Elsevier

Cellular Immunology

Volume 350, April 2020, 104086
Cellular Immunology

Research paper
Reprint of “Multi-modal image cytometry approach – From dynamic to whole organ imaging”

https://doi.org/10.1016/j.cellimm.2020.104086Get rights and content

Highlights

  • Multi-modal imaging provides different types of biological and spatial information.

  • Technical aspects to consider for general image cytometry.

  • Image cytometry for dynamic and whole mount imaging datasets.

Abstract

Optical imaging is a valuable tool to visualise biological processes in the context of the tissue. Each imaging modality provides the biologist with different types of information – cell dynamics and migration over time can be tracked with time-lapse imaging (e.g. intra-vital imaging); an overview of whole tissues can be acquired using optical clearing in conjunction with light sheet microscopy; finer details such as cellular morphology and fine nerve tortuosity can be imaged at higher resolution using the confocal microscope. Multi-modal imaging combined with image cytometry – a form of quantitative analysis of image datasets – provides an objective basis for comparing between sample groups. Here, we provide an overview of technical aspects to look out for in an image cytometry workflow, and discuss issues related to sample preparation, image post-processing and analysis for intra-vital and whole organ imaging.

Introduction

In comparison to other techniques such as flow cytometry, mass cytometry, RNA sequencing or enzyme-linked immunosorbent assays, imaging experiments can be comparatively more tedious, have a lower throughput and requires specific technical know-how and trained personnel to carry out. Why then is imaging valuable?

The key advantage of imaging is the capability to visualise cellular components in the context of the entire tissue. Tissue dissociation or lysis is typically required to release cells or genetic material from the tissue for analysis, but in this process, information such as cell–cell interactions, cellular behaviour and localization is lost. Tissue dissociation protocols can also vary between laboratories, and enzymatic digestion using trypsin or Type IV collagenase is also known to cause cleavage of some surface receptors [1]. Imaging is thus a valuable tool for visualising cellular distribution in situ without disrupting the architecture of the tissue, preserving the spatial information.

Imaging modalities for biological discovery are ultimately dependent on technological developments in imaging hardware and software. Antonie van Leeuwenhoek and Robert Hooke, widely regarded as the pioneers of the field of microbiology for their observations of protozoa, were able to make their discoveries by designing the most primitive form of the microscope using glass lenses [2]. Recent technological advances in the field of optical imaging have provided the biologist with a plethora of techniques such as: intra-vital two-photon microscopy to track cellular activity and changes over space and time; light sheet microscopy for whole organ imaging coupled with optical clearing to provide an overview of any changes in the tissue; and confocal microscopy to interrogate specific regions within the tissue at higher resolution. Together, these techniques can be used in combination in an imaging workflow to provide a multi-faceted answer for biological questions (Fig. 1A).

While imaging results are typically assessed visually to provide “yes or no” answers to determine if any differences exist, quantitative analyses of imaging experiments ultimately provide more objective insights into the data acquired. Image cytometry began with this initial purpose, to quantitatively describe cellular phenotypes together with spatial localization and morphology in two-dimensional images in an automated manner [3]. Such analytical methods have been successfully applied for simple nuclear stained cytospin preparations [4], haematoxylin and eosin [5], immunohistochemically stained tissues and fluorescent in situ hybridization (FISH) preparations [6] for the last twenty years. Flow cytometry-style analysis of three-dimensional lymph node static image datasets, termed “histocytometry” [7], dynamic in situ cytometry (DISC) of intra-vital imaging datasets in spleen [8]; as well as correlated dynamic and static images obtained from the same tissue using “intra-vital dynamics-immunosignal correlative (iDISC) microscopy” [9] in recent years have further popularised this form of analysis in a tissue-based context. In this review, we highlight key technical aspects for which to pay attention, for the reader who may be interested to start image cytometry. We also discuss issues linked to sample preparation, image post-processing and analysis for intra-vital imaging and whole organ imaging.

