1
|
Hartsock I, Park E, Toppen J, Bubenik P, Dimitrova ES, Kemp ML, Cruz DA. Topological data analysis of pattern formation of human induced pluripotent stem cell colonies. Sci Rep 2025; 15:11544. [PMID: 40185811 PMCID: PMC11971356 DOI: 10.1038/s41598-025-90592-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 02/13/2025] [Indexed: 04/07/2025] Open
Abstract
Understanding the multicellular organization of stem cells is vital for determining the mechanisms that coordinate cell fate decision-making during differentiation; these mechanisms range from neighbor-to-neighbor communication to tissue-level biochemical gradients. Current methods for quantifying multicellular patterning tend to capture the spatial properties of cell colonies at a fixed scale and typically rely on human annotation. We present a computational pipeline that utilizes topological data analysis to generate quantitative, multiscale descriptors which capture the shape of data extracted from 2D multichannel microscopy images. By applying our pipeline to certain stem cell colonies, we detected subtle differences in patterning that reflect distinct spatial organization associated with loss of pluripotency. These results yield insight into putative directed cellular organization and morphogen-mediated, neighbor-to-neighbor signaling. Because of its broad applicability to immunofluorescence microscopy images, our pipeline is well-positioned to serve as a general-purpose tool for the quantitative study of multicellular pattern formation.
Collapse
Affiliation(s)
- Iryna Hartsock
- Department of Machine Learning, H. Lee Moffitt Cancer Center & Research Institute, Tampa, 33612, US
| | - Eunbi Park
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, 30332, US
| | - Jack Toppen
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, 30332, US
- Program in Computational and Systems Biology, Massachusetts Institute of Technology, Cambridge, 02139, US
| | - Peter Bubenik
- Department of Mathematics, University of Florida, Gainesville, 32611, US
| | - Elena S Dimitrova
- Mathematics Department, California Polytechnic State University, San Luis Obispo, 93407, US
| | - Melissa L Kemp
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, 30332, US
| | - Daniel A Cruz
- Department of Medicine, University of Florida, Gainesville, 32611, US.
| |
Collapse
|
2
|
Ali M, Benfante V, Basirinia G, Alongi P, Sperandeo A, Quattrocchi A, Giannone AG, Cabibi D, Yezzi A, Di Raimondo D, Tuttolomondo A, Comelli A. Applications of Artificial Intelligence, Deep Learning, and Machine Learning to Support the Analysis of Microscopic Images of Cells and Tissues. J Imaging 2025; 11:59. [PMID: 39997561 PMCID: PMC11856378 DOI: 10.3390/jimaging11020059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 02/08/2025] [Accepted: 02/12/2025] [Indexed: 02/26/2025] Open
Abstract
Artificial intelligence (AI) transforms image data analysis across many biomedical fields, such as cell biology, radiology, pathology, cancer biology, and immunology, with object detection, image feature extraction, classification, and segmentation applications. Advancements in deep learning (DL) research have been a critical factor in advancing computer techniques for biomedical image analysis and data mining. A significant improvement in the accuracy of cell detection and segmentation algorithms has been achieved as a result of the emergence of open-source software and innovative deep neural network architectures. Automated cell segmentation now enables the extraction of quantifiable cellular and spatial features from microscope images of cells and tissues, providing critical insights into cellular organization in various diseases. This review aims to examine the latest AI and DL techniques for cell analysis and data mining in microscopy images, aid the biologists who have less background knowledge in AI and machine learning (ML), and incorporate the ML models into microscopy focus images.
Collapse
Affiliation(s)
- Muhammad Ali
- Ri.MED Foundation, Via Bandiera 11, 90133 Palermo, Italy; (M.A.); (G.B.)
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, Molecular and Clinical Medicine, University of Palermo, 90127 Palermo, Italy; (D.D.R.); (A.T.)
| | - Viviana Benfante
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, Molecular and Clinical Medicine, University of Palermo, 90127 Palermo, Italy; (D.D.R.); (A.T.)
- Advanced Diagnostic Imaging—INNOVA Project, Department of Radiological Sciences, A.R.N.A.S. Civico, Di Cristina e Benfratelli Hospitals, P.zza N. Leotta 4, 90127 Palermo, Italy;
- Pharmaceutical Factory, La Maddalena S.P.A., Via San Lorenzo Colli, 312/d, 90146 Palermo, Italy;
| | - Ghazal Basirinia
- Ri.MED Foundation, Via Bandiera 11, 90133 Palermo, Italy; (M.A.); (G.B.)
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, Molecular and Clinical Medicine, University of Palermo, 90127 Palermo, Italy; (D.D.R.); (A.T.)
| | - Pierpaolo Alongi
- Advanced Diagnostic Imaging—INNOVA Project, Department of Radiological Sciences, A.R.N.A.S. Civico, Di Cristina e Benfratelli Hospitals, P.zza N. Leotta 4, 90127 Palermo, Italy;
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (BiND), University of Palermo, 90127 Palermo, Italy
| | - Alessandro Sperandeo
- Pharmaceutical Factory, La Maddalena S.P.A., Via San Lorenzo Colli, 312/d, 90146 Palermo, Italy;
| | - Alberto Quattrocchi
- Pathologic Anatomy Unit, Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, 90127 Palermo, Italy; (A.Q.); (A.G.G.); (D.C.)
| | - Antonino Giulio Giannone
- Pathologic Anatomy Unit, Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, 90127 Palermo, Italy; (A.Q.); (A.G.G.); (D.C.)
| | - Daniela Cabibi
- Pathologic Anatomy Unit, Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, 90127 Palermo, Italy; (A.Q.); (A.G.G.); (D.C.)
| | - Anthony Yezzi
- Department of Electrical and Computer Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA;
| | - Domenico Di Raimondo
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, Molecular and Clinical Medicine, University of Palermo, 90127 Palermo, Italy; (D.D.R.); (A.T.)
| | - Antonino Tuttolomondo
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, Molecular and Clinical Medicine, University of Palermo, 90127 Palermo, Italy; (D.D.R.); (A.T.)
| | - Albert Comelli
- Ri.MED Foundation, Via Bandiera 11, 90133 Palermo, Italy; (M.A.); (G.B.)
| |
Collapse
|
3
|
Perochon T, Krsnik Z, Massimo M, Ruchiy Y, Romero AL, Mohammadi E, Li X, Long KR, Parkkinen L, Blomgren K, Lagache T, Menassa DA, Holcman D. Unraveling microglial spatial organization in the developing human brain with DeepCellMap, a deep learning approach coupled with spatial statistics. Nat Commun 2025; 16:1577. [PMID: 39948387 PMCID: PMC11825