding of polyphenols to ordered domains and the resulting disruption of membrane rafts structure might be a fundamental mechanism by which polyphenols inhibited the differentiation of 3T3-L1 preadipocytes.
Collapse
Affiliation(s)
- Ruifeng Wang
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Xin Shi
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Chunmei Li
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan, Hubei 430070, China
- Key Laboratory of Environment Correlative Food Science, Ministry of Education, Wuhan, Hubei 430070, China
| |
Collapse
|
15
|
Boyang H, Yangyanqiu W, Wenting R, Chenxin Y, Jian C, Zhanbo Q, Yanjun Y, Qiang Y, Shuwen H. Application and progress of highcontent imaging in molecular biology. Biotechnol J 2023; 18:e2300170. [PMID: 37639283 DOI: 10.1002/biot.202300170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 08/03/2023] [Accepted: 08/22/2023] [Indexed: 08/29/2023]
Abstract
Humans have adopted many different methods to explore matter imaging, among which high content imaging (HCI) could conduct automated imaging analysis of cells while maintaining its structural and functional integrity. Meanwhile, as one of the most important research tools for diagnosing human diseases, HCI is widely used in the frontier of medical research, and its future application has attracted researchers' great interests. Here, the meaning of HCI was briefly explained, the history of optical imaging and the birth of HCI were described, and the experimental methods of HCI were described. Furthermore, the directions of the application of HCI were highlighted in five aspects: protein localization changes, gene identification, chemical and genetic analysis, microbiology, and drug discovery. Most importantly, some challenges and future directions of HCI were discussed, and the application and optimization of HCI were expected to be further explored.
Collapse
Affiliation(s)
- Hu Boyang
- Huzhou Hospital of Zhejiang University, Affiliated Central Hospital Huzhou University, Huzhou, China
| | - Wang Yangyanqiu
- Huzhou Hospital of Zhejiang University, Affiliated Central Hospital Huzhou University, Huzhou, China
| | - Rui Wenting
- Huzhou Hospital of Zhejiang University, Affiliated Central Hospital Huzhou University, Huzhou, China
| | - Yan Chenxin
- Shulan International Medical School, Zhejiang Shuren University, Hangzhou, China
| | - Chu Jian
- Fifth Affiliated Clinical Medical College of Zhejiang Chinese Medical University, Huzhou Central Hospital, Huzhou, China
| | - Qu Zhanbo
- Fifth Affiliated Clinical Medical College of Zhejiang Chinese Medical University, Huzhou Central Hospital, Huzhou, China
| | - Yao Yanjun
- Huzhou Hospital of Zhejiang University, Affiliated Central Hospital Huzhou University, Huzhou, China
| | - Yan Qiang
- Huzhou Hospital of Zhejiang University, Affiliated Central Hospital Huzhou University, Huzhou, China
| | - Han Shuwen
- Huzhou Hospital of Zhejiang University, Affiliated Central Hospital Huzhou University, Huzhou, China
- Key Laboratory of Multiomics Research and Clinical Transformation of Digestive Cancer of Huzhou, Huzhou, China
| |
Collapse
|
16
|
Antonelli L, Polverino F, Albu A, Hada A, Asteriti IA, Degrassi F, Guarguaglini G, Maddalena L, Guarracino MR. ALFI: Cell cycle phenotype annotations of label-free time-lapse imaging data from cultured human cells. Sci Data 2023; 10:677. [PMID: 37794110 PMCID: PMC10551030 DOI: 10.1038/s41597-023-02540-1] [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: 02/21/2023] [Accepted: 09/05/2023] [Indexed: 10/06/2023] Open
Abstract
Detecting and tracking multiple moving objects in a video is a challenging task. For living cells, the task becomes even more arduous as cells change their morphology over time, can partially overlap, and mitosis leads to new cells. Differently from fluorescence microscopy, label-free techniques can be easily applied to almost all cell lines, reducing sample preparation complexity and phototoxicity. In this study, we present ALFI, a dataset of images and annotations for label-free microscopy, made publicly available to the scientific community, that notably extends the current panorama of expertly labeled data for detection and tracking of cultured living nontransformed and cancer human cells. It consists of 29 time-lapse image sequences from HeLa, U2OS, and hTERT RPE-1 cells under different experimental conditions, acquired by differential interference contrast microscopy, for a total of 237.9 hours. It contains various annotations (pixel-wise segmentation masks, object-wise bounding boxes, tracking information). The dataset is useful for testing and comparing methods for identifying interphase and mitotic events and reconstructing their lineage, and for discriminating different cellular phenotypes.
Collapse
Affiliation(s)
- Laura Antonelli
- ICAR, Institute for High-Performance Computing and Networking, National Research Council, Naples, Italy
| | - Federica Polverino
- IBPM, Institute of Molecular Biology and Pathology, National Research Council, Rome, Italy
| | - Alexandra Albu
- Department of Economics and Law, University of Cassino and Southern Lazio, Cassino, Italy
| | - Aroj Hada
- Department of Economics and Law, University of Cassino and Southern Lazio, Cassino, Italy
| | - Italia A Asteriti
- IBPM, Institute of Molecular Biology and Pathology, National Research Council, Rome, Italy
| | - Francesca Degrassi
- IBPM, Institute of Molecular Biology and Pathology, National Research Council, Rome, Italy
| | - Giulia Guarguaglini
- IBPM, Institute of Molecular Biology and Pathology, National Research Council, Rome, Italy.
| | - Lucia Maddalena
- ICAR, Institute for High-Performance Computing and Networking, National Research Council, Naples, Italy.
| | - Mario R Guarracino
- Department of Economics and Law, University of Cassino and Southern Lazio, Cassino, Italy
- Laboratory of Algorithms and Technologies for Networks Analysis, National Research University Higher School of Economics, Moscow, Russia
| |
Collapse
|
17
|
Beriashvili D, Yao R, D'Amico F, Krafčíková M, Gurinov A, Safeer A, Cai X, Mulder MPC, Liu Y, Folkers GE, Baldus M. A high-field cellular DNP-supported solid-state NMR approach to study proteins with sub-cellular specificity. Chem Sci 2023; 14:9892-9899. [PMID: 37736634 PMCID: PMC10510770 DOI: 10.1039/d3sc02117c] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 08/24/2023] [Indexed: 09/23/2023] Open
Abstract
Studying the structural aspects of proteins within sub-cellular compartments is of growing interest. Dynamic nuclear polarization supported solid-state NMR (DNP-ssNMR) is uniquely suited to provide such information, but critically lacks the desired sensitivity and resolution. Here we utilize SNAPol-1, a novel biradical, to conduct DNP-ssNMR at high-magnetic fields (800 MHz/527 GHz) inside HeLa cells and isolated cell nuclei electroporated with [13C,15N] labeled ubiquitin. We report that SNAPol-1 passively diffuses and homogenously distributes within whole cells and cell nuclei providing ubiquitin spectra of high sensitivity and remarkably improved spectral resolution. For cell nuclei, physical enrichment facilitates a further 4-fold decrease in measurement time and provides an exclusive structural view of the nuclear ubiquitin pool. Taken together, these advancements enable atomic interrogation of protein conformational plasticity at atomic resolution and with sub-cellular specificity.
Collapse
Affiliation(s)
- David Beriashvili
- NMR Spectroscopy, Bijvoet Center for Biomolecular Research, Utrecht University Padualaan 8 3584 CH Utrecht The Netherlands
| | - Ru Yao
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University Tianjin 300070 P. R. China
| | - Francesca D'Amico
- Department of Cell and Chemical Biology, Leiden University Medical Center (LUMC) Einthovenweg 20 2333 ZC Leiden The Netherlands
| | - Michaela Krafčíková
- NMR Spectroscopy, Bijvoet Center for Biomolecular Research, Utrecht University Padualaan 8 3584 CH Utrecht The Netherlands
| | - Andrei Gurinov
- NMR Spectroscopy, Bijvoet Center for Biomolecular Research, Utrecht University Padualaan 8 3584 CH Utrecht The Netherlands
| | - Adil Safeer
- NMR Spectroscopy, Bijvoet Center for Biomolecular Research, Utrecht University Padualaan 8 3584 CH Utrecht The Netherlands
| | - Xinyi Cai
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University Tianjin 300070 P. R. China
| | - Monique P C Mulder
- Department of Cell and Chemical Biology, Leiden University Medical Center (LUMC) Einthovenweg 20 2333 ZC Leiden The Netherlands
| | - Yangping Liu
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University Tianjin 300070 P. R. China
| | - Gert E Folkers
- NMR Spectroscopy, Bijvoet Center for Biomolecular Research, Utrecht University Padualaan 8 3584 CH Utrecht The Netherlands
| | - Marc Baldus
- NMR Spectroscopy, Bijvoet Center for Biomolecular Research, Utrecht University Padualaan 8 3584 CH Utrecht The Netherlands
| |
Collapse
|
18
|
Xing X, Sun M, Guo Z, Zhao Y, Cai Y, Zhou P, Wang H, Gao W, Li P, Yang H. Functional annotation map of natural compounds in traditional Chinese medicines library: TCMs with myocardial protection as a case. Acta Pharm Sin B 2023; 13:3802-3816. [PMID: 37719385 PMCID: PMC10502289 DOI: 10.1016/j.apsb.2023.06.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 05/14/2023] [Accepted: 05/31/2023] [Indexed: 09/19/2023] Open
Abstract
The chemical complexity of traditional Chinese medicines (TCMs) makes the active and functional annotation of natural compounds challenging. Herein, we developed the TCMs-Compounds Functional Annotation platform (TCMs-CFA) for large-scale predicting active compounds with potential mechanisms from TCM complex system, without isolating and activity testing every single compound one by one. The platform was established based on the integration of TCMs knowledge base, chemome profiling, and high-content imaging. It mainly included: (1) selection of herbal drugs of target based on TCMs knowledge base; (2) chemome profiling of TCMs extract library by LC‒MS; (3) cytological profiling of TCMs extract library by high-content cell-based imaging; (4) active compounds discovery by combining each mass signal and multi-parametric cell phenotypes; (5) construction of functional annotation map for predicting the potential mechanisms of lead compounds. In this stud TCMs with myocardial protection were applied as a case study, and validated for the feasibility and utility of the platform. Seven frequently used herbal drugs (Ginseng, etc.) were screened from 100,000 TCMs formulas for myocardial protection and subsequently prepared as a library of 700 extracts. By using TCMs-CFA platform, 81 lead compounds, including 10 novel bioactive ones, were quickly identified by correlating 8089 mass signals with 170,100 cytological parameters from an extract library. The TCMs-CFA platform described a new evidence-led tool for the rapid discovery process by data mining strategies, which is valuable for novel lead compounds from TCMs. All computations are done through Python and are publicly available on GitHub.