Section snippets

Technical aspects of image cytometry

Prior to starting the experiment, it is crucial to consider the following question “Is imaging even the right tool to answer the biological question?” While imaging is a powerful tool to visually illustrate biological phenomenon, and great imaging data has the capability to blow audiences off their feet, it may not be worthwhile to do imaging solely for the “wow factor” it evokes. In many situations, alternatives such as flow cytometry could be more rapid and reliable for answering the question

Image cytometry for dynamic imaging

Intra-vital imaging provides the scientist with the capability to track cellular behaviour, cell–cell interaction, cell proliferation or cell death within a living mouse over time and space. The generation of reporter mice where specific populations of cell types are labelled with fluorescent proteins was an important step forward for intra-vital imaging, as it made it possible to make observations with minimal perturbation of the biological system [32]. Mice expressing photoconvertible

Image cytometry for whole mount imaging

Immunofluorescence imaging typically focuses on a limited region of interest, with a limited penetration depth. In order to obtain clear and sharp images with minimal signal loss, tissues are typically sectioned into thin 10 µm sections for imaging. However, this makes it difficult to obtain an accurate three-dimensional representation of anatomical structures such as nerves or blood vessels which exist as a three-dimensional network [67]. One strategy to resolve this issue since the 1970s was

Conclusion and future perspectives

Interrogating a piece of tissue using a multi-modal imaging workflow quantitatively allows the biologist to correlate both temporal and spatial information of differing resolutions to provide an objective picture of ongoing biological processes. Recent development of newer techniques for multiplex imaging such as the Opal Vectra system by Perkin Elmer, tissue-based cyclic immunofluorescence (t-CyCIF) [106], co-detection by indexing (CODEX) [107] and imaging mass cytometry [108] make it possible

Acknowledgements

The authors would like to thank Leonard De Li Tan and Keith Weng Kit Leong for providing some of the images for the figure, Dr. Jackson Li Liang Yao who was instrumental in helping to set-up the image cytometry workflow in the Lai Guan Ng lab, as well as members of the lab for providing feedback on the manuscript.

Funding

This work was supported by the National Medical Research Council grant (NMRC/CSA-INV/0023/2017). This article is supported by Singapore Immunology Network (A*STAR) core funding to L.G.N. N.H. is supported by the A*STAR Graduate Scholarship (Singapore).

References (118)

  • R.W. Ware

    Three-dimensional reconstruction from serial sections

    Int. Rev. Cytol.

    (1975)
  • D.S. Richardson et al.

    Clarifying Tissue Clearing

    Cell

    (2015)
  • N. Renier et al.

    iDISCO: a simple, rapid method to immunolabel large tissue samples for volume imaging

    Cell

    (2014)
  • M.T. Ke et al.

    Super-Resolution Mapping of Neuronal Circuitry With an Index-Optimized Clearing Agent

    Cell reports

    (2016)
  • E.A. Susaki et al.

    Whole-brain imaging with single-cell resolution using chemical cocktails and computational analysis

    Cell

    (2014)
  • K. Tainaka et al.

    Whole-body imaging with single-cell resolution by tissue decolorization

    Cell

    (2014)
  • B. Yang et al.

    Single-cell phenotyping within transparent intact tissue through whole-body clearing

    Cell

    (2014)
  • M. Monici

    Cell and tissue autofluorescence research and diagnostic applications

    Biotechnol. Annual Rev.

    (2005)
  • E.A. Susaki et al.

    Whole-body and Whole-Organ Clearing and Imaging Techniques with Single-Cell Resolution: Toward Organism-Level Systems Biology in Mammals

    Cell Chem. Biol.

    (2016)
  • R.A. Botting et al.

    Phenotypic and functional consequences of different isolation protocols on skin mononuclear phagocytes

    J. Leukoc. Biol.

    (2017)
  • H. Gest

    The discovery of microorganisms by Robert Hooke and Antoni Van Leeuwenhoek, fellows of the Royal Society

    Notes and records of the Royal Society of London

    (2004)
  • R.C. Ecker et al.

    Microscopy-based multicolor tissue cytometry at the single-cell level

    Cytometry. Part A : the journal of the International Society for Analytical Cytology

    (2004)
  • G. Mehes et al.

    Quantitative analysis of disseminated tumor cells in the bone marrow by automated fluorescence image analysis

    Cytometry

    (2000)
  • J. Smolle et al.

    Tissue counter analysis of histologic sections of melanoma: influence of mask size and shape, feature selection, statistical methods and tissue preparation

    Analytical cellular pathology : the journal of the European Society for Analytical Cellular Pathology

    (2002)
  • L.A. Kamentsky et al.

