1
|
Kesapragada M, Sun YH, Zhu K, Recendez C, Fregoso D, Yang HY, Rolandi M, Isseroff R, Zhao M, Gomez M. A data-driven approach to establishing cell motility patterns as predictors of macrophage subtypes and their relation to cell morphology. PLoS One 2024; 19:e0315023. [PMID: 39739899 DOI: 10.1371/journal.pone.0315023] [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: 09/06/2024] [Accepted: 11/18/2024] [Indexed: 01/02/2025] Open
Abstract
The motility of macrophages in response to microenvironment stimuli is a hallmark of innate immunity, where macrophages play pro-inflammatory or pro-reparatory roles depending on their activation status during wound healing. Cell size and shape have been informative in defining macrophage subtypes. Studies show pro and anti-inflammatory macrophages exhibit distinct migratory behaviors, in vitro, in 3D and in vivo but this link has not been rigorously studied. We apply both morphology and motility-based image processing approaches to analyze live cell images consisting of macrophage phenotypes. Macrophage subtypes are differentiated from primary murine bone marrow derived macrophages using a potent lipopolysaccharide (LPS) or cytokine interleukin-4 (IL-4). We show that morphology is tightly linked to motility, which leads to our hypothesis that motility analysis could be used alone or in conjunction with morphological features for improved prediction of macrophage subtypes. We train a support vector machine (SVM) classifier to predict macrophage subtypes based on morphology alone, motility alone, and both morphology and motility combined. We show that motility has comparable predictive capabilities as morphology. However, using both measures can enhance predictive capabilities. While motility and morphological features can be individually ambiguous identifiers, together they provide significantly improved prediction accuracies (75%) from a training dataset of 1000 cells tracked over time using only phase contrast time-lapse microscopy. Thus, the approach combining cell motility and cell morphology information can lead to methods that accurately assess functionally diverse macrophage phenotypes quickly and efficiently. This can support the development of cost efficient and high through-put methods for screening biochemicals targeting macrophage polarization.
Collapse
Affiliation(s)
- Manasa Kesapragada
- Department of Applied Mathematics, University of California, Santa Cruz, Santa Cruz, CA, United States of America
| | - Yao-Hui Sun
- Department of Ophthalmology & Vision Science, School of Medicine, University of California, Davis, Sacramento, CA, United States of America
| | - Kan Zhu
- Department of Ophthalmology & Vision Science, School of Medicine, University of California, Davis, Sacramento, CA, United States of America
| | - Cynthia Recendez
- Department of Ophthalmology & Vision Science, School of Medicine, University of California, Davis, Sacramento, CA, United States of America
| | - Daniel Fregoso
- Department of Dermatology, School of Medicine, University of California, Davis, Sacramento, CA, United States of America
| | - Hsin-Ya Yang
- Department of Dermatology, School of Medicine, University of California, Davis, Sacramento, CA, United States of America
| | - Marco Rolandi
- Department of Electrical and Computer Engineering, University of California, Santa Cruz, Santa Cruz, CA, United States of America
| | - Rivkah Isseroff
- Department of Dermatology, School of Medicine, University of California, Davis, Sacramento, CA, United States of America
| | - Min Zhao
- Department of Ophthalmology & Vision Science, School of Medicine, University of California, Davis, Sacramento, CA, United States of America
- Department of Dermatology, School of Medicine, University of California, Davis, Sacramento, CA, United States of America
| | - Marcella Gomez
- Department of Applied Mathematics, University of California, Santa Cruz, Santa Cruz, CA, United States of America
| |
Collapse
|
2
|
Verma A, Yu C, Bachl S, Lopez I, Schwartz M, Moen E, Kale N, Ching C, Miller G, Dougherty T, Pao E, Graf W, Ward C, Jena S, Marson A, Carnevale J, Van Valen D, Engelhardt BE. Cellular behavior analysis from live-cell imaging of TCR T cell-cancer cell interactions. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.19.624390. [PMID: 39605616 PMCID: PMC11601648 DOI: 10.1101/2024.11.19.624390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
T cell therapies, such as chimeric antigen receptor (CAR) T cells and T cell receptor (TCR) T cells, are a growing class of anti-cancer treatments. However, expansion to novel indications and beyond last-line treatment requires engineering cells' dynamic population behaviors. Here we develop the tools for cellular behavior analysis of T cells from live-cell imaging, a common and inexpensive experimental setup used to evaluate engineered T cells. We first develop a state-of-the-art segmentation and tracking pipeline, Caliban, based on human-in-the-loop deep learning. We then build the Occident pipeline to collect a catalog of phenotypes that characterize cell populations, morphology, movement, and interactions in co-cultures of modified T cells and antigen-presenting tumor cells. We use Caliban and Occident to interrogate how interactions between T cells and cancer cells differ when beneficial knock-outs of RASA2 and CUL5 are introduced into TCR T cells. We apply spatiotemporal models to quantify T cell recruitment and proliferation after interactions with cancer cells. We discover that, compared to a safe harbor knockout control, RASA2 knockout T cells have longer interaction times with cancer cells leading to greater T cell activation and killing efficacy, while CUL5 knockout T cells have increased proliferation rates leading to greater numbers of T cells for hunting. Together, segmentation and tracking from Caliban and phenotype quantification from Occident enable cellular behavior analysis to better engineer T cell therapies for improved cancer treatment.
Collapse
Affiliation(s)
- Archit Verma
- Institute of Data Science and Biotechnology, Gladstone Institutes, San Francisco, CA, USA
| | - Changhua Yu
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Stefanie Bachl
- School of Medicine, University of California, San Francisco, San Francisco,CA, USA
- Gladstone-UCSF Institute of Genomic Immunology, San Francisco, CA, USA
| | - Ivan Lopez
- School of Medicine, Stanford University, Stanford, California, USA
- Department of Biomedical Data Science, Stanford University, Stanford, CA, USA
| | - Morgan Schwartz
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Erick Moen
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Nupura Kale
- School of Medicine, University of California, San Francisco, San Francisco,CA, USA
- Gladstone-UCSF Institute of Genomic Immunology, San Francisco, CA, USA
| | - Carter Ching
- School of Medicine, University of California, San Francisco, San Francisco,CA, USA
- Gladstone-UCSF Institute of Genomic Immunology, San Francisco, CA, USA
| | - Geneva Miller
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Tom Dougherty
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Ed Pao
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - William Graf
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Carl Ward
- Gladstone-UCSF Institute of Genomic Immunology, San Francisco, CA, USA
| | - Siddhartha Jena
- Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts, USA
| | - Alex Marson
- School of Medicine, University of California, San Francisco, San Francisco,CA, USA
- Gladstone-UCSF Institute of Genomic Immunology, San Francisco, CA, USA
| | - Julia Carnevale
- School of Medicine, University of California, San Francisco, San Francisco,CA, USA
- Gladstone-UCSF Institute of Genomic Immunology, San Francisco, CA, USA
| | - David Van Valen
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Barbara E Engelhardt
- Institute of Data Science and Biotechnology, Gladstone Institutes, San Francisco, CA, USA
- Department of Biomedical Data Science, Stanford University, Stanford, CA, USA
| |
Collapse
|
3
|
Chai B, Efstathiou C, Yue H, Draviam VM. Opportunities and challenges for deep learning in cell dynamics research. Trends Cell Biol 2024; 34:955-967. [PMID: 38030542 DOI: 10.1016/j.tcb.2023.10.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 09/30/2023] [Accepted: 10/13/2023] [Indexed: 12/01/2023]
Abstract
The growth of artificial intelligence (AI) has led to an increase in the adoption of computer vision and deep learning (DL) techniques for the evaluation of microscopy images and movies. This adoption has not only addressed hurdles in quantitative analysis of dynamic cell biological processes but has also started to support advances in drug development, precision medicine, and genome-phenome mapping. We survey existing AI-based techniques and tools, as well as open-source datasets, with a specific focus on the computational tasks of segmentation, classification, and tracking of cellular and subcellular structures and dynamics. We summarise long-standing challenges in microscopy video analysis from a computational perspective and review emerging research frontiers and innovative applications for DL-guided automation in cell dynamics research.
Collapse
Affiliation(s)
- Binghao Chai
- School of Biological and Behavioural Sciences, Queen Mary University of London (QMUL), London E1 4NS, UK
| | - Christoforos Efstathiou
- School of Biological and Behavioural Sciences, Queen Mary University of London (QMUL), London E1 4NS, UK
| | - Haoran Yue
- School of Biological and Behavioural Sciences, Queen Mary University of London (QMUL), London E1 4NS, UK
| | - Viji M Draviam
- School of Biological and Behavioural Sciences, Queen Mary University of London (QMUL), London E1 4NS, UK; The Alan Turing Institute, London NW1 2DB, UK.
| |
Collapse
|
4
|
Alahmari SS, Goldgof D, Hall LO, Mouton PR. A Review of Nuclei Detection and Segmentation on Microscopy Images Using Deep Learning With Applications to Unbiased Stereology Counting. IEEE TRANSACTIONS ON NEURAL NETWORKS AND LEARNING SYSTEMS 2024; 35:7458-7477. [PMID: 36327184 DOI: 10.1109/tnnls.2022.3213407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
The detection and segmentation of stained cells and nuclei are essential prerequisites for subsequent quantitative research for many diseases. Recently, deep learning has shown strong performance in many computer vision problems, including solutions for medical image analysis. Furthermore, accurate stereological quantification of microscopic structures in stained tissue sections plays a critical role in understanding human diseases and developing safe and effective treatments. In this article, we review the most recent deep learning approaches for cell (nuclei) detection and segmentation in cancer and Alzheimer's disease with an emphasis on deep learning approaches combined with unbiased stereology. Major challenges include accurate and reproducible cell detection and segmentation of microscopic images from stained sections. Finally, we discuss potential improvements and future trends in deep learning applied to cell detection and segmentation.
Collapse
|
5
|
Ryu J, Nejatbakhsh A, Torkashvand M, Gangadharan S, Seyedolmohadesin M, Kim J, Paninski L, Venkatachalam V. Versatile multiple object tracking in sparse 2D/3D videos via deformable image registration. PLoS Comput Biol 2024; 20:e1012075. [PMID: 38768230 PMCID: PMC11142724 DOI: 10.1371/journal.pcbi.1012075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 05/31/2024] [Accepted: 04/14/2024] [Indexed: 05/22/2024] Open
Abstract
Tracking body parts in behaving animals, extracting fluorescence signals from cells embedded in deforming tissue, and analyzing cell migration patterns during development all require tracking objects with partially correlated motion. As dataset sizes increase, manual tracking of objects becomes prohibitively inefficient and slow, necessitating automated and semi-automated computational tools. Unfortunately, existing methods for multiple object tracking (MOT) are either developed for specific datasets and hence do not generalize well to other datasets, or require large amounts of training data that are not readily available. This is further exacerbated when tracking fluorescent sources in moving and deforming tissues, where the lack of unique features and sparsely populated images create a challenging environment, especially for modern deep learning techniques. By leveraging technology recently developed for spatial transformer networks, we propose ZephIR, an image registration framework for semi-supervised MOT in 2D and 3D videos. ZephIR can generalize to a wide range of biological systems by incorporating adjustable parameters that encode spatial (sparsity, texture, rigidity) and temporal priors of a given data class. We demonstrate the accuracy and versatility of our approach in a variety of applications, including tracking the body parts of a behaving mouse and neurons in the brain of a freely moving C. elegans. We provide an open-source package along with a web-based graphical user interface that allows users to provide small numbers of annotations to interactively improve tracking results.
Collapse
Affiliation(s)
- James Ryu
- Department of Physics, Northeastern University, Boston, Massachusetts, United States of America
| | - Amin Nejatbakhsh
- Department of Neuroscience, Columbia University, New York, New York, United States of America
| | - Mahdi Torkashvand
- Department of Physics, Northeastern University, Boston, Massachusetts, United States of America
| | - Sahana Gangadharan
- Department of Physics, Northeastern University, Boston, Massachusetts, United States of America
| | - Maedeh Seyedolmohadesin
- Department of Physics, Northeastern University, Boston, Massachusetts, United States of America
| | - Jinmahn Kim
- Department of Physics, Northeastern University, Boston, Massachusetts, United States of America
| | - Liam Paninski
- Department of Neuroscience, Columbia University, New York, New York, United States of America
| | - Vivek Venkatachalam
- Department of Physics, Northeastern University, Boston, Massachusetts, United States of America
| |
Collapse
|
6
|
Zhao Y, Chen KL, Shen XY, Li MK, Wan YJ, Yang C, Yu RJ, Long YT, Yan F, Ying YL. HFM-Tracker: a cell tracking algorithm based on hybrid feature matching. Analyst 2024; 149:2629-2636. [PMID: 38563459 DOI: 10.1039/d4an00199k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Cell migration is known to be a fundamental biological process, playing an essential role in development, homeostasis, and diseases. This paper introduces a cell tracking algorithm named HFM-Tracker (Hybrid Feature Matching Tracker) that automatically identifies cell migration behaviours in consecutive images. It combines Contour Attention (CA) and Adaptive Confusion Matrix (ACM) modules to accurately capture cell contours in each image and track the dynamic behaviors of migrating cells in the field of view. Cells are firstly located and identified via the CA module-based cell detection network, and then associated and tracked via a cell tracking algorithm employing a hybrid feature-matching strategy. This proposed HFM-Tracker exhibits superiorities in cell detection and tracking, achieving 75% in MOTA (Multiple Object Tracking Accuracy) and 65% in IDF1 (ID F1 score). It provides quantitative analysis of the cell morphology and migration features, which could further help in understanding the complicated and diverse cell migration processes.
Collapse
Affiliation(s)
- Yan Zhao
- School of Information Science and Engineering, East China University of Science and Technology, 130 Meilong Road, 200237 Shanghai, P. R. China.
| | - Ke-Le Chen
- School of Chemistry and Chemical Engineering, Molecular Sensing and Imaging Center (MSIC), Nanjing University, Nanjing 210023, P. R. China.
| | - Xin-Yu Shen
- School of Electronic Sciences and Engineering, Nanjing University, Nanjing, 210023, China
| | - Ming-Kang Li
- School of Chemistry and Chemical Engineering, Molecular Sensing and Imaging Center (MSIC), Nanjing University, Nanjing 210023, P. R. China.
| | - Yong-Jing Wan
- School of Information Science and Engineering, East China University of Science and Technology, 130 Meilong Road, 200237 Shanghai, P. R. China.
| | - Cheng Yang
- School of Electronic Sciences and Engineering, Nanjing University, Nanjing, 210023, China
| | - Ru-Jia Yu
- School of Chemistry and Chemical Engineering, Molecular Sensing and Imaging Center (MSIC), Nanjing University, Nanjing 210023, P. R. China.
| | - Yi-Tao Long
- School of Chemistry and Chemical Engineering, Molecular Sensing and Imaging Center (MSIC), Nanjing University, Nanjing 210023, P. R. China.
| | - Feng Yan
- School of Electronic Sciences and Engineering, Nanjing University, Nanjing, 210023, China
| | - Yi-Lun Ying
- School of Information Science and Engineering, East China University of Science and Technology, 130 Meilong Road, 200237 Shanghai, P. R. China.