Collapse
Affiliation(s)
- Xudong Xing
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Mengru Sun
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Zifan Guo
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Yongjuan Zhao
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Yuru Cai
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Ping Zhou
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Huiying Wang
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Wen Gao
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Ping Li
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Hua Yang
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| |
Collapse
|
19
|
André O, Kumra Ahnlide J, Norlin N, Swaminathan V, Nordenfelt P. Data-driven microscopy allows for automated context-specific acquisition of high-fidelity image data. CELL REPORTS METHODS 2023; 3:100419. [PMID: 37056378 PMCID: PMC10088093 DOI: 10.1016/j.crmeth.2023.100419] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Revised: 12/20/2022] [Accepted: 02/10/2023] [Indexed: 04/15/2023]
Abstract
Light microscopy is a powerful single-cell technique that allows for quantitative spatial information at subcellular resolution. However, unlike flow cytometry and single-cell sequencing techniques, microscopy has issues achieving high-quality population-wide sample characterization while maintaining high resolution. Here, we present a general framework, data-driven microscopy (DDM) that uses real-time population-wide object characterization to enable data-driven high-fidelity imaging of relevant phenotypes based on the population context. DDM combines data-independent and data-dependent steps to synergistically enhance data acquired using different imaging modalities. As a proof of concept, we develop and apply DDM with plugins for improved high-content screening and live adaptive microscopy for cell migration and infection studies that capture events of interest, rare or common, with high precision and resolution. We propose that DDM can reduce human bias, increase reproducibility, and place single-cell characteristics in the context of the sample population when interpreting microscopy data, leading to an increase in overall data fidelity.
Collapse
Affiliation(s)
- Oscar André
- Department of Clinical Sciences, Faculty of Medicine, Lund University, Lund, Sweden
| | | | - Nils Norlin
- Department of Experimental Medical Science, Lund University, Lund, Sweden
- Lund University Bioimaging Centre, Lund, Sweden
| | - Vinay Swaminathan
- Department of Clinical Sciences, Wallenberg Centre for Molecular Medicine, Division of Oncology, Lund University, Lund, Sweden
| | - Pontus Nordenfelt
- Department of Clinical Sciences, Faculty of Medicine, Lund University, Lund, Sweden
| |
Collapse
|
20
|
Dual roles of TRIM3 in colorectal cancer by retaining p53 in the cytoplasm to decrease its nuclear expression. Cell Death Discov 2023; 9:85. [PMID: 36894560 PMCID: PMC9998637 DOI: 10.1038/s41420-023-01386-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 02/22/2023] [Accepted: 02/27/2023] [Indexed: 03/11/2023] Open
Abstract
Colorectal cancer is a very heterogeneous disease caused by the interaction of genetic and environmental factors. P53, as a frequent mutation gene, plays a critical role in the adenoma-carcinoma transition during the tumorous pathological process. Our team discovered TRIM3 as a tumor-associated gene in CRC by high-content screening techniques. TRIM3 demonstrated both tumor-suppressive and tumorigenic features in cell experiments dependent on the cell status of wild or mutant p53. TRIM3 could directly interact with the C terminus of p53 (residues 320 to 393), a common segment of wtp53 and mutp53. Moreover, TRIM3 could exert different neoplastic features by retaining p53 in the cytoplasm to decrease its nuclear expression in a wtp53 or mutp53-dependent pathway. Chemotherapy resistance develops in nearly all patients with advanced CRC and seriously limits the therapeutic efficacies of anticancer drugs. TRIM3 could reverse the chemotherapy resistance of oxaliplatin in mutp53 CRC cells by degradation of mutp53 in the nuclei to downregulate the multidrug resistance gene. Therefore, TRIM3 could be a potential therapeutic strategy to improve the survival of CRC patients with mutp53.
Collapse
|
21
|
Yu N, Zhang F, Tang X, Liu Y, Zhang J, Yang B, Wang Q. Hierarchical hydrogel microarrays fabricated based on a microfluidic printing platform for high-throughput screening of stem cell lineage specification. Acta Biomater 2023; 161:144-153. [PMID: 36868445 DOI: 10.1016/j.actbio.2023.02.036] [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: 10/21/2022] [Revised: 02/02/2023] [Accepted: 02/23/2023] [Indexed: 03/05/2023]
Abstract
2D cell cultures are suitable for rapid exploration of the factors in the extracellular matrix affecting the development of cells. The technology of the micrometre-sized hydrogel array provides a feasible, miniaturized, and high-throughput strategy for the process. However, current microarray devices lack a handy and parallelized methodology in sample treatment, which makes the process of high-throughput cell screening (HTCS) expensive and inefficient. Here, based on the functionalization of micro-nano structures and the fluid control capability of microfluidic chips, we build a microfluidic spotting-screening platform (MSSP). The MSSP can print 20000 microdroplet spots within 5 min, coupled with a simple strategy for parallel addition of compound libraries. Compared with open microdroplet arrays, the MSSP can control the evaporation rate of nanoliter droplets, providing a stable fabrication platform for hydrogel-microarray-based materials. As a proof-of-concept demonstration, the MSSP successfully controlled the adhesion, adipogenic, and osteogenic differentiation behavior of mesenchymal stem cells by rationally designing the substrate stiffness, adhesion area, and cell density. We anticipate that the MSSP may provide an accessible and promising tool for hydrogel-based HTCS. STATEMENT OF SIGNIFICANCE: High-throughput screening of cells is a common approach to improve the efficiency of biological experiments, and one challenge of the existing technologies is to achieve rapid and precise cell screening with a low-cost and simple strategy. Through the integration of the microfluidic and micro-nanostructure technologies, we fabricated a microfluidic spotting-screening platforms. Benefiting from the flexible control of the fluids, the device can print 20000 microdroplet spots within 5 min, coupled with a simple procedure for parallel addition of compound libraries. High-throughput screening of stem cell lineage specification has also been achieved by the platform, which provides a high-throughput, high-content information extraction strategy for cell-biomaterial interaction research.
Collapse
Affiliation(s)
- Nianzuo Yu
- Department of Gastrointestinal Surgery, The First Hospital of Jilin University, Changchun, 130031, PR China; Joint Laboratory of Opto-Functional Theranostics in Medicine and Chemistry, The First Hospital of Jilin University, Changchun, 130031, PR China; State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, 130012, PR China
| | - Feiran Zhang
- Joint Laboratory of Opto-Functional Theranostics in Medicine and Chemistry, The First Hospital of Jilin University, Changchun, 130031, PR China
| | - Xiaoduo Tang
- Joint Laboratory of Opto-Functional Theranostics in Medicine and Chemistry, The First Hospital of Jilin University, Changchun, 130031, PR China
| | - Yongshun Liu
- State Key Laboratory of Integrated Optoelectronics, College of Electronic Science and Engineering, Jilin University, Changchun, 130012, PR China.
| | - Junhu Zhang
- Joint Laboratory of Opto-Functional Theranostics in Medicine and Chemistry, The First Hospital of Jilin University, Changchun, 130031, PR China; State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, 130012, PR China.
| | - Bai Yang
- Joint Laboratory of Opto-Functional Theranostics in Medicine and Chemistry, The First Hospital of Jilin University, Changchun, 130031, PR China
| | - Quan Wang
- Department of Gastrointestinal Surgery, The First Hospital of Jilin University, Changchun, 130031, PR China.
| |
Collapse
|
22
|
Razdaibiedina A, Brechalov A, Friesen H, Usaj MM, Masinas MPD, Suresh HG, Wang K, Boone C, Ba J, Andrews B. PIFiA: Self-supervised Approach for Protein Functional Annotation from Single-Cell Imaging Data. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.24.529975. [PMID: 36909656 PMCID: PMC10002629 DOI: 10.1101/2023.02.24.529975] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/03/2023]
Abstract
Fluorescence microscopy data describe protein localization patterns at single-cell resolution and have the potential to reveal whole-proteome functional information with remarkable precision. Yet, extracting biologically meaningful representations from cell micrographs remains a major challenge. Existing approaches often fail to learn robust and noise-invariant features or rely on supervised labels for accurate annotations. We developed PIFiA, (Protein Image-based Functional Annotation), a self-supervised approach for protein functional annotation from single-cell imaging data. We imaged the global yeast ORF-GFP collection and applied PIFiA to generate protein feature profiles from single-cell images of fluorescently tagged proteins. We show that PIFiA outperforms existing approaches for molecular representation learning and describe a range of downstream analysis tasks to explore the information content of the feature profiles. Specifically, we cluster extracted features into a hierarchy of functional organization, study cell population heterogeneity, and develop techniques to distinguish multi-localizing proteins and identify functional modules. Finally, we confirm new PIFiA predictions using a colocalization assay, suggesting previously unappreciated biological roles for several proteins. Paired with a fully interactive website (https://thecellvision.org/pifia/), PIFiA is a resource for the quantitative analysis of protein organization within the cell.