    Slide-based laser scanning cytometry

    Acta Cytol.

    (1997)
  • G. Chodaczek et al.

    Body-barrier surveillance by epidermal gammadelta TCRs

    Nat. Immunol.

    (2012)
  • H. Peng

    Bioimage informatics: a new area of engineering biology

    Bioinformatics

    (2008)
  • E. Meijering

    Cell Segmentation: 50 Years Down the Road [Life Sciences]

    IEEE Signal Process Mag.

    (2012)
  • S. Dimopoulos et al.

    Accurate cell segmentation in microscopy images using membrane patterns

    Bioinformatics

    (2014)
  • V. Ljosa et al.

    Introduction to the quantitative analysis of two-dimensional fluorescence microscopy images for cell-based screening

    PLoS Comput. Biol.

    (2009)
  • T.J. Lambert

    FPbase: a community-editable fluorescent protein database

    Nat. Methods

    (2019)
  • A. Joyner et al.

    Immunohistochemistry of whole-mount mouse embryos

    CSH protocols

    (2008 (2008))
  • J.J. White et al.

    Wholemount immunohistochemistry for revealing complex brain topography

    J. visualized experiments : JoVE

    (2012)
  • J. Jonkman et al.

    Any Way You Slice It-A Comparison of Confocal Microscopy Techniques

    J. biomolecular techniques : JBT

    (2015)
  • A.J. North

    Seeing is believing? A beginners' guide to practical pitfalls in image acquisition

    J. Cell Biol.

    (2006)
  • K. Smith et al.

    CIDRE: an illumination-correction method for optical microscopy

    Nat. Methods

    (2015)
  • T. Peng et al.

    A BaSiC tool for background and shading correction of optical microscopy images

    Nat. Commun.

    (2017)
  • S. Singh et al.

    Pipeline for illumination correction of images for high-throughput microscopy

    J. Microsc.

    (2014)
  • S. Preibisch et al.

    Globally optimal stitching of tiled 3D microscopic image acquisitions

    Bioinformatics

    (2009)
  • J. Chalfoun et al.

    MIST: Accurate and Scalable Microscopy Image Stitching Tool with Stage Modeling and Error Minimization

    Sci. Rep.

    (2017)
  • C. Sommer et al.

    Ilastik: Interactive learning and segmentation toolkit, in

    IEEE International Symposium on Biomedical Imaging: From Nano to Macro

    (2011)
  • H. Peng et al.

    V3D enables real-time 3D visualization and quantitative analysis of large-scale biological image data sets

    Nat. Biotechnol.

    (2010)
  • H. Peng et al.

    Extensible visualization and analysis for multidimensional images using Vaa3D

    Nat. Protoc.

    (2014)
  • I. Belevich et al.

    Microscopy Image Browser: A Platform for Segmentation and Analysis of Multidimensional Datasets

    PLoS Biol.

    (2016)
  • C. McQuin et al.

    Cell Profiler 3.0: Next-generation image processing for biology

    PLoS Biol.

    (2018)
  • Y. Tan et al.

    Streamlining volumetric multi-channel image cytometry using hue-saturation-brightness-based surface creation

    Commun. Biol.

    (2018)
  • T. Abe et al.

    Reporter mouse lines for fluorescence imaging

    Dev. Growth Differ.

    (2013)
  • S.L. Griswold et al.

    Generation and characterization of iUBC-KikGR photoconvertible transgenic mice for live time-lapse imaging during development

    Genesis

    (2011)
  • M. Tomura et al.

    Monitoring cellular movement in vivo with photoconvertible fluorescence protein “Kaede” transgenic mice

    PNAS

    (2008)
  • D.M. Chudakov et al.

    Tracking intracellular protein movements using photoswitchable fluorescent proteins PS-CFP2 and Dendra2

    Nat. Protoc.

    (2007)
  • A publisher’s error resulted in this article appearing in the wrong issue. The article is reprinted here for the reader’s convenience and for the continuity of the special issue. For citation purposes, please use the original publication details; Cell. Immunol, 344, 103946.**DOI of original item: http://dx.doi.org/10.1016/j.cellimm.2019.103946.

    View full text