1
|
Scheufen A, Moreno-Andrés D. Quantitative Live-Cell Imaging to Study Chromatin Segregation and Nuclear Reformation. Methods Mol Biol 2025; 2874:47-60. [PMID: 39614046 DOI: 10.1007/978-1-0716-4236-8_5] [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: 12/01/2024]
Abstract
Live-cell imaging is a powerful tool for the investigation of different steps of the life and fate of single cells and cell populations. In this chapter, we describe how to perform live-cell imaging in tissue culture cells and the subsequent image analysis to precisely characterize the cytological events occurring during mitotic exit and nuclear reformation.
Collapse
Affiliation(s)
- Anja Scheufen
- Institute of Biochemistry and Molecular Cell Biology, Medical School, RWTH Aachen University, Aachen, Germany
| | - Daniel Moreno-Andrés
- Institute of Biochemistry and Molecular Cell Biology, Medical School, RWTH Aachen University, Aachen, Germany.
| |
Collapse
|
2
|
Copperman J, Mclean IC, Gross SM, Singh J, Chang YH, Zuckerman DM, Heiser LM. Single-cell morphodynamical trajectories enable prediction of gene expression accompanying cell state change. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.18.576248. [PMID: 38293173 PMCID: PMC10827140 DOI: 10.1101/2024.01.18.576248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2024]
Abstract
Extracellular signals induce changes to molecular programs that modulate multiple cellular phenotypes, including proliferation, motility, and differentiation status. The connection between dynamically adapting phenotypic states and the molecular programs that define them is not well understood. Here we develop data-driven models of single-cell phenotypic responses to extracellular stimuli by linking gene transcription levels to "morphodynamics" - changes in cell morphology and motility observable in time-lapse image data. We adopt a dynamics-first view of cell state by grouping single-cell trajectories into states with shared morphodynamic responses. The single-cell trajectories enable development of a first-of-its-kind computational approach to map live-cell dynamics to snapshot gene transcript levels, which we term MMIST, Molecular and Morphodynamics-Integrated Single-cell Trajectories. The key conceptual advance of MMIST is that cell behavior can be quantified based on dynamically defined states and that extracellular signals alter the overall distribution of cell states by altering rates of switching between states. We find a cell state landscape that is bound by epithelial and mesenchymal endpoints, with distinct sequences of epithelial to mesenchymal transition (EMT) and mesenchymal to epithelial transition (MET) intermediates. The analysis yields predictions for gene expression changes consistent with curated EMT gene sets and provides a prediction of thousands of RNA transcripts through extracellular signal-induced EMT and MET with near-continuous time resolution. The MMIST framework leverages true single-cell dynamical behavior to generate molecular-level omics inferences and is broadly applicable to other biological domains, time-lapse imaging approaches and molecular snapshot data.
Collapse
Affiliation(s)
- Jeremy Copperman
- Cancer Early Detection Advanced Research Center, Oregon Health and Science University, Portland OR 97239, U.S.A
| | - Ian C. Mclean
- Department of Biomedical Engineering, Oregon Health and Science University, Portland OR 97239, U.S.A
| | | | - Jalim Singh
- Knight Cancer Institute, Oregon Health and Science University, Portland OR 97239, U.S.A
| | - Young Hwan Chang
- Department of Biomedical Engineering, Oregon Health and Science University, Portland OR 97239, U.S.A
- Knight Cancer Institute, Oregon Health and Science University, Portland OR 97239, U.S.A
| | - Daniel M. Zuckerman
- Department of Biomedical Engineering, Oregon Health and Science University, Portland OR 97239, U.S.A
- Knight Cancer Institute, Oregon Health and Science University, Portland OR 97239, U.S.A
| | - Laura M. Heiser
- Department of Biomedical Engineering, Oregon Health and Science University, Portland OR 97239, U.S.A
- Knight Cancer Institute, Oregon Health and Science University, Portland OR 97239, U.S.A
- Cancer Early Detection Advanced Research Center, Oregon Health and Science University, Portland OR 97239, U.S.A
| |
Collapse
|
3
|
Jose A, Roy R, Moreno-Andrés D, Stegmaier J. Automatic detection of cell-cycle stages using recurrent neural networks. PLoS One 2024; 19:e0297356. [PMID: 38466708 PMCID: PMC10927108 DOI: 10.1371/journal.pone.0297356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 01/02/2024] [Indexed: 03/13/2024] Open
Abstract
Mitosis is the process by which eukaryotic cells divide to produce two similar daughter cells with identical genetic material. Research into the process of mitosis is therefore of critical importance both for the basic understanding of cell biology and for the clinical approach to manifold pathologies resulting from its malfunctioning, including cancer. In this paper, we propose an approach to study mitotic progression automatically using deep learning. We used neural networks to predict different mitosis stages. We extracted video sequences of cells undergoing division and trained a Recurrent Neural Network (RNN) to extract image features. The use of RNN enabled better extraction of features. The RNN-based approach gave better performance compared to classifier based feature extraction methods which do not use time information. Evaluation of precision, recall, and F-score indicates the superiority of the proposed model compared to the baseline. To study the loss in performance due to confusion between adjacent classes, we plotted the confusion matrix as well. In addition, we visualized the feature space to understand why RNNs are better at classifying the mitosis stages than other classifier models, which indicated the formation of strong clusters for the different classes, clearly confirming the advantage of the proposed RNN-based approach.
Collapse
Affiliation(s)
- Abin Jose
- Institute of Imaging and Computer Vision, RWTH Aachen University, Aachen, Germany
| | - Rijo Roy
- Institute of Imaging and Computer Vision, RWTH Aachen University, Aachen, Germany
| | - Daniel Moreno-Andrés
- 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
| |
Collapse
|
4
|
Pierzynska-Mach A, Czada C, Vogel C, Gwosch E, Osswald X, Bartoschek D, Diaspro A, Kappes F, Ferrando-May E. DEK oncoprotein participates in heterochromatin replication via SUMO-dependent nuclear bodies. J Cell Sci 2023; 136:jcs261329. [PMID: 37997922 PMCID: PMC10753498 DOI: 10.1242/jcs.261329] [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: 05/12/2023] [Accepted: 11/17/2023] [Indexed: 11/25/2023] Open
Abstract
The correct inheritance of chromatin structure is key for maintaining genome function and cell identity and preventing cellular transformation. DEK, a conserved non-histone chromatin protein, has recognized tumor-promoting properties, its overexpression being associated with poor prognosis in various cancer types. At the cellular level, DEK displays pleiotropic functions, influencing differentiation, apoptosis and stemness, but a characteristic oncogenic mechanism has remained elusive. Here, we report the identification of DEK bodies, focal assemblies of DEK that regularly occur at specific, yet unidentified, sites of heterochromatin replication exclusively in late S-phase. In these bodies, DEK localizes in direct proximity to active replisomes in agreement with a function in the early maturation of heterochromatin. A high-throughput siRNA screen, supported by mutational and biochemical analyses, identifies SUMO as one regulator of DEK body formation, linking DEK to the complex SUMO protein network that controls chromatin states and cell fate. This work combines and refines our previous data on DEK as a factor essential for heterochromatin integrity and facilitating replication under stress, and delineates an avenue of further study for unraveling the contribution of DEK to cancer development.
Collapse
Affiliation(s)
| | - Christina Czada
- Department of Biology, Bioimaging Center, University of Konstanz, Konstanz 78464, Germany
| | - Christopher Vogel
- Department of Biology, Bioimaging Center, University of Konstanz, Konstanz 78464, Germany
| | - Eva Gwosch
- Department of Biology, Bioimaging Center, University of Konstanz, Konstanz 78464, Germany
| | - Xenia Osswald
- Department of Biology, Bioimaging Center, University of Konstanz, Konstanz 78464, Germany
| | - Denis Bartoschek
- Department of Biology, Bioimaging Center, University of Konstanz, Konstanz 78464, Germany
| | - Alberto Diaspro
- Nanoscopy & NIC@IIT, Istituto Italiano di Tecnologia, Genoa 16152, Italy
- DIFILAB, Department of Physics, University of Genoa, Genoa 16146, Italy
| | - Ferdinand Kappes
- Duke Kunshan University, Division of Natural and Applied Sciences, Kunshan 215316, People's Republic of China
| | - Elisa Ferrando-May
- Department of Biology, Bioimaging Center, University of Konstanz, Konstanz 78464, Germany
- German Cancer Research Center, Heidelberg 69120, Germany
| |
Collapse
|
5
|
Stossi F, Singh PK, Safari K, Marini M, Labate D, Mancini MA. High throughput microscopy and single cell phenotypic image-based analysis in toxicology and drug discovery. Biochem Pharmacol 2023; 216:115770. [PMID: 37660829 DOI: 10.1016/j.bcp.2023.115770] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 08/23/2023] [Accepted: 08/25/2023] [Indexed: 09/05/2023]
Abstract
Measuring single cell responses to the universe of chemicals (drugs, natural products, environmental toxicants etc.) is of paramount importance to human health as phenotypic variability in sensing stimuli is a hallmark of biology that is considered during high throughput screening. One of the ways to approach this problem is via high throughput, microscopy-based assays coupled with multi-dimensional single cell analysis methods. Here, we will summarize some of the efforts in this vast and growing field, focusing on phenotypic screens (e.g., Cell Painting), single cell analytics and quality control, with particular attention to environmental toxicology and drug screening. We will discuss advantages and limitations of high throughput assays with various end points and levels of complexity.
Collapse
Affiliation(s)
- Fabio Stossi
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA; GCC Center for Advanced Microscopy and Image Informatics, Houston, TX, USA.
| | - Pankaj K Singh
- GCC Center for Advanced Microscopy and Image Informatics, Houston, TX, USA; Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M University, Houston, TX, USA
| | - Kazem Safari
- GCC Center for Advanced Microscopy and Image Informatics, Houston, TX, USA; Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M University, Houston, TX, USA
| | - Michela Marini
- GCC Center for Advanced Microscopy and Image Informatics, Houston, TX, USA; Department of Mathematics, University of Houston, Houston, TX, USA
| | - Demetrio Labate
- GCC Center for Advanced Microscopy and Image Informatics, Houston, TX, USA; Department of Mathematics, University of Houston, Houston, TX, USA
| | - Michael A Mancini
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA; GCC Center for Advanced Microscopy and Image Informatics, Houston, TX, USA; Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M University, Houston, TX, USA
| |
Collapse
|
6
|
Copperman J, Gross SM, Chang YH, Heiser LM, Zuckerman DM. Morphodynamical cell state description via live-cell imaging trajectory embedding. Commun Biol 2023; 6:484. [PMID: 37142678 PMCID: PMC10160022 DOI: 10.1038/s42003-023-04837-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 04/10/2023] [Indexed: 05/06/2023] Open
Abstract
Time-lapse imaging is a powerful approach to gain insight into the dynamic responses of cells, but the quantitative analysis of morphological changes over time remains challenging. Here, we exploit the concept of "trajectory embedding" to analyze cellular behavior using morphological feature trajectory histories-that is, multiple time points simultaneously, rather than the more common practice of examining morphological feature time courses in single timepoint (snapshot) morphological features. We apply this approach to analyze live-cell images of MCF10A mammary epithelial cells after treatment with a panel of microenvironmental perturbagens that strongly modulate cell motility, morphology, and cell cycle behavior. Our morphodynamical trajectory embedding analysis constructs a shared cell state landscape revealing ligand-specific regulation of cell state transitions and enables quantitative and descriptive models of single-cell trajectories. Additionally, we show that incorporation of trajectories into single-cell morphological analysis enables (i) systematic characterization of cell state trajectories, (ii) better separation of phenotypes, and (iii) more descriptive models of ligand-induced differences as compared to snapshot-based analysis. This morphodynamical trajectory embedding is broadly applicable to the quantitative analysis of cell responses via live-cell imaging across many biological and biomedical applications.
Collapse
Affiliation(s)
- Jeremy Copperman
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, OR, 97239, USA.
| | - Sean M Gross
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, OR, 97239, USA
| | - Young Hwan Chang
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, OR, 97239, USA
- Knight Cancer Institute, Oregon Health and Science University, Portland, OR, 97239, USA
| | - Laura M Heiser
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, OR, 97239, USA.
- Knight Cancer Institute, Oregon Health and Science University, Portland, OR, 97239, USA.
| | - Daniel M Zuckerman
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, OR, 97239, USA.