Collapse
Affiliation(s)
- Anastasia Razdaibiedina
- Department of Molecular Genetics, University of Toronto, Toronto ON, Canada
- The Donnelly Centre, University of Toronto, Toronto ON, Canada
- Vector Institute for Artificial Intelligence, Toronto ON, Canada
| | - Alexander Brechalov
- Department of Molecular Genetics, University of Toronto, Toronto ON, Canada
- The Donnelly Centre, University of Toronto, Toronto ON, Canada
| | - Helena Friesen
- The Donnelly Centre, University of Toronto, Toronto ON, Canada
| | | | | | | | - Kyle Wang
- Department of Molecular Genetics, University of Toronto, Toronto ON, Canada
- The Donnelly Centre, University of Toronto, Toronto ON, Canada
| | - Charles Boone
- Department of Molecular Genetics, University of Toronto, Toronto ON, Canada
- The Donnelly Centre, University of Toronto, Toronto ON, Canada
- RIKEN Center for Sustainable Resource Science, 2-1 Hirosawa, Wako, Saitama, Japan
| | - Jimmy Ba
- Department of Computer Science, University of Toronto, Toronto ON, Canada
- Vector Institute for Artificial Intelligence, Toronto ON, Canada
| | - Brenda Andrews
- Department of Molecular Genetics, University of Toronto, Toronto ON, Canada
- The Donnelly Centre, University of Toronto, Toronto ON, Canada
| |
Collapse
|
23
|
Zhang L, Liang X, Takáč T, Komis G, Li X, Zhang Y, Ovečka M, Chen Y, Šamaj J. Spatial proteomics of vesicular trafficking: coupling mass spectrometry and imaging approaches in membrane biology. PLANT BIOTECHNOLOGY JOURNAL 2023; 21:250-269. [PMID: 36204821 PMCID: PMC9884029 DOI: 10.1111/pbi.13929] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 08/14/2022] [Accepted: 09/08/2022] [Indexed: 06/16/2023]
Abstract
In plants, membrane compartmentalization requires vesicle trafficking for communication among distinct organelles. Membrane proteins involved in vesicle trafficking are highly dynamic and can respond rapidly to changes in the environment and to cellular signals. Capturing their localization and dynamics is thus essential for understanding the mechanisms underlying vesicular trafficking pathways. Quantitative mass spectrometry and imaging approaches allow a system-wide dissection of the vesicular proteome, the characterization of ligand-receptor pairs and the determination of secretory, endocytic, recycling and vacuolar trafficking pathways. In this review, we highlight major proteomics and imaging methods employed to determine the location, distribution and abundance of proteins within given trafficking routes. We focus in particular on methodologies for the elucidation of vesicle protein dynamics and interactions and their connections to downstream signalling outputs. Finally, we assess their biological applications in exploring different cellular and subcellular processes.
Collapse
Affiliation(s)
- Liang Zhang
- State Key Laboratory of Plant Physiology and Biochemistry, College of Biological SciencesChina Agricultural UniversityBeijingChina
- College of Life ScienceHenan Normal UniversityXinxiangChina
| | - Xinlin Liang
- State Key Laboratory of Plant Physiology and Biochemistry, College of Biological SciencesChina Agricultural UniversityBeijingChina
| | - Tomáš Takáč
- Department of Biotechnology, Faculty of SciencePalacky University OlomoucOlomoucCzech Republic
| | - George Komis
- Department of Cell Biology, Centre of the Region Hana for Biotechnological and Agricultural Research, Faculty of SciencePalacky University OlomoucOlomoucCzech Republic
| | - Xiaojuan Li
- College of Biological Sciences and TechnologyBeijing Forestry UniversityBeijingChina
| | - Yuan Zhang
- College of Biological Sciences and TechnologyBeijing Forestry UniversityBeijingChina
| | - Miroslav Ovečka
- Department of Biotechnology, Faculty of SciencePalacky University OlomoucOlomoucCzech Republic
| | - Yanmei Chen
- State Key Laboratory of Plant Physiology and Biochemistry, College of Biological SciencesChina Agricultural UniversityBeijingChina
| | - Jozef Šamaj
- Department of Biotechnology, Faculty of SciencePalacky University OlomoucOlomoucCzech Republic
| |
Collapse
|
24
|
El Hage K, Babault N, Maciejak O, Desforges B, Craveur P, Steiner E, Rengifo-Gonzalez JC, Henrie H, Clement MJ, Joshi V, Bouhss A, Wang L, Bauvais C, Pastré D. Targeting RNA:protein interactions with an integrative approach leads to the identification of potent YBX1 inhibitors. eLife 2023; 12:e80387. [PMID: 36651723 PMCID: PMC9928419 DOI: 10.7554/elife.80387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 01/17/2023] [Indexed: 01/19/2023] Open
Abstract
RNA-protein interactions (RPIs) are promising targets for developing new molecules of therapeutic interest. Nevertheless, challenges arise from the lack of methods and feedback between computational and experimental techniques during the drug discovery process. Here, we tackle these challenges by developing a drug screening approach that integrates chemical, structural and cellular data from both advanced computational techniques and a method to score RPIs in cells for the development of small RPI inhibitors; and we demonstrate its robustness by targeting Y-box binding protein 1 (YB-1), a messenger RNA-binding protein involved in cancer progression and resistance to chemotherapy. This approach led to the identification of 22 hits validated by molecular dynamics (MD) simulations and nuclear magnetic resonance (NMR) spectroscopy of which 11 were found to significantly interfere with the binding of messenger RNA (mRNA) to YB-1 in cells. One of our leads is an FDA-approved poly(ADP-ribose) polymerase 1 (PARP-1) inhibitor. This work shows the potential of our integrative approach and paves the way for the rational development of RPI inhibitors.
Collapse
Affiliation(s)
- Krystel El Hage
- Université Paris-Saclay, INSERM U1204, Univ Evry, Structure-Activité des Biomolécules Normales et Pathologiques (SABNP)EvryFrance
| | | | - Olek Maciejak
- Université Paris-Saclay, INSERM U1204, Univ Evry, Structure-Activité des Biomolécules Normales et Pathologiques (SABNP)EvryFrance
| | - Bénédicte Desforges
- Université Paris-Saclay, INSERM U1204, Univ Evry, Structure-Activité des Biomolécules Normales et Pathologiques (SABNP)EvryFrance
| | | | - Emilie Steiner
- Université Paris-Saclay, INSERM U1204, Univ Evry, Structure-Activité des Biomolécules Normales et Pathologiques (SABNP)EvryFrance
| | - Juan Carlos Rengifo-Gonzalez
- Université Paris-Saclay, INSERM U1204, Univ Evry, Structure-Activité des Biomolécules Normales et Pathologiques (SABNP)EvryFrance
| | - Hélène Henrie
- Université Paris-Saclay, INSERM U1204, Univ Evry, Structure-Activité des Biomolécules Normales et Pathologiques (SABNP)EvryFrance
| | - Marie-Jeanne Clement
- Université Paris-Saclay, INSERM U1204, Univ Evry, Structure-Activité des Biomolécules Normales et Pathologiques (SABNP)EvryFrance
| | - Vandana Joshi
- Université Paris-Saclay, INSERM U1204, Univ Evry, Structure-Activité des Biomolécules Normales et Pathologiques (SABNP)EvryFrance
| | - Ahmed Bouhss
- Université Paris-Saclay, INSERM U1204, Univ Evry, Structure-Activité des Biomolécules Normales et Pathologiques (SABNP)EvryFrance
| | - Liya Wang
- Université Paris-Saclay, INSERM U1204, Univ Evry, Structure-Activité des Biomolécules Normales et Pathologiques (SABNP)EvryFrance
| | | | - David Pastré
- Université Paris-Saclay, INSERM U1204, Univ Evry, Structure-Activité des Biomolécules Normales et Pathologiques (SABNP)EvryFrance
| |
Collapse
|
25
|
Pelicci S, Furia L, Pelicci PG, Faretta M. Correlative Multi-Modal Microscopy: A Novel Pipeline for Optimizing Fluorescence Microscopy Resolutions in Biological Applications. Cells 2023; 12:cells12030354. [PMID: 36766696 PMCID: PMC9913119 DOI: 10.3390/cells12030354] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/05/2023] [Accepted: 01/16/2023] [Indexed: 01/20/2023] Open
Abstract
The modern fluorescence microscope is the convergence point of technologies with different performances in terms of statistical sampling, number of simultaneously analyzed signals, and spatial resolution. However, the best results are usually obtained by maximizing only one of these parameters and finding a compromise for the others, a limitation that can become particularly significant when applied to cell biology and that can reduce the spreading of novel optical microscopy tools among research laboratories. Super resolution microscopy and, in particular, molecular localization-based approaches provide a spatial resolution and a molecular localization precision able to explore the scale of macromolecular complexes in situ. However, its use is limited to restricted regions, and consequently few cells, and frequently no more than one or two parameters. Correlative microscopy, obtained by the fusion of different optical technologies, can consequently surpass this barrier by merging results from different spatial scales. We discuss here the use of an acquisition and analysis correlative microscopy pipeline to obtain high statistical sampling, high content, and maximum spatial resolution by combining widefield, confocal, and molecular localization microscopy.