- Chemistry and Biomedicine Innovation Center, Nanjing University, Nanjing 210023, P. R. China
| |
Collapse
|
7
|
Hu F, Hu H, Xu H, Xu J, Chen Q. Dilated Heterogeneous Convolution for Cell Detection and Segmentation Based on Mask R-CNN. SENSORS (BASEL, SWITZERLAND) 2024; 24:2424. [PMID: 38676041 PMCID: PMC11053869 DOI: 10.3390/s24082424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Revised: 04/06/2024] [Accepted: 04/07/2024] [Indexed: 04/28/2024]
Abstract
Owing to the variable shapes, large size difference, uneven grayscale, and dense distribution among biological cells in an image, it is very difficult to accurately detect and segment cells. Especially, it is a serious challenge for some microscope imaging devices with limited resources owing to a large number of learning parameters and computational burden when using the standard Mask R-CNN. In this work, we propose a mask R-DHCNN for cell detection and segmentation. More specifically, Dilation Heterogeneous Convolution (DHConv) is proposed by designing a novel convolutional kernel structure (i.e., DHConv), which integrates the strengths of the heterogeneous kernel structure and dilated convolution. Then, the traditional homogeneous convolution structure of the standard Mask R-CNN is replaced with the proposed DHConv module to it adapt to shape and size differences encountered in cell detection and segmentation tasks. Finally, a series of comparison and ablation experiments are conducted on various biological cell datasets (such as U373, GoTW1, SIM+, and T24) to verify the effectiveness of the proposed method. The results show that the proposed method can obtain better performance than some state-of-the-art methods in multiple metrics (including AP, Precision, Recall, Dice, and PQ) while maintaining competitive FLOPs and FPS.
Collapse
Affiliation(s)
| | | | | | - Jinshan Xu
- College of Computer Science and Technology, Zhejiang University of Technology, Hangzhou 310014, China; (F.H.); (H.H.); (H.X.)
| | - Qi Chen
- College of Computer Science and Technology, Zhejiang University of Technology, Hangzhou 310014, China; (F.H.); (H.H.); (H.X.)
| |
Collapse
|
8
|
REINKE ANNIKA, TIZABI MINUD, BAUMGARTNER MICHAEL, EISENMANN MATTHIAS, HECKMANN-NÖTZEL DOREEN, KAVUR AEMRE, RÄDSCH TIM, SUDRE CAROLEH, ACION LAURA, ANTONELLI MICHELA, ARBEL TAL, BAKAS SPYRIDON, BENIS ARRIEL, BLASCHKO MATTHEWB, BUETTNER FLORIAN, CARDOSO MJORGE, CHEPLYGINA VERONIKA, CHEN JIANXU, CHRISTODOULOU EVANGELIA, CIMINI BETHA, COLLINS GARYS, FARAHANI KEYVAN, FERRER LUCIANA, GALDRAN ADRIAN, VAN GINNEKEN BRAM, GLOCKER BEN, GODAU PATRICK, HAASE ROBERT, HASHIMOTO DANIELA, HOFFMAN MICHAELM, HUISMAN MEREL, ISENSEE FABIAN, JANNIN PIERRE, KAHN CHARLESE, KAINMUELLER DAGMAR, KAINZ BERNHARD, KARARGYRIS ALEXANDROS, KARTHIKESALINGAM ALAN, KENNGOTT HANNES, KLEESIEK JENS, KOFLER FLORIAN, KOOI THIJS, KOPP-SCHNEIDER ANNETTE, KOZUBEK MICHAL, KRESHUK ANNA, KURC TAHSIN, LANDMAN BENNETTA, LITJENS GEERT, MADANI AMIN, MAIER-HEIN KLAUS, MARTEL ANNEL, MATTSON PETER, MEIJERING ERIK, MENZE BJOERN, MOONS KARELG, MÜLLER HENNING, NICHYPORUK BRENNAN, NICKEL FELIX, PETERSEN JENS, RAFELSKI SUSANNEM, RAJPOOT NASIR, REYES MAURICIO, RIEGLER MICHAELA, RIEKE NICOLA, SAEZ-RODRIGUEZ JULIO, SÁNCHEZ CLARAI, SHETTY SHRAVYA, SUMMERS RONALDM, TAHA ABDELA, TIULPIN ALEKSEI, TSAFTARIS SOTIRIOSA, VAN CALSTER BEN, VAROQUAUX GAËL, YANIV ZIVR, JÄGER PAULF, MAIER-HEIN LENA. Understanding metric-related pitfalls in image analysis validation. ARXIV 2024:arXiv:2302.01790v4. [PMID: 36945687 PMCID: PMC10029046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 03/23/2023]
Abstract
Validation metrics are key for the reliable tracking of scientific progress and for bridging the current chasm between artificial intelligence (AI) research and its translation into practice. However, increasing evidence shows that particularly in image analysis, metrics are often chosen inadequately in relation to the underlying research problem. This could be attributed to a lack of accessibility of metric-related knowledge: While taking into account the individual strengths, weaknesses, and limitations of validation metrics is a critical prerequisite to making educated choices, the relevant knowledge is currently scattered and poorly accessible to individual researchers. Based on a multi-stage Delphi process conducted by a multidisciplinary expert consortium as well as extensive community feedback, the present work provides the first reliable and comprehensive common point of access to information on pitfalls related to validation metrics in image analysis. Focusing on biomedical image analysis but with the potential of transfer to other fields, the addressed pitfalls generalize across application domains and are categorized according to a newly created, domain-agnostic taxonomy. To facilitate comprehension, illustrations and specific examples accompany each pitfall. As a structured body of information accessible to researchers of all levels of expertise, this work enhances global comprehension of a key topic in image analysis validation.
Collapse
Affiliation(s)
- ANNIKA REINKE
- German Cancer Research Center (DKFZ) Heidelberg, Division of Intelligent Medical Systems and HI Helmholtz Imaging, Germany and Faculty of Mathematics and Computer Science, Heidelberg University, Heidelberg, Germany
| | - MINU D. TIZABI
- German Cancer Research Center (DKFZ) Heidelberg, Division of Intelligent Medical Systems, Germany and National Center for Tumor Diseases (NCT), NCT Heidelberg, a partnership between DKFZ and University Medical Center Heidelberg, Germany
| | - MICHAEL BAUMGARTNER
- German Cancer Research Center (DKFZ) Heidelberg, Division of Medical Image Computing, Germany
| | - MATTHIAS EISENMANN
- German Cancer Research Center (DKFZ) Heidelberg, Division of Intelligent Medical Systems, Germany
| | - DOREEN HECKMANN-NÖTZEL
- German Cancer Research Center (DKFZ) Heidelberg, Division of Intelligent Medical Systems, Germany and National Center for Tumor Diseases (NCT), NCT Heidelberg, a partnership between DKFZ and University Medical Center Heidelberg, Germany
| | - A. EMRE KAVUR
- HI Applied Computer Vision Lab, Division of Medical Image Computing; German Cancer Research Center (DKFZ) Heidelberg, Division of Intelligent Medical Systems, Germany
| | - TIM RÄDSCH
- German Cancer Research Center (DKFZ) Heidelberg, Division of Intelligent Medical Systems and HI Helmholtz Imaging, Germany
| | - CAROLE H. SUDRE
- MRC Unit for Lifelong Health and Ageing at UCL and Centre for Medical Image Computing, Department of Computer Science, University College London, London, UK and School of Biomedical Engineering and Imaging Science, King’s College London, London, UK
| | - LAURA ACION
- Instituto de Cálculo, CONICET – Universidad de Buenos Aires, Buenos Aires, Argentina
| | - MICHELA ANTONELLI
- School of Biomedical Engineering and Imaging Science, King’s College London, London, UK and Centre for Medical Image Computing, University College London, London, UK
| | - TAL ARBEL
- Centre for Intelligent Machines and MILA (Quebec Artificial Intelligence Institute), McGill University, Montreal, Canada
| | - SPYRIDON BAKAS
- Division of Computational Pathology, Dept of Pathology & Laboratory Medicine, Indiana University School of Medicine, IU Health Information and Translational Sciences Building, Indianapolis, USA and Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Richards Medical Research Laboratories FL7, Philadelphia, PA, USA
| | - ARRIEL BENIS
- Department of Digital Medical Technologies, Holon Institute of Technology, Holon, Israel and European Federation for Medical Informatics, Le Mont-sur-Lausanne, Switzerland
| | - MATTHEW B. BLASCHKO
- Center for Processing Speech and Images, Department of Electrical Engineering, KU Leuven, Leuven, Belgium
| | - FLORIAN BUETTNER
- German Cancer Consortium (DKTK), partner site Frankfurt/Mainz, a partnership between DKFZ and UCT Frankfurt-Marburg, Germany, German Cancer Research Center (DKFZ) Heidelberg, Germany, Goethe University Frankfurt, Department of Medicine, Germany, Goethe University Frankfurt, Department of Informatics, Germany, and Frankfurt Cancer Insititute, Germany
| | - M. JORGE CARDOSO
- School of Biomedical Engineering and Imaging Science, King’s College London, London, UK
| | - VERONIKA CHEPLYGINA
- Department of Computer Science, IT University of Copenhagen, Copenhagen, Denmark
| | - JIANXU CHEN
- Leibniz-Institut für Analytische Wissenschaften – ISAS – e.V., Dortmund, Germany
| | - EVANGELIA CHRISTODOULOU
- German Cancer Research Center (DKFZ) Heidelberg, Division of Intelligent Medical Systems, Germany
| | - BETH A. CIMINI
- Imaging Platform, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - GARY S. COLLINS
- Centre for Statistics in Medicine, University of Oxford, Oxford, UK
| | - KEYVAN FARAHANI
- Center for Biomedical Informatics and Information Technology, National Cancer Institute, Bethesda, MD, USA
| | - LUCIANA FERRER
- Instituto de Investigación en Ciencias de la Computación (ICC), CONICET-UBA, Ciudad Universitaria, Ciudad Autónoma de Buenos Aires, Argentina
| | - ADRIAN GALDRAN
- Universitat Pompeu Fabra, Barcelona, Spain and University of Adelaide, Adelaide, Australia
| | - BRAM VAN GINNEKEN
- Fraunhofer MEVIS, Bremen, Germany and Radboud Institute for Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - BEN GLOCKER
- Department of Computing, Imperial College London, London, UK
| | - PATRICK GODAU
- German Cancer Research Center (DKFZ) Heidelberg, Division of Intelligent Medical Systems, Germany, Faculty of Mathematics and Computer Science, Heidelberg University, Heidelberg, Germany, and National Center for Tumor Diseases (NCT), NCT Heidelberg, a partnership between DKFZ and University Medical Center Heidelberg, Germany
| | - ROBERT HAASE
- Now with: Center for Scalable Data Analytics and Artificial Intelligence (ScaDS.AI), Leipzig University, Leipzig, Germany, DFG Cluster of Excellence “Physics of Life”, Technische Universität (TU) Dresden, Dresden, Germany, and Center for Systems Biology , Dresden, Germany
| | - DANIEL A. HASHIMOTO
- Department of Surgery, Perelman School of Medicine, Philadelphia, PA, USA and General Robotics Automation Sensing and Perception Laboratory, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, USA
| | - MICHAEL M. HOFFMAN
- Princess Margaret Cancer Centre, University Health Network, Toronto, Canada, Department of Medical Biophysics, University of Toronto, Toronto, Canada, Department of Computer Science, University of Toronto, Toronto, Canada, and Vector Institute for Artificial Intelligence, Toronto, Canada
| | - MEREL HUISMAN
- Department of Radiology and Nuclear Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - FABIAN ISENSEE
- German Cancer Research Center (DKFZ) Heidelberg, Division of Medical Image Computing and HI Applied Computer Vision Lab, Germany
| | - PIERRE JANNIN
- Laboratoire Traitement du Signal et de l’Image – UMR_S 1099, Université de Rennes 1, Rennes, France and INSERM, Paris Cedex, France
| | - CHARLES E. KAHN
- Department of Radiology and Institute for Biomedical Informatics, University of Pennsylvania, Philadelphia, PA, USA
| | - DAGMAR KAINMUELLER
- Max-Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Biomedical Image Analysis and HI Helmholtz Imaging, Berlin, Germany and University of Potsdam, Digital Engineering Faculty, Potsdam, Germany
| | - BERNHARD KAINZ
- Department of Computing, Faculty of Engineering, Imperial College London, London, UK and Department AIBE, Friedrich-Alexander-Universität (FAU), Erlangen-Nürnberg, Germany
| | | | | | - HANNES KENNGOTT
- Department of General, Visceral and Transplantation Surgery, Heidelberg University Hospital, Heidelberg, Germany
| | - JENS KLEESIEK
- Translational Image-guided Oncology (TIO), Institute for AI in Medicine (IKIM), University Medicine Essen, Essen, Germany
| | | | | | | | - MICHAL KOZUBEK
- Centre for Biomedical Image Analysis and Faculty of Informatics, Masaryk University, Brno, Czech Republic
| | - ANNA KRESHUK
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - TAHSIN KURC
- Department of Biomedical Informatics, Stony Brook University, Stony Brook, NY, USA
| | | | - GEERT LITJENS
- Department of Pathology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - AMIN MADANI
- Department of Surgery, University Health Network, Philadelphia, PA, Canada
| | - KLAUS MAIER-HEIN
- German Cancer Research Center (DKFZ) Heidelberg, Division of Medical Image Computing and HI Helmholtz Imaging, Germany and Pattern Analysis and Learning Group, Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany
| | - ANNE L. MARTEL
- Physical Sciences, Sunnybrook Research Institute, Toronto, Canada and Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | | | - ERIK MEIJERING
- School of Computer Science and Engineering, University of New South Wales, Sydney, Australia
| | - BJOERN MENZE
- Department of Quantitative Biomedicine, University of Zurich, Zurich, Switzerland
| | - KAREL G.M. MOONS
- Julius Center for Health Sciences and Primary Care, UMC Utrecht, Utrecht University, Utrecht, The Netherlands
| | - HENNING MÜLLER
- Information Systems Institute, University of Applied Sciences Western Switzerland (HES-SO), Sierre, Switzerland and Medical Faculty, University of Geneva, Geneva, Switzerland
| | | | - FELIX NICKEL
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - JENS PETERSEN
- German Cancer Research Center (DKFZ) Heidelberg, Division of Medical Image Computing, Germany
| | | | - NASIR RAJPOOT
- Tissue Image Analytics Laboratory, Department of Computer Science, University of Warwick, Coventry, UK
| | - MAURICIO REYES
- ARTORG Center for Biomedical Engineering Research, University of Bern, Bern, Switzerland and Department of Radiation Oncology, University Hospital Bern, University of Bern, Bern, Switzerland
| | - MICHAEL A. RIEGLER
- Simula Metropolitan Center for Digital Engineering, Oslo, Norway and UiT The Arctic University of Norway, Tromsø, Norway
| | | | - JULIO SAEZ-RODRIGUEZ
- Institute for Computational Biomedicine, Heidelberg University, Heidelberg. Germany and Faculty of Medicine, Heidelberg University Hospital, Heidelberg, Germany
| | - CLARA I. SÁNCHEZ
- Informatics Institute, Faculty of Science, University of Amsterdam, Amsterdam, The Netherlands
| | | | | | - ABDEL A. TAHA
- Institute of Information Systems Engineering, TU Wien, Vienna, Austria
| | - ALEKSEI TIULPIN
- Research Unit of Health Sciences and Technology, Faculty of Medicine, University of Oulu, Oulu, Finland and Neurocenter Oulu, Oulu University Hospital, Oulu, Finland
| | | | - BEN VAN CALSTER
- Department of Development and Regeneration and EPI-centre, KU Leuven, Leuven, Belgium and Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, The Netherlands
| | - GAËL VAROQUAUX
- Parietal project team, INRIA Saclay-Île de France, Palaiseau, France
| | - ZIV R. YANIV
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - PAUL F. JÄGER
- German Cancer Research Center (DKFZ) Heidelberg, Interactive Machine Learning Group and HI Helmholtz Imaging, Germany
| | - LENA MAIER-HEIN
- German Cancer Research Center (DKFZ) Heidelberg, Division of Intelligent Medical Systems and HI Helmholtz Imaging, Germany, Faculty of Mathematics and Computer Science and Medical Faculty, Heidelberg University, Heidelberg, Germany, and National Center for Tumor Diseases (NCT), NCT Heidelberg, a partnership between DKFZ and University Medical Center Heidelberg, Germany
| |
Collapse
|
9
|
Zhang G, Li X, Zhang Y, Han X, Li X, Yu J, Liu B, Wu J, Yu L, Dai Q. Bio-friendly long-term subcellular dynamic recording by self-supervised image enhancement microscopy. Nat Methods 2023; 20:1957-1970. [PMID: 37957429 PMCID: PMC10703694 DOI: 10.1038/s41592-023-02058-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 09/29/2023] [Indexed: 11/15/2023]
Abstract
Fluorescence microscopy has become an indispensable tool for revealing the dynamic regulation of cells and organelles. However, stochastic noise inherently restricts optical interrogation quality and exacerbates observation fidelity when balancing the joint demands of high frame rate, long-term recording and low phototoxicity. Here we propose DeepSeMi, a self-supervised-learning-based denoising framework capable of increasing signal-to-noise ratio by over 12 dB across various conditions. With the introduction of newly designed eccentric blind-spot convolution filters, DeepSeMi effectively denoises images with no loss of spatiotemporal resolution. In combination with confocal microscopy, DeepSeMi allows for recording organelle interactions in four colors at high frame rates across tens of thousands of frames, monitoring migrasomes and retractosomes over a half day, and imaging ultra-phototoxicity-sensitive Dictyostelium cells over thousands of frames. Through comprehensive validations across various samples and instruments, we prove DeepSeMi to be a versatile and biocompatible tool for breaking the shot-noise limit.