- Knight Cancer Institute, Oregon Health and Science University, Portland, OR, 97239, USA.
| |
Collapse
|
7
|
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
|
8
|
Guo Z, Gu J, Zhang M, Su F, Su W, Xie Y. NMR-Based Metabolomics to Analyze the Effects of a Series of Monoamine Oxidases-B Inhibitors on U251 Cells. Biomolecules 2023; 13:biom13040600. [PMID: 37189348 DOI: 10.3390/biom13040600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 03/17/2023] [Accepted: 03/22/2023] [Indexed: 03/29/2023] Open
Abstract
Alzheimer’s disease (AD) is a typical progressive neurodegenerative disorder, and with multiple possible pathogenesis. Among them, coumarin derivatives could be used as potential drugs as monoamine oxidase-B (MAO-B) inhibitors. Our lab has designed and synthesized coumarin derivatives based on MAO-B. In this study, we used nuclear magnetic resonance (NMR)-based metabolomics to accelerate the pharmacodynamic evaluation of candidate drugs for coumarin derivative research and development. We detailed alterations in the metabolic profiles of nerve cells with various coumarin derivatives. In total, we identified 58 metabolites and calculated their relative concentrations in U251 cells. In the meantime, the outcomes of multivariate statistical analysis showed that when twelve coumarin compounds were treated with U251cells, the metabolic phenotypes were distinct. In the treatment of different coumarin derivatives, there several metabolic pathways changed, including aminoacyl-tRNA biosynthesis, D-glutamine and D-glutamate metabolism, glycine, serine and threonine metabolism, taurine and hypotaurine metabolism, arginine biosynthesis, alanine, aspartate and glutamate metabolism, phenylalanine, tyrosine and tryptophan biosynthesis, glutathione metabolism and valine, leucine and isoleucine biosynthesis. Our work documented how our coumarin derivatives affected the metabolic phenotype of nerve cells in vitro. We believe that these NMR-based metabolomics might accelerate the process of drug research in vitro and in vivo.
Collapse
|
9
|
Azeroglu B, Ozbun L, Pegoraro G, Lazzerini Denchi E. Native FISH: A low- and high-throughput assay to analyze the alternative lengthening of telomere (ALT) pathway. Methods Cell Biol 2022; 182:265-284. [PMID: 38359982 DOI: 10.1016/bs.mcb.2022.10.010] [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: 11/29/2022]
Abstract
Alternative lengthening of telomeres (ALT) is a telomerase-independent and recombination-based mechanism used by approximately 15% of human cancers to maintain telomere length and to sustain proliferation. ALT-positive cells display unique features that could be exploited for tailored cancer therapies. A key limitation for the development of ALT-specific treatments is the lack of an assay to detect ALT-positive cells that is easy to perform and that can be scaled up. One of the most broadly used assays for ALT detection, CCA (C-circle assay), does not provide single-cell information and it is not amenable to High-Throughput Screening (HTS). To overcome these limitations, we developed Native-FISH (N-FISH) as an alternative method to visualize ALT-specific single-stranded telomeric DNA. N-FISH produces single-cell data, can be applied to fixed tissues, does not require DNA isolation or amplification steps, and it can be miniaturized in a 384-well format. This protocol details the steps to perform N-FISH protocol both in a low- and high-throughput format to analyze ALT. While low-throughput N-FISH is useful to assay the ALT state of cell lines, we expect that the miniaturized N-FISH assay coupled with high-throughput imaging will be useful in functional genomics and chemical screens to identify novel cellular factors that regulate ALT and potential ALT therapeutic targets for cancer therapies directed against ALT-positive tumors, respectively.
Collapse
Affiliation(s)
- Benura Azeroglu
- Laboratory of Genome Integrity, National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD, United States.
| | - Laurent Ozbun
- High-Throughput Imaging Facility (HiTIF), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD, United States
| | - Gianluca Pegoraro
- High-Throughput Imaging Facility (HiTIF), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD, United States
| | - Eros Lazzerini Denchi
- Laboratory of Genome Integrity, National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD, United States
| |
Collapse
|
10
|
Zhang X, Kupczyk E, Schmitt-Kopplin P, Mueller C. Current and future approaches for in vitro hit discovery in diabetes mellitus. Drug Discov Today 2022; 27:103331. [PMID: 35926826 DOI: 10.1016/j.drudis.2022.07.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 06/10/2022] [Accepted: 07/26/2022] [Indexed: 12/15/2022]
Abstract
Type 2 diabetes mellitus (T2DM) is a serious public health problem. In this review, we discuss current and promising future drugs, targets, in vitro assays and emerging omics technologies in T2DM. Importantly, we open the perspective to image-based high-content screening (HCS), with the focus of combining it with metabolomics or lipidomics. HCS has become a strong technology in phenotypic screens because it allows comprehensive screening for the cell-modulatory activity of small molecules. Metabolomics and lipidomics screen for perturbations at the molecular level. The combination of these data-intensive comprehensive technologies is enabled by the rapid development of artificial intelligence. It promises a deep cellular and molecular phenotyping directly linked to chemical information about the applied drug candidates or complex mixtures.
Collapse
Affiliation(s)
- Xin Zhang
- Research Unit Analytical BioGeoChemistry, Helmholtz Zentrum München, Ingolstaedter Landstr. 1, 85764 Neuherberg, Germany
| | - Erwin Kupczyk
- Research Unit Analytical BioGeoChemistry, Helmholtz Zentrum München, Ingolstaedter Landstr. 1, 85764 Neuherberg, Germany; Comprehensive Foodomics Platform, Chair of Analytical Food Chemistry, TUM School of Life Sciences, Technical University of Munich, Maximus-von-Imhof-Forum 2, 85354 Freising, Germany
| | - Philippe Schmitt-Kopplin
- Research Unit Analytical BioGeoChemistry, Helmholtz Zentrum München, Ingolstaedter Landstr. 1, 85764 Neuherberg, Germany; Comprehensive Foodomics Platform, Chair of Analytical Food Chemistry, TUM School of Life Sciences, Technical University of Munich, Maximus-von-Imhof-Forum 2, 85354 Freising, Germany.
| | - Constanze Mueller
- Research Unit Analytical BioGeoChemistry, Helmholtz Zentrum München, Ingolstaedter Landstr. 1, 85764 Neuherberg, Germany.
| |
Collapse
|
11
|
Jung J, Khan MM, Landry J, Halavatyi A, Machado P, Reiss M, Pepperkok R. Regulation of the COPII secretory machinery via focal adhesions and extracellular matrix signaling. J Cell Biol 2022; 221:213351. [PMID: 35829701 PMCID: PMC9284426 DOI: 10.1083/jcb.202110081] [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: 10/15/2021] [Revised: 05/10/2022] [Accepted: 06/24/2022] [Indexed: 12/19/2022] Open
Abstract
Proteins that enter the secretory pathway are transported from their place of synthesis in the endoplasmic reticulum to the Golgi complex by COPII-coated carriers. The networks of proteins that regulate these components in response to extracellular cues have remained largely elusive. Using high-throughput microscopy, we comprehensively screened 378 cytoskeleton-associated and related proteins for their functional interaction with the coat protein complex II (COPII) components SEC23A and SEC23B. Among these, we identified a group of proteins associated with focal adhesions (FERMT2, MACF1, MAPK8IP2, NGEF, PIK3CA, and ROCK1) that led to the downregulation of SEC23A when depleted by siRNA. Changes in focal adhesions induced by plating cells on ECM also led to the downregulation of SEC23A and decreases in VSVG transport from ER to Golgi. Both the expression of SEC23A and the transport defect could be rescued by treatment with a focal adhesion kinase inhibitor. Altogether, our results identify a network of cytoskeleton-associated proteins connecting focal adhesions and ECM-related signaling with the gene expression of the COPII secretory machinery and trafficking.
Collapse
Affiliation(s)
- Juan Jung
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Muzamil Majid Khan
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, Heidelberg, Germany.,Translational Lung Research Center Heidelberg, German Center for Lung Research, Heidelberg, Germany
| | - Jonathan Landry
- Core Facilities Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Aliaksandr Halavatyi
- Core Facilities Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Pedro Machado
- Core Facilities Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Miriam Reiss
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Rainer Pepperkok
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, Heidelberg, Germany.,Core Facilities Unit, European Molecular Biology Laboratory, Heidelberg, Germany.,Translational Lung Research Center Heidelberg, German Center for Lung Research, Heidelberg, Germany
| |
Collapse
|
12
|
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
|
13
|
Image-Based Annotation of Chemogenomic Libraries for Phenotypic Screening. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27041439. [PMID: 35209227 PMCID: PMC8878468 DOI: 10.3390/molecules27041439] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 02/15/2022] [Accepted: 02/16/2022] [Indexed: 12/26/2022]
Abstract
Phenotypical screening is a widely used approach in drug discovery for the identification of small molecules with cellular activities. However, functional annotation of identified hits often poses a challenge. The development of small molecules with narrow or exclusive target selectivity such as chemical probes and chemogenomic (CG) libraries, greatly diminishes this challenge, but non-specific effects caused by compound toxicity or interference with basic cellular functions still pose a problem to associate phenotypic readouts with molecular targets. Hence, each compound should ideally be comprehensively characterized regarding its effects on general cell functions. Here, we report an optimized live-cell multiplexed assay that classifies cells based on nuclear morphology, presenting an excellent indicator for cellular responses such as early apoptosis and necrosis. This basic readout in combination with the detection of other general cell damaging activities of small molecules such as changes in cytoskeletal morphology, cell cycle and mitochondrial health provides a comprehensive time-dependent characterization of the effect of small molecules on cellular health in a single experiment. The developed high-content assay offers multi-dimensional comprehensive characterization that can be used to delineate generic effects regarding cell functions and cell viability, allowing an assessment of compound suitability for subsequent detailed phenotypic and mechanistic studies.
Collapse
|
14
|
Tirumala MG, Anchi P, Raja S, Rachamalla M, Godugu C. Novel Methods and Approaches for Safety Evaluation of Nanoparticle Formulations: A Focus Towards In Vitro Models and Adverse Outcome Pathways. Front Pharmacol 2021; 12:612659. [PMID: 34566630 PMCID: PMC8458898 DOI: 10.3389/fphar.2021.612659] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 08/05/2021] [Indexed: 12/18/2022] Open
Abstract
Nanotoxicology is an emerging field employed in the assessment of unintentional hazardous effects produced by nanoparticles (NPs) impacting human health and the environment. The nanotoxicity affects the range between induction of cellular stress and cytotoxicity. The reasons so far reported for these toxicological effects are due to their variable sizes with high surface areas, shape, charge, and physicochemical properties, which upon interaction with the biological components may influence their functioning and result in adverse outcomes (AO). Thus, understanding the risk produced by these materials now is an important safety concern for the development of nanotechnology and nanomedicine. Since the time nanotoxicology has evolved, the methods employed have been majorly relied on in vitro cell-based evaluations, while these simple methods may not predict the complexity involved in preclinical and clinical conditions concerning pharmacokinetics, organ toxicity, and toxicities evidenced through multiple cellular levels. The safety profiles of nanoscale nanomaterials and nanoformulations in the delivery of drugs and therapeutic applications are of considerable concern. In addition, the safety assessment for new nanomedicine formulas lacks regulatory standards. Though the in vivo studies are greatly needed, the end parameters used for risk assessment are not predicting the possible toxic effects produced by various nanoformulations. On the other side, due to increased restrictions on animal usage and demand for the need for high-throughput assays, there is a need for developing and exploring novel methods to evaluate NPs safety concerns. The progress made in molecular biology and the availability of several modern techniques may offer novel and innovative methods to evaluate the toxicological behavior of different NPs by using single cells, cell population, and whole organisms. This review highlights the recent novel methods developed for the evaluation of the safety impacts of NPs and attempts to solve the problems that come with risk assessment. The relevance of investigating adverse outcome pathways (AOPs) in nanotoxicology has been stressed in particular.
Collapse
Affiliation(s)
- Mounika Gayathri Tirumala
- Department of Regulatory Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Pratibha Anchi
- Department of Regulatory Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Susmitha Raja
- Department of Regulatory Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Mahesh Rachamalla
- Department of Biology, University of Saskatchewan, Saskatoon, SK, Canada
| | - Chandraiah Godugu
- Department of Regulatory Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| |
Collapse
|
15
|
Schilling M, Prusty AB, Boysen B, Oppermann FS, Riedel YL, Husedzinovic A, Rasouli H, König A, Ramanathan P, Reymann J, Erfle H, Daub H, Fischer U, Gruss OJ. TOR signaling regulates liquid phase separation of the SMN complex governing snRNP biogenesis. Cell Rep 2021; 35:109277. [PMID: 34161763 DOI: 10.1016/j.celrep.2021.109277] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 04/23/2021] [Accepted: 05/27/2021] [Indexed: 01/10/2023] Open
Abstract
The activity of the SMN complex in promoting the assembly of pre-mRNA processing UsnRNPs correlates with condensation of the complex in nuclear Cajal bodies. While mechanistic details of its activity have been elucidated, the molecular basis for condensation remains unclear. High SMN complex phosphorylation suggests extensive regulation. Here, we report on systematic siRNA-based screening for modulators of the capacity of SMN to condense in Cajal bodies and identify mTOR and ribosomal protein S6 kinase β-1 as key regulators. Proteomic analysis reveals TOR-dependent phosphorylations in SMN complex subunits. Using stably expressed or optogenetically controlled phospho mutants, we demonstrate that serine 49 and 63 phosphorylation of human SMN controls the capacity of the complex to condense in Cajal bodies via liquid-liquid phase separation. Our findings link SMN complex condensation and UsnRNP biogenesis to cellular energy levels and suggest modulation of TOR signaling as a rational concept for therapy of the SMN-linked neuromuscular disorder spinal muscular atrophy.