Collapse
Affiliation(s)
- Simone Pelicci
- Department of Experimental Oncology, European Institute of Oncology IRCCS, 20139 Milan, Italy
| | - Laura Furia
- Department of Experimental Oncology, European Institute of Oncology IRCCS, 20139 Milan, Italy
| | - Pier Giuseppe Pelicci
- Department of Experimental Oncology, European Institute of Oncology IRCCS, 20139 Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy
| | - Mario Faretta
- Department of Experimental Oncology, European Institute of Oncology IRCCS, 20139 Milan, Italy
- Correspondence:
| |
Collapse
|
26
|
Krentzel D, Shorte SL, Zimmer C. Deep learning in image-based phenotypic drug discovery. Trends Cell Biol 2023:S0962-8924(22)00262-8. [PMID: 36623998 DOI: 10.1016/j.tcb.2022.11.011] [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: 08/25/2022] [Revised: 11/26/2022] [Accepted: 11/29/2022] [Indexed: 01/08/2023]
Abstract
Modern drug discovery approaches often use high-content imaging to systematically study the effect on cells of large libraries of chemical compounds. By automatically screening thousands or millions of images to identify specific drug-induced cellular phenotypes, for example, altered cellular morphology, these approaches can reveal 'hit' compounds offering therapeutic promise. In the past few years, artificial intelligence (AI) methods based on deep learning (DL) [a family of machine learning (ML) techniques] have disrupted virtually all image analysis tasks, from image classification to segmentation. These powerful methods also promise to impact drug discovery by accelerating the identification of effective drugs and their modes of action. In this review, we highlight applications and adaptations of ML, especially DL methods for cell-based phenotypic drug discovery (PDD).
Collapse
Affiliation(s)
- Daniel Krentzel
- Institut Pasteur, Université Paris Cité, Imaging and Modeling Unit, F-75015 Paris, France; Institut Pasteur, Joint International Unit Artificial Intelligence for Image-based Drug Discovery & Development (PIU-Ai3D), F-75015 Paris, France.
| | - Spencer L Shorte
- Institut Pasteur, Joint International Unit Artificial Intelligence for Image-based Drug Discovery & Development (PIU-Ai3D), F-75015 Paris, France; Institut Pasteur, Université Paris Cité, Photonic Bio-Imaging, Centre de Ressources et Recherches Technologiques (UTechS-PBI, C2RT), F-75015 Paris, France
| | - Christophe Zimmer
- Institut Pasteur, Université Paris Cité, Imaging and Modeling Unit, F-75015 Paris, France; Institut Pasteur, Joint International Unit Artificial Intelligence for Image-based Drug Discovery & Development (PIU-Ai3D), F-75015 Paris, France.
| |
Collapse
|
27
|
Mou M, Pan Z, Lu M, Sun H, Wang Y, Luo Y, Zhu F. Application of Machine Learning in Spatial Proteomics. J Chem Inf Model 2022; 62:5875-5895. [PMID: 36378082 DOI: 10.1021/acs.jcim.2c01161] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Spatial proteomics is an interdisciplinary field that investigates the localization and dynamics of proteins, and it has gained extensive attention in recent years, especially the subcellular proteomics. Numerous evidence indicate that the subcellular localization of proteins is associated with various cellular processes and disease progression. Mass spectrometry (MS)-based and imaging-based experimental approaches have been developed to acquire large-scale spatial proteomic data. To allow the reliable analysis of increasingly complex spatial proteomics data, machine learning (ML) methods have been widely used in both MS-based and imaging-based spatial proteomic data analysis pipelines. Here, we comprehensively survey the applications of ML in spatial proteomics from following aspects: (1) data resources for spatial proteome are comprehensively introduced; (2) the roles of different ML algorithms in data analysis pipelines are elaborated; (3) successful applications of spatial proteomics and several analytical tools integrating ML methods are presented; (4) challenges existing in modern ML-based spatial proteomics studies are discussed. This review provides guidelines for researchers seeking to apply ML methods to analyze spatial proteomic data and can facilitate insightful understanding of cell biology as well as the future research in medical and drug discovery communities.
Collapse
Affiliation(s)
- Minjie Mou
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Ziqi Pan
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Mingkun Lu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Huaicheng Sun
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yunxia Wang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yongchao Luo
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Feng Zhu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
28
|
The unperturbed picture: Label-free real-time optical monitoring of cells and extracellular vesicles for therapy. CURRENT OPINION IN BIOMEDICAL ENGINEERING 2022. [DOI: 10.1016/j.cobme.2022.100414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
|
29
|
Bettauer V, Costa ACBP, Omran RP, Massahi S, Kirbizakis E, Simpson S, Dumeaux V, Law C, Whiteway M, Hallett MT. A Deep Learning Approach to Capture the Essence of Candida albicans Morphologies. Microbiol Spectr 2022; 10:e0147222. [PMID: 35972285 PMCID: PMC9604015 DOI: 10.1128/spectrum.01472-22] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 07/25/2022] [Indexed: 12/31/2022] Open
Abstract
We present deep learning-based approaches for exploring the complex array of morphologies exhibited by the opportunistic human pathogen Candida albicans. Our system, entitled Candescence, automatically detects C. albicans cells from differential image contrast microscopy and labels each detected cell with one of nine morphologies. This ranges from yeast white and opaque forms to hyphal and pseudohyphal filamentous morphologies. The software is based upon a fully convolutional one-stage (FCOS) object detector, a deep learning technique that uses an extensive set of images that we manually annotated with the location and morphology of each cell. We developed a novel cumulative curriculum-based learning strategy that stratifies our images by difficulty from simple yeast forms to complex filamentous architectures. Candescence achieves very good performance (~85% recall; 81% precision) on this difficult learning set, where some images contain hundreds of cells with substantial intermixing between the predicted classes. To capture the essence of each C. albicans morphology and how they intermix, we used a second technique from deep learning entitled generative adversarial networks. The resultant models allow us to identify and explore technical variables, developmental trajectories, and morphological switches. Importantly, the model allows us to quantitatively capture morphological plasticity observed with genetically modified strains or strains grown in different media and environments. We envision Candescence as a community meeting point for quantitative explorations of C. albicans morphology. IMPORTANCE The fungus Candida albicans can "shape shift" between 12 morphologies in response to environmental variables. The cytoprotective capacity provided by this polymorphism makes C. albicans a formidable pathogen to treat clinically. Microscopy images of C. albicans colonies can contain hundreds of cells in different morphological states. Manual annotation of images can be difficult, especially as a result of densely packed and filamentous colonies and of technical artifacts from the microscopy itself. Manual annotation is inherently subjective, depending on the experience and opinion of annotators. Here, we built a deep learning approach entitled Candescence to parse images in an automated, quantitative, and objective fashion: each cell in an image is located and labeled with its morphology. Candescence effectively replaces simple rules based on visual phenotypes (size, shape, and shading) with neural circuitry capable of capturing subtle but salient features in images that may be too complex for human annotators.
Collapse
Affiliation(s)
- Van Bettauer
- Department of Computer Science and Software Engineering, Concordia University, Montreal, Quebec, Canada
| | | | | | - Samira Massahi
- Department of Biology, Concordia University, Montreal, Quebec, Canada
| | | | - Shawn Simpson
- Department of Computer Science and Software Engineering, Concordia University, Montreal, Quebec, Canada
| | - Vanessa Dumeaux
- Department of Anatomy and Cell Biology, Western University, London, Ontario, Canada
| | - Chris Law
- Centre for Microscopy and Cellular Imaging, Concordia University, Montreal, Quebec, Canada
| | - Malcolm Whiteway
- Department of Biology, Concordia University, Montreal, Quebec, Canada
| | - Michael T. Hallett
- Department of Biology, Concordia University, Montreal, Quebec, Canada
- Department of Biochemistry, Western University, London, Ontario, Canada
| |
Collapse
|
30
|
Wong KS, Zhong X, Low CSL, Kanchanawong P. Self-supervised classification of subcellular morphometric phenotypes reveals extracellular matrix-specific morphological responses. Sci Rep 2022; 12:15329. [PMID: 36097150 PMCID: PMC9468179 DOI: 10.1038/s41598-022-19472-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 08/30/2022] [Indexed: 11/17/2022] Open
Abstract
Cell morphology is profoundly influenced by cellular interactions with microenvironmental factors such as the extracellular matrix (ECM). Upon adhesion to specific ECM, various cell types are known to exhibit different but distinctive morphologies, suggesting that ECM-dependent cell morphological responses may harbour rich information on cellular signalling states. However, the inherent morphological complexity of cellular and subcellular structures has posed an ongoing challenge for automated quantitative analysis. Since multi-channel fluorescence microscopy provides robust molecular specificity important for the biological interpretations of observed cellular architecture, here we develop a deep learning-based analysis pipeline for the classification of cell morphometric phenotypes from multi-channel fluorescence micrographs, termed SE-RNN (residual neural network with squeeze-and-excite blocks). We demonstrate SERNN-based classification of distinct morphological signatures observed when fibroblasts or epithelial cells are presented with different ECM. Our results underscore how cell shapes are non-random and established the framework for classifying cell shapes into distinct morphological signature in a cell-type and ECM-specific manner.