Collapse
Affiliation(s)
- Guoxun Zhang
- Department of Automation, Tsinghua University, Beijing, China
- Institute for Brain and Cognitive Sciences, Tsinghua University, Beijing, China
| | - Xiaopeng Li
- State Key Laboratory of Membrane Biology, Tsinghua University-Peking University Joint Center for Life Sciences, Beijing Frontier Research Center for Biological Structure, School of Life Sciences, Tsinghua University, Beijing, China
| | - Yuanlong Zhang
- Department of Automation, Tsinghua University, Beijing, China
- Institute for Brain and Cognitive Sciences, Tsinghua University, Beijing, China
| | - Xiaofei Han
- Department of Automation, Tsinghua University, Beijing, China
- Institute for Brain and Cognitive Sciences, Tsinghua University, Beijing, China
| | - Xinyang Li
- Department of Automation, Tsinghua University, Beijing, China
- Institute for Brain and Cognitive Sciences, Tsinghua University, Beijing, China
- Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, China
| | - Jinqiang Yu
- State Key Laboratory of Membrane Biology, Tsinghua University-Peking University Joint Center for Life Sciences, Beijing Frontier Research Center for Biological Structure, School of Life Sciences, Tsinghua University, Beijing, China
| | - Boqi Liu
- State Key Laboratory of Membrane Biology, Tsinghua University-Peking University Joint Center for Life Sciences, Beijing Frontier Research Center for Biological Structure, School of Life Sciences, Tsinghua University, Beijing, China
| | - Jiamin Wu
- Department of Automation, Tsinghua University, Beijing, China.
- Institute for Brain and Cognitive Sciences, Tsinghua University, Beijing, China.
- Shanghai AI Laboratory, Shanghai, China.
| | - Li Yu
- State Key Laboratory of Membrane Biology, Tsinghua University-Peking University Joint Center for Life Sciences, Beijing Frontier Research Center for Biological Structure, School of Life Sciences, Tsinghua University, Beijing, China.
| | - Qionghai Dai
- Department of Automation, Tsinghua University, Beijing, China.
- Institute for Brain and Cognitive Sciences, Tsinghua University, Beijing, China.
| |
Collapse
|
10
|
Stine JS, Aziere N, Harper BJ, Harper SL. A Novel Approach for Identifying Nanoplastics by Assessing Deformation Behavior with Scanning Electron Microscopy. MICROMACHINES 2023; 14:1903. [PMID: 37893340 PMCID: PMC10609349 DOI: 10.3390/mi14101903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Revised: 09/27/2023] [Accepted: 10/04/2023] [Indexed: 10/29/2023]
Abstract
As plastic production continues to increase globally, plastic waste accumulates and degrades into smaller plastic particles. Through chemical and biological processes, nanoscale plastic particles (nanoplastics) are formed and are expected to exist in quantities of several orders of magnitude greater than those found for microplastics. Due to their small size and low mass, nanoplastics remain challenging to detect in the environment using most standard analytical methods. The goal of this research is to adapt existing tools to address the analytical challenges posed by the identification of nanoplastics. Given the unique and well-documented properties of anthropogenic plastics, we hypothesized that nanoplastics could be differentiated by polymer type using spatiotemporal deformation data collected through irradiation with scanning electron microscopy (SEM). We selected polyvinyl chloride (PVC), polyethylene terephthalate (PET), and high-density polyethylene (HDPE) to capture a range of thermodynamic properties and molecular structures encompassed by commercially available plastics. Pristine samples of each polymer type were chosen and individually milled to generate micro and nanoscale particles for SEM analysis. To test the hypothesis that polymers could be differentiated from other constituents in complex samples, the polymers were compared against proxy materials common in environmental media, i.e., algae, kaolinite clay, and nanocellulose. Samples for SEM analysis were prepared uncoated to enable observation of polymer deformation under set electron beam parameters. For each sample type, particles approximately 1 µm in diameter were chosen, and videos of particle deformation were recorded and studied. Blinded samples were also prepared with mixtures of the aforementioned materials to test the viability of this method for identifying near-nanoscale plastic particles in environmental media. Based on the evidence collected, deformation patterns between plastic particles and particles present in common environmental media show significant differences. A computer vision algorithm was also developed and tested against manual measurements to improve the usefulness and efficiency of this method further.
Collapse
Affiliation(s)
- Jared S. Stine
- School of Chemical, Biological and Environmental Engineering, Oregon State University, Corvallis, OR 97331, USA;
- Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, OR 97331, USA;
| | - Nicolas Aziere
- School of Electrical Engineering and Computer Science, Oregon State University, Corvallis, OR 97331, USA;
| | - Bryan J. Harper
- Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, OR 97331, USA;
| | - Stacey L. Harper
- School of Chemical, Biological and Environmental Engineering, Oregon State University, Corvallis, OR 97331, USA;
- Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, OR 97331, USA;
| |
Collapse
|
11
|
Ramos AP, Szalapak A, Ferme LC, Modes CD. From cells to form: A roadmap to study shape emergence in vivo. Biophys J 2023; 122:3587-3599. [PMID: 37243338 PMCID: PMC10541488 DOI: 10.1016/j.bpj.2023.05.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Revised: 04/25/2023] [Accepted: 05/18/2023] [Indexed: 05/28/2023] Open
Abstract
Organogenesis arises from the collective arrangement of cells into progressively 3D-shaped tissue. The acquisition of a correctly shaped organ is then the result of a complex interplay between molecular cues, responsible for differentiation and patterning, and the mechanical properties of the system, which generate the necessary forces that drive correct shape emergence. Nowadays, technological advances in the fields of microscopy, molecular biology, and computer science are making it possible to see and record such complex interactions in incredible, unforeseen detail within the global context of the developing embryo. A quantitative and interdisciplinary perspective of developmental biology becomes then necessary for a comprehensive understanding of morphogenesis. Here, we provide a roadmap to quantify the events that lead to morphogenesis from imaging to image analysis, quantification, and modeling, focusing on the discrete cellular and tissue shape changes, as well as their mechanical properties.
Collapse
Affiliation(s)
| | - Alicja Szalapak
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany; Center for Systems Biology Dresden, Dresden, Germany
| | | | - Carl D Modes
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany; Center for Systems Biology Dresden, Dresden, Germany; Cluster of Excellence Physics of Life, TU Dresden, Dresden, Germany
| |
Collapse
|
12
|
Maška M, Ulman V, Delgado-Rodriguez P, Gómez-de-Mariscal E, Nečasová T, Guerrero Peña FA, Ren TI, Meyerowitz EM, Scherr T, Löffler K, Mikut R, Guo T, Wang Y, Allebach JP, Bao R, Al-Shakarji NM, Rahmon G, Toubal IE, Palaniappan K, Lux F, Matula P, Sugawara K, Magnusson KEG, Aho L, Cohen AR, Arbelle A, Ben-Haim T, Raviv TR, Isensee F, Jäger PF, Maier-Hein KH, Zhu Y, Ederra C, Urbiola A, Meijering E, Cunha A, Muñoz-Barrutia A, Kozubek M, Ortiz-de-Solórzano C. The Cell Tracking Challenge: 10 years of objective benchmarking. Nat Methods 2023:10.1038/s41592-023-01879-y. [PMID: 37202537 PMCID: PMC10333123 DOI: 10.1038/s41592-023-01879-y] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 04/13/2023] [Indexed: 05/20/2023]
Abstract
The Cell Tracking Challenge is an ongoing benchmarking initiative that has become a reference in cell segmentation and tracking algorithm development. Here, we present a significant number of improvements introduced in the challenge since our 2017 report. These include the creation of a new segmentation-only benchmark, the enrichment of the dataset repository with new datasets that increase its diversity and complexity, and the creation of a silver standard reference corpus based on the most competitive results, which will be of particular interest for data-hungry deep learning-based strategies. Furthermore, we present the up-to-date cell segmentation and tracking leaderboards, an in-depth analysis of the relationship between the performance of the state-of-the-art methods and the properties of the datasets and annotations, and two novel, insightful studies about the generalizability and the reusability of top-performing methods. These studies provide critical practical conclusions for both developers and users of traditional and machine learning-based cell segmentation and tracking algorithms.
Collapse
Affiliation(s)
- Martin Maška
- Centre for Biomedical Image Analysis, Faculty of Informatics, Masaryk University, Brno, Czech Republic
| | - Vladimír Ulman
- Centre for Biomedical Image Analysis, Faculty of Informatics, Masaryk University, Brno, Czech Republic
- IT4Innovations National Supercomputing Center, VSB - Technical University of Ostrava, Ostrava, Czech Republic
| | - Pablo Delgado-Rodriguez
- Bioengineering Department, Universidad Carlos III de Madrid, Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | - Estibaliz Gómez-de-Mariscal
- Bioengineering Department, Universidad Carlos III de Madrid, Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
- Optical Cell Biology, Instituto Gulbenkian de Ciência, Oeiras, Portugal
| | - Tereza Nečasová
- Centre for Biomedical Image Analysis, Faculty of Informatics, Masaryk University, Brno, Czech Republic
| | - Fidel A Guerrero Peña
- Centro de Informatica, Universidade Federal de Pernambuco, Recife, Brazil
- Center for Advanced Methods in Biological Image Analysis, Beckman Institute, California Institute of Technology, Pasadena, CA, USA
| | - Tsang Ing Ren
- Centro de Informatica, Universidade Federal de Pernambuco, Recife, Brazil
| | - Elliot M Meyerowitz
- Division of Biology and Biological Engineering and Howard Hughes Medical Institute, California Institute of Technology, Pasadena, CA, USA
| | - Tim Scherr
- Institute for Automation and Applied Informatics, Karlsruhe Institute of Technology, Eggenstein-Leopoldshafen, Germany
| | - Katharina Löffler
- Institute for Automation and Applied Informatics, Karlsruhe Institute of Technology, Eggenstein-Leopoldshafen, Germany
| | - Ralf Mikut
- Institute for Automation and Applied Informatics, Karlsruhe Institute of Technology, Eggenstein-Leopoldshafen, Germany
| | - Tianqi Guo
- The Elmore Family School of Electrical and Computer Engineering, Purdue University, West Lafayette, IN, USA
| | - Yin Wang
- The Elmore Family School of Electrical and Computer Engineering, Purdue University, West Lafayette, IN, USA
| | - Jan P Allebach
- The Elmore Family School of Electrical and Computer Engineering, Purdue University, West Lafayette, IN, USA
| | - Rina Bao
- Boston Children's Hospital and Harvard Medical School, Boston, MA, USA
- CIVA Lab, Department of Electrical Engineering and Computer Science, University of Missouri, Columbia, MO, USA
| | - Noor M Al-Shakarji
- CIVA Lab, Department of Electrical Engineering and Computer Science, University of Missouri, Columbia, MO, USA
| | - Gani Rahmon
- CIVA Lab, Department of Electrical Engineering and Computer Science, University of Missouri, Columbia, MO, USA
| | - Imad Eddine Toubal
- CIVA Lab, Department of Electrical Engineering and Computer Science, University of Missouri, Columbia, MO, USA
| | - Kannappan Palaniappan
- CIVA Lab, Department of Electrical Engineering and Computer Science, University of Missouri, Columbia, MO, USA
| | - Filip Lux
- Centre for Biomedical Image Analysis, Faculty of Informatics, Masaryk University, Brno, Czech Republic
| | - Petr Matula
- Centre for Biomedical Image Analysis, Faculty of Informatics, Masaryk University, Brno, Czech Republic
| | - Ko Sugawara
- Institut de Génomique Fonctionnelle de Lyon (IGFL), École Normale Supérieure de Lyon, Lyon, France
- Centre National de la Recherche Scientifique (CNRS), Paris, France
| | | | - Layton Aho
- Department of Electrical and Computer Engineering, Drexel University, Philadelphia, PA, USA
| | - Andrew R Cohen
- Department of Electrical and Computer Engineering, Drexel University, Philadelphia, PA, USA
| | - Assaf Arbelle
- School of Electrical and Computer Engineering, Ben-Gurion University of the Negev, Beersheba, Israel
| | - Tal Ben-Haim
- School of Electrical and Computer Engineering, Ben-Gurion University of the Negev, Beersheba, Israel
| | - Tammy Riklin Raviv
- School of Electrical and Computer Engineering, Ben-Gurion University of the Negev, Beersheba, Israel
| | - Fabian Isensee
- Division of Medical Image Computing, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Helmholtz Imaging, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Paul F Jäger
- Helmholtz Imaging, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Interactive Machine Learning Group, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Klaus H Maier-Hein
- Division of Medical Image Computing, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Pattern Analysis and Learning Group, Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany
| | - Yanming Zhu
- School of Computer Science and Engineering, University of New South Wales, Sydney, New South Wales, Australia
- Griffith University, Nathan, Queensland, Australia
| | - Cristina Ederra
- Biomedical Engineering Program and Ciberonc, Center for Applied Medical Research, Universidad de Navarra, Pamplona, Spain
| | - Ainhoa Urbiola
- Biomedical Engineering Program and Ciberonc, Center for Applied Medical Research, Universidad de Navarra, Pamplona, Spain
| | - Erik Meijering
- School of Computer Science and Engineering, University of New South Wales, Sydney, New South Wales, Australia
| | - Alexandre Cunha
- Center for Advanced Methods in Biological Image Analysis, Beckman Institute, California Institute of Technology, Pasadena, CA, USA
| | - Arrate Muñoz-Barrutia
- Bioengineering Department, Universidad Carlos III de Madrid, Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | - Michal Kozubek
- Centre for Biomedical Image Analysis, Faculty of Informatics, Masaryk University, Brno, Czech Republic.
| | - Carlos Ortiz-de-Solórzano
- Biomedical Engineering Program and Ciberonc, Center for Applied Medical Research, Universidad de Navarra, Pamplona, Spain.
| |
Collapse
|
13
|
Czolbe S, Pegios P, Krause O, Feragen A. Semantic similarity metrics for image registration. Med Image Anal 2023; 87:102830. [PMID: 37172390 DOI: 10.1016/j.media.2023.102830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 01/19/2023] [Accepted: 04/20/2023] [Indexed: 05/15/2023]
Abstract
Image registration aims to find geometric transformations that align images. Most algorithmic and deep learning-based methods solve the registration problem by minimizing a loss function, consisting of a similarity metric comparing the aligned images, and a regularization term ensuring smoothness of the transformation. Existing similarity metrics like Euclidean Distance or Normalized Cross-Correlation focus on aligning pixel intensity values or correlations, giving difficulties with low intensity contrast, noise, and ambiguous matching. We propose a semantic similarity metric for image registration, focusing on aligning image areas based on semantic correspondence instead. Our approach learns dataset-specific features that drive the optimization of a learning-based registration model. We train both an unsupervised approach extracting features with an auto-encoder, and a semi-supervised approach using supplemental segmentation data. We validate the semantic similarity metric using both deep-learning-based and algorithmic image registration methods. Compared to existing methods across four different image modalities and applications, the method achieves consistently high registration accuracy and smooth transformation fields.