Collapse
Affiliation(s)
- Maximilian Schilling
- Institut für Genetik, Rheinische Friedrich-Wilhelms Universität Bonn, 53115 Bonn, Germany
| | - Archana B Prusty
- Theodor Boveri Institute, Biocenter of the University of Würzburg, 97074 Würzburg, Germany
| | - Björn Boysen
- Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH), Im Neuenheimer Feld 282, 69120 Heidelberg, Germany
| | | | - Yannick L Riedel
- Institut für Genetik, Rheinische Friedrich-Wilhelms Universität Bonn, 53115 Bonn, Germany
| | - Alma Husedzinovic
- Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH), Im Neuenheimer Feld 282, 69120 Heidelberg, Germany
| | - Homa Rasouli
- Evotec SE, Am Klopferspitz 19a, 82152 Martinsried, Germany
| | - Angelika König
- Institut für Genetik, Rheinische Friedrich-Wilhelms Universität Bonn, 53115 Bonn, Germany
| | - Pradhipa Ramanathan
- Theodor Boveri Institute, Biocenter of the University of Würzburg, 97074 Würzburg, Germany
| | - Jürgen Reymann
- Advanced Biological Screening Facility, BioQuant Centre, Universität Heidelberg, Im Neuenheimer Feld 267, 69120 Heidelberg, Germany
| | - Holger Erfle
- Advanced Biological Screening Facility, BioQuant Centre, Universität Heidelberg, Im Neuenheimer Feld 267, 69120 Heidelberg, Germany
| | - Henrik Daub
- Evotec SE, Am Klopferspitz 19a, 82152 Martinsried, Germany
| | - Utz Fischer
- Theodor Boveri Institute, Biocenter of the University of Würzburg, 97074 Würzburg, Germany
| | - Oliver J Gruss
- Institut für Genetik, Rheinische Friedrich-Wilhelms Universität Bonn, 53115 Bonn, Germany; Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH), Im Neuenheimer Feld 282, 69120 Heidelberg, Germany.
| |
Collapse
|
16
|
Wilson EL, Metzakopian E. ER-mitochondria contact sites in neurodegeneration: genetic screening approaches to investigate novel disease mechanisms. Cell Death Differ 2021; 28:1804-1821. [PMID: 33335290 PMCID: PMC8185109 DOI: 10.1038/s41418-020-00705-8] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 11/26/2020] [Accepted: 11/30/2020] [Indexed: 12/26/2022] Open
Abstract
Mitochondria-ER contact sites (MERCS) are known to underpin many important cellular homoeostatic functions, including mitochondrial quality control, lipid metabolism, calcium homoeostasis, the unfolded protein response and ER stress. These functions are known to be dysregulated in neurodegenerative diseases, including Parkinson's disease (PD), Alzheimer's disease (AD) and amyloid lateral sclerosis (ALS), and the number of disease-related proteins and genes being associated with MERCS is increasing. However, many details regarding MERCS and their role in neurodegenerative diseases remain unknown. In this review, we aim to summarise the current knowledge regarding the structure and function of MERCS, and to update the field on current research in PD, AD and ALS. Furthermore, we will evaluate high-throughput screening techniques, including RNAi vs CRISPR/Cas9, pooled vs arrayed formats and how these could be combined with current techniques to visualise MERCS. We will consider the advantages and disadvantages of each technique and how it can be utilised to uncover novel protein pathways involved in MERCS dysfunction in neurodegenerative diseases.
Collapse
Affiliation(s)
- Emma Louise Wilson
- UK Dementia Research Institute, Department of Clinical Neuroscience, University of Cambridge, Cambridge, CB2 0AH, UK.
- Open Targets, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, UK.
| | - Emmanouil Metzakopian
- UK Dementia Research Institute, Department of Clinical Neuroscience, University of Cambridge, Cambridge, CB2 0AH, UK.
| |
Collapse
|
17
|
Gubas A, Karantanou C, Popovic D, Tascher G, Hoffmann ME, Platzek A, Dawe N, Dikic I, Krause DS, McEwan DG. The endolysosomal adaptor PLEKHM1 is a direct target for both mTOR and MAPK pathways. FEBS Lett 2021; 595:864-880. [PMID: 33452816 DOI: 10.1002/1873-3468.14041] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 12/23/2020] [Accepted: 12/28/2020] [Indexed: 12/21/2022]
Abstract
The lysosome is a cellular signalling hub at the point of convergence of endocytic and autophagic pathways, where the contents are degraded and recycled. Pleckstrin homology domain-containing family member 1 (PLEKHM1) acts as an adaptor to facilitate the fusion of endocytic and autophagic vesicles with the lysosome. However, it is unclear how PLEKHM1 function at the lysosome is controlled. Herein, we show that PLEKHM1 coprecipitates with, and is directly phosphorylated by, mTOR. Using a phosphospecific antibody against Ser432/S435 of PLEKHM1, we show that the same motif is a direct target for ERK2-mediated phosphorylation in a growth factor-dependent manner. This dual regulation of PLEKHM1 at a highly conserved region points to a convergence of both growth factor- and amino acid-sensing pathways, placing PLEKHM1 at a critical juncture of cellular metabolism.
Collapse
Affiliation(s)
- Andrea Gubas
- Faculty of Medicine, Institute of Biochemistry II, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Christina Karantanou
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Medicine, Frankfurt, Germany.,Goethe University Frankfurt, Frankfurt, Germany
| | - Doris Popovic
- Faculty of Medicine, Institute of Biochemistry II, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Georg Tascher
- Faculty of Medicine, Institute of Biochemistry II, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Marina E Hoffmann
- Faculty of Medicine, Institute of Biochemistry II, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Anna Platzek
- Faculty of Medicine, Institute of Biochemistry II, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Nina Dawe
- Division of Cell Signalling & Immunology, School of Life Sciences, University of Dundee, UK
| | - Ivan Dikic
- Faculty of Medicine, Institute of Biochemistry II, Goethe University Frankfurt, Frankfurt am Main, Germany.,Buchmann Institute for Molecular Life Sciences, Goethe University Frankfurt, Frankfurt am Main, Germany.,Max Planck Institute of Biophysics, Frankfurt am Main, Germany
| | - Daniela S Krause
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Medicine, Frankfurt, Germany.,Goethe University Frankfurt, Frankfurt, Germany
| | - David G McEwan
- Division of Cell Signalling & Immunology, School of Life Sciences, University of Dundee, UK.,Cancer Research UK Beatson Institute, Garscube Estate, Glasgow, UK
| |
Collapse
|
18
|
Liimatainen K, Huttunen R, Latonen L, Ruusuvuori P. Convolutional Neural Network-Based Artificial Intelligence for Classification of Protein Localization Patterns. Biomolecules 2021; 11:biom11020264. [PMID: 33670112 PMCID: PMC7916854 DOI: 10.3390/biom11020264] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 01/15/2021] [Accepted: 01/19/2021] [Indexed: 12/22/2022] Open
Abstract
Identifying localization of proteins and their specific subpopulations associated with certain cellular compartments is crucial for understanding protein function and interactions with other macromolecules. Fluorescence microscopy is a powerful method to assess protein localizations, with increasing demand of automated high throughput analysis methods to supplement the technical advancements in high throughput imaging. Here, we study the applicability of deep neural network-based artificial intelligence in classification of protein localization in 13 cellular subcompartments. We use deep learning-based on convolutional neural network and fully convolutional network with similar architectures for the classification task, aiming at achieving accurate classification, but importantly, also comparison of the networks. Our results show that both types of convolutional neural networks perform well in protein localization classification tasks for major cellular organelles. Yet, in this study, the fully convolutional network outperforms the convolutional neural network in classification of images with multiple simultaneous protein localizations. We find that the fully convolutional network, using output visualizing the identified localizations, is a very useful tool for systematic protein localization assessment.
Collapse
Affiliation(s)
- Kaisa Liimatainen
- Faculty of Medicine and Health Technology, Tampere University, FI-33520 Tampere, Finland; (K.L.); (R.H.)
| | - Riku Huttunen
- Faculty of Medicine and Health Technology, Tampere University, FI-33520 Tampere, Finland; (K.L.); (R.H.)
- Department of Applied Physics, University of Eastern Finland, FI-70211 Kuopio, Finland
| | - Leena Latonen
- Institute of Biomedicine, University of Eastern Finland, FI-70211 Kuopio, Finland;
| | - Pekka Ruusuvuori
- Faculty of Medicine and Health Technology, Tampere University, FI-33520 Tampere, Finland; (K.L.); (R.H.)
- Institute of Biomedicine, University of Turku, FI-20014 Turku, Finland
- Correspondence: ; Tel.: +358-50-318-2407
| |
Collapse
|
19
|
Hagemeijer MC, Vonk AM, Awatade NT, Silva IAL, Tischer C, Hilsenstein V, Beekman JM, Amaral MD, Botelho HM. An open-source high-content analysis workflow for CFTR function measurements using the forskolin-induced swelling assay. Bioinformatics 2020; 36:5686-5694. [PMID: 33367496 DOI: 10.1093/bioinformatics/btaa1073] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Revised: 11/13/2020] [Accepted: 12/14/2020] [Indexed: 01/20/2023] Open
Abstract
MOTIVATION The forskolin-induced swelling (FIS) assay has become the preferential assay to predict the efficacy of approved and investigational CFTR-modulating drugs for individuals with cystic fibrosis (CF). Currently, no standardized quantification method of FIS data exists thereby hampering inter-laboratory reproducibility. RESULTS We developed a complete open-source workflow for standardized high-content analysis of CFTR function measurements in intestinal organoids using raw microscopy images as input. The workflow includes tools for (i) file and metadata handling; (ii) image quantification and (iii) statistical analysis. Our workflow reproduced results generated by published proprietary analysis protocols and enables standardized CFTR function measurements in CF organoids. AVAILABILITY All workflow components are open-source and freely available: the htmrenamer R package for file handling https://github.com/hmbotelho/htmrenamer; CellProfiler and ImageJ analysis scripts/pipelines https://github.com/hmbotelho/FIS_image_analysis; the Organoid Analyst application for statistical analysis https://github.com/hmbotelho/organoid_analyst; detailed usage instructions and a demonstration dataset https://github.com/hmbotelho/FIS_analysis. Distributed under GPL v3.0. SUPPLEMENTARY INFORMATION Supplementary information and a stepwise guide for software installation and data analysis for training purposes are available at Bioinformatics online.
Collapse
Affiliation(s)
- Marne C Hagemeijer
- Department of Pediatric Pulmonology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, The Netherlands; Regenerative Medicine Center Utrecht, University Medical Centre Utrecht, 3584 EA, Utrecht, The Netherlands.,Department of Clinical Genetics, Erasmus Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Annelotte M Vonk
- Department of Pediatric Pulmonology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, The Netherlands; Regenerative Medicine Center Utrecht, University Medical Centre Utrecht, 3584 EA, Utrecht, The Netherlands
| | - Nikhil T Awatade
- BioISI-Biosystems & Integrative Sciences Institute, Faculty of Sciences, University of Lisboa, 1749-016, Lisboa, Portugal.,>School of Women's & Children's Health, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
| | - Iris A L Silva
- BioISI-Biosystems & Integrative Sciences Institute, Faculty of Sciences, University of Lisboa, 1749-016, Lisboa, Portugal
| | - Christian Tischer
- European Molecular Biology Laboratory (EMBL), Centre for Bioimage Analysis (CBA), 69117, Heidelberg, Germany
| | - Volker Hilsenstein
- European Molecular Biology Laboratory (EMBL), Advanced Light Microscopy Facility (ALMF), 69117, Heidelberg, Germany.,Monash Micro Imaging, Monash University, Melbourne, Australia
| | - Jeffrey M Beekman
- Department of Pediatric Pulmonology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, The Netherlands; Regenerative Medicine Center Utrecht, University Medical Centre Utrecht, 3584 EA, Utrecht, The Netherlands
| | - Margarida D Amaral
- BioISI-Biosystems & Integrative Sciences Institute, Faculty of Sciences, University of Lisboa, 1749-016, Lisboa, Portugal
| | - Hugo M Botelho
- BioISI-Biosystems & Integrative Sciences Institute, Faculty of Sciences, University of Lisboa, 1749-016, Lisboa, Portugal
| |
Collapse
|
20
|
Tsuzuki Y, Sanami S, Sugimoto K, Fujita S. Pseudo-nuclear staining of cells by deep learning improves the accuracy of automated cell counting in a label-free cellular population. J Biosci Bioeng 2020; 131:213-218. [PMID: 33077361 DOI: 10.1016/j.jbiosc.2020.09.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 08/21/2020] [Accepted: 09/22/2020] [Indexed: 11/18/2022]
Abstract
Deep learning has emerged as a breakthrough tool for the segmentation of images without supporting human experts. Here, we propose an automated approach that uses deep learning to generate pseudo-nuclear staining of cells from phase contrast images. Our proposed approach, which has the feature to generate pseudo-nuclear stained images by simple DNN, showed remarkable advantages over existing approaches in the cell-detection and the detection of the relative position of cells for various cell densities, as well as in counting the exact cell numbers. Pseudo-nuclear staining of cells by deep learning will improve the accuracy of automated cell counting in a label-free cellular population on phase contrast images.