Collapse
Affiliation(s)
- Kin Sun Wong
- Department of Biomedical Engineering, National University of Singapore, Singapore, 117411, Republic of Singapore
| | - Xueying Zhong
- Mechanobiology Institute, National University of Singapore, Singapore, 117411, Republic of Singapore
| | - Christine Siok Lan Low
- Mechanobiology Institute, National University of Singapore, Singapore, 117411, Republic of Singapore
| | - Pakorn Kanchanawong
- Department of Biomedical Engineering, National University of Singapore, Singapore, 117411, Republic of Singapore. .,Mechanobiology Institute, National University of Singapore, Singapore, 117411, Republic of Singapore.
| |
Collapse
|
31
|
Chand S. Semantic segmentation of human cell nucleus using deep U-Net and other versions of U-Net models. NETWORK (BRISTOL, ENGLAND) 2022; 33:167-186. [PMID: 35822269 DOI: 10.1080/0954898x.2022.2096938] [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: 11/06/2021] [Revised: 04/04/2022] [Accepted: 06/27/2022] [Indexed: 06/15/2023]
Abstract
The deep learning models play an essential role in many areas, including medical image analysis. These models extract important features without human intervention. In this paper, we propose a deep convolution neural network, named as deep U-Net model, for the segmentation of the cell nucleus, a critical functional unit that determines the function and structure of the body. The nucleus contains all kinds of DNA, RNA, chromosomes, and genes governing all life activities, and its disorder may lead to different types of diseases such as cancer, heart disease, diabetes, Alzheimer's, etc. If the nucleus structure is known correctly, diseases due to nucleus disorder may be detected early. It may also reduce the drug discovery time if the shape and size of the nucleus are known. We evaluate the performance of the proposed models on the nucleus segmentation data set used by the Data Science Bowl 2018 competition hosted by Kaggle. We compare its performance with that of the U-Net, Attention U-Net, R2U-Net, Attention R2U-Net, and both versions of the U-Net++ with and without supervision, in terms of loss, dice coefficient, dice loss, intersection over union, and accuracy. Our model performs better than the existing models.
Collapse
Affiliation(s)
- Satish Chand
- School of Computer and Systems Sciences, Jawaharlal Nehru Univesity, New Delhi, India
| |
Collapse
|
32
|
Xu M, Cui Q, Su W, Zhang D, Pan J, Liu X, Pang Z, Zhu Q. High-content screening of active components of Traditional Chinese Medicine inhibiting TGF-β-induced cell EMT. Heliyon 2022; 8:e10238. [PMID: 36042745 PMCID: PMC9420491 DOI: 10.1016/j.heliyon.2022.e10238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 05/13/2022] [Accepted: 08/05/2022] [Indexed: 11/30/2022] Open
Abstract
The epithelial mesenchymal transition (EMT) has roles in metastasis and invasion during fibrotic diseases and cancer progression. Some Traditional Chinese Medicines (TCMs) have shown inhibitory effects with respect to the EMT. The current study attempted to establish a multiparametric high-content method to screen for active monomeric compounds in TCM with the ability to target cellular EMT by assessing phenotypic changes. A total of 306 monomeric compounds from the MedChemExpress (MCE) compound library were screened by the high-content screening (HCS) system and 5 compounds with anti-EMT activity, including camptothecin (CPT), dimethyl curcumin (DMC), artesunate (ART), sinapine (SNP) and berberine (BER) were identified. To confirm anti-EMT activity, expression of EMT markers was assessed by qRT-PCR and Western blotting, and cell adhesion and migration measured by cell function assays. The results revealed that CPT, DMC, ART, SNP and BER inhibited transforming growth factor-β1 (TGF-β1)-induced expression of vimentin and α-SMA, upregulated expression of E-cadherin, increased cell adhesion and reduced cell migration. In summary, by quantifying the cell morphological changes during TGF-β1-induced EMT through multi-parametric analysis, TCM compounds with anti-EMT activity were successfully screened using the HCS system, a faster and more economical approach than conventional methods.
Collapse
Affiliation(s)
- Mengzhen Xu
- College of Pharmaceutical Science, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Qinghua Cui
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Wen Su
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Dan Zhang
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Jiaxu Pan
- College of Pharmaceutical Science, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Xiangqi Liu
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Zheng Pang
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Qingjun Zhu
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China.,Key Laboratory of Traditional Chinese Medicine Classical Theory, Ministry of Education, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| |
Collapse
|
33
|
Li Y, Sheng H, Yan Z, Guan B, Qiang S, Qian J, Wang Y. Bilirubin stabilizes the mitochondrial membranes during NLRP3 inflammasome activation. Biochem Pharmacol 2022; 203:115204. [DOI: 10.1016/j.bcp.2022.115204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 08/01/2022] [Accepted: 08/02/2022] [Indexed: 11/28/2022]
|
34
|
High-content imaging of human hepatic spheroids for researching the mechanism of duloxetine-induced hepatotoxicity. Cell Death Dis 2022; 13:669. [PMID: 35915074 PMCID: PMC9343405 DOI: 10.1038/s41419-022-05042-x] [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: 02/15/2022] [Revised: 06/12/2022] [Accepted: 06/27/2022] [Indexed: 01/21/2023]
Abstract
Duloxetine (DLX) has been approved for the successful treatment of psychiatric diseases, including major depressive disorder, diabetic neuropathy, fibromyalgia and generalized anxiety disorder. However, since the usage of DLX carries a manufacturer warning of hepatotoxicity given its implication in numerous cases of drug-induced liver injuries (DILI), it is not recommended for patients with chronic liver diseases. In our previous study, we developed an enhanced human-simulated hepatic spheroid (EHS) imaging model system for performing drug hepatotoxicity evaluation using the human hepatoma cell line HepaRG and the support of a pulverized liver biomatrix scaffold, which demonstrated much improved hepatic-specific functions. In the current study, we were able to use this robust model to demonstrate that the DLX-DILI is a human CYP450 specific, metabolism-dependent, oxidative stress triggered complex hepatic injury. High-content imaging analysis (HCA) of organoids exposed to DLX showed that the potential toxicophore, naphthyl ring in DLX initiated oxidative stress which ultimately led to mitochondrial dysfunction in the hepatic organoids, and vice versa. Furthermore, DLX-induced hepatic steatosis and cholestasis was also detected in the exposed EHSs. We also discovered that a novel compound S-071031B, which replaced DLX's naphthyl ring with benzodioxole, showed dramatically lower hepatotoxicities through reducing oxidative stress. Thus, we conclusively present the human-relevant EHS model as an ideal, highly competent system for evaluating DLX induced hepatotoxicity and exploring related mechanisms in vitro. Moreover, HCA use on functional hepatic organoids has promising application prospects for guiding compound structural modifications and optimization in order to improve drug development by reducing hepatotoxicity.
Collapse
|
35
|
Moreno-Andrés D, Bhattacharyya A, Scheufen A, Stegmaier J. LiveCellMiner: A new tool to analyze mitotic progression. PLoS One 2022; 17:e0270923. [PMID: 35797385 PMCID: PMC9262191 DOI: 10.1371/journal.pone.0270923] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 06/17/2022] [Indexed: 11/21/2022] Open
Abstract
Live-cell imaging has become state of the art to accurately identify the nature of mitotic and cell cycle defects. Low- and high-throughput microscopy setups have yield huge data amounts of cells recorded in different experimental and pathological conditions. Tailored semi-automated and automated image analysis approaches allow the analysis of high-content screening data sets, saving time and avoiding bias. However, they were mostly designed for very specific experimental setups, which restricts their flexibility and usability. The general need for dedicated experiment-specific user-annotated training sets and experiment-specific user-defined segmentation parameters remains a major bottleneck for fully automating the analysis process. In this work we present LiveCellMiner, a highly flexible open-source software tool to automatically extract, analyze and visualize both aggregated and time-resolved image features with potential biological relevance. The software tool allows analysis across high-content data sets obtained in different platforms, in a quantitative and unbiased manner. As proof of principle application, we analyze here the dynamic chromatin and tubulin cytoskeleton features in human cells passing through mitosis highlighting the versatile and flexible potential of this tool set.
Collapse
Affiliation(s)
- Daniel Moreno-Andrés
- Institute of Biochemistry and Molecular Cell Biology, Medical School, RWTH Aachen University, Aachen, Germany
- * E-mail: (DMA), (JS)
| | - Anuk Bhattacharyya
- Institute of Imaging and Computer Vision, RWTH Aachen University, Aachen, Germany
| | - Anja Scheufen
- Institute of Biochemistry and Molecular Cell Biology, Medical School, RWTH Aachen University, Aachen, Germany
| | - Johannes Stegmaier
- Institute of Imaging and Computer Vision, RWTH Aachen University, Aachen, Germany
- * E-mail: (DMA), (JS)
| |
Collapse
|
36
|
Seo Y, Bang S, Son J, Kim D, Jeong Y, Kim P, Yang J, Eom JH, Choi N, Kim HN. Brain physiome: A concept bridging in vitro 3D brain models and in silico models for predicting drug toxicity in the brain. Bioact Mater 2022; 13:135-148. [PMID: 35224297 PMCID: PMC8843968 DOI: 10.1016/j.bioactmat.2021.11.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 11/01/2021] [Accepted: 11/06/2021] [Indexed: 12/12/2022] Open
Abstract
In the last few decades, adverse reactions to pharmaceuticals have been evaluated using 2D in vitro models and animal models. However, with increasing computational power, and as the key drivers of cellular behavior have been identified, in silico models have emerged. These models are time-efficient and cost-effective, but the prediction of adverse reactions to unknown drugs using these models requires relevant experimental input. Accordingly, the physiome concept has emerged to bridge experimental datasets with in silico models. The brain physiome describes the systemic interactions of its components, which are organized into a multilevel hierarchy. Because of the limitations in obtaining experimental data corresponding to each physiome component from 2D in vitro models and animal models, 3D in vitro brain models, including brain organoids and brain-on-a-chip, have been developed. In this review, we present the concept of the brain physiome and its hierarchical organization, including cell- and tissue-level organizations. We also summarize recently developed 3D in vitro brain models and link them with the elements of the brain physiome as a guideline for dataset collection. The connection between in vitro 3D brain models and in silico modeling will lead to the establishment of cost-effective and time-efficient in silico models for the prediction of the safety of unknown drugs.