Collapse
Affiliation(s)
- Steffen Czolbe
- Department of Computer Science, University of Copenhagen, Denmark.
| | | | - Oswin Krause
- Department of Computer Science, University of Copenhagen, Denmark
| | - Aasa Feragen
- DTU Compute, Technical University of Denmark, Denmark
| |
Collapse
|
14
|
Schulte A, Lohner H, Degenbeck J, Segebarth D, Rittner HL, Blum R, Aue A. Unbiased analysis of the dorsal root ganglion after peripheral nerve injury: no neuronal loss, no gliosis, but satellite glial cell plasticity. Pain 2023; 164:728-740. [PMID: 35969236 PMCID: PMC10026836 DOI: 10.1097/j.pain.0000000000002758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Revised: 07/13/2022] [Accepted: 07/26/2022] [Indexed: 11/26/2022]
Abstract
ABSTRACT Pain syndromes are often accompanied by complex molecular and cellular changes in dorsal root ganglia (DRG). However, the evaluation of cellular plasticity in the DRG is often performed by heuristic manual analysis of a small number of representative microscopy image fields. In this study, we introduce a deep learning-based strategy for objective and unbiased analysis of neurons and satellite glial cells (SGCs) in the DRG. To validate the approach experimentally, we examined serial sections of the rat DRG after spared nerve injury (SNI) or sham surgery. Sections were stained for neurofilament, glial fibrillary acidic protein (GFAP), and glutamine synthetase (GS) and imaged using high-resolution large-field (tile) microscopy. After training of deep learning models on consensus information of different experts, thousands of image features in DRG sections were analyzed. We used known (GFAP upregulation), controversial (neuronal loss), and novel (SGC phenotype switch) changes to evaluate the method. In our data, the number of DRG neurons was similar 14 d after SNI vs sham. In GFAP-positive subareas, the percentage of neurons in proximity to GFAP-positive cells increased after SNI. In contrast, GS-positive signals, and the percentage of neurons in proximity to GS-positive SGCs decreased after SNI. Changes in GS and GFAP levels could be linked to specific DRG neuron subgroups of different size. Hence, we could not detect gliosis but plasticity changes in the SGC marker expression. Our objective analysis of DRG tissue after peripheral nerve injury shows cellular plasticity responses of SGCs in the whole DRG but neither injury-induced neuronal death nor gliosis.
Collapse
Affiliation(s)
- Annemarie Schulte
- Department of Neurology, University Hospital of Würzburg, Würzburg, Germany
| | - Hannah Lohner
- Department of Anesthesiology, Center for Interdisciplinary Pain Medicine, Intensive Care, Emergency Medicine and Pain Therapy, University Hospital of Würzburg, Würzburg, Germany
| | - Johannes Degenbeck
- Department of Anesthesiology, Center for Interdisciplinary Pain Medicine, Intensive Care, Emergency Medicine and Pain Therapy, University Hospital of Würzburg, Würzburg, Germany
| | - Dennis Segebarth
- Institute of Clinical Neurobiology, University Hospital of Würzburg, Würzburg, Germany
| | - Heike L. Rittner
- Department of Anesthesiology, Center for Interdisciplinary Pain Medicine, Intensive Care, Emergency Medicine and Pain Therapy, University Hospital of Würzburg, Würzburg, Germany
| | - Robert Blum
- Department of Neurology, University Hospital of Würzburg, Würzburg, Germany
| | - Annemarie Aue
- Department of Anesthesiology, Center for Interdisciplinary Pain Medicine, Intensive Care, Emergency Medicine and Pain Therapy, University Hospital of Würzburg, Würzburg, Germany
| |
Collapse
|
15
|
Jiang J, Khan A, Shailja S, Belteton SA, Goebel M, Szymanski DB, Manjunath BS. Segmentation, tracking, and sub-cellular feature extraction in 3D time-lapse images. Sci Rep 2023; 13:3483. [PMID: 36859457 PMCID: PMC9977871 DOI: 10.1038/s41598-023-29149-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 01/31/2023] [Indexed: 03/03/2023] Open
Abstract
This paper presents a method for time-lapse 3D cell analysis. Specifically, we consider the problem of accurately localizing and quantitatively analyzing sub-cellular features, and for tracking individual cells from time-lapse 3D confocal cell image stacks. The heterogeneity of cells and the volume of multi-dimensional images presents a major challenge for fully automated analysis of morphogenesis and development of cells. This paper is motivated by the pavement cell growth process, and building a quantitative morphogenesis model. We propose a deep feature based segmentation method to accurately detect and label each cell region. An adjacency graph based method is used to extract sub-cellular features of the segmented cells. Finally, the robust graph based tracking algorithm using multiple cell features is proposed for associating cells at different time instances. We also demonstrate the generality of our tracking method on C. elegans fluorescent nuclei imagery. Extensive experiment results are provided and demonstrate the robustness of the proposed method. The code is available on GitHub and the method is available as a service through the BisQue portal.
Collapse
Affiliation(s)
- Jiaxiang Jiang
- Department of Electrical and Computer Engineering, University of California, Santa Barbara, USA.
| | - Amil Khan
- grid.133342.40000 0004 1936 9676Department of Electrical and Computer Engineering, University of California, Santa Barbara, USA
| | - S. Shailja
- grid.133342.40000 0004 1936 9676Department of Electrical and Computer Engineering, University of California, Santa Barbara, USA
| | - Samuel A. Belteton
- grid.169077.e0000 0004 1937 2197Department of Botany and Plant Pathology, Purdue University, West Lafayette, USA ,grid.24805.3b0000 0001 0687 2182Molecular Biology Program, New Mexico State University, Las Cruces, USA
| | - Michael Goebel
- grid.133342.40000 0004 1936 9676Department of Electrical and Computer Engineering, University of California, Santa Barbara, USA
| | - Daniel B. Szymanski
- grid.169077.e0000 0004 1937 2197Department of Botany and Plant Pathology, Purdue University, West Lafayette, USA
| | - B. S. Manjunath
- grid.133342.40000 0004 1936 9676Department of Electrical and Computer Engineering, University of California, Santa Barbara, USA
| |
Collapse
|
16
|
Kostrykin L, Rohr K. Superadditivity and Convex Optimization for Globally Optimal Cell Segmentation Using Deformable Shape Models. IEEE TRANSACTIONS ON PATTERN ANALYSIS AND MACHINE INTELLIGENCE 2023; 45:3831-3847. [PMID: 35737620 DOI: 10.1109/tpami.2022.3185583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Cell nuclei segmentation is challenging due to shape variation and closely clustered or partially overlapping objects. Most previous methods are not globally optimal, limited to elliptical models, or are computationally expensive. In this work, we introduce a globally optimal approach based on deformable shape models and global energy minimization for cell nuclei segmentation and cluster splitting. We propose an implicit parameterization of deformable shape models and show that it leads to a convex energy. Convex energy minimization yields the global solution independently of the initialization, is fast, and robust. To jointly perform cell nuclei segmentation and cluster splitting, we developed a novel iterative global energy minimization method, which leverages the inherent property of superadditivity of the convex energy. This property exploits the lower bound of the energy of the union of the models and improves the computational efficiency. Our method provably determines a solution close to global optimality. In addition, we derive a closed-form solution of the proposed global minimization based on the superadditivity property for non-clustered cell nuclei. We evaluated our method using fluorescence microscopy images of five different cell types comprising various challenges, and performed a quantitative comparison with previous methods. Our method achieved state-of-the-art or improved performance.
Collapse
|
17
|
Mou T, Liang J, Vu TN, Tian M, Gao Y. A Comprehensive Landscape of Imaging Feature-Associated RNA Expression Profiles in Human Breast Tissue. SENSORS (BASEL, SWITZERLAND) 2023; 23:1432. [PMID: 36772473 PMCID: PMC9921444 DOI: 10.3390/s23031432] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 01/15/2023] [Accepted: 01/20/2023] [Indexed: 06/18/2023]
Abstract
The expression abundance of transcripts in nondiseased breast tissue varies among individuals. The association study of genotypes and imaging phenotypes may help us to understand this individual variation. Since existing reports mainly focus on tumors or lesion areas, the heterogeneity of pathological image features and their correlations with RNA expression profiles for nondiseased tissue are not clear. The aim of this study is to discover the association between the nucleus features and the transcriptome-wide RNAs. We analyzed both microscopic histology images and RNA-sequencing data of 456 breast tissues from the Genotype-Tissue Expression (GTEx) project and constructed an automatic computational framework. We classified all samples into four clusters based on their nucleus morphological features and discovered feature-specific gene sets. The biological pathway analysis was performed on each gene set. The proposed framework evaluates the morphological characteristics of the cell nucleus quantitatively and identifies the associated genes. We found image features that capture population variation in breast tissue associated with RNA expressions, suggesting that the variation in expression pattern affects population variation in the morphological traits of breast tissue. This study provides a comprehensive transcriptome-wide view of imaging-feature-specific RNA expression for healthy breast tissue. Such a framework could also be used for understanding the connection between RNA expression and morphology in other tissues and organs. Pathway analysis indicated that the gene sets we identified were involved in specific biological processes, such as immune processes.
Collapse
Affiliation(s)
- Tian Mou
- School of Biomedical Engineering, Medical School, Shenzhen University, Shenzhen 518000, China
| | - Jianwen Liang
- School of Biomedical Engineering, Medical School, Shenzhen University, Shenzhen 518000, China
| | - Trung Nghia Vu
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, SE 17177 Stockholm, Sweden
| | - Mu Tian
- School of Biomedical Engineering, Medical School, Shenzhen University, Shenzhen 518000, China
| | - Yi Gao
- School of Biomedical Engineering, Medical School, Shenzhen University, Shenzhen 518000, China
| |
Collapse
|
18
|
Togninalli M, Ho ATV, Madl CM, Holbrook CA, Wang YX, Magnusson KEG, Kirillova A, Chang A, Blau HM. Machine learning-based classification of dual fluorescence signals reveals muscle stem cell fate transitions in response to regenerative niche factors. NPJ Regen Med 2023; 8:4. [PMID: 36639373 PMCID: PMC9839750 DOI: 10.1038/s41536-023-00277-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 01/03/2023] [Indexed: 01/15/2023] Open
Abstract
The proper regulation of muscle stem cell (MuSC) fate by cues from the niche is essential for regeneration of skeletal muscle. How pro-regenerative niche factors control the dynamics of MuSC fate decisions remains unknown due to limitations of population-level endpoint assays. To address this knowledge gap, we developed a dual fluorescence imaging time lapse (Dual-FLIT) microscopy approach that leverages machine learning classification strategies to track single cell fate decisions with high temporal resolution. Using two fluorescent reporters that read out maintenance of stemness and myogenic commitment, we constructed detailed lineage trees for individual MuSCs and their progeny, classifying each division event as symmetric self-renewing, asymmetric, or symmetric committed. Our analysis reveals that treatment with the lipid metabolite, prostaglandin E2 (PGE2), accelerates the rate of MuSC proliferation over time, while biasing division events toward symmetric self-renewal. In contrast, the IL6 family member, Oncostatin M (OSM), decreases the proliferation rate after the first generation, while blocking myogenic commitment. These insights into the dynamics of MuSC regulation by niche cues were uniquely enabled by our Dual-FLIT approach. We anticipate that similar binary live cell readouts derived from Dual-FLIT will markedly expand our understanding of how niche factors control tissue regeneration in real time.