Collapse
Affiliation(s)
- Yuji Tsuzuki
- Advance Business Center, ICT Business Development Division, Dai Nippon Printing Co., Ltd., 1-1-1 Ichigaya Kaga-cho, Shinjuku-ku, Tokyo 162-8001, Japan
| | - Sho Sanami
- Advance Business Center, ICT Business Development Division, Dai Nippon Printing Co., Ltd., 1-1-1 Ichigaya Kaga-cho, Shinjuku-ku, Tokyo 162-8001, Japan
| | - Kenji Sugimoto
- Live Cell Imaging Institute, University-Originated Venture, Osaka Prefecture University, 1-1 Gakuen-cho, Naka-ku, Sakai, Osaka 599-8531, Japan
| | - Satoshi Fujita
- Advanced Photonics and Biosensing Open Innovation Laboratory, National Institute of Advanced Industrial Science and Technology (AIST), 2-1 Yamada-Oka, Suita, Osaka 565-0871, Japan; Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), 1-8-31 Midorioka, Ikeda, Osaka 563-0026, Japan.
| |
Collapse
|
21
|
Shakirova KM, Ovchinnikova VY, Dashinimaev EB. Cell Reprogramming With CRISPR/Cas9 Based Transcriptional Regulation Systems. Front Bioeng Biotechnol 2020; 8:882. [PMID: 32850737 PMCID: PMC7399070 DOI: 10.3389/fbioe.2020.00882] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 07/09/2020] [Indexed: 12/22/2022] Open
Abstract
The speed of reprogramming technologies evolution is rising dramatically in modern science. Both the scientific community and health workers depend on such developments due to the lack of safe autogenic cells and tissues for regenerative medicine, genome editing tools and reliable screening techniques. To perform experiments efficiently and to propel the fundamental science it is important to keep up with novel modifications and techniques that are being discovered almost weekly. One of them is CRISPR/Cas9 based genome and transcriptome editing. The aim of this article is to summarize currently existing CRISPR/Cas9 applications for cell reprogramming, mainly, to compare them with other non-CRISPR approaches and to highlight future perspectives and opportunities.
Collapse
Affiliation(s)
- Ksenia M Shakirova
- Faculty of Biology, Lomonosov Moscow State University, Moscow, Russia.,Koltzov Institute of Developmental Biology, Russian Academy of Sciences, Moscow, Russia.,Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Moscow, Russia
| | - Viktoriia Y Ovchinnikova
- Faculty of Biology, Lomonosov Moscow State University, Moscow, Russia.,Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Moscow, Russia
| | - Erdem B Dashinimaev
- Koltzov Institute of Developmental Biology, Russian Academy of Sciences, Moscow, Russia.,Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Moscow, Russia
| |
Collapse
|
22
|
Liu Y, Tronser T, Peravali R, Reischl M, Levkin PA. High‐Throughput Screening of Cell Transfection Enhancers Using Miniaturized Droplet Microarrays. ACTA ACUST UNITED AC 2020; 4:e1900257. [DOI: 10.1002/adbi.201900257] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 12/04/2019] [Indexed: 11/07/2022]
Affiliation(s)
- Yanxi Liu
- Institute of Toxicology and Genetics (ITG)Karlsruhe Institute of Technology (KIT) Hermann‐von Helmholtz‐Platz 1 Eggenstein‐Leopoldshafen 76344 Germany
| | - Tina Tronser
- Institute of Toxicology and Genetics (ITG)Karlsruhe Institute of Technology (KIT) Hermann‐von Helmholtz‐Platz 1 Eggenstein‐Leopoldshafen 76344 Germany
| | - Ravindra Peravali
- Institute of Toxicology and Genetics (ITG)Karlsruhe Institute of Technology (KIT) Hermann‐von Helmholtz‐Platz 1 Eggenstein‐Leopoldshafen 76344 Germany
| | - Markus Reischl
- Institute for Automation and Applied Informatics (IAI)Karlsruhe Institute of Technology (KIT) Hermann‐von Helmholtz‐Platz 1 Eggenstein‐Leopoldshafen 76344 Germany
| | - Pavel A. Levkin
- Institute of Toxicology and Genetics (ITG)Karlsruhe Institute of Technology (KIT) Hermann‐von Helmholtz‐Platz 1 Eggenstein‐Leopoldshafen 76344 Germany
- Institute of Organic ChemistryKarlsruhe Institute of Technology (KIT) Fritz‐Haber‐Weg 6 Karlsruhe 76131 Germany
| |
Collapse
|
23
|
Yin L, Siracusa JS, Measel E, Guan X, Edenfield C, Liang S, Yu X. High-Content Image-Based Single-Cell Phenotypic Analysis for the Testicular Toxicity Prediction Induced by Bisphenol A and Its Analogs Bisphenol S, Bisphenol AF, and Tetrabromobisphenol A in a Three-Dimensional Testicular Cell Co-culture Model. Toxicol Sci 2020; 173:313-335. [PMID: 31750923 PMCID: PMC6986343 DOI: 10.1093/toxsci/kfz233] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Emerging data indicate that structural analogs of bisphenol A (BPA) such as bisphenol S (BPS), tetrabromobisphenol A (TBBPA), and bisphenol AF (BPAF) have been introduced into the market as substitutes for BPA. Our previous study compared in vitro testicular toxicity using murine C18-4 spermatogonial cells and found that BPAF and TBBPA exhibited higher spermatogonial toxicities as compared with BPA and BPS. Recently, we developed a novel in vitro three-dimensional (3D) testicular cell co-culture model, enabling the classification of reproductive toxic substances. In this study, we applied the testicular cell co-culture model and employed a high-content image (HCA)-based single-cell analysis to further compare the testicular toxicities of BPA and its analogs. We also developed a machine learning (ML)-based HCA pipeline to examine the complex phenotypic changes associated with testicular toxicities. We found dose- and time-dependent changes in a wide spectrum of adverse endpoints, including nuclear morphology, DNA synthesis, DNA damage, and cytoskeletal structure in a single-cell-based analysis. The co-cultured testicular cells were more sensitive than the C18 spermatogonial cells in response to BPA and its analogs. Unlike conventional population-averaged assays, single-cell-based assays not only showed the levels of the averaged population, but also revealed changes in the sub-population. Machine learning-based phenotypic analysis revealed that treatment of BPA and its analogs resulted in the loss of spatial cytoskeletal structure, and an accumulation of M phase cells in a dose- and time-dependent manner. Furthermore, treatment of BPAF-induced multinucleated cells, which were associated with altered DNA damage response and impaired cellular F-actin filaments. Overall, we demonstrated a new and effective means to evaluate multiple toxic endpoints in the testicular co-culture model through the combination of ML and high-content image-based single-cell analysis. This approach provided an in-depth analysis of the multi-dimensional HCA data and provided an unbiased quantitative analysis of the phenotypes of interest.
Collapse
Affiliation(s)
- Lei Yin
- ReproTox Biotech LLC, Athens, Georgia 30602
| | - Jacob Steven Siracusa
- Department of Environmental Health Science, College of Public Health, University of Georgia, Athens, Georgia
| | - Emily Measel
- Department of Environmental Health Science, College of Public Health, University of Georgia, Athens, Georgia
| | | | - Clayton Edenfield
- Department of Environmental Health Science, College of Public Health, University of Georgia, Athens, Georgia
| | - Shenxuan Liang
- Department of Environmental Health Science, College of Public Health, University of Georgia, Athens, Georgia
| | - Xiaozhong Yu
- Department of Environmental Health Science, College of Public Health, University of Georgia, Athens, Georgia
| |
Collapse
|
24
|
Mitra S, Bodor DL, David AF, Abdul-Zani I, Mata JF, Neumann B, Reither S, Tischer C, Jansen LET. Genetic screening identifies a SUMO protease dynamically maintaining centromeric chromatin. Nat Commun 2020; 11:501. [PMID: 31980633 PMCID: PMC6981222 DOI: 10.1038/s41467-019-14276-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 12/18/2019] [Indexed: 12/27/2022] Open
Abstract
Centromeres are defined by a self-propagating chromatin structure based on stable inheritance of CENP-A containing nucleosomes. Here, we present a genetic screen coupled to pulse-chase labeling that allow us to identify proteins selectively involved in deposition of nascent CENP-A or in long-term transmission of chromatin-bound CENP-A. These include factors with known roles in DNA replication, repair, chromatin modification, and transcription, revealing a broad set of chromatin regulators that impact on CENP-A dynamics. We further identify the SUMO-protease SENP6 as a key factor, not only controlling CENP-A stability but virtually the entire centromere and kinetochore. Loss of SENP6 results in hyper-SUMOylation of CENP-C and CENP-I but not CENP-A itself. SENP6 activity is required throughout the cell cycle, suggesting that a dynamic SUMO cycle underlies a continuous surveillance of the centromere complex that in turn ensures stable transmission of CENP-A chromatin.
Collapse
Affiliation(s)
- Sreyoshi Mitra
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford, OX1 3QU, UK
- Instituto Gulbenkian de Ciência, 2780-156, Oeiras, Portugal
| | - Dani L Bodor
- Instituto Gulbenkian de Ciência, 2780-156, Oeiras, Portugal
- MRC-Laboratory for Molecular Cell Biology, UCL, London, WC1E 6BT, UK
| | - Ana F David
- Instituto Gulbenkian de Ciência, 2780-156, Oeiras, Portugal
- Institute of Molecular Biotechnology, Dr. Bohr-Gasse 3, 1030, Vienna, Austria
| | - Izma Abdul-Zani
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford, OX1 3QU, UK
| | - João F Mata
- Instituto Gulbenkian de Ciência, 2780-156, Oeiras, Portugal
| | - Beate Neumann
- European Molecular Biology Laboratory (EMBL), Meyerhofstrasse 1, D-69117, Heidelberg, Germany
| | - Sabine Reither
- European Molecular Biology Laboratory (EMBL), Meyerhofstrasse 1, D-69117, Heidelberg, Germany
| | - Christian Tischer
- European Molecular Biology Laboratory (EMBL), Meyerhofstrasse 1, D-69117, Heidelberg, Germany
| | - Lars E T Jansen
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford, OX1 3QU, UK.
- Instituto Gulbenkian de Ciência, 2780-156, Oeiras, Portugal.
| |
Collapse
|
25
|
Rappaz B, Jourdain P, Banfi D, Kuttler F, Marquet P, Turcatti G. Image-Based Marker-Free Screening of GABA A Agonists, Antagonists, and Modulators. SLAS DISCOVERY 2019; 25:458-470. [PMID: 31779505 PMCID: PMC7243081 DOI: 10.1177/2472555219887142] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
The ionotropic GABAA receptors represent the main target for different groups of widely used drugs having hypnotic and anxiolytic effects. So far, most approaches used to assess GABA activity involve invasive low -throughput electrophysiological techniques or rely on fluorescent dyes, preventing the ability to conduct noninvasive and thus nonperturbing screens. To address this limitation, we have developed an automated marker-free cell imaging method, based on digital holographic microscopy (DHM). This technology allows the automatically screening of compounds in multiple plates without having to label the cells or use special plates. This methodological approach was first validated by screening the GABAA receptor expressed in HEK cells using a selection of active compounds in agonist, antagonist, and modulator modes. Then, in a second blind screen of a library of 3041 compounds (mostly composed of natural products), 5 compounds having a specific agonist action on the GABAA receptor were identified. The hits validated from this unbiased screen were the natural products muscimol, neurosteroid alphaxalone, and three compounds belonging to the avermectin family, all known for having an agonistic effect on the GABAA receptor. The results obtained were exempt from false negatives (structurally similar unassigned hits), and false-positive hits were detected and discarded without the need for performing electrophysiological measurements. The outcome of the screen demonstrates the applicability of our screening by imaging method for the discovery of new chemical structures, particularly regarding chemicals interacting with the ionotropic GABAA receptor and more generally with any ligand-gated ion channels and transporters.