Collapse
Affiliation(s)
- Yoojin Seo
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Seokyoung Bang
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Jeongtae Son
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Dongsup Kim
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Yong Jeong
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Pilnam Kim
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Jihun Yang
- Next&Bio Inc., Seoul, 02841, Republic of Korea
| | - Joon-Ho Eom
- Medical Device Research Division, National Institute of Food and Drug Safety Evaluation, Cheongju, 28159, Republic of Korea
| | - Nakwon Choi
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
- Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology (UST), Seoul, 02792, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, Republic of Korea
| | - Hong Nam Kim
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
- Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology (UST), Seoul, 02792, Republic of Korea
- School of Mechanical Engineering, Yonsei University, Seoul, 03722, Republic of Korea
- Yonsei-KIST Convergence Research Institute, Yonsei University, Seoul, 03722, Republic of Korea
| |
Collapse
|
37
|
Bajusz D, Keserű GM. Maximizing the integration of virtual and experimental screening in hit discovery. Expert Opin Drug Discov 2022; 17:629-640. [PMID: 35671403 DOI: 10.1080/17460441.2022.2085685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Experimental and virtual screening contributes to the discovery of more than 50% of clinical candidates. Considering the similar concept and goals, early-phase drug discovery would benefit from the effective integration of these approaches. AREAS COVERED After reviewing the recent trends in both experimental and virtual screening, the authors discuss different integration strategies from parallel, focused, sequential, and iterative screening. Strategic considerations are demonstrated in a number of real-life case studies. EXPERT OPINION Experimental and virtual screening are complementary approaches that should be integrated in lead discovery settings. Virtual screening can access extremely large synthetically feasible chemical space that can be effectively searched on GPU clusters or cloud architectures. Experimental screening provides reliable datasets by quantitative HTS applications, and DNA-encoded libraries (DEL) have enlarged the chemical space covered by these technologies. These developments, together with the use of artificial intelligence methods, represent new options for their efficient integration. The case studies discussed here demonstrate the benefits of complementary strategies, such as focused and iterative screening.
Collapse
Affiliation(s)
- Dávid Bajusz
- Medicinal Chemistry Research Group, Research Centre for Natural Sciences, Budapest, Hungary
| | - György M Keserű
- Medicinal Chemistry Research Group, Research Centre for Natural Sciences, Budapest, Hungary
| |
Collapse
|
38
|
Dawson J, Pandey S, Yu Q, Schaub P, Wüst F, Moradi AB, Dovzhenko O, Palme K, Welsch R. Determination of protoplast growth properties using quantitative single-cell tracking analysis. PLANT METHODS 2022; 18:64. [PMID: 35585602 PMCID: PMC9118701 DOI: 10.1186/s13007-022-00895-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 05/01/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND Although quantitative single-cell analysis is frequently applied in animal systems, e.g. to identify novel drugs, similar applications on plant single cells are largely missing. We have exploited the applicability of high-throughput microscopic image analysis on plant single cells using tobacco leaf protoplasts, cell-wall free single cells isolated by lytic digestion. Protoplasts regenerate their cell wall within several days after isolation and have the potential to expand and proliferate, generating microcalli and finally whole plants after the application of suitable regeneration conditions. RESULTS High-throughput automated microscopy coupled with the development of image processing pipelines allowed to quantify various developmental properties of thousands of protoplasts during the initial days following cultivation by immobilization in multi-well-plates. The focus on early protoplast responses allowed to study cell expansion prior to the initiation of proliferation and without the effects of shape-compromising cell walls. We compared growth parameters of wild-type tobacco cells with cells expressing the antiapoptotic protein Bcl2-associated athanogene 4 from Arabidopsis (AtBAG4). CONCLUSIONS AtBAG4-expressing protoplasts showed a higher proportion of cells responding with positive area increases than the wild type and showed increased growth rates as well as increased proliferation rates upon continued cultivation. These features are associated with reported observations on a BAG4-mediated increased resilience to various stress responses and improved cellular survival rates following transformation approaches. Moreover, our single-cell expansion results suggest a BAG4-mediated, cell-independent increase of potassium channel abundance which was hitherto reported for guard cells only. The possibility to explain plant phenotypes with single-cell properties, extracted with the single-cell processing and analysis pipeline developed, allows to envision novel biotechnological screening strategies able to determine improved plant properties via single-cell analysis.
Collapse
Affiliation(s)
- Jonathan Dawson
- Institute of Biology II, Faculty of Biology, Albert-Ludwigs-University of Freiburg, Schänzlestrasse 1, 79104, Freiburg, Germany
- Institute of General Electrical Engineering, University of Rostock, Albert-Einstein-Str. 2, 18059, Rostock, Germany
- Augusta University, 1201 Goss Ln, Augusta, GA, 30912, USA
| | - Saurabh Pandey
- Institute of Biology II, Faculty of Biology, Albert-Ludwigs-University of Freiburg, Schänzlestrasse 1, 79104, Freiburg, Germany
| | - Qiuju Yu
- Institute of Biology II, Faculty of Biology, Albert-Ludwigs-University of Freiburg, Schänzlestrasse 1, 79104, Freiburg, Germany
- ScreenSYS GmbH, Engesserstr. 4, 79108, Freiburg, Germany
| | - Patrick Schaub
- Institute of Biology II, Faculty of Biology, Albert-Ludwigs-University of Freiburg, Schänzlestrasse 1, 79104, Freiburg, Germany
- ScreenSYS GmbH, Engesserstr. 4, 79108, Freiburg, Germany
| | - Florian Wüst
- Institute of Biology II, Faculty of Biology, Albert-Ludwigs-University of Freiburg, Schänzlestrasse 1, 79104, Freiburg, Germany
- ScreenSYS GmbH, Engesserstr. 4, 79108, Freiburg, Germany
| | - Amir Bahram Moradi
- Institute of Biology II, Faculty of Biology, Albert-Ludwigs-University of Freiburg, Schänzlestrasse 1, 79104, Freiburg, Germany
| | - Oleksandr Dovzhenko
- Institute of Biology II, Faculty of Biology, Albert-Ludwigs-University of Freiburg, Schänzlestrasse 1, 79104, Freiburg, Germany
- ScreenSYS GmbH, Engesserstr. 4, 79108, Freiburg, Germany
| | - Klaus Palme
- Institute of Biology II, Faculty of Biology, Albert-Ludwigs-University of Freiburg, Schänzlestrasse 1, 79104, Freiburg, Germany
- ScreenSYS GmbH, Engesserstr. 4, 79108, Freiburg, Germany
- BIOSS Center for Biological Signaling Studies, Albert-Ludwigs-University of Freiburg, Schänzlestrasse 1, 79104, Freiburg, Germany
- State Key Laboratory of Crop Biology, College of Life Sciences, Shandong Agricultural University, Daizong Street 61, Tai'an, 271018, China
| | - Ralf Welsch
- Institute of Biology II, Faculty of Biology, Albert-Ludwigs-University of Freiburg, Schänzlestrasse 1, 79104, Freiburg, Germany.
- ScreenSYS GmbH, Engesserstr. 4, 79108, Freiburg, Germany.
| |
Collapse
|
39
|
Liu W, Liu C, Ren PG, Chu J, Wang L. An Improved Genetically Encoded Fluorescent cAMP Indicator for Sensitive cAMP Imaging and Fast Drug Screening. Front Pharmacol 2022; 13:902290. [PMID: 35694242 PMCID: PMC9175130 DOI: 10.3389/fphar.2022.902290] [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: 03/23/2022] [Accepted: 04/18/2022] [Indexed: 11/24/2022] Open
Abstract
Cyclic adenosine 3',5'-monophosphate (cAMP) is an important intracellular second messenger molecule downstream of many G protein-coupled receptors (GPCRs). Fluorescence imaging with bright and sensitive cAMP indicators allows not only dissecting the spatiotemporal dynamics of intracellular cAMP, but also high-content screening of compounds against GPCRs. We previously reported the high-performance circularly permuted GFP (cpGFP)-based cAMP indicator G-Flamp1. Here, we developed improved G-Flamp1 variants G-Flamp2 and G-Flamp2b. Compared to G-Flamp1, G-Flamp2 exhibited increased baseline fluorescence (1.6-fold) and larger fluorescence change (ΔF/F0) (1,300% vs. 1,100%) in HEK293T cells, while G-Flamp2b showed increased baseline fluorescence (3.1-fold) and smaller ΔF/F0 (400% vs. 1,100%). Furthermore, live cell imaging of mitochondrial matrix-targeted G-Flamp2 confirmed cytosolic cAMP was able to enter the mitochondrial matrix. G-Flamp2 imaging also showed that adipose tissue extract activated the Gi protein-coupled orphan GPCR GPR50 in HEK293T cells. Taken together, our results showed that the high-performance of G-Flamp2 would facilitate sensitive intracellular cAMP imaging and activity measurement of compounds targeting GPCR-cAMP signaling pathway during early drug development.
Collapse
Affiliation(s)
- Wenfeng Liu
- Guangdong Provincial Key Laboratory of Biomedical Optical Imaging Technology & Center for Biomedical Optics and Molecular Imaging & CAS Key Laboratory of Health Informatics, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Chang Liu
- Center for Energy Metabolism and Reproduction, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- Shenzhen College of Advanced Technology, University of Chinese Academy of Sciences, Shenzhen, China
| | - Pei-Gen Ren
- Center for Energy Metabolism and Reproduction, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- Shenzhen College of Advanced Technology, University of Chinese Academy of Sciences, Shenzhen, China
| | - Jun Chu
- Guangdong Provincial Key Laboratory of Biomedical Optical Imaging Technology & Center for Biomedical Optics and Molecular Imaging & CAS Key Laboratory of Health Informatics, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen, China
| | - Liang Wang
- Guangdong Provincial Key Laboratory of Biomedical Optical Imaging Technology & Center for Biomedical Optics and Molecular Imaging & CAS Key Laboratory of Health Informatics, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| |
Collapse
|
40
|
Valerio HP, Ravagnani FG, Yaya Candela AP, Dias Carvalho da Costa B, Ronsein GE, Di Mascio P. Spatial proteomics reveals subcellular reorganization in human keratinocytes exposed to UVA light. iScience 2022; 25:104093. [PMID: 35372811 PMCID: PMC8971936 DOI: 10.1016/j.isci.2022.104093] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 11/16/2021] [Accepted: 03/14/2022] [Indexed: 12/16/2022] Open
Abstract
The effects of UV light on the skin have been extensively investigated. However, systematic information about how the exposure to ultraviolet-A (UVA) light, the least energetic but the most abundant UV radiation reaching the Earth, shapes the subcellular organization of proteins is lacking. Using subcellular fractionation, mass-spectrometry-based proteomics, machine learning algorithms, immunofluorescence, and functional assays, we mapped the subcellular reorganization of the proteome of human keratinocytes in response to UVA light. Our workflow quantified and assigned subcellular localization for over 1,600 proteins, of which about 200 were found to redistribute upon UVA exposure. Reorganization of the proteome affected modulators of signaling pathways, cellular metabolism, and DNA damage response. Strikingly, mitochondria were identified as one of the main targets of UVA-induced stress. Further investigation demonstrated that UVA induces mitochondrial fragmentation, up-regulates redox-responsive proteins, and attenuates respiratory rates. These observations emphasize the role of this radiation as a potent metabolic stressor in the skin.
Collapse
Affiliation(s)
- Hellen Paula Valerio
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo 05508-000, Brazil
| | - Felipe Gustavo Ravagnani
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo 05508-000, Brazil
| | - Angela Paola Yaya Candela
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo 05508-000, Brazil
| | | | - Graziella Eliza Ronsein
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo 05508-000, Brazil
| | - Paolo Di Mascio
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo 05508-000, Brazil
| |
Collapse
|
41
|
Super-resolution imaging reveals the subcellular distribution of dextran at the nanoscale in living cells. CHINESE CHEM LETT 2022. [DOI: 10.1016/j.cclet.2021.10.025] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
42
|
Ghanegolmohammadi F, Ohnuki S, Ohya Y. Assignment of unimodal probability distribution models for quantitative morphological phenotyping. BMC Biol 2022; 20:81. [PMID: 35361198 PMCID: PMC8969357 DOI: 10.1186/s12915-022-01283-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Accepted: 03/17/2022] [Indexed: 01/02/2023] Open
Abstract
Background Cell morphology is a complex and integrative readout, and therefore, an attractive measurement for assessing the effects of genetic and chemical perturbations to cells. Microscopic images provide rich information on cell morphology; therefore, subjective morphological features are frequently extracted from digital images. However, measured datasets are fundamentally noisy; thus, estimation of the true values is an ultimate goal in quantitative morphological phenotyping. Ideal image analyses require precision, such as proper probability distribution analyses to detect subtle morphological changes, recall to minimize artifacts due to experimental error, and reproducibility to confirm the results. Results Here, we present UNIMO (UNImodal MOrphological data), a reliable pipeline for precise detection of subtle morphological changes by assigning unimodal probability distributions to morphological features of the budding yeast cells. By defining the data type, followed by validation using the model selection method, examination of 33 probability distributions revealed nine best-fitting probability distributions. The modality of the distribution was then clarified for each morphological feature using a probabilistic mixture model. Using a reliable and detailed set of experimental log data of wild-type morphological replicates, we considered the effects of confounding factors. As a result, most of the yeast morphological parameters exhibited unimodal distributions that can be used as basic tools for powerful downstream parametric analyses. The power of the proposed pipeline was confirmed by reanalyzing morphological changes in non-essential yeast mutants and detecting 1284 more mutants with morphological defects compared with a conventional approach (Box–Cox transformation). Furthermore, the combined use of canonical correlation analysis permitted global views on the cellular network as well as new insights into possible gene functions. Conclusions Based on statistical principles, we showed that UNIMO offers better predictions of the true values of morphological measurements. We also demonstrated how these concepts can provide biologically important information. This study draws attention to the necessity of employing a proper approach to do more with less. Supplementary Information The online version contains supplementary material available at 10.1186/s12915-022-01283-6.
Collapse
Affiliation(s)
- Farzan Ghanegolmohammadi
- Department of Integrated Biosciences, Graduate School of Frontier Sciences, The University of Tokyo, Bldg. FSB-101, 5-1-5 Kashiwanoha, Kashiwa, Chiba Prefecture, 277-8562, Japan.,Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Shinsuke Ohnuki
- Department of Integrated Biosciences, Graduate School of Frontier Sciences, The University of Tokyo, Bldg. FSB-101, 5-1-5 Kashiwanoha, Kashiwa, Chiba Prefecture, 277-8562, Japan
| | - Yoshikazu Ohya
- Department of Integrated Biosciences, Graduate School of Frontier Sciences, The University of Tokyo, Bldg. FSB-101, 5-1-5 Kashiwanoha, Kashiwa, Chiba Prefecture, 277-8562, Japan. .,Collaborative Research Institute for Innovative Microbiology, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo, 113-8657, Japan.
| |
Collapse
|
43
|
Christopher JA, Geladaki A, Dawson CS, Vennard OL, Lilley KS. Subcellular Transcriptomics and Proteomics: A Comparative Methods Review. Mol Cell Proteomics 2022; 21:100186. [PMID: 34922010 PMCID: PMC8864473 DOI: 10.1016/j.mcpro.2021.100186] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 11/16/2021] [Accepted: 12/13/2021] [Indexed: 12/23/2022] Open
Abstract
The internal environment of cells is molecularly crowded, which requires spatial organization via subcellular compartmentalization. These compartments harbor specific conditions for molecules to perform their biological functions, such as coordination of the cell cycle, cell survival, and growth. This compartmentalization is also not static, with molecules trafficking between these subcellular neighborhoods to carry out their functions. For example, some biomolecules are multifunctional, requiring an environment with differing conditions or interacting partners, and others traffic to export such molecules. Aberrant localization of proteins or RNA species has been linked to many pathological conditions, such as neurological, cancer, and pulmonary diseases. Differential expression studies in transcriptomics and proteomics are relatively common, but the majority have overlooked the importance of subcellular information. In addition, subcellular transcriptomics and proteomics data do not always colocate because of the biochemical processes that occur during and after translation, highlighting the complementary nature of these fields. In this review, we discuss and directly compare the current methods in spatial proteomics and transcriptomics, which include sequencing- and imaging-based strategies, to give the reader an overview of the current tools available. We also discuss current limitations of these strategies as well as future developments in the field of spatial -omics.
Collapse
Affiliation(s)
- Josie A Christopher
- Department of Biochemistry, Cambridge Centre for Proteomics, University of Cambridge, Cambridge, UK; Milner Therapeutics Institute, Jeffrey Cheah Biomedical Centre, Cambridge, UK
| | - Aikaterini Geladaki
- Department of Biochemistry, Cambridge Centre for Proteomics, University of Cambridge, Cambridge, UK; Department of Genetics, University of Cambridge, Cambridge, UK
| | - Charlotte S Dawson
- Department of Biochemistry, Cambridge Centre for Proteomics, University of Cambridge, Cambridge, UK; Milner Therapeutics Institute, Jeffrey Cheah Biomedical Centre, Cambridge, UK
| | - Owen L Vennard
- Department of Biochemistry, Cambridge Centre for Proteomics, University of Cambridge, Cambridge, UK; Milner Therapeutics Institute, Jeffrey Cheah Biomedical Centre, Cambridge, UK
| | - Kathryn S Lilley
- Department of Biochemistry, Cambridge Centre for Proteomics, University of Cambridge, Cambridge, UK; Milner Therapeutics Institute, Jeffrey Cheah Biomedical Centre, Cambridge, UK.
| |
Collapse
|
44
|
Yang JW, Lin YR, Chu YL, Chung JHY, Lu HE, Chen GY. Tissue-level alveolar epithelium model for recapitulating SARS-CoV-2 infection and cellular plasticity. Commun Biol 2022; 5:70. [PMID: 35046486 PMCID: PMC8770515 DOI: 10.1038/s42003-022-03026-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 12/27/2021] [Indexed: 12/11/2022] Open
Abstract
AbstractPulmonary sequelae following COVID-19 pneumonia have been emerging as a challenge; however, suitable cell sources for studying COVID-19 mechanisms and therapeutics are currently lacking. In this paper, we present a standardized primary alveolar cell culture method for establishing a human alveolar epithelium model that can recapitulate viral infection and cellular plasticity. The alveolar model is infected with a SARS-CoV-2 pseudovirus, and the clinically relevant features of the viral entry into the alveolar type-I/II cells, cytokine production activation, and pulmonary surfactant destruction are reproduced. For this damaged alveolar model, we find that the inhibition of Wnt signaling via XAV939 substantially improves alveolar repair function and prevents subsequent pulmonary fibrosis. Thus, the proposed alveolar cell culture strategy exhibits potential for the identification of pathogenesis and therapeutics in basic and translational research.