Collapse
Affiliation(s)
- Matteo Togninalli
- Baxter Laboratory for Stem Cell Biology, Department of Microbiology and Immunology, Institute for Stem Cell Biology and Regenerative Medicine, Stanford School of Medicine, Stanford, CA, 94305-5175, USA
| | - Andrew T V Ho
- Baxter Laboratory for Stem Cell Biology, Department of Microbiology and Immunology, Institute for Stem Cell Biology and Regenerative Medicine, Stanford School of Medicine, Stanford, CA, 94305-5175, USA
- Department of Functional and Adaptive Biology - UMR 8251 CNRS, Université Paris Cité, 75013, Paris, France
| | - Christopher M Madl
- Baxter Laboratory for Stem Cell Biology, Department of Microbiology and Immunology, Institute for Stem Cell Biology and Regenerative Medicine, Stanford School of Medicine, Stanford, CA, 94305-5175, USA
- Department of Materials Science and Engineering, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Colin A Holbrook
- Baxter Laboratory for Stem Cell Biology, Department of Microbiology and Immunology, Institute for Stem Cell Biology and Regenerative Medicine, Stanford School of Medicine, Stanford, CA, 94305-5175, USA
| | - Yu Xin Wang
- Baxter Laboratory for Stem Cell Biology, Department of Microbiology and Immunology, Institute for Stem Cell Biology and Regenerative Medicine, Stanford School of Medicine, Stanford, CA, 94305-5175, USA
- Center for Genetic Disorders and Aging, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, 92037, USA
| | - Klas E G Magnusson
- Baxter Laboratory for Stem Cell Biology, Department of Microbiology and Immunology, Institute for Stem Cell Biology and Regenerative Medicine, Stanford School of Medicine, Stanford, CA, 94305-5175, USA
- Department of Signal Processing, ACCESS Linnaeus Centre, KTH Royal Institute of Technology, 100 44, Stockholm, Sweden
| | - Anna Kirillova
- Baxter Laboratory for Stem Cell Biology, Department of Microbiology and Immunology, Institute for Stem Cell Biology and Regenerative Medicine, Stanford School of Medicine, Stanford, CA, 94305-5175, USA
| | - Andrew Chang
- Baxter Laboratory for Stem Cell Biology, Department of Microbiology and Immunology, Institute for Stem Cell Biology and Regenerative Medicine, Stanford School of Medicine, Stanford, CA, 94305-5175, USA
| | - Helen M Blau
- Baxter Laboratory for Stem Cell Biology, Department of Microbiology and Immunology, Institute for Stem Cell Biology and Regenerative Medicine, Stanford School of Medicine, Stanford, CA, 94305-5175, USA.
| |
Collapse
|
19
|
Hradecka L, Wiesner D, Sumbal J, Koledova ZS, Maska M. Segmentation and Tracking of Mammary Epithelial Organoids in Brightfield Microscopy. IEEE TRANSACTIONS ON MEDICAL IMAGING 2023; 42:281-290. [PMID: 36170389 DOI: 10.1109/tmi.2022.3210714] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
We present an automated and deep-learning-based workflow to quantitatively analyze the spatiotemporal development of mammary epithelial organoids in two-dimensional time-lapse (2D+t) sequences acquired using a brightfield microscope at high resolution. It involves a convolutional neural network (U-Net), purposely trained using computer-generated bioimage data created by a conditional generative adversarial network (pix2pixHD), to infer semantic segmentation, adaptive morphological filtering to identify organoid instances, and a shape-similarity-constrained, instance-segmentation-correcting tracking procedure to reliably cherry-pick the organoid instances of interest in time. By validating it using real 2D+t sequences of mouse mammary epithelial organoids of morphologically different phenotypes, we clearly demonstrate that the workflow achieves reliable segmentation and tracking performance, providing a reproducible and laborless alternative to manual analyses of the acquired bioimage data.
Collapse
|
20
|
Scherr T, Seiffarth J, Wollenhaupt B, Neumann O, Schilling MP, Kohlheyer D, Scharr H, Nöh K, Mikut R. microbeSEG: A deep learning software tool with OMERO data management for efficient and accurate cell segmentation. PLoS One 2022; 17:e0277601. [PMID: 36445903 PMCID: PMC9707790 DOI: 10.1371/journal.pone.0277601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Accepted: 11/01/2022] [Indexed: 12/02/2022] Open
Abstract
In biotechnology, cell growth is one of the most important properties for the characterization and optimization of microbial cultures. Novel live-cell imaging methods are leading to an ever better understanding of cell cultures and their development. The key to analyzing acquired data is accurate and automated cell segmentation at the single-cell level. Therefore, we present microbeSEG, a user-friendly Python-based cell segmentation tool with a graphical user interface and OMERO data management. microbeSEG utilizes a state-of-the-art deep learning-based segmentation method and can be used for instance segmentation of a wide range of cell morphologies and imaging techniques, e.g., phase contrast or fluorescence microscopy. The main focus of microbeSEG is a comprehensible, easy, efficient, and complete workflow from the creation of training data to the final application of the trained segmentation model. We demonstrate that accurate cell segmentation results can be obtained within 45 minutes of user time. Utilizing public segmentation datasets or pre-labeling further accelerates the microbeSEG workflow. This opens the door for accurate and efficient data analysis of microbial cultures.
Collapse
Affiliation(s)
- Tim Scherr
- Institute for Automation and Applied Informatics, Karlsruhe Institute of Technology, Eggenstein-Leopoldshafen, Germany
- * E-mail: (TS); (KN); (RM)
| | - Johannes Seiffarth
- Institute of Bio- and Geosciences, IBG-1: Biotechnology, Forschungszentrum Jülich GmbH, Jülich, Germany
- Computational Systems Biology (AVT.CSB), RWTH Aachen University, Aachen, Germany
| | - Bastian Wollenhaupt
- Institute of Bio- and Geosciences, IBG-1: Biotechnology, Forschungszentrum Jülich GmbH, Jülich, Germany
| | - Oliver Neumann
- Institute for Automation and Applied Informatics, Karlsruhe Institute of Technology, Eggenstein-Leopoldshafen, Germany
| | - Marcel P. Schilling
- Institute for Automation and Applied Informatics, Karlsruhe Institute of Technology, Eggenstein-Leopoldshafen, Germany
| | - Dietrich Kohlheyer
- Institute of Bio- and Geosciences, IBG-1: Biotechnology, Forschungszentrum Jülich GmbH, Jülich, Germany
| | - Hanno Scharr
- Institute of Bio- and Geosciences, IBG-2: Plant Sciences, Forschungszentrum Jülich GmbH, Jülich, Germany
- Institute for Advanced Simulation, IAS-8: Data Analytics and Machine Learning, Forschungszentrum Jülich GmbH, Jülich, Germany
| | - Katharina Nöh
- Institute of Bio- and Geosciences, IBG-1: Biotechnology, Forschungszentrum Jülich GmbH, Jülich, Germany
- * E-mail: (TS); (KN); (RM)
| | - Ralf Mikut
- Institute for Automation and Applied Informatics, Karlsruhe Institute of Technology, Eggenstein-Leopoldshafen, Germany
- * E-mail: (TS); (KN); (RM)
| |
Collapse
|
21
|
Qureshi MH, Ozlu N, Bayraktar H. Adaptive tracking algorithm for trajectory analysis of cells and layer-by-layer assessment of motility dynamics. Comput Biol Med 2022; 150:106193. [PMID: 37859286 DOI: 10.1016/j.compbiomed.2022.106193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 09/26/2022] [Accepted: 10/08/2022] [Indexed: 11/03/2022]
Abstract
Tracking biological objects such as cells or subcellular components imaged with time-lapse microscopy enables us to understand the molecular principles about the dynamics of cell behaviors. However, automatic object detection, segmentation and extracting trajectories remain as a rate-limiting step due to intrinsic challenges of video processing. This paper presents an adaptive tracking algorithm (Adtari) that automatically finds the optimum search radius and cell linkages to determine trajectories in consecutive frames. A critical assumption in most tracking studies is that displacement remains unchanged throughout the movie and cells in a few frames are usually analyzed to determine its magnitude. Tracking errors and inaccurate association of cells may occur if the user does not correctly evaluate the value or prior knowledge is not present on cell movement. The key novelty of our method is that minimum intercellular distance and maximum displacement of cells between frames are dynamically computed and used to determine the threshold distance. Since the space between cells is highly variable in a given frame, our software recursively alters the magnitude to determine all plausible matches in the trajectory analysis. Our method therefore eliminates a major preprocessing step where a constant distance was used to determine the neighbor cells in tracking methods. Cells having multiple overlaps and splitting events were further evaluated by using the shape attributes including perimeter, area, ellipticity and distance. The features were applied to determine the closest matches by minimizing the difference in their magnitudes. Finally, reporting section of our software were used to generate instant maps by overlaying cell features and trajectories. Adtari was validated by using videos with variable signal-to-noise, contrast ratio and cell density. We compared the adaptive tracking with constant distance and other methods to evaluate performance and its efficiency. Our algorithm yields reduced mismatch ratio, increased ratio of whole cell track, higher frame tracking efficiency and allows layer-by-layer assessment of motility to characterize single-cells. Adaptive tracking provides a reliable, accurate, time efficient and user-friendly open source software that is well suited for analysis of 2D fluorescence microscopy video datasets.
Collapse
Affiliation(s)
- Mohammad Haroon Qureshi
- Department of Molecular Biology and Genetics, Koç University, Rumelifeneri Yolu, Sariyer, 34450, Istanbul, Turkey; Center for Translational Research, Koç University, Rumelifeneri Yolu, Sariyer, 34450, Istanbul, Turkey
| | - Nurhan Ozlu
- Department of Molecular Biology and Genetics, Koç University, Rumelifeneri Yolu, Sariyer, 34450, Istanbul, Turkey
| | - Halil Bayraktar
- Department of Molecular Biology and Genetics, Istanbul Technical University, Maslak, Sariyer, 34467, Istanbul, Turkey.
| |
Collapse
|
22
|
Liang P, Zhang Y, Ding Y, Chen J, Madukoma CS, Weninger T, Shrout JD, Chen DZ. H-EMD: A Hierarchical Earth Mover's Distance Method for Instance Segmentation. IEEE TRANSACTIONS ON MEDICAL IMAGING 2022; 41:2582-2597. [PMID: 35446762 DOI: 10.1109/tmi.2022.3169449] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Deep learning (DL) based semantic segmentation methods have achieved excellent performance in biomedical image segmentation, producing high quality probability maps to allow extraction of rich instance information to facilitate good instance segmentation. While numerous efforts were put into developing new DL semantic segmentation models, less attention was paid to a key issue of how to effectively explore their probability maps to attain the best possible instance segmentation. We observe that probability maps by DL semantic segmentation models can be used to generate many possible instance candidates, and accurate instance segmentation can be achieved by selecting from them a set of "optimized" candidates as output instances. Further, the generated instance candidates form a well-behaved hierarchical structure (a forest), which allows selecting instances in an optimized manner. Hence, we propose a novel framework, called hierarchical earth mover's distance (H-EMD), for instance segmentation in biomedical 2D+time videos and 3D images, which judiciously incorporates consistent instance selection with semantic-segmentation-generated probability maps. H-EMD contains two main stages: (1) instance candidate generation: capturing instance-structured information in probability maps by generating many instance candidates in a forest structure; (2) instance candidate selection: selecting instances from the candidate set for final instance segmentation. We formulate a key instance selection problem on the instance candidate forest as an optimization problem based on the earth mover's distance (EMD), and solve it by integer linear programming. Extensive experiments on eight biomedical video or 3D datasets demonstrate that H-EMD consistently boosts DL semantic segmentation models and is highly competitive with state-of-the-art methods.
Collapse
|
23
|
Komuro J, Tokuoka Y, Seki T, Kusumoto D, Hashimoto H, Katsuki T, Nakamura T, Akiba Y, Kuoka T, Kimura M, Yamada T, Fukuda K, Funahashi A, Yuasa S. Development of non-bias phenotypic drug screening for cardiomyocyte hypertrophy by image segmentation using deep learning. Biochem Biophys Res Commun 2022; 632:181-188. [DOI: 10.1016/j.bbrc.2022.09.108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 09/15/2022] [Accepted: 09/27/2022] [Indexed: 11/02/2022]
|
24
|
Cuny AP, Ponti A, Kündig T, Rudolf F, Stelling J. Cell region fingerprints enable highly precise single-cell tracking and lineage reconstruction. Nat Methods 2022; 19:1276-1285. [PMID: 36138173 DOI: 10.1038/s41592-022-01603-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 08/02/2022] [Indexed: 11/09/2022]
Abstract
Experimental studies of cell growth, inheritance and their associated processes by microscopy require accurate single-cell observations of sufficient duration to reconstruct the genealogy. However, cell tracking-assigning identical cells on consecutive images to a track-is often challenging, resulting in laborious manual verification. Here, we propose fingerprints to identify problematic assignments rapidly. A fingerprint distance compares the structural information contained in the low frequencies of a Fourier transform to measure the similarity between cells in two consecutive images. We show that fingerprints are broadly applicable across cell types and image modalities, provided the image has sufficient structural information. Our tracker (TracX) uses fingerprints to reject unlikely assignments, thereby increasing tracking performance on published and newly generated long-term data sets. For Saccharomyces cerevisiae, we propose a comprehensive model for cell size control at the single-cell and population level centered on the Whi5 regulator, demonstrating how precise tracking can help uncover previously undescribed single-cell biology.
Collapse
Affiliation(s)
- Andreas P Cuny
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland.,Swiss Institute of Bioinformatics, Basel, Switzerland
| | - Aaron Ponti
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
| | - Tomas Kündig
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
| | - Fabian Rudolf
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland.,Swiss Institute of Bioinformatics, Basel, Switzerland
| | - Jörg Stelling
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland. .,Swiss Institute of Bioinformatics, Basel, Switzerland.
| |
Collapse
|
25
|
Jiang Q, Sudalagunta P, Silva MC, Canevarolo RR, Zhao X, Ahmed KT, Alugubelli RR, DeAvila G, Tungesvik A, Perez L, Gatenby RA, Gillies RJ, Baz R, Meads MB, Shain KH, Silva AS, Zhang W. CancerCellTracker: a brightfield time-lapse microscopy framework for cancer drug sensitivity estimation. Bioinformatics 2022; 38:4002-4010. [PMID: 35751591 PMCID: PMC9991899 DOI: 10.1093/bioinformatics/btac417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 05/18/2022] [Accepted: 06/22/2022] [Indexed: 12/24/2022] Open
Abstract
MOTIVATION Time-lapse microscopy is a powerful technique that relies on images of live cells cultured ex vivo that are captured at regular intervals of time to describe and quantify their behavior under certain experimental conditions. This imaging method has great potential in advancing the field of precision oncology by quantifying the response of cancer cells to various therapies and identifying the most efficacious treatment for a given patient. Digital image processing algorithms developed so far require high-resolution images involving very few cells originating from homogeneous cell line populations. We propose a novel framework that tracks cancer cells to capture their behavior and quantify cell viability to inform clinical decisions in a high-throughput manner. RESULTS The brightfield microscopy images a large number of patient-derived cells in an ex vivo reconstruction of the tumor microenvironment treated with 31 drugs for up to 6 days. We developed a robust and user-friendly pipeline CancerCellTracker that detects cells in co-culture, tracks these cells across time and identifies cell death events using changes in cell attributes. We validated our computational pipeline by comparing the timing of cell death estimates by CancerCellTracker from brightfield images and a fluorescent channel featuring ethidium homodimer. We benchmarked our results using a state-of-the-art algorithm implemented in ImageJ and previously published in the literature. We highlighted CancerCellTracker's efficiency in estimating the percentage of live cells in the presence of bone marrow stromal cells. AVAILABILITY AND IMPLEMENTATION https://github.com/compbiolabucf/CancerCellTracker. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Qibing Jiang
- Department of Computer Science, University of Central Florida, Orlando, FL 32816, USA
| | - Praneeth Sudalagunta
- Departments of Malignant Hematology and Chemical Biology and Molecular Medicine, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - Maria C Silva
- Departments of Malignant Hematology and Chemical Biology and Molecular Medicine, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - Rafael R Canevarolo
- Departments of Malignant Hematology and Chemical Biology and Molecular Medicine, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - Xiaohong Zhao
- Departments of Malignant Hematology and Chemical Biology and Molecular Medicine, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | | | - Raghunandan Reddy Alugubelli
- Departments of Malignant Hematology and Chemical Biology and Molecular Medicine, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - Gabriel DeAvila
- Departments of Malignant Hematology and Chemical Biology and Molecular Medicine, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - Alexandre Tungesvik
- Departments of Malignant Hematology and Chemical Biology and Molecular Medicine, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - Lia Perez
- Departments of Malignant Hematology and Chemical Biology and Molecular Medicine, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - Robert A Gatenby
- Departments of Malignant Hematology and Chemical Biology and Molecular Medicine, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - Robert J Gillies
- Departments of Malignant Hematology and Chemical Biology and Molecular Medicine, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - Rachid Baz
- Departments of Malignant Hematology and Chemical Biology and Molecular Medicine, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - Mark B Meads
- Departments of Malignant Hematology and Chemical Biology and Molecular Medicine, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - Kenneth H Shain
- Departments of Malignant Hematology and Chemical Biology and Molecular Medicine, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - Ariosto S Silva
- Departments of Malignant Hematology and Chemical Biology and Molecular Medicine, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - Wei Zhang
- Department of Computer Science, University of Central Florida, Orlando, FL 32816, USA
| |
Collapse
|
26
|
Ershov D, Phan MS, Pylvänäinen JW, Rigaud SU, Le Blanc L, Charles-Orszag A, Conway JRW, Laine RF, Roy NH, Bonazzi D, Duménil G, Jacquemet G, Tinevez JY. TrackMate 7: integrating state-of-the-art segmentation algorithms into tracking pipelines. Nat Methods 2022; 19:829-832. [PMID: 35654950 DOI: 10.1038/s41592-022-01507-1] [Citation(s) in RCA: 343] [Impact Index Per Article: 114.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 04/25/2022] [Indexed: 11/09/2022]
Abstract
TrackMate is an automated tracking software used to analyze bioimages and is distributed as a Fiji plugin. Here, we introduce a new version of TrackMate. TrackMate 7 is built to address the broad spectrum of modern challenges researchers face by integrating state-of-the-art segmentation algorithms into tracking pipelines. We illustrate qualitatively and quantitatively that these new capabilities function effectively across a wide range of bio-imaging experiments.
Collapse
Affiliation(s)
- Dmitry Ershov
- Institut Pasteur, Université de Paris Cité, Image Analysis Hub, Paris, France.,Institut Pasteur, Université de Paris Cité, Biostatistics and Bioinformatic Hub, Paris, France
| | - Minh-Son Phan
- Institut Pasteur, Université de Paris Cité, Image Analysis Hub, Paris, France
| | - Joanna W Pylvänäinen
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland.,Åbo Akademi University, Faculty of Science and Engineering, Biosciences, Turku, Finland.,Turku Bioimaging, University of Turku and Åbo Akademi University, Turku, Finland
| | - Stéphane U Rigaud
- Institut Pasteur, Université de Paris Cité, Image Analysis Hub, Paris, France
| | - Laure Le Blanc
- Institut Pasteur, Université Paris Cité, INSERM UMR1225, Pathogenesis of Vascular Infections unit, Paris, France
| | - Arthur Charles-Orszag
- Institut Pasteur, Université Paris Cité, INSERM UMR1225, Pathogenesis of Vascular Infections unit, Paris, France
| | - James R W Conway
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
| | - Romain F Laine
- MRC Laboratory for Molecular Cell Biology, University College London, London, UK.,The Francis Crick Institute, London, UK.,Micrographia Bio, Translation and Innovation Hub, London, UK
| | - Nathan H Roy
- Department of Microbiology and Immunology, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Daria Bonazzi
- Institut Pasteur, Université Paris Cité, INSERM UMR1225, Pathogenesis of Vascular Infections unit, Paris, France
| | - Guillaume Duménil
- Institut Pasteur, Université Paris Cité, INSERM UMR1225, Pathogenesis of Vascular Infections unit, Paris, France
| | - Guillaume Jacquemet
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland. .,Åbo Akademi University, Faculty of Science and Engineering, Biosciences, Turku, Finland. .,Turku Bioimaging, University of Turku and Åbo Akademi University, Turku, Finland.
| | - Jean-Yves Tinevez
- Institut Pasteur, Université de Paris Cité, Image Analysis Hub, Paris, France.
| |
Collapse
|
27
|
Tahk MJ, Torp J, Ali MAS, Fishman D, Parts L, Grätz L, Müller C, Keller M, Veiksina S, Laasfeld T, Rinken A. Live-cell microscopy or fluorescence anisotropy with budded baculoviruses-which way to go with measuring ligand binding to M 4 muscarinic receptors? Open Biol 2022; 12:220019. [PMID: 35674179 PMCID: PMC9175271 DOI: 10.1098/rsob.220019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 04/27/2022] [Indexed: 01/04/2023] Open
Abstract
M4 muscarinic acetylcholine receptor is a G protein-coupled receptor (GPCR) that has been associated with alcohol and cocaine abuse, Alzheimer's disease, and schizophrenia which makes it an interesting drug target. For many GPCRs, the high-affinity fluorescence ligands have expanded the options for high-throughput screening of drug candidates and serve as useful tools in fundamental receptor research. Here, we explored two TAMRA-labelled fluorescence ligands, UR-MK342 and UR-CG072, for development of assays for studying ligand-binding properties to M4 receptor. Using budded baculovirus particles as M4 receptor preparation and fluorescence anisotropy method, we measured the affinities and binding kinetics of both fluorescence ligands. Using the fluorescence ligands as reporter probes, the binding affinities of unlabelled ligands could be determined. Based on these results, we took a step towards a more natural system and developed a method using live CHO-K1-hM4R cells and automated fluorescence microscopy suitable for the routine determination of unlabelled ligand affinities. For quantitative image analysis, we developed random forest and deep learning-based pipelines for cell segmentation. The pipelines were integrated into the user-friendly open-source Aparecium software. Both image analysis methods were suitable for measuring fluorescence ligand saturation binding and kinetics as well as for screening binding affinities of unlabelled ligands.
Collapse
Affiliation(s)
- Maris-Johanna Tahk
- Institute of Chemistry, University of Tartu, Ravila 14a, 50411 Tartu, Estonia
| | - Jane Torp
- Institute of Chemistry, University of Tartu, Ravila 14a, 50411 Tartu, Estonia
| | - Mohammed A. S. Ali
- Department of Computer Science, University of Tartu, Narva Street 20, 51009 Tartu, Estonia
| | - Dmytro Fishman
- Department of Computer Science, University of Tartu, Narva Street 20, 51009 Tartu, Estonia
| | - Leopold Parts
- Department of Computer Science, University of Tartu, Narva Street 20, 51009 Tartu, Estonia
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridgeshire, UK
| | - Lukas Grätz
- Institute of Pharmacy, Faculty of Chemistry and Pharmacy, University of Regensburg, Universitätsstrasse 31, 93053 Regensburg, Germany
| | - Christoph Müller
- Institute of Pharmacy, Faculty of Chemistry and Pharmacy, University of Regensburg, Universitätsstrasse 31, 93053 Regensburg, Germany
| | - Max Keller
- Institute of Pharmacy, Faculty of Chemistry and Pharmacy, University of Regensburg, Universitätsstrasse 31, 93053 Regensburg, Germany
| | - Santa Veiksina
- Institute of Chemistry, University of Tartu, Ravila 14a, 50411 Tartu, Estonia
| | - Tõnis Laasfeld
- Institute of Chemistry, University of Tartu, Ravila 14a, 50411 Tartu, Estonia
- Department of Computer Science, University of Tartu, Narva Street 20, 51009 Tartu, Estonia
| | - Ago Rinken
- Institute of Chemistry, University of Tartu, Ravila 14a, 50411 Tartu, Estonia
| |
Collapse
|
28
|
Hoorali F, Khosravi H, Moradi B. Automatic microscopic diagnosis of diseases using an improved UNet++ architecture. Tissue Cell 2022; 76:101816. [DOI: 10.1016/j.tice.2022.101816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 05/02/2022] [Accepted: 05/04/2022] [Indexed: 12/01/2022]
|
29
|
Marzec M, Piórkowski A, Gertych A. Efficient automatic 3D segmentation of cell nuclei for high-content screening. BMC Bioinformatics 2022; 23:203. [PMID: 35641922 DOI: 10.1186/s12859-022-04737-4] [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: 09/03/2021] [Accepted: 05/17/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND High-content screening (HCS) is a pre-clinical approach for the assessment of drug efficacy. On modern platforms, it involves fluorescent image capture using three-dimensional (3D) scanning microscopy. Segmentation of cell nuclei in 3D images is an essential prerequisite to quantify captured fluorescence in cells for screening. However, this segmentation is challenging due to variabilities in cell confluency, drug-induced alterations in cell morphology, and gradual degradation of fluorescence with the depth of scanning. Despite advances in algorithms for segmenting nuclei for HCS, robust 3D methods that are insensitive to these conditions are still lacking. RESULTS We have developed an algorithm which first generates a 3D nuclear mask in the original images. Next, an iterative 3D marker-controlled watershed segmentation is applied to downsized images to segment adjacent nuclei under the mask. In the last step, borders of segmented nuclei are adjusted in the original images based on local nucleus and background intensities. The method was developed using a set of 10 3D images. Extensive tests on a separate set of 27 3D images containing 2,367 nuclei demonstrated that our method, in comparison with 6 reference methods, achieved the highest precision (PR = 0.97), recall (RE = 0.88) and F1-score (F1 = 0.93) of nuclei detection. The Jaccard index (JI = 0.83), which reflects the accuracy of nuclei delineation, was similar to that yielded by all reference approaches. Our method was on average more than twice as fast as the reference method that produced the best results. Additional tests carried out on three stacked 3D images comprising heterogenous nuclei yielded average PR = 0.96, RE = 0.84, F1 = 0.89, and JI = 0.80. CONCLUSIONS The high-performance metrics yielded by the proposed approach suggest that it can be used to reliably delineate nuclei in 3D images of monolayered and stacked cells exposed to cytotoxic drugs.
Collapse
Affiliation(s)
- Mariusz Marzec
- Faculty of Science and Technology, Institute of Biomedical Engineering, University of Silesia, Bedzinska St. 39, 41-200, Sosnowiec, Poland.
| | - Adam Piórkowski
- Department of Biocybernetics and Biomedical Engineering, AGH University of Science and Technology, Mickiewicza 30, 30-059, Cracow, Poland
| | - Arkadiusz Gertych
- Department of Surgery, Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA.,Faculty of Biomedical Engineering, Silesian University of Technology, Roosvelta 40, 41-800, Zabrze, Poland
| |
Collapse
|
30
|
Gwatimba A, Rosenow T, Stick SM, Kicic A, Iosifidis T, Karpievitch YV. AI-Driven Cell Tracking to Enable High-Throughput Drug Screening Targeting Airway Epithelial Repair for Children with Asthma. J Pers Med 2022; 12:jpm12050809. [PMID: 35629232 PMCID: PMC9146422 DOI: 10.3390/jpm12050809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 05/08/2022] [Accepted: 05/13/2022] [Indexed: 11/16/2022] Open
Abstract
The airway epithelium of children with asthma is characterized by aberrant repair that may be therapeutically modifiable. The development of epithelial-targeting therapeutics that enhance airway repair could provide a novel treatment avenue for childhood asthma. Drug discovery efforts utilizing high-throughput live cell imaging of patient-derived airway epithelial culture-based wound repair assays can be used to identify compounds that modulate airway repair in childhood asthma. Manual cell tracking has been used to determine cell trajectories and wound closure rates, but is time consuming, subject to bias, and infeasible for high-throughput experiments. We therefore developed software, EPIC, that automatically tracks low-resolution low-framerate cells using artificial intelligence, analyzes high-throughput drug screening experiments and produces multiple wound repair metrics and publication-ready figures. Additionally, unlike available cell trackers that perform cell segmentation, EPIC tracks cells using bounding boxes and thus has simpler and faster training data generation requirements for researchers working with other cell types. EPIC outperformed publicly available software in our wound repair datasets by achieving human-level cell tracking accuracy in a fraction of the time. We also showed that EPIC is not limited to airway epithelial repair for children with asthma but can be applied in other cellular contexts by outperforming the same software in the Cell Tracking with Mitosis Detection Challenge (CTMC) dataset. The CTMC is the only established cell tracking benchmark dataset that is designed for cell trackers utilizing bounding boxes. We expect our open-source and easy-to-use software to enable high-throughput drug screening targeting airway epithelial repair for children with asthma.
Collapse
Affiliation(s)
- Alphons Gwatimba
- Wal-Yan Respiratory Research Centre, Telethon Kids Institute, University of Western Australia, Perth, WA 6009, Australia; (T.R.); (S.M.S.); (A.K.); (T.I.); (Y.V.K.)
- School of Computer Science and Software Engineering, University of Western Australia, Nedlands, WA 6009, Australia
- Correspondence:
| | - Tim Rosenow
- Wal-Yan Respiratory Research Centre, Telethon Kids Institute, University of Western Australia, Perth, WA 6009, Australia; (T.R.); (S.M.S.); (A.K.); (T.I.); (Y.V.K.)
- Centre for Microscopy, Characterisation and Analysis, University of Western Australia, Nedlands, WA 6009, Australia
| | - Stephen M. Stick
- Wal-Yan Respiratory Research Centre, Telethon Kids Institute, University of Western Australia, Perth, WA 6009, Australia; (T.R.); (S.M.S.); (A.K.); (T.I.); (Y.V.K.)
- Division of Paediatrics, Medical School, University of Western Australia, Nedlands, WA 6009, Australia
- Department of Respiratory and Sleep Medicine, Perth Children’s Hospital, Nedlands, WA 6009, Australia
- Centre for Cell Therapy and Regenerative Medicine, School of Medicine, University of Western Australia, Nedlands, WA 6009, Australia
| | - Anthony Kicic
- Wal-Yan Respiratory Research Centre, Telethon Kids Institute, University of Western Australia, Perth, WA 6009, Australia; (T.R.); (S.M.S.); (A.K.); (T.I.); (Y.V.K.)
- Division of Paediatrics, Medical School, University of Western Australia, Nedlands, WA 6009, Australia
- Centre for Cell Therapy and Regenerative Medicine, School of Medicine, University of Western Australia, Nedlands, WA 6009, Australia
- School of Population Health, Curtin University, Bentley, WA 6102, Australia
| | - Thomas Iosifidis
- Wal-Yan Respiratory Research Centre, Telethon Kids Institute, University of Western Australia, Perth, WA 6009, Australia; (T.R.); (S.M.S.); (A.K.); (T.I.); (Y.V.K.)
- Centre for Cell Therapy and Regenerative Medicine, School of Medicine, University of Western Australia, Nedlands, WA 6009, Australia
- School of Population Health, Curtin University, Bentley, WA 6102, Australia
| | - Yuliya V. Karpievitch
- Wal-Yan Respiratory Research Centre, Telethon Kids Institute, University of Western Australia, Perth, WA 6009, Australia; (T.R.); (S.M.S.); (A.K.); (T.I.); (Y.V.K.)
- School of Biomedical Sciences, University of Western Australia, Nedlands, WA 6009, Australia
| |
Collapse
|
31
|
Rasal T, Veerakumar T, Subudhi BN, Esakkirajan S. A novel approach for reduction of the noise from microscopy images using Fourier decomposition. Biocybern Biomed Eng 2022. [DOI: 10.1016/j.bbe.2022.05.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
|
32
|
Le AV, Fenech M. Image-Based Experimental Measurement Techniques to Characterize Velocity Fields in Blood Microflows. Front Physiol 2022; 13:886675. [PMID: 35574441 PMCID: PMC9099138 DOI: 10.3389/fphys.2022.886675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 04/04/2022] [Indexed: 11/13/2022] Open
Abstract
Predicting blood microflow in both simple and complex geometries is challenging because of the composition and behavior of the blood at microscale. However, characterization of the velocity in microchannels is the key for gaining insights into cellular interactions at the microscale, mechanisms of diseases, and efficacy of therapeutic solutions. Image-based measurement techniques are a subset of methods for measuring the local flow velocity that typically utilize tracer particles for flow visualization. In the most basic form, a high-speed camera and microscope setup are the only requirements for data acquisition; however, the development of image processing algorithms and equipment has made current image-based techniques more sophisticated. This mini review aims to provide a succinct and accessible overview of image-based experimental measurement techniques to characterize the velocity field of blood microflow. The following techniques are introduced: cell tracking velocimetry, kymographs, micro-particle velocimetry, and dual-slit photometry as entry techniques for measuring various velocity fields either in vivo or in vitro.
Collapse
Affiliation(s)
- Andy Vinh Le
- Department of Mechanical Engineering, University of Ottawa, Ottawa, ON, Canada
- Centre de Biochimie Structurale, CNRS UMR 5048—INSERM UMR 1054, University of Montpellier, Montpellier, France
| | - Marianne Fenech
- Department of Mechanical Engineering, University of Ottawa, Ottawa, ON, Canada
- *Correspondence: Marianne Fenech,
| |
Collapse
|
33
|
Establishing trajectories of moving objects without identities: The intricacies of cell tracking and a solution. INFORM SYST 2022. [DOI: 10.1016/j.is.2021.101955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
34
|
|
35
|
Aragaki H, Ogoh K, Kondo Y, Aoki K. LIM Tracker: a software package for cell tracking and analysis with advanced interactivity. Sci Rep 2022; 12:2702. [PMID: 35177675 PMCID: PMC8854686 DOI: 10.1038/s41598-022-06269-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 01/24/2022] [Indexed: 11/24/2022] Open
Abstract
Cell tracking is one of the most critical tools for time-lapse image analysis to observe cell behavior and cell lineages over a long period of time. However, the accompanying graphical user interfaces are often difficult to use and do not incorporate seamless manual correction, data analysis tools, or simple training set design tools if it is machine learning based. In this paper, we introduce our cell tracking software "LIM Tracker". This software has a conventional tracking function consisting of recognition processing and link processing, a sequential search-type tracking function based on pattern matching, and a manual tracking function. LIM Tracker enables the seamless use of these functions. In addition, the system incorporates a highly interactive and interlocking data visualization method, which displays analysis result in real time, making it possible to flexibly correct the data and reduce the burden of tracking work. Moreover, recognition functions with deep learning (DL) are also available, which can be used for a wide range of targets including stain-free images. LIM Tracker allows researchers to track living objects with good usability and high versatility for various targets. We present a tracking case study based on fluorescence microscopy images (NRK-52E/EKAREV-NLS cells or MCF-10A/H2B-iRFP-P2A-mScarlet-I-hGem-P2A-PIP-NLS-mNeonGreen cells) and phase contrast microscopy images (Glioblastoma-astrocytoma U373 cells). LIM Tracker is implemented as a plugin for ImageJ/Fiji. The software can be downloaded from https://github.com/LIMT34/LIM-Tracker .
Collapse
Affiliation(s)
- Hideya Aragaki
- Innovation and Core Technology Management, Olympus Corporation, Kuboyama 2-3, Hachioji, Tokyo, 192-8512, Japan.
| | - Katsunori Ogoh
- Innovation and Core Technology Management, Olympus Corporation, Kuboyama 2-3, Hachioji, Tokyo, 192-8512, Japan
| | - Yohei Kondo
- Quantitative Biology Research Group, Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, 5-1 Higashiyama, Myodaiji-cho, Okazaki, Aichi, 444-8787, Japan
- Division of Quantitative Biology, National Institute for Basic Biology, National Institutes of Natural Sciences, 5-1 Higashiyama, Myodaiji-cho, Okazaki, Aichi, 444-8787, Japan
- Department of Basic Biology, School of Life Science, SOKENDAI (The Graduate University for Advanced Studies), 5-1 Higashiyama, Myodaiji-cho, Okazaki, Aichi, 444-8787, Japan
| | - Kazuhiro Aoki
- Quantitative Biology Research Group, Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, 5-1 Higashiyama, Myodaiji-cho, Okazaki, Aichi, 444-8787, Japan
- Division of Quantitative Biology, National Institute for Basic Biology, National Institutes of Natural Sciences, 5-1 Higashiyama, Myodaiji-cho, Okazaki, Aichi, 444-8787, Japan
- Department of Basic Biology, School of Life Science, SOKENDAI (The Graduate University for Advanced Studies), 5-1 Higashiyama, Myodaiji-cho, Okazaki, Aichi, 444-8787, Japan
| |
Collapse
|
36
|
Bioimaging approaches for quantification of individual cell behavior during cell fate decisions. Biochem Soc Trans 2022; 50:513-527. [PMID: 35166330 DOI: 10.1042/bst20210534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 01/10/2022] [Accepted: 01/24/2022] [Indexed: 11/17/2022]
Abstract
Tracking individual cells has allowed a new understanding of cellular behavior in human health and disease by adding a dynamic component to the already complex heterogeneity of single cells. Technically, despite countless advances, numerous experimental variables can affect data collection and interpretation and need to be considered. In this review, we discuss the main technical aspects and biological findings in the analysis of the behavior of individual cells. We discuss the most relevant contributions provided by these approaches in clinically relevant human conditions like embryo development, stem cells biology, inflammation, cancer and microbiology, along with the cellular mechanisms and molecular pathways underlying these conditions. We also discuss the key technical aspects to be considered when planning and performing experiments involving the analysis of individual cells over long periods. Despite the challenges in automatic detection, features extraction and long-term tracking that need to be tackled, the potential impact of single-cell bioimaging is enormous in understanding the pathogenesis and development of new therapies in human pathophysiology.
Collapse
|
37
|
Arzt M, Deschamps J, Schmied C, Pietzsch T, Schmidt D, Tomancak P, Haase R, Jug F. LABKIT: Labeling and Segmentation Toolkit for Big Image Data. FRONTIERS IN COMPUTER SCIENCE 2022. [DOI: 10.3389/fcomp.2022.777728] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
We present LABKIT, a user-friendly Fiji plugin for the segmentation of microscopy image data. It offers easy to use manual and automated image segmentation routines that can be rapidly applied to single- and multi-channel images as well as to timelapse movies in 2D or 3D. LABKIT is specifically designed to work efficiently on big image data and enables users of consumer laptops to conveniently work with multiple-terabyte images. This efficiency is achieved by using ImgLib2 and BigDataViewer as well as a memory efficient and fast implementation of the random forest based pixel classification algorithm as the foundation of our software. Optionally we harness the power of graphics processing units (GPU) to gain additional runtime performance. LABKIT is easy to install on virtually all laptops and workstations. Additionally, LABKIT is compatible with high performance computing (HPC) clusters for distributed processing of big image data. The ability to use pixel classifiers trained in LABKIT via the ImageJ macro language enables our users to integrate this functionality as a processing step in automated image processing workflows. Finally, LABKIT comes with rich online resources such as tutorials and examples that will help users to familiarize themselves with available features and how to best use LABKIT in a number of practical real-world use-cases.
Collapse
|
38
|
No Cell Left behind: Automated, Stochastic, Physics-Based Tracking of Every Cell in a Dense, Growing Colony. ALGORITHMS 2022. [DOI: 10.3390/a15020051] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Motivation: Precise tracking of individual cells—especially tracking the family lineage, for example in a developing embryo—has widespread applications in biology and medicine. Due to significant noise in microscope images, existing methods have difficulty precisely tracking cell activities. These difficulties often require human intervention to resolve. Humans are helpful because our brain naturally and automatically builds a simulation “model” of any scene that we observe. Because we understand simple truths about the world—for example cells can move and divide, but they cannot instantaneously move vast distances—this model “in our heads” helps us to severely constrain the possible interpretations of what we see, allowing us to easily distinguish signal from noise, and track the motion of cells even in the presence of extreme levels of noise that would completely confound existing automated methods. Results: Here, we mimic the ability of the human brain by building an explicit computer simulation model of the scene. Our simulated cells are programmed to allow movement and cell division consistent with reality. At each video frame, we stochastically generate millions of nearby “Universes” and evolve them stochastically to the next frame. We then find and fit the best universes to reality by minimizing the residual between the real image frame and a synthetic image of the simulation. The rule-based simulation puts extremely stringent constraints on possible interpretations of the data, allowing our system to perform far better than existing methods even in the presense of extreme levels of image noise. We demonstrate the viability of this method by accurately tracking every cell in a colony that grows from 4 to over 300 individuals, doing about as well as a human can in the difficult task of tracking cell lineages.
Collapse
|
39
|
Sugawara K, Çevrim Ç, Averof M. Tracking cell lineages in 3D by incremental deep learning. eLife 2022; 11:e69380. [PMID: 34989675 PMCID: PMC8741210 DOI: 10.7554/elife.69380] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 12/07/2021] [Indexed: 11/13/2022] Open
Abstract
Deep learning is emerging as a powerful approach for bioimage analysis. Its use in cell tracking is limited by the scarcity of annotated data for the training of deep-learning models. Moreover, annotation, training, prediction, and proofreading currently lack a unified user interface. We present ELEPHANT, an interactive platform for 3D cell tracking that addresses these challenges by taking an incremental approach to deep learning. ELEPHANT provides an interface that seamlessly integrates cell track annotation, deep learning, prediction, and proofreading. This enables users to implement cycles of incremental learning starting from a few annotated nuclei. Successive prediction-validation cycles enrich the training data, leading to rapid improvements in tracking performance. We test the software's performance against state-of-the-art methods and track lineages spanning the entire course of leg regeneration in a crustacean over 1 week (504 timepoints). ELEPHANT yields accurate, fully-validated cell lineages with a modest investment in time and effort.
Collapse
Affiliation(s)
- Ko Sugawara
- Institut de Génomique Fonctionnelle de Lyon (IGFL), École Normale Supérieure de LyonLyonFrance
- Centre National de la Recherche Scientifique (CNRS)ParisFrance
| | - Çağrı Çevrim
- Institut de Génomique Fonctionnelle de Lyon (IGFL), École Normale Supérieure de LyonLyonFrance
- Centre National de la Recherche Scientifique (CNRS)ParisFrance
| | - Michalis Averof
- Institut de Génomique Fonctionnelle de Lyon (IGFL), École Normale Supérieure de LyonLyonFrance
- Centre National de la Recherche Scientifique (CNRS)ParisFrance
| |
Collapse
|
40
|
Zhao T, Yin Z. Weakly Supervised Cell Segmentation by Point Annotation. IEEE TRANSACTIONS ON MEDICAL IMAGING 2021; 40:2736-2747. [PMID: 33347404 DOI: 10.1109/tmi.2020.3046292] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
We propose weakly supervised training schemes to train end-to-end cell segmentation networks that only require a single point annotation per cell as the training label and generate a high-quality segmentation mask close to those fully supervised methods using mask annotation on cells. Three training schemes are investigated to train cell segmentation networks, using the point annotation. First, self-training is performed to learn additional information near the annotated points. Next, co-training is applied to learn more cell regions using multiple networks that supervise each other. Finally, a hybrid-training scheme is proposed to leverage the advantages of both self-training and co-training. During the training process, we propose a divergence loss to avoid the overfitting and a consistency loss to enforce the consensus among multiple co-trained networks. Furthermore, we propose weakly supervised learning with human in the loop, aiming at achieving high segmentation accuracy and annotation efficiency simultaneously. Evaluated on two benchmark datasets, our proposal achieves high-quality cell segmentation results comparable to the fully supervised methods, but with much less amount of human annotation effort.
Collapse
|
41
|
Löffler K, Scherr T, Mikut R. A graph-based cell tracking algorithm with few manually tunable parameters and automated segmentation error correction. PLoS One 2021; 16:e0249257. [PMID: 34492015 PMCID: PMC8423278 DOI: 10.1371/journal.pone.0249257] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 08/03/2021] [Indexed: 11/29/2022] Open
Abstract
Automatic cell segmentation and tracking enables to gain quantitative insights into the processes driving cell migration. To investigate new data with minimal manual effort, cell tracking algorithms should be easy to apply and reduce manual curation time by providing automatic correction of segmentation errors. Current cell tracking algorithms, however, are either easy to apply to new data sets but lack automatic segmentation error correction, or have a vast set of parameters that needs either manual tuning or annotated data for parameter tuning. In this work, we propose a tracking algorithm with only few manually tunable parameters and automatic segmentation error correction. Moreover, no training data is needed. We compare the performance of our approach to three well-performing tracking algorithms from the Cell Tracking Challenge on data sets with simulated, degraded segmentation—including false negatives, over- and under-segmentation errors. Our tracking algorithm can correct false negatives, over- and under-segmentation errors as well as a mixture of the aforementioned segmentation errors. On data sets with under-segmentation errors or a mixture of segmentation errors our approach performs best. Moreover, without requiring additional manual tuning, our approach ranks several times in the top 3 on the 6th edition of the Cell Tracking Challenge.
Collapse
Affiliation(s)
- Katharina Löffler
- Institute for Automation and Applied Informatics, Karlsruhe Institute of Technology, Eggenstein-Leopoldshafen, Germany
- Institute of Biological and Chemical Systems - Biological Information Processing, Karlsruhe Institute of Technology, Eggenstein-Leopoldshafen, Germany
- * E-mail:
| | - Tim Scherr
- Institute for Automation and Applied Informatics, Karlsruhe Institute of Technology, Eggenstein-Leopoldshafen, Germany
| | - Ralf Mikut
- Institute for Automation and Applied Informatics, Karlsruhe Institute of Technology, Eggenstein-Leopoldshafen, Germany
| |
Collapse
|
42
|
Xu B, Shi J, Lu M, Cong J, Wang L, Nener B. An Automated Cell Tracking Approach With Multi-Bernoulli Filtering and Ant Colony Labor Division. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2021; 18:1850-1863. [PMID: 31751247 DOI: 10.1109/tcbb.2019.2954502] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
In this article, we take as inspiration the labor division into scouts and workers in an ant colony and propose a novel approach for automated cell tracking in the framework of multi-Bernoulli random finite sets. To approximate the Bernoulli parameter sets, we first define an existence probability of an ant colony as well as its discrete density distribution. During foraging, the behavior of scouts is modeled as a chaotic movement to produce a set of potential candidates. Afterwards, a group of workers, i.e., a worker ant colony, is recruited for each candidate, which then embark on gathering heuristic information in a self-organized way. Finally, the pheromone field is formed by the corresponding worker ant colony, from which the Bernoulli parameter is derived and the state of the cell is estimated accordingly to be associated with the existing tracks. Performance comparisons with other previous approaches are conducted on both simulated and real cell image sequences and show the superiority of this algorithm.
Collapse
|
43
|
Hu H, Liu A, Zhou Q, Guan Q, Li X, Chen Q. An adaptive learning method of anchor shape priors for biological cells detection and segmentation. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2021; 208:106260. [PMID: 34273675 DOI: 10.1016/j.cmpb.2021.106260] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 06/24/2021] [Indexed: 06/13/2023]
Abstract
BACKGROUND AND OBJECTIVE Owing to the variable shapes, large size difference, uneven grayscale and dense distribution among biological cells in an image, it is still a challenging task for the standard Mask R-CNN to accurately detect and segment cells. Especially, the state-of-the-art anchor-based methods fail to generate the anchors of sufficient scales effectively according to the various sizes and shapes of cells, thereby hardly covering all scales of cells. METHODS We propose an adaptive approach to learn the anchor shape priors from data samples, and the aspect ratios and the number of anchor boxes can be dynamically adjusted by using ISODATA clustering algorithm instead of human prior knowledge in this work. To solve the identification difficulties for small objects owing to the multiple down-samplings in a deep learning-based method, a densification strategy of candidate anchors is presented to enhance the effects of identifying tinny size cells. Finally, a series of comparative experiments are conducted on various datasets to select appropriate a network structure and verify the effectiveness of the proposed methods. RESULTS The results show that the ResNet-50-FPN combining the ISODATA method and densification strategy can obtain better performance than other methods in multiple metrics (including AP, Precision, Recall, Dice and PQ) for various biological cell datasets, such as U373, GoTW1, SIM+ and T24. CONCLUSIONS Our adaptive algorithm could effectively learn the anchor shape priors from the various sizes and shapes of cells. It is promising and encouraging for a real-world anchor-based detection and segmentation application of biomedical engineering in the future.
Collapse
Affiliation(s)
- Haigen Hu
- College of Computer Science and Technology, Zhejiang University of Technology, Hangzhou 310023, PR China; Key Laboratory of Visual Media Intelligent Processing Technology of Zhejiang Province, Hangzhou 310023, PR China.
| | - Aizhu Liu
- College of Computer Science and Technology, Zhejiang University of Technology, Hangzhou 310023, PR China; Key Laboratory of Visual Media Intelligent Processing Technology of Zhejiang Province, Hangzhou 310023, PR China
| | - Qianwei Zhou
- College of Computer Science and Technology, Zhejiang University of Technology, Hangzhou 310023, PR China; Key Laboratory of Visual Media Intelligent Processing Technology of Zhejiang Province, Hangzhou 310023, PR China
| | - Qiu Guan
- College of Computer Science and Technology, Zhejiang University of Technology, Hangzhou 310023, PR China; Key Laboratory of Visual Media Intelligent Processing Technology of Zhejiang Province, Hangzhou 310023, PR China
| | - Xiaoxin Li
- College of Computer Science and Technology, Zhejiang University of Technology, Hangzhou 310023, PR China; Key Laboratory of Visual Media Intelligent Processing Technology of Zhejiang Province, Hangzhou 310023, PR China
| | - Qi Chen
- College of Computer Science and Technology, Zhejiang University of Technology, Hangzhou 310023, PR China; Key Laboratory of Visual Media Intelligent Processing Technology of Zhejiang Province, Hangzhou 310023, PR China.
| |
Collapse
|
44
|
Liu Q, Gaeta IM, Zhao M, Deng R, Jha A, Millis BA, Mahadevan-Jansen A, Tyska MJ, Huo Y. ASIST: Annotation-free synthetic instance segmentation and tracking by adversarial simulations. Comput Biol Med 2021; 134:104501. [PMID: 34107436 PMCID: PMC8263511 DOI: 10.1016/j.compbiomed.2021.104501] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 05/14/2021] [Accepted: 05/15/2021] [Indexed: 10/21/2022]
Abstract
BACKGROUND The quantitative analysis of microscope videos often requires instance segmentation and tracking of cellular and subcellular objects. The traditional method consists of two stages: (1) performing instance object segmentation of each frame, and (2) associating objects frame-by-frame. Recently, pixel-embedding-based deep learning approaches these two steps simultaneously as a single stage holistic solution. Pixel-embedding-based learning forces similar feature representation of pixels from the same object, while maximizing the difference of feature representations from different objects. However, such deep learning methods require consistent annotations not only spatially (for segmentation), but also temporally (for tracking). In computer vision, annotated training data with consistent segmentation and tracking is resource intensive, the severity of which is multiplied in microscopy imaging due to (1) dense objects (e.g., overlapping or touching), and (2) high dynamics (e.g., irregular motion and mitosis). Adversarial simulations have provided successful solutions to alleviate the lack of such annotations in dynamics scenes in computer vision, such as using simulated environments (e.g., computer games) to train real-world self-driving systems. METHODS In this paper, we propose an annotation-free synthetic instance segmentation and tracking (ASIST) method with adversarial simulation and single-stage pixel-embedding based learning. CONTRIBUTION The contribution of this paper is three-fold: (1) the proposed method aggregates adversarial simulations and single-stage pixel-embedding based deep learning (2) the method is assessed with both the cellular (i.e., HeLa cells); and subcellular (i.e., microvilli) objects; and (3) to the best of our knowledge, this is the first study to explore annotation-free instance segmentation and tracking study for microscope videos. RESULTS The ASIST method achieved an important step forward, when compared with fully supervised approaches: ASIST shows 7%-11% higher segmentation, detection and tracking performance on microvilli relative to fully supervised methods, and comparable performance on Hela cell videos.
Collapse
Affiliation(s)
- Quan Liu
- Vanderbilt University, Computer Science, Nashville, TN, 37215, USA
| | - Isabella M Gaeta
- Vanderbilt University, Cell and Developmental Biology, Nashville, TN, 37215, USA
| | - Mengyang Zhao
- Tufts University, Computer Science, Medford, MA, 02155, USA
| | - Ruining Deng
- Vanderbilt University, Computer Science, Nashville, TN, 37215, USA
| | - Aadarsh Jha
- Vanderbilt University, Computer Science, Nashville, TN, 37215, USA
| | - Bryan A Millis
- Vanderbilt University, Cell and Developmental Biology, Nashville, TN, 37215, USA
| | | | - Matthew J Tyska
- Vanderbilt University, Cell and Developmental Biology, Nashville, TN, 37215, USA
| | - Yuankai Huo
- Vanderbilt University, Computer Science, Nashville, TN, 37215, USA.
| |
Collapse
|
45
|
Xu B, Lu M, Shi J, Cong J, Nener B. A Joint Tracking Approach via Ant Colony Evolution for Quantitative Cell Cycle Analysis. IEEE J Biomed Health Inform 2021; 25:2338-2349. [PMID: 33079687 DOI: 10.1109/jbhi.2020.3032592] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
In this paper, we use an ant colony heuristic method to tackle the integration of data association and state estimation in the presence of cell mitosis, morphological change and uncertainty of measurement. Our approach first models the scouting behavior of an unlabeled ant colony as a chaotic process to generate a set of cell candidates in the current frame, then a labeled ant colony foraging process is modeled to construct an interframe matching between previously estimated cell states and current cell candidates through minimizing the optimal sub-pattern assignment metric for track (OSPA-T). The states of cells in the current frame are finally estimated using labeled ant colonies via a multi-Bernoulli parameter set approximated by individual food pheromone fields and heuristic information within the same region of support, the resulting trail pheromone fields over frames constitutes the cell lineage trees of the tracks. A four-stage track recovery strategy is proposed to monitor the history of all established tracks to reconstruct broken tracks in a computationally economic way. The labeling method used in this work is an improvement on previous techniques. The method has been evaluated on publicly available, challenging cell image sequences, and a satisfied performance improvement is achieved in contrast to the state-of-the-art methods.
Collapse
|
46
|
Belyaev I, Praetorius JP, Medyukhina A, Figge MT. Enhanced segmentation of label-free cells for automated migration and interaction tracking. Cytometry A 2021; 99:1218-1229. [PMID: 34060210 DOI: 10.1002/cyto.a.24466] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 05/25/2021] [Indexed: 02/01/2023]
Abstract
In biomedical research, the migration behavior of cells and interactions between various cell types are frequently studied subjects. An automated and quantitative analysis of time-lapse microscopy data is an essential component of these studies, especially when characteristic migration patterns need to be identified. Plenty of software tools have been developed to serve this need. However, the majority of algorithms is designed for fluorescently labeled cells, even though it is well-known that fluorescent labels can substantially interfere with the physiological behavior of interacting cells. We here present a fully revised version of our algorithm for migration and interaction tracking (AMIT), which includes a novel segmentation approach. This approach allows segmenting label-free cells with high accuracy and also enables detecting almost all cells within the field of view. With regard to cell tracking, we designed and implemented a new method for cluster detection and splitting. This method does not rely on any geometrical characteristics of individual objects inside a cluster but relies on monitoring the events of cell-cell fusion from and cluster fission into single cells forward and backward in time. We demonstrate that focusing on these events provides accurate splitting of transient clusters. Furthermore, the substantially improved quantitative analysis of cell migration by the revised version of AMIT is more than two orders of magnitude faster than the previous implementation, which makes it feasible to process video data at higher spatial and temporal resolutions.
Collapse
Affiliation(s)
- Ivan Belyaev
- Applied Systems Biology, Leibniz Institute for Natural Product Research and Infection Biology - Hans Knöll Institute (HKI), Jena, Germany.,Faculty of Biological Sciences, Friedrich Schiller University Jena, Jena, Germany
| | - Jan-Philipp Praetorius
- Applied Systems Biology, Leibniz Institute for Natural Product Research and Infection Biology - Hans Knöll Institute (HKI), Jena, Germany.,Faculty of Biological Sciences, Friedrich Schiller University Jena, Jena, Germany
| | - Anna Medyukhina
- Applied Systems Biology, Leibniz Institute for Natural Product Research and Infection Biology - Hans Knöll Institute (HKI), Jena, Germany.,Center for Bioimage Informatics, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Marc Thilo Figge
- Applied Systems Biology, Leibniz Institute for Natural Product Research and Infection Biology - Hans Knöll Institute (HKI), Jena, Germany.,Institute of Microbiology, Faculty of Biological Sciences, Friedrich Schiller University Jena, Jena, Germany
| |
Collapse
|
47
|
Tian C, Yang C, Spencer SL. EllipTrack: A Global-Local Cell-Tracking Pipeline for 2D Fluorescence Time-Lapse Microscopy. Cell Rep 2021; 32:107984. [PMID: 32755578 DOI: 10.1016/j.celrep.2020.107984] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 05/29/2020] [Accepted: 07/09/2020] [Indexed: 12/12/2022] Open
Abstract
Time-lapse microscopy provides an unprecedented opportunity to monitor single-cell dynamics. However, tracking cells for long periods remains a technical challenge, especially for multi-day, large-scale movies with rapid cell migration, high cell density, and drug treatments that alter cell morphology/behavior. Here, we present EllipTrack, a global-local cell-tracking pipeline optimized for tracking such movies. EllipTrack first implements a global track-linking algorithm to construct tracks that maximize the probability of cell lineages. Tracking mistakes are then corrected with a local track-correction module in which tracks generated by the global algorithm are systematically examined and amended if a more probable alternative can be found. Through benchmarking, we show that EllipTrack outperforms state-of-the-art cell trackers and generates nearly error-free cell lineages for multiple large-scale movies. In addition, EllipTrack can adapt to time- and cell-density-dependent changes in cell migration speeds and requires minimal training datasets. EllipTrack is available at https://github.com/tianchengzhe/EllipTrack.
Collapse
Affiliation(s)
- Chengzhe Tian
- Department of Biochemistry, University of Colorado Boulder, Boulder, CO 80303, USA; BioFrontiers Institute, University of Colorado Boulder, Boulder, CO 80303, USA.
| | - Chen Yang
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado Boulder, Boulder, CO 80303, USA; BioFrontiers Institute, University of Colorado Boulder, Boulder, CO 80303, USA
| | - Sabrina L Spencer
- Department of Biochemistry, University of Colorado Boulder, Boulder, CO 80303, USA; BioFrontiers Institute, University of Colorado Boulder, Boulder, CO 80303, USA.
| |
Collapse
|
48
|
Heryanto YD, Cheng CY, Uchida Y, Mimura K, Ishii M, Yamada R. Integrated analysis of cell shape and movement in moving frame. Biol Open 2021; 10:bio058512. [PMID: 33664097 PMCID: PMC8015248 DOI: 10.1242/bio.058512] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Accepted: 02/11/2021] [Indexed: 11/20/2022] Open
Abstract
The cell's movement and morphological change are two interrelated cellular processes. An integrated analysis is needed to explore the relationship between them. However, it has been challenging to investigate them as a whole. The cell's trajectory can be described by its speed, curvature, and torsion. On the other hand, the three-dimensional (3D) cell shape can be studied by using a shape descriptor such as spherical harmonic (SH) descriptor, which is an extension of a Fourier transform in 3D space. We propose a novel method using parallel-transport (PT) to integrate these shape-movement data by using moving frames as the 3D-shape coordinate system. This moving frame is purely determined by the velocity vector. On this moving frame, the movement change will influence the coordinate system for shape analysis. By analyzing the change of the SH coefficients over time in the moving frame, we can observe the relationship between shape and movement. We illustrate the application of our approach using simulated and real datasets in this paper.
Collapse
Affiliation(s)
- Yusri Dwi Heryanto
- Unit of Statistical Genetics, Center for Genomic Medicine, Graduate School of Medicine Kyoto University, Kyoto, 606-8507, Japan
| | - Chin-Yi Cheng
- Unit of Statistical Genetics, Center for Genomic Medicine, Graduate School of Medicine Kyoto University, Kyoto, 606-8507, Japan
| | - Yutaka Uchida
- Department of Immunology and Cell Biology, Graduate School of Medicine and Frontier Biosciences, Osaka University, Osaka, 565-0871, Japan
| | - Kazushi Mimura
- Department of Intelligent Systems, Graduate School of Information Sciences, Hiroshima City University, Hiroshima, 731-3194 Japan
| | - Masaru Ishii
- Department of Immunology and Cell Biology, Graduate School of Medicine and Frontier Biosciences, Osaka University, Osaka, 565-0871, Japan
| | - Ryo Yamada
- Unit of Statistical Genetics, Center for Genomic Medicine, Graduate School of Medicine Kyoto University, Kyoto, 606-8507, Japan
| |
Collapse
|
49
|
Isensee F, Jaeger PF, Kohl SAA, Petersen J, Maier-Hein KH. nnU-Net: a self-configuring method for deep learning-based biomedical image segmentation. Nat Methods 2021. [DOI: 10.1038/s41592-020-01008-z 10.1038/s41592-020-01008-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
|
50
|
Lutton EJ, Collier S, Bretschneider T. A Curvature-Enhanced Random Walker Segmentation Method for Detailed Capture of 3D Cell Surface Membranes. IEEE TRANSACTIONS ON MEDICAL IMAGING 2021; 40:514-526. [PMID: 33052849 DOI: 10.1109/tmi.2020.3031029] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
High-resolution 3D microscopy is a fast advancing field and requires new techniques in image analysis to handle these new datasets. In this work, we focus on detailed 3D segmentation of Dictyostelium cells undergoing macropinocytosis captured on an iSPIM microscope. We propose a novel random walker-based method with a curvature-based enhancement term, with the aim of capturing fine protrusions, such as filopodia and deep invaginations, such as macropinocytotic cups, on the cell surface. We tested our method on both real and synthetic 3D image volumes, demonstrating that the inclusion of the curvature enhancement term can improve the segmentation of the aforementioned features. We show that our method performs better than other state of the art segmentation methods in 3D images of Dictyostelium cells, and performs competitively against CNN-based methods in two Cell Tracking Challenge datasets, demonstrating the ability to obtain accurate segmentations without the requirement of large training datasets. We also present an automated seeding method for microscopy data, which, combined with the curvature-enhanced random walker method, enables the segmentation of large time series with minimal input from the experimenter.
Collapse
|