Collapse
Affiliation(s)
- Benjamin Rappaz
- Biomolecular Screening Facility (BSF), Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Pascal Jourdain
- Joint International Research Unit in Child Psychiatry, Département de Psychiatrie CHUV, University of Lausanne, Switzerland.,Université Laval, Québec, QC, Canada
| | - Damiano Banfi
- Biomolecular Screening Facility (BSF), Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Fabien Kuttler
- Biomolecular Screening Facility (BSF), Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Pierre Marquet
- Joint International Research Unit in Child Psychiatry, Département de Psychiatrie CHUV, University of Lausanne, Switzerland.,Université Laval, Québec, QC, Canada.,Centre de Recherche CERVO, Québec, QC, Canada
| | - Gerardo Turcatti
- Biomolecular Screening Facility (BSF), Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| |
Collapse
|
26
|
Zhou FY, Ruiz-Puig C, Owen RP, White MJ, Rittscher J, Lu X. Characterization of Biological Motion Using Motion Sensing Superpixels. Bio Protoc 2019; 9:e3365. [PMID: 33654862 DOI: 10.21769/bioprotoc.3365] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 08/05/2019] [Accepted: 08/19/2019] [Indexed: 11/02/2022] Open
Abstract
Precise spatiotemporal regulation is the foundation for the healthy development and maintenance of living organisms. All cells must correctly execute their function in the right place at the right time. Cellular motion is thus an important dynamic readout of signaling in key disease-relevant molecular pathways. However despite the rapid advancement of imaging technology, a comprehensive quantitative description of motion imaged under different imaging modalities at all spatiotemporal scales; molecular, cellular and tissue-level is still lacking. Generally, cells move either 'individually' or 'collectively' as a group with nearby cells. Current computational tools specifically focus on one or the other regime, limiting their general applicability. To address this, we recently developed and reported a new computational framework, Motion Sensing Superpixels (MOSES). Incorporating the individual advantages of single cell trackers for individual cell and particle image velocimetry (PIV) for collective cell motion analyses, MOSES enables 'mesoscale' analysis of both single-cell and collective motion over arbitrarily long times. At the same time, MOSES readily complements existing single-cell tracking workflows with additional characterization of global motion patterns and interaction analysis between cells and also operates directly on PIV extracted motion fields to yield rich motion trajectories analogous for single-cell tracks suitable for high-throughput motion phenotyping. This protocol provides a step-by-step practical guide for those interested in applying MOSES to their own datasets. The protocol highlights the salient features of a MOSES analysis and demonstrates the ease-of-use and wide applicability of MOSES to biological imaging through demo experimental analyses with ready-to-use code snippets of four datasets from different microscope modalities; phase-contrast, fluorescent, lightsheet and intra-vital microscopy. In addition we discuss critical points of consideration in the analysis.
Collapse
Affiliation(s)
- Felix Y Zhou
- Ludwig Institute for Cancer Research, University of Oxford, Oxford, United Kingdom
| | - Carlos Ruiz-Puig
- Ludwig Institute for Cancer Research, University of Oxford, Oxford, United Kingdom
| | - Richard P Owen
- Ludwig Institute for Cancer Research, University of Oxford, Oxford, United Kingdom
| | - Michael J White
- Ludwig Institute for Cancer Research, University of Oxford, Oxford, United Kingdom
| | - Jens Rittscher
- Ludwig Institute for Cancer Research, University of Oxford, Oxford, United Kingdom.,Institute of Biomedical Engineering, University of Oxford, Oxford, United Kingdom.,Big Data Institute, University of Oxford, Oxford, United Kingdom
| | - Xin Lu
- Ludwig Institute for Cancer Research, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
27
|
Krzysztoń R, Woschée D, Reiser A, Schwake G, Strey HH, Rädler JO. Single-cell kinetics of siRNA-mediated mRNA degradation. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2019; 21:102077. [PMID: 31400572 DOI: 10.1016/j.nano.2019.102077] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Revised: 07/26/2019] [Accepted: 07/29/2019] [Indexed: 12/26/2022]
Abstract
RNA interference (RNAi) enables the therapeutic use of small interfering RNAs (siRNAs) to silence disease-related genes. The efficiency of silencing is commonly assessed by measuring expression levels of the target protein at a given time point post-transfection. Here, we determine the siRNA-induced fold change in mRNA degradation kinetics from single-cell fluorescence time-courses obtained using live-cell imaging on single-cell arrays (LISCA). After simultaneous transfection of mRNAs encoding eGFP (target) and CayRFP (reference), the eGFP expression is silenced by siRNA. The single-cell time-courses are fitted using a mathematical model of gene expression. Analysis yields best estimates of related kinetic rate constants, including mRNA degradation constants. We determine the siRNA-induced changes in kinetic rates and their correlations between target and reference protein expression. Assessment of mRNA degradation constants using single-cell time-lapse imaging is fast (<30 h) and returns an accurate, time-independent measure of siRNA-induced silencing, thus allowing the exact evaluation of siRNA therapeutics.
Collapse
Affiliation(s)
- Rafał Krzysztoń
- Faculty of Physics, Ludwig-Maximilians-Universität Munich (LMU), Munich, Germany; Graduate School of Quantitative Biosciences (QBM), Ludwig-Maximilians-Universität Munich (LMU), Munich, Germany; Nano systems Initiative Munich (NIM) and Center for NanoScience (CeNS), Munich, Germany.
| | - Daniel Woschée
- Faculty of Physics, Ludwig-Maximilians-Universität Munich (LMU), Munich, Germany; Nano systems Initiative Munich (NIM) and Center for NanoScience (CeNS), Munich, Germany
| | - Anita Reiser
- Faculty of Physics, Ludwig-Maximilians-Universität Munich (LMU), Munich, Germany; Graduate School of Quantitative Biosciences (QBM), Ludwig-Maximilians-Universität Munich (LMU), Munich, Germany; Nano systems Initiative Munich (NIM) and Center for NanoScience (CeNS), Munich, Germany
| | - Gerlinde Schwake
- Faculty of Physics, Ludwig-Maximilians-Universität Munich (LMU), Munich, Germany
| | - Helmut H Strey
- Department of Biomedical Engineering and Laufer Center for Quantitative Biology, Stony Brook University, Stony Brook, NY
| | - Joachim O Rädler
- Faculty of Physics, Ludwig-Maximilians-Universität Munich (LMU), Munich, Germany; Graduate School of Quantitative Biosciences (QBM), Ludwig-Maximilians-Universität Munich (LMU), Munich, Germany; Nano systems Initiative Munich (NIM) and Center for NanoScience (CeNS), Munich, Germany.
| |
Collapse
|
28
|
Whole-Cell Multiparameter Assay for Ricin and Abrin Activity-Based Digital Holographic Microscopy. Toxins (Basel) 2019; 11:toxins11030174. [PMID: 30909438 PMCID: PMC6468687 DOI: 10.3390/toxins11030174] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 03/11/2019] [Accepted: 03/15/2019] [Indexed: 01/25/2023] Open
Abstract
Ricin and abrin are ribosome-inactivating proteins leading to inhibition of protein synthesis and cell death. These toxins are considered some of the most potent and lethal toxins against which there is no available antidote. Digital holographic microscopy (DHM) is a time-lapse, label-free, and noninvasive imaging technique that can provide phase information on morphological features of cells. In this study, we employed DHM to evaluate the morphological changes of cell lines during ricin and abrin intoxication. We showed that the effect of these toxins is characterized by a decrease in cell confluence and changes in morphological parameters such as cell area, perimeter, irregularity, and roughness. In addition, changes in optical parameters such as phase-shift, optical thickness, and effective-calculated volume were observed. These effects were completely inhibited by specific neutralizing antibodies. An enhanced intoxication effect was observed for preadherent compared to adherent cells, as was detected in early morphology changes and confirmed by annexin V/propidium iodide (PI) apoptosis assay. Detection of the dynamic changes in cell morphology at initial stages of cell intoxication by DHM emphasizes the highly sensitive and rapid nature of this method, allowing the early detection of active toxins.
Collapse
|
29
|
Zhou FY, Ruiz-Puig C, Owen RP, White MJ, Rittscher J, Lu X. Motion sensing superpixels (MOSES) is a systematic computational framework to quantify and discover cellular motion phenotypes. eLife 2019; 8:e40162. [PMID: 30803483 PMCID: PMC6391079 DOI: 10.7554/elife.40162] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Accepted: 01/11/2019] [Indexed: 12/12/2022] Open
Abstract
Correct cell/cell interactions and motion dynamics are fundamental in tissue homeostasis, and defects in these cellular processes cause diseases. Therefore, there is strong interest in identifying factors, including drug candidates that affect cell/cell interactions and motion dynamics. However, existing quantitative tools for systematically interrogating complex motion phenotypes in timelapse datasets are limited. We present Motion Sensing Superpixels (MOSES), a computational framework that measures and characterises biological motion with a unique superpixel 'mesh' formulation. Using published datasets, MOSES demonstrates single-cell tracking capability and more advanced population quantification than Particle Image Velocimetry approaches. From > 190 co-culture videos, MOSES motion-mapped the interactions between human esophageal squamous epithelial and columnar cells mimicking the esophageal squamous-columnar junction, a site where Barrett's esophagus and esophageal adenocarcinoma often arise clinically. MOSES is a powerful tool that will facilitate unbiased, systematic analysis of cellular dynamics from high-content time-lapse imaging screens with little prior knowledge and few assumptions.
Collapse
Affiliation(s)
- Felix Y Zhou
- Ludwig Institute for Cancer Research, Nuffield Department of Clinical MedicineUniversity of OxfordOxfordUnited Kingdom
| | - Carlos Ruiz-Puig
- Ludwig Institute for Cancer Research, Nuffield Department of Clinical MedicineUniversity of OxfordOxfordUnited Kingdom
| | - Richard P Owen
- Ludwig Institute for Cancer Research, Nuffield Department of Clinical MedicineUniversity of OxfordOxfordUnited Kingdom
| | - Michael J White
- Ludwig Institute for Cancer Research, Nuffield Department of Clinical MedicineUniversity of OxfordOxfordUnited Kingdom
| | - Jens Rittscher
- Ludwig Institute for Cancer Research, Nuffield Department of Clinical MedicineUniversity of OxfordOxfordUnited Kingdom
- Institute of Biomedical Engineering, Department of EngineeringUniversity of OxfordOxfordUnited Kingdom
- Big Data Institute, Li Ka Shing Centre for Health Information and DiscoveryUniversity of OxfordOxfordUnited Kingdom
| | - Xin Lu
- Ludwig Institute for Cancer Research, Nuffield Department of Clinical MedicineUniversity of OxfordOxfordUnited Kingdom
| |
Collapse
|
30
|
Starkuviene V, Kallenberger SM, Beil N, Lisauskas T, Schumacher BSS, Bulkescher R, Wajda P, Gunkel M, Beneke J, Erfle H. High-Density Cell Arrays for Genome-Scale Phenotypic Screening. SLAS DISCOVERY 2019; 24:274-283. [PMID: 30682322 DOI: 10.1177/2472555218818757] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Due to high associated costs and considerable time investments of cell-based screening, there is a strong demand for new technologies that enable preclinical development and tests of diverse biologicals in a cost-saving and time-efficient manner. For those reasons we developed the high-density cell array (HD-CA) platform, which miniaturizes cell-based screening in the form of preprinted and ready-to-run screening arrays. With the HD-CA technology, up to 24,576 samples can be tested in a single experiment, thereby saving costs and time for microscopy-based screening by 75%. Experiments on the scale of the entire human genome can be addressed in a real parallel manner, with screening campaigns becoming more comfortable and devoid of robotics infrastructure on the user side. The high degree of miniaturization enables working with expensive reagents and rare and difficult-to-obtain cell lines. We have also optimized an automated imaging procedure for HD-CA and demonstrate the applicability of HD-CA to CRISPR-Cas9- and RNAi-mediated phenotypic assessment of the gene function.
Collapse
Affiliation(s)
- Vytaute Starkuviene
- 1 BioQuant, Heidelberg University, Heidelberg, Germany.,2 Institute of Biosciences, Vilnius University Life Sciences Center, Vilnius, Lithuania
| | - Stefan M Kallenberger
- 1 BioQuant, Heidelberg University, Heidelberg, Germany.,3 Division of Theoretical Bioinformatics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Nina Beil
- 1 BioQuant, Heidelberg University, Heidelberg, Germany
| | | | | | | | - Piotr Wajda
- 1 BioQuant, Heidelberg University, Heidelberg, Germany
| | - Manuel Gunkel
- 1 BioQuant, Heidelberg University, Heidelberg, Germany
| | - Jürgen Beneke
- 1 BioQuant, Heidelberg University, Heidelberg, Germany
| | - Holger Erfle
- 1 BioQuant, Heidelberg University, Heidelberg, Germany
| |
Collapse
|
31
|
Ford K, McDonald D, Mali P. Functional Genomics via CRISPR-Cas. J Mol Biol 2019; 431:48-65. [PMID: 29959923 PMCID: PMC6309720 DOI: 10.1016/j.jmb.2018.06.034] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 06/02/2018] [Accepted: 06/14/2018] [Indexed: 12/22/2022]
Abstract
RNA-guided CRISPR (clustered regularly interspaced short palindromic repeat)-associated Cas proteins have recently emerged as versatile tools to investigate and engineer the genome. The programmability of CRISPR-Cas has proven especially useful for probing genomic function in high-throughput. Facile single-guide RNA library synthesis allows CRISPR-Cas screening to rapidly investigate the functional consequences of genomic, transcriptomic, and epigenomic perturbations. Furthermore, by combining CRISPR-Cas perturbations with downstream single-cell analyses (flow cytometry, expression profiling, etc.), forward screens can generate robust data sets linking genotypes to complex cellular phenotypes. In the following review, we highlight recent advances in CRISPR-Cas genomic screening while outlining protocols and pitfalls associated with screen implementation. Finally, we describe current challenges limiting the utility of CRISPR-Cas screening as well as future research needed to resolve these impediments. As CRISPR-Cas technologies develop, so too will their clinical applications. Looking ahead, patient centric functional screening in primary cells will likely play a greater role in disease management and therapeutic development.
Collapse
Affiliation(s)
- Kyle Ford
- Department of Bioengineering, University of California, San Diego, San Diego, CA 92093, USA
| | - Daniella McDonald
- Biomedical Sciences Graduate Program, University of California, San Diego, San Diego, CA 92093, USA
| | - Prashant Mali
- Department of Bioengineering, University of California, San Diego, San Diego, CA 92093, USA.
| |
Collapse
|
32
|
Zou F, Bai L. Using time-lapse fluorescence microscopy to study gene regulation. Methods 2018; 159-160:138-145. [PMID: 30599195 DOI: 10.1016/j.ymeth.2018.12.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2018] [Revised: 12/20/2018] [Accepted: 12/27/2018] [Indexed: 12/20/2022] Open
Abstract
Time-lapse fluorescence microscopy is a powerful tool to study gene regulation. By probing fluorescent signals in single cells over extended period of time, this method can be used to study the dynamics, noise, movement, memory, inheritance, and coordination, of gene expression during cell growth, development, and differentiation. In combination with a flow-cell device, it can also measure gene regulation by external stimuli. Due to the single cell nature and the spatial/temporal capacity, this method can often provide information that is hard to get using other methods. Here, we review the standard experimental procedures and new technical developments in this field.
Collapse
Affiliation(s)
- Fan Zou
- Department of Physics, The Pennsylvania State University, University Park, PA 16802, United States; Center for Eukaryotic Gene Regulation, The Pennsylvania State University, University Park, PA 16802, United States
| | - Lu Bai
- Department of Physics, The Pennsylvania State University, University Park, PA 16802, United States; Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, PA 16802, United States; Center for Eukaryotic Gene Regulation, The Pennsylvania State University, University Park, PA 16802, United States.
| |
Collapse
|
33
|
Matsui TS, Wu H, Deguchi S. Deformable 96-well cell culture plate compatible with high-throughput screening platforms. PLoS One 2018; 13:e0203448. [PMID: 30188938 PMCID: PMC6126838 DOI: 10.1371/journal.pone.0203448] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Accepted: 07/03/2018] [Indexed: 12/22/2022] Open
Abstract
Adherent cells such as endothelial cells sense applied mechanical stretch to adapt to changes in their surrounding mechanical environment. Despite numerous studies, signaling pathways underlying the cellular mechanosensing and adaptation remain to be fully elucidated partly because of the lack of tools that allow for a comprehensive screening approach. Conventionally, multi-well cell culture plates of standard configurations are used for comprehensive analyses in cell biology study to identify key molecules in a high-throughput manner. Given that situation, here we design a 96-well cell culture plate made of elastic silicone and mechanically stretchable using a motorized device. Computational analysis suggested that highly uniform stretch can be applied to each of the wells other than the peripheral wells. Elastic image registration-based experimental evaluation on stretch distributions within individual wells revealed the presence of larger variations among wells compared to those in the computational analysis, but a stretch level of 10%–that has been employed in conventional studies on cellular response to stretch—was almost achieved with our setup. We exposed vascular smooth muscle cells to cyclic stretch using the device to demonstrate morphological repolarization of the cells, i.e. typical cellular response to cyclic stretch. Because the deformable multi-well plate validated here is compatible with other high-throughput screening-oriented technologies, we expect this novel system to be utilized for future comprehensive analyses of stretch-related signaling pathways.
Collapse
Affiliation(s)
- Tsubasa S Matsui
- Division of Bioengineering, Graduate School of Engineering Science, Osaka University, Toyonaka, Osaka, Japan.,Department of Nanopharmaceutical Sciences, Nagoya Institute of Technology, Nagoya, Aichi, Japan
| | - Hugejile Wu
- Department of Nanopharmaceutical Sciences, Nagoya Institute of Technology, Nagoya, Aichi, Japan
| | - Shinji Deguchi
- Division of Bioengineering, Graduate School of Engineering Science, Osaka University, Toyonaka, Osaka, Japan.,Department of Nanopharmaceutical Sciences, Nagoya Institute of Technology, Nagoya, Aichi, Japan
| |
Collapse
|
34
|
Scheeder C, Heigwer F, Boutros M. Machine learning and image-based profiling in drug discovery. CURRENT OPINION IN SYSTEMS BIOLOGY 2018; 10:43-52. [PMID: 30159406 PMCID: PMC6109111 DOI: 10.1016/j.coisb.2018.05.004] [Citation(s) in RCA: 94] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The increase in imaging throughput, new analytical frameworks and high-performance computational resources open new avenues for data-rich phenotypic profiling of small molecules in drug discovery. Image-based profiling assays assessing single-cell phenotypes have been used to explore mechanisms of action, target efficacy and toxicity of small molecules. Technological advances to generate large data sets together with new machine learning approaches for the analysis of high-dimensional profiling data create opportunities to improve many steps in drug discovery. In this review, we will discuss how recent studies applied machine learning approaches in functional profiling workflows with a focus on chemical genetics. While their utility in image-based screening and profiling is predictably evident, examples of novel insights beyond the status quo based on the applications of machine learning approaches are just beginning to emerge. To enable discoveries, future studies also need to develop methodologies that lower the entry barriers to high-throughput profiling experiments by streamlining image-based profiling assays and providing applications for advanced learning technologies such as easy to deploy deep neural networks.
Collapse
Affiliation(s)
| | | | - Michael Boutros
- German Cancer Research Center (DKFZ), Division Signaling and Functional Genomics and Heidelberg University, Department of Cell and Molecular Biology, Medical Faculty Mannheim, D-69120 Heidelberg, Germany
| |
Collapse
|
35
|
Sinha H, Quach ABV, Vo PQN, Shih SCC. An automated microfluidic gene-editing platform for deciphering cancer genes. LAB ON A CHIP 2018; 18:2300-2312. [PMID: 29989627 DOI: 10.1039/c8lc00470f] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/09/2023]
Abstract
Gene-editing techniques such as RNA-guided endonuclease systems are becoming increasingly popular for phenotypic screening. Such screens are normally conducted in arrayed or pooled formats. There has been considerable interest in recent years to find new technological methods for conducting these gene-editing assays. We report here the first digital microfluidic method that can automate arrayed gene-editing in mammalian cells. Specifically, this method was useful in culturing lung cancer cells for up to six days, as well as implementing automated gene transfection and knockout procedures. In addition, a standardized imaging pipeline to analyse fluorescently labelled cells was also designed and implemented during these procedures. A gene editing assay for interrogating the MAPK/ERK pathway was performed to show the utility of our platform and to determine the effects of knocking out the RAF1 gene in lung cancer cells. In addition to gene knockout, we also treated the cells with an inhibitor, Sorafenib Tosylate, to determine the effects of enzymatic inhibition. The combination of enzymatic inhibition and guide targeting on device resulted in lower drug concentrations for achieving half-inhibitory effects (IC50) compared to cells treated only with the inhibitor, confirming that lung cancer cells are being successfully edited on the device. We propose that this system will be useful for other types of gene-editing assays and applications related to personalized medicine.
Collapse
Affiliation(s)
- Hugo Sinha
- Department of Electrical and Computer Engineering, Concordia University, Montréal, Québec, Canada.
| | | | | | | |
Collapse
|
36
|
Stojic L, Lun AT, Mangei J, Mascalchi P, Quarantotti V, Barr AR, Bakal C, Marioni JC, Gergely F, Odom DT. Specificity of RNAi, LNA and CRISPRi as loss-of-function methods in transcriptional analysis. Nucleic Acids Res 2018; 46:5950-5966. [PMID: 29860520 PMCID: PMC6093183 DOI: 10.1093/nar/gky437] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Revised: 05/03/2018] [Accepted: 05/09/2018] [Indexed: 02/06/2023] Open
Abstract
Loss-of-function (LOF) methods such as RNA interference (RNAi), antisense oligonucleotides or CRISPR-based genome editing provide unparalleled power for studying the biological function of genes of interest. However, a major concern is non-specific targeting, which involves depletion of transcripts other than those intended. Little work has been performed to characterize the off-target effects of these common LOF methods at the whole-transcriptome level. Here, we experimentally compared the non-specific activity of RNAi, antisense oligonucleotides and CRISPR interference (CRISPRi). All three methods yielded non-negligible off-target effects in gene expression, with CRISPRi also exhibiting strong clonal effects. As an illustrative example, we evaluated the performance of each method for determining the role of an uncharacterized long noncoding RNA (lncRNA). Several LOF methods successfully depleted the candidate lncRNA but yielded different sets of differentially expressed genes as well as a different cellular phenotype upon depletion. Similar discrepancies between methods were observed with a protein-coding gene (Ch-TOG/CKAP5) and another lncRNA (MALAT1). We suggest that the differences between methods arise due to method-specific off-target effects and provide guidelines for mitigating such effects in functional studies. Our recommendations provide a framework with which off-target effects can be managed to improve functional characterization of genes of interest.
Collapse
Affiliation(s)
- Lovorka Stojic
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge CB2 0RE, UK
| | - Aaron T L Lun
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge CB2 0RE, UK
| | - Jasmin Mangei
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge CB2 0RE, UK
| | - Patrice Mascalchi
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge CB2 0RE, UK
| | - Valentina Quarantotti
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge CB2 0RE, UK
| | - Alexis R Barr
- Institute of Cancer Research, 237 Fulham Road London SW3 6JB, UK
| | - Chris Bakal
- Institute of Cancer Research, 237 Fulham Road London SW3 6JB, UK
| | - John C Marioni
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge CB2 0RE, UK
- European Bioinformatics Institute, European Molecular Biology Laboratory (EMBL-EBI), Wellcome Genome Campus, Hinxton, Cambridgeshire CB10 1SD, UK
- Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridgeshire CB10 1SA, UK
| | - Fanni Gergely
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge CB2 0RE, UK
| | - Duncan T Odom
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge CB2 0RE, UK
| |
Collapse
|
37
|
Rosas-Salvans M, Cavazza T, Espadas G, Sabido E, Vernos I. Proteomic Profiling of Microtubule Self-organization in M-phase. Mol Cell Proteomics 2018; 17:1991-2004. [PMID: 29970457 DOI: 10.1074/mcp.ra118.000745] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Revised: 06/11/2018] [Indexed: 01/08/2023] Open
Abstract
Microtubules (MTs) and associated proteins can self-organize into complex structures such as the bipolar spindle, a process in which RanGTP plays a major role. Addition of RanGTP to M-phase Xenopus egg extracts promotes the nucleation and self-organization of MTs into asters and bipolar-like structures in the absence of centrosomes or chromosomes. We show here that the complex proteome of these RanGTP-induced MT assemblies is similar to that of mitotic spindles. Using proteomic profiling we show that MT self-organization in the M-phase cytoplasm involves the non-linear and non-stoichiometric recruitment of proteins from specific functional groups. Our study provides for the first time a temporal understanding of the protein dynamics driving MT self-organization in M-phase.
Collapse
Affiliation(s)
- Miquel Rosas-Salvans
- From the ‡Cell and Developmental Biology Programme, Centre for Genomic Regulation, Barcelona Institute of Science and Technology, Dr. Aiguader 88, 08003 Barcelona, Spain
| | - Tommaso Cavazza
- From the ‡Cell and Developmental Biology Programme, Centre for Genomic Regulation, Barcelona Institute of Science and Technology, Dr. Aiguader 88, 08003 Barcelona, Spain
| | - Guadalupe Espadas
- **Proteomics Unit, Centre for Genomic Regulation (CRG), The Barcelona Institute for Science and Technology, Dr. Aiguader 88, 08003 Barcelona, Spain.,§Universitat Pompeu Fabra, Dr. Aiguader 88, 08003 Barcelona, Spain
| | - Eduard Sabido
- **Proteomics Unit, Centre for Genomic Regulation (CRG), The Barcelona Institute for Science and Technology, Dr. Aiguader 88, 08003 Barcelona, Spain.,§Universitat Pompeu Fabra, Dr. Aiguader 88, 08003 Barcelona, Spain
| | - Isabelle Vernos
- From the ‡Cell and Developmental Biology Programme, Centre for Genomic Regulation, Barcelona Institute of Science and Technology, Dr. Aiguader 88, 08003 Barcelona, Spain; .,§Universitat Pompeu Fabra, Dr. Aiguader 88, 08003 Barcelona, Spain.,‡‡Institució Catalana de Recerca I Estudis Avançats (ICREA), Passeig de Lluis Companys 23, 08010 Barcelona, Spain
| |
Collapse
|
38
|
Becker AK, Erfle H, Gunkel M, Beil N, Kaderali L, Starkuviene V. Comparison of Cell Arrays and Multi-Well Plates in Microscopy-Based Screening. High Throughput 2018; 7:ht7020013. [PMID: 29762489 PMCID: PMC6023461 DOI: 10.3390/ht7020013] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Revised: 05/08/2018] [Accepted: 05/09/2018] [Indexed: 01/30/2023] Open
Abstract
Multi-well plates and cell arrays enable microscopy-based screening assays in which many samples can be analysed in parallel. Each of the formats possesses its own strengths and weaknesses, but reference comparisons between these platforms and their application rationale is lacking. We aim to fill this gap by comparing two RNA interference (RNAi)-mediated fluorescence microscopy-based assays, namely epidermal growth factor (EGF) internalization and cell cycle progression, on both platforms. Quantitative analysis revealed that both platforms enabled the generation of data with the appearance of the expected phenotypes significantly distinct from the negative controls. The measurements of cell cycle progression were less variable in multi-well plates. The result can largely be attributed to higher cell numbers resulting in less data variability when dealing with the assay generating phenotypic cell subpopulations. The EGF internalization assay with a uniform phenotype over nearly the whole cell population performed better on cell arrays than in multi-well plates. The result was achieved by scoring five times less cells on cell arrays than in multi-well plates, indicating the efficiency of the cell array format. Our data indicate that the choice of the screening platform primarily depends on the type of the cellular assay to achieve a maximum data quality and screen efficiency.
Collapse
Affiliation(s)
- Ann-Kristin Becker
- Institute of Bioinformatics, University Medicine Greifswald, 17475 Greifswald, Germany.
| | - Holger Erfle
- BioQuant, Heidelberg University, 69120 Heidelberg, Germany.
| | - Manuel Gunkel
- BioQuant, Heidelberg University, 69120 Heidelberg, Germany.
| | - Nina Beil
- BioQuant, Heidelberg University, 69120 Heidelberg, Germany.
| | - Lars Kaderali
- Institute of Bioinformatics, University Medicine Greifswald, 17475 Greifswald, Germany.
| | - Vytaute Starkuviene
- BioQuant, Heidelberg University, 69120 Heidelberg, Germany.
- Institute of Biosciences, Vilnius University Life Sciences Center, LT-10257 Vilnius, Lithuania.
| |
Collapse
|
39
|
McKinley KL. Employing CRISPR/Cas9 genome engineering to dissect the molecular requirements for mitosis. Methods Cell Biol 2018; 144:75-105. [PMID: 29804684 DOI: 10.1016/bs.mcb.2018.03.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/05/2023]
Abstract
The faithful execution of cell division requires the coordinated action of hundreds of gene products. Precisely perturbing these gene products in cells is central to understanding their functions during normal cell division, and the contributions of their disruption to disease. Here, we describe experimental approaches for using CRISPR/Cas9 for gene disruption and modification, with a focus on human cell culture. We describe strategies for inducible gene disruption to generate acute knockouts of essential cell division genes, which can be modified for the chronic elimination of nonessential genes. We also describe strategies for modifying the genome to generate protein fusions to report on and modify protein behavior. These tools facilitate investigation of protein function, dissection of protein assembly networks, and analyses of disease-associated mutations.
Collapse
Affiliation(s)
- Kara L McKinley
- Department of Cellular and Molecular Pharmacology, University of California San Francisco, San Francisco, CA, United States.
| |
Collapse
|
40
|
Hasan MR, Hassan N, Khan R, Kim YT, Iqbal SM. Classification of cancer cells using computational analysis of dynamic morphology. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2018; 156:105-112. [PMID: 29428061 DOI: 10.1016/j.cmpb.2017.12.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Revised: 11/09/2017] [Accepted: 12/05/2017] [Indexed: 06/08/2023]
Abstract
BACKGROUND AND OBJECTIVE Detection of metastatic tumor cells is important for early diagnosis and staging of cancer. However, such cells are exceedingly difficult to detect from blood or biopsy samples at the disease onset. It is reported that cancer cells, and especially metastatic tumor cells, show very distinctive morphological behavior compared to their healthy counterparts on aptamer functionalized substrates. The ability to quickly analyze the data and quantify the cell morphology for an instant real-time feedback can certainly contribute to early cancer diagnosis. A supervised machine learning approach is presented for identification and classification of cancer cell gestures for early diagnosis. METHODS We quantified the morphologically distinct behavior of metastatic cells and their healthy counterparts captured on aptamer-functionalized glass substrates from time-lapse optical micrographs. As a proof of concept, the morphologies of human glioblastoma (hGBM) and astrocyte cells were used. The cells were captured and imaged with an optical microscope. Multiple feature vectors were extracted to quantify and differentiate the complex physical gestures of cancerous and non-cancerous cells. Three different classifier models, Support Vector Machine (SVM), Random Forest Tree (RFT), and Naïve Bayes Classifier (NBC) were trained with the known dataset using machine learning algorithms. The performances of the classifiers were compared for accuracy, precision, and recall measurements using five-fold cross-validation technique. RESULTS All the classifier models detected the cancer cells with an average accuracy of at least 82%. The NBC performed the best among the three classifiers in terms of Precision (0.91), Recall (0.9), and F1-score (0.89) for the existing dataset. CONCLUSIONS This paper presents a standalone system built on machine learning techniques for cancer screening based on cell gestures. The system offers rapid, efficient, and novel identification of hGBM brain tumor cells and can be extended to define single cell analysis metrics for many other types of tumor cells.
Collapse
Affiliation(s)
- Mohammad R Hasan
- Nano-Bio Lab, University of Texas at Arlington, Arlington, TX 76019, USA; Nanotechnology Research Center, University of Texas at Arlington, Arlington, TX 76019, USA; Department of Electrical Engineering, University of Texas at Arlington, Arlington, TX 76019, USA
| | - Naeemul Hassan
- Department of Computer and Information Science, University of Mississippi, University, MS 38677, USA
| | - Rayan Khan
- Nano-Bio Lab, University of Texas at Arlington, Arlington, TX 76019, USA; Nanotechnology Research Center, University of Texas at Arlington, Arlington, TX 76019, USA; Department of Electrical Engineering, University of Texas at Arlington, Arlington, TX 76019, USA
| | - Young-Tae Kim
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX 76019, USA; Department of Urology, University of Texas Southwestern Medical Center at Dallas, Dallas, TX 75235, USA
| | - Samir M Iqbal
- Nano-Bio Lab, University of Texas at Arlington, Arlington, TX 76019, USA; Department of Electrical Engineering, University of Texas Rio Grande Valley, Edinburg, TX 78539, USA; School of Medicine, University of Texas Rio Grande Valley, Edinburg, TX 78539, USA.
| |
Collapse
|
41
|
Abstract
Applying the right acquisition method in a fluorescence imaging-based screening context is of great importance to obtain an appropriate readout and to select the right scale of the screen. In order to save imaging time and data, we have developed routines for multiscale targeted imaging, providing both a broad overview of a sample and additional in-depth information for targets of interest identified within the screen. These objects can be identified and acquired on-the-fly by an interconnection of image acquisition and image analysis.
Collapse
|
42
|
Bun P, Dmitrieff S, Belmonte JM, Nédélec FJ, Lénárt P. A disassembly-driven mechanism explains F-actin-mediated chromosome transport in starfish oocytes. eLife 2018; 7:31469. [PMID: 29350616 PMCID: PMC5788506 DOI: 10.7554/elife.31469] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Accepted: 01/18/2018] [Indexed: 12/12/2022] Open
Abstract
While contraction of sarcomeric actomyosin assemblies is well understood, this is not the case for disordered networks of actin filaments (F-actin) driving diverse essential processes in animal cells. For example, at the onset of meiosis in starfish oocytes a contractile F-actin network forms in the nuclear region transporting embedded chromosomes to the assembling microtubule spindle. Here, we addressed the mechanism driving contraction of this 3D disordered F-actin network by comparing quantitative observations to computational models. We analyzed 3D chromosome trajectories and imaged filament dynamics to monitor network behavior under various physical and chemical perturbations. We found no evidence of myosin activity driving network contractility. Instead, our observations are well explained by models based on a disassembly-driven contractile mechanism. We reconstitute this disassembly-based contractile system in silico revealing a simple architecture that robustly drives chromosome transport to prevent aneuploidy in the large oocyte, a prerequisite for normal embryonic development.
Collapse
Affiliation(s)
- Philippe Bun
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Serge Dmitrieff
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Julio M Belmonte
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - François J Nédélec
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Péter Lénárt
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| |
Collapse
|
43
|
Sharif NA. iDrugs and iDevices Discovery Research: Preclinical Assays, Techniques, and Animal Model Studies for Ocular Hypotensives and Neuroprotectants. J Ocul Pharmacol Ther 2018; 34:7-39. [PMID: 29323613 DOI: 10.1089/jop.2017.0125] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Discovery ophthalmic research is centered around delineating the molecular and cellular basis of ocular diseases and finding and exploiting molecular and genetic pathways associated with them. From such studies it is possible to determine suitable intervention points to address the disease process and hopefully to discover therapeutics to treat them. An investigational new drug (IND) filing for a new small-molecule drug, peptide, antibody, genetic treatment, or a device with global health authorities requires a number of preclinical studies to provide necessary safety and efficacy data. Specific regulatory elements needed for such IND-enabling studies are beyond the scope of this article. However, to enhance the overall data packages for such entities and permit high-quality foundation-building publications for medical affairs, additional research and development studies are always desirable. This review aims to provide examples of some target localization/verification, ocular drug discovery processes, and mechanistic and portfolio-enhancing exploratory investigations for candidate drugs and devices for the treatment of ocular hypertension and glaucomatous optic neuropathy (neurodegeneration of retinal ganglion cells and their axons). Examples of compound screening assays, use of various technologies and techniques, deployment of animal models, and data obtained from such studies are also presented.
Collapse
Affiliation(s)
- Najam A Sharif
- 1 Global Alliances & External Research , Santen Incorporated, Emeryville, California.,2 Department of Pharmaceutical Sciences, Texas Southern University , Houston, Texas.,3 Department of Pharmacology and Neuroscience, University of North Texas Health Sciences Center , Fort Worth, Texas
| |
Collapse
|
44
|
Lérias JR, Pinto MC, Botelho HM, Awatade NT, Quaresma MC, Silva IAL, Wanitchakool P, Schreiber R, Pepperkok R, Kunzelmann K, Amaral MD. A novel microscopy-based assay identifies extended synaptotagmin-1 (ESYT1) as a positive regulator of anoctamin 1 traffic. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2017; 1865:421-431. [PMID: 29154949 DOI: 10.1016/j.bbamcr.2017.11.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Revised: 11/02/2017] [Accepted: 11/14/2017] [Indexed: 01/14/2023]
Abstract
An attractive possibility to treat Cystic Fibrosis (CF), a severe condition caused by dysfunctional CFTR, an epithelial anion channel, is through the activation of alternative (non-CFTR) anion channels. Anoctamin 1 (ANO1) was demonstrated to be a Ca2+-activated chloride channel (CaCC) and thus of high potential to replace CFTR. Despite that ANO1 is expressed in human lung CF tissue, it is present at the cell surface at very low levels. In addition, little is known about regulation of ANO1 traffic, namely which factors promote its plasma membrane (PM) localization. Here, we generated a novel cellular model, expressing an inducible 3HA-ANO1-eGFP construct, and validated its usage as a microscopy tool to monitor for ANO1 traffic. We demonstrate the robustness and specificity of this cell-based assay, by the identification of siRNAs acting both as ANO1 traffic enhancer and inhibitor, targeting respectively COPB1 and ESYT1 (extended synaptotagmin-1), the latter involved in coupling of the endoplasmic reticulum to the PM at specific microdomains. We further show that knockdown of ESYT1 (and family members ESYT2 and ESYT3) significantly decreased ANO1 current density. This ANO1 cell-based assay constitutes an important tool to be further used in high-throughput screens and drug discovery of high relevance for CF and cancer.
Collapse
Affiliation(s)
- Joana R Lérias
- University of Lisboa, Faculty of Sciences, BioISI - Biosystems & Integrative Sciences Institute, Campo Grande, C8, 1749-016 Lisboa, Portugal; Department of Physiology, University of Regensburg, Universitätsstrasse 31, 93053 Regensburg, Germany
| | - Madalena C Pinto
- University of Lisboa, Faculty of Sciences, BioISI - Biosystems & Integrative Sciences Institute, Campo Grande, C8, 1749-016 Lisboa, Portugal; Department of Physiology, University of Regensburg, Universitätsstrasse 31, 93053 Regensburg, Germany; Cell Biology and Biophysics Unit and Advanced Light Microscopy Facility, European Molecular Biology Laboratory (EMBL), Meyerhofstraße 1, 69117 Heidelberg, Germany
| | - Hugo M Botelho
- University of Lisboa, Faculty of Sciences, BioISI - Biosystems & Integrative Sciences Institute, Campo Grande, C8, 1749-016 Lisboa, Portugal
| | - Nikhil T Awatade
- University of Lisboa, Faculty of Sciences, BioISI - Biosystems & Integrative Sciences Institute, Campo Grande, C8, 1749-016 Lisboa, Portugal
| | - Margarida C Quaresma
- University of Lisboa, Faculty of Sciences, BioISI - Biosystems & Integrative Sciences Institute, Campo Grande, C8, 1749-016 Lisboa, Portugal
| | - Iris A L Silva
- University of Lisboa, Faculty of Sciences, BioISI - Biosystems & Integrative Sciences Institute, Campo Grande, C8, 1749-016 Lisboa, Portugal
| | - Podchanart Wanitchakool
- Department of Physiology, University of Regensburg, Universitätsstrasse 31, 93053 Regensburg, Germany
| | - Rainer Schreiber
- Department of Physiology, University of Regensburg, Universitätsstrasse 31, 93053 Regensburg, Germany
| | - Rainer Pepperkok
- Cell Biology and Biophysics Unit and Advanced Light Microscopy Facility, European Molecular Biology Laboratory (EMBL), Meyerhofstraße 1, 69117 Heidelberg, Germany
| | - Karl Kunzelmann
- Department of Physiology, University of Regensburg, Universitätsstrasse 31, 93053 Regensburg, Germany
| | - Margarida D Amaral
- University of Lisboa, Faculty of Sciences, BioISI - Biosystems & Integrative Sciences Institute, Campo Grande, C8, 1749-016 Lisboa, Portugal; Cell Biology and Biophysics Unit and Advanced Light Microscopy Facility, European Molecular Biology Laboratory (EMBL), Meyerhofstraße 1, 69117 Heidelberg, Germany.
| |
Collapse
|
45
|
Sommer C, Hoefler R, Samwer M, Gerlich DW. A deep learning and novelty detection framework for rapid phenotyping in high-content screening. Mol Biol Cell 2017; 28:3428-3436. [PMID: 28954863 PMCID: PMC5687041 DOI: 10.1091/mbc.e17-05-0333] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 08/31/2017] [Accepted: 09/18/2017] [Indexed: 11/16/2022] Open
Abstract
Supervised machine learning is a powerful and widely used method for analyzing high-content screening data. Despite its accuracy, efficiency, and versatility, supervised machine learning has drawbacks, most notably its dependence on a priori knowledge of expected phenotypes and time-consuming classifier training. We provide a solution to these limitations with CellCognition Explorer, a generic novelty detection and deep learning framework. Application to several large-scale screening data sets on nuclear and mitotic cell morphologies demonstrates that CellCognition Explorer enables discovery of rare phenotypes without user training, which has broad implications for improved assay development in high-content screening.
Collapse
Affiliation(s)
- Christoph Sommer
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna Biocenter (VBC), 1030 Vienna, Austria
| | - Rudolf Hoefler
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna Biocenter (VBC), 1030 Vienna, Austria
| | - Matthias Samwer
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna Biocenter (VBC), 1030 Vienna, Austria
| | - Daniel W Gerlich
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna Biocenter (VBC), 1030 Vienna, Austria
| |
Collapse
|
46
|
Localization-based super-resolution imaging meets high-content screening. Nat Methods 2017; 14:1184-1190. [PMID: 29083400 DOI: 10.1038/nmeth.4486] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Accepted: 09/26/2017] [Indexed: 11/08/2022]
Abstract
Single-molecule localization microscopy techniques have proven to be essential tools for quantitatively monitoring biological processes at unprecedented spatial resolution. However, these techniques are very low throughput and are not yet compatible with fully automated, multiparametric cellular assays. This shortcoming is primarily due to the huge amount of data generated during imaging and the lack of software for automation and dedicated data mining. We describe an automated quantitative single-molecule-based super-resolution methodology that operates in standard multiwell plates and uses analysis based on high-content screening and data-mining software. The workflow is compatible with fixed- and live-cell imaging and allows extraction of quantitative data like fluorophore photophysics, protein clustering or dynamic behavior of biomolecules. We demonstrate that the method is compatible with high-content screening using 3D dSTORM and DNA-PAINT based super-resolution microscopy as well as single-particle tracking.
Collapse
|
47
|
Burri O, Wolf B, Seitz A, Gönczy P. TRACMIT: An effective pipeline for tracking and analyzing cells on micropatterns through mitosis. PLoS One 2017; 12:e0179752. [PMID: 28746386 PMCID: PMC5528263 DOI: 10.1371/journal.pone.0179752] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2017] [Accepted: 06/02/2017] [Indexed: 01/01/2023] Open
Abstract
The use of micropatterns has transformed investigations of dynamic biological processes by enabling the reproducible analysis of live cells using time-lapse fluorescence microscopy. With micropatterns, thousands of individual cells can be efficiently imaged in parallel, rendering the approach well suited for screening projects. Despite being powerful, such screens remain challenging in terms of data handling and analysis. Typically, only a fraction of micropatterns is occupied in a manner suitable to monitor a given phenotypic output. Moreover, the presence of dying or otherwise compromised cells complicates the analysis. Therefore, focusing strictly on relevant cells in such large time-lapse microscopy dataset poses interesting analysis challenges that are not readily met by existing software packages. This motivated us to develop an image analysis pipeline that handles all necessary image processing steps within one open-source platform to detect and analyze individual cells seeded on micropatterns through mitosis. We introduce a comprehensive image analysis pipeline running on Fiji termed TRACMIT (pipeline for TRACking and analyzing cells on micropatterns through MITosis). TRACMIT was developed to rapidly and accurately assess the orientation of the mitotic spindle during metaphase in time-lapse fluorescence microscopy of human cells expressing mCherry::histone 2B and plated on L-shaped micropatterns. This solution enables one to perform the entire analysis from the raw data, avoiding the need to save intermediate images, thereby decreasing data volume and thus reducing the data that needs to be processed. We first select micropatterns containing a single cell and then identify anaphase figures in the time-lapse recording. Next, TRACMIT tracks back in time until metaphase, when the angle of the mitotic spindle with respect to the micropattern is assessed. We designed the pipeline to allow for manual validation of selected cells with a simple user interface, and to enable analysis of cells plated on micropatterns of different shapes. For ease of use, the entire pipeline is provided as a series of Fiji/ImageJ macros, grouped into an ActionBar. In conclusion, the open source TRACMIT pipeline enables high-throughput analysis of single mitotic cells on micropatterns, thus accurately and efficiently allowing automatic determination of spindle positioning from time-lapse recordings.
Collapse
Affiliation(s)
- Olivier Burri
- Bioimaging and Optics Platform (BIOP), School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland
| | - Benita Wolf
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland
| | - Arne Seitz
- Bioimaging and Optics Platform (BIOP), School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland
| | - Pierre Gönczy
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland
| |
Collapse
|
48
|
Pegoraro G, Misteli T. High-Throughput Imaging for the Discovery of Cellular Mechanisms of Disease. Trends Genet 2017; 33:604-615. [PMID: 28732598 DOI: 10.1016/j.tig.2017.06.005] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Revised: 06/19/2017] [Accepted: 06/20/2017] [Indexed: 12/23/2022]
Abstract
High-throughput imaging (HTI) is a powerful tool in the discovery of cellular disease mechanisms. While traditional approaches to identify disease pathways often rely on knowledge of the causative genetic defect, HTI-based screens offer an unbiased discovery approach based on any morphological or functional defects of disease cells or tissues. In this review, we provide an overview of the use of HTI for the study of human disease mechanisms. We discuss key technical aspects of HTI and highlight representative examples of its practical applications for the discovery of molecular mechanisms of disease, focusing on infectious diseases and host-pathogen interactions, cancer, and rare genetic diseases. We also present some of the current challenges and possible solutions offered by novel cell culture systems and genome engineering approaches.
Collapse
Affiliation(s)
- Gianluca Pegoraro
- NCI High-Throughput Imaging Facility, Bethesda, MD 20892, USA; Center for Cancer Research, National Cancer Institute/NIH, Bethesda, MD 20892, USA.
| | - Tom Misteli
- Center for Cancer Research, National Cancer Institute/NIH, Bethesda, MD 20892, USA.
| |
Collapse
|
49
|
Fetz V, Prochnow H, Brönstrup M, Sasse F. Target identification by image analysis. Nat Prod Rep 2017; 33:655-67. [PMID: 26777141 DOI: 10.1039/c5np00113g] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Covering: 1997 to the end of 2015Each biologically active compound induces phenotypic changes in target cells that are characteristic for its mode of action. These phenotypic alterations can be directly observed under the microscope or made visible by labelling structural elements or selected proteins of the cells with dyes. A comparison of the cellular phenotype induced by a compound of interest with the phenotypes of reference compounds with known cellular targets allows predicting its mode of action. While this approach has been successfully applied to the characterization of natural products based on a visual inspection of images, recent studies used automated microscopy and analysis software to increase speed and to reduce subjective interpretation. In this review, we give a general outline of the workflow for manual and automated image analysis, and we highlight natural products whose bacterial and eucaryotic targets could be identified through such approaches.
Collapse
Affiliation(s)
- V Fetz
- Helmholtz Centre for Infection Research, Department of Chemical Biology, Inhoffenstrasse 7, D-38124 Braunschweig, Germany. and German Centre for Infection Research (DZIF), Partner Site Hannover-Braunschweig, Germany and School of Engineering and Science, Jacobs University Bremen, Germany
| | - H Prochnow
- Helmholtz Centre for Infection Research, Department of Chemical Biology, Inhoffenstrasse 7, D-38124 Braunschweig, Germany.
| | - M Brönstrup
- Helmholtz Centre for Infection Research, Department of Chemical Biology, Inhoffenstrasse 7, D-38124 Braunschweig, Germany. and German Centre for Infection Research (DZIF), Partner Site Hannover-Braunschweig, Germany
| | - F Sasse
- Helmholtz Centre for Infection Research, Department of Chemical Biology, Inhoffenstrasse 7, D-38124 Braunschweig, Germany.
| |
Collapse
|
50
|
Rostam HM, Reynolds PM, Alexander MR, Gadegaard N, Ghaemmaghami AM. Image based Machine Learning for identification of macrophage subsets. Sci Rep 2017; 7:3521. [PMID: 28615717 PMCID: PMC5471192 DOI: 10.1038/s41598-017-03780-z] [Citation(s) in RCA: 110] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Accepted: 05/03/2017] [Indexed: 11/29/2022] Open
Abstract
Macrophages play a crucial rule in orchestrating immune responses against pathogens and foreign materials. Macrophages have remarkable plasticity in response to environmental cues and are able to acquire a spectrum of activation status, best exemplified by pro-inflammatory (M1) and anti-inflammatory (M2) phenotypes at the two ends of the spectrum. Characterisation of M1 and M2 subsets is usually carried out by quantification of multiple cell surface markers, transcription factors and cytokine profiles. These approaches are time-consuming, require large numbers of cells and are resource intensive. In this study, we used machine learning algorithms to develop a simple and fast imaging-based approach that enables automated identification of different macrophage functional phenotypes using their cell size and morphology. Fluorescent microscopy was used to assess cell morphology of different cell types which were stained for nucleus and actin distribution using DAPI and phalloidin respectively. By only analysing their morphology we were able to identify M1 and M2 phenotypes effectively and could distinguish them from naïve macrophages and monocytes with an average accuracy of 90%. Thus we suggest high-content and automated image analysis can be used for fast phenotyping of functionally diverse cell populations with reasonable accuracy and without the need for using multiple markers.
Collapse
Affiliation(s)
- Hassan M Rostam
- Division of Immunology, School of Life Sciences, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham, NG7 2RD, UK.,Department of Biology, University of Garmian, Kalar, Kurdistan, Iraq
| | - Paul M Reynolds
- Division of Biomedical Engineering, School of Engineering, University of Glasgow, Glasgow, G12 8LT, UK
| | - Morgan R Alexander
- Advanced Materials and Healthcare Technologies Division, School of Pharmacy, University of Nottingham, Nottingham, NG7 2RD, UK
| | - Nikolaj Gadegaard
- Division of Biomedical Engineering, School of Engineering, University of Glasgow, Glasgow, G12 8LT, UK.
| | - Amir M Ghaemmaghami
- Division of Immunology, School of Life Sciences, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham, NG7 2RD, UK.
| |
Collapse
|