Collapse
|
45
|
Watson ER, Taherian Fard A, Mar JC. Computational Methods for Single-Cell Imaging and Omics Data Integration. Front Mol Biosci 2022; 8:768106. [PMID: 35111809 PMCID: PMC8801747 DOI: 10.3389/fmolb.2021.768106] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 11/29/2021] [Indexed: 12/12/2022] Open
Abstract
Integrating single cell omics and single cell imaging allows for a more effective characterisation of the underlying mechanisms that drive a phenotype at the tissue level, creating a comprehensive profile at the cellular level. Although the use of imaging data is well established in biomedical research, its primary application has been to observe phenotypes at the tissue or organ level, often using medical imaging techniques such as MRI, CT, and PET. These imaging technologies complement omics-based data in biomedical research because they are helpful for identifying associations between genotype and phenotype, along with functional changes occurring at the tissue level. Single cell imaging can act as an intermediary between these levels. Meanwhile new technologies continue to arrive that can be used to interrogate the genome of single cells and its related omics datasets. As these two areas, single cell imaging and single cell omics, each advance independently with the development of novel techniques, the opportunity to integrate these data types becomes more and more attractive. This review outlines some of the technologies and methods currently available for generating, processing, and analysing single-cell omics- and imaging data, and how they could be integrated to further our understanding of complex biological phenomena like ageing. We include an emphasis on machine learning algorithms because of their ability to identify complex patterns in large multidimensional data.
Collapse
Affiliation(s)
| | - Atefeh Taherian Fard
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, Australia
| | - Jessica Cara Mar
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
46
|
Wang A, Zhang Q, Han Y, Megason S, Hormoz S, Mosaliganti KR, Lam JCK, Li VOK. A novel deep learning-based 3D cell segmentation framework for future image-based disease detection. Sci Rep 2022; 12:342. [PMID: 35013443 PMCID: PMC8748745 DOI: 10.1038/s41598-021-04048-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Accepted: 12/09/2021] [Indexed: 11/12/2022] Open
Abstract
Cell segmentation plays a crucial role in understanding, diagnosing, and treating diseases. Despite the recent success of deep learning-based cell segmentation methods, it remains challenging to accurately segment densely packed cells in 3D cell membrane images. Existing approaches also require fine-tuning multiple manually selected hyperparameters on the new datasets. We develop a deep learning-based 3D cell segmentation pipeline, 3DCellSeg, to address these challenges. Compared to the existing methods, our approach carries the following novelties: (1) a robust two-stage pipeline, requiring only one hyperparameter; (2) a light-weight deep convolutional neural network (3DCellSegNet) to efficiently output voxel-wise masks; (3) a custom loss function (3DCellSeg Loss) to tackle the clumped cell problem; and (4) an efficient touching area-based clustering algorithm (TASCAN) to separate 3D cells from the foreground masks. Cell segmentation experiments conducted on four different cell datasets show that 3DCellSeg outperforms the baseline models on the ATAS (plant), HMS (animal), and LRP (plant) datasets with an overall accuracy of 95.6%, 76.4%, and 74.7%, respectively, while achieving an accuracy comparable to the baselines on the Ovules (plant) dataset with an overall accuracy of 82.2%. Ablation studies show that the individual improvements in accuracy is attributable to 3DCellSegNet, 3DCellSeg Loss, and TASCAN, with the 3DCellSeg demonstrating robustness across different datasets and cell shapes. Our results suggest that 3DCellSeg can serve a powerful biomedical and clinical tool, such as histo-pathological image analysis, for cancer diagnosis and grading.
Collapse
Affiliation(s)
- Andong Wang
- Department of Electrical and Electronic Engineering, The University of Hong Kong, Hong Kong, China
| | - Qi Zhang
- Department of Electrical and Electronic Engineering, The University of Hong Kong, Hong Kong, China
| | - Yang Han
- Department of Electrical and Electronic Engineering, The University of Hong Kong, Hong Kong, China
| | - Sean Megason
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA
| | - Sahand Hormoz
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA
| | | | - Jacqueline C K Lam
- Department of Electrical and Electronic Engineering, The University of Hong Kong, Hong Kong, China.
| | - Victor O K Li
- Department of Electrical and Electronic Engineering, The University of Hong Kong, Hong Kong, China.
| |
Collapse
|
47
|
Bagheri N, Carpenter AE, Lundberg E, Plant AL, Horwitz R. The new era of quantitative cell imaging—challenges and opportunities. Mol Cell 2022; 82:241-247. [DOI: 10.1016/j.molcel.2021.12.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 12/16/2021] [Accepted: 12/17/2021] [Indexed: 11/24/2022]
|
48
|
From imaging a single cell to implementing precision medicine: an exciting new era. Emerg Top Life Sci 2021; 5:837-847. [PMID: 34889448 PMCID: PMC8786301 DOI: 10.1042/etls20210219] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 11/24/2021] [Accepted: 11/24/2021] [Indexed: 11/17/2022]
Abstract
In the age of high-throughput, single-cell biology, single-cell imaging has evolved not only in terms of technological advancements but also in its translational applications. The synchronous advancements of imaging and computational biology have produced opportunities of merging the two, providing the scientific community with tools towards observing, understanding, and predicting cellular and tissue phenotypes and behaviors. Furthermore, multiplexed single-cell imaging and machine learning algorithms now enable patient stratification and predictive diagnostics of clinical specimens. Here, we provide an overall summary of the advances in single-cell imaging, with a focus on high-throughput microscopy phenomics and multiplexed proteomic spatial imaging platforms. We also review various computational tools that have been developed in recent years for image processing and downstream applications used in biomedical sciences. Finally, we discuss how harnessing systems biology approaches and data integration across disciplines can further strengthen the exciting applications and future implementation of single-cell imaging on precision medicine.
Collapse
|
49
|
Liu L, Bi M, Wang Y, Liu J, Jiang X, Xu Z, Zhang X. Artificial intelligence-powered microfluidics for nanomedicine and materials synthesis. NANOSCALE 2021; 13:19352-19366. [PMID: 34812823 DOI: 10.1039/d1nr06195j] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
Artificial intelligence (AI) is an emerging technology with great potential, and its robust calculation and analysis capabilities are unmatched by traditional calculation tools. With the promotion of deep learning and open-source platforms, the threshold of AI has also become lower. Combining artificial intelligence with traditional fields to create new fields of high research and application value has become a trend. AI has been involved in many disciplines, such as medicine, materials, energy, and economics. The development of AI requires the support of many kinds of data, and microfluidic systems can often mine object data on a large scale to support AI. Due to the excellent synergy between the two technologies, excellent research results have emerged in many fields. In this review, we briefly review AI and microfluidics and introduce some applications of their combination, mainly in nanomedicine and material synthesis. Finally, we discuss the development trend of the combination of the two technologies.
Collapse
Affiliation(s)
- Linbo Liu
- John A. Paulson School of Engineering and Applied Science, Harvard University, Cambridge, MA 02138, USA
| | - Mingcheng Bi
- Institute of Process Equipment, College of Energy Engineering, Zhejiang University, Hangzhou 310027, P.R. China
| | - Yunhua Wang
- John A. Paulson School of Engineering and Applied Science, Harvard University, Cambridge, MA 02138, USA
| | - Junfeng Liu
- Institute of Process Equipment, College of Energy Engineering, Zhejiang University, Hangzhou 310027, P.R. China
| | - Xiwen Jiang
- College of Biological Science and Engineering, Fuzhou university, Fuzhou 350108, P.R. China
| | - Zhongbin Xu
- Institute of Process Equipment, College of Energy Engineering, Zhejiang University, Hangzhou 310027, P.R. China
| | - Xingcai Zhang
- John A. Paulson School of Engineering and Applied Science, Harvard University, Cambridge, MA 02138, USA
- School of Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| |
Collapse
|
50
|
A quantitative analysis of cell bridging kinetics on a scaffold using computer vision algorithms. Acta Biomater 2021; 136:429-440. [PMID: 34571272 DOI: 10.1016/j.actbio.2021.09.042] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 09/19/2021] [Accepted: 09/21/2021] [Indexed: 01/01/2023]
Abstract
Tissue engineering involves the seeding of cells into a structural scaffolding to regenerate the architecture of damaged or diseased tissue. To effectively design a scaffold, an understanding of how cells collectively sense and react to the geometry of their local environment is needed. Advances in the development of melt electro-writing have allowed micron and submicron polymeric fibres to be accurately printed into porous, complex and three-dimensional structures. By using melt electrowriting, we created a geometrically relevant in vitro scaffold model to study cellular spatial-temporal kinetics. These scaffolds were paired with custom computer vision algorithms to investigate cell nuclei, cell membrane actin and scaffold fibres over different pore sizes (200-600 µm) and time points (28 days). We find that cells proliferated much faster in the smaller (200 µm) pores which halved the time until confluence versus larger (500 and 600 µm) pores. Our analysis of stained actin fibres revealed that cells were highly aligned to the fibres and the leading edge of the pore filling front, and we found that cells behind the leading edge were not aligned in any particular direction. This study provides a systematic understanding of cellular spatial temporal kinetics within a 3D in vitro model to inform the design of more effective synthetic tissue engineering scaffolds for tissue regeneration. STATEMENT OF SIGNIFICANCE: Advances in the development of melt electro-writing have allowed micron and submicron polymeric fibres to be accurately printed into porous, complex and three-dimensional structures. By using melt electrowriting, we created a geometrically relevant in vitro model to study cellular spatial-temporal kinetics to provide a systematic understanding of cellular spatial temporal kinetics within a 3D in vitro model. The insights presented in this work help to inform the design of more effective synthetic tissue engineering scaffolds by reducing cell culture time; which is valuable information for the implant or lab-grown-meat industries.
Collapse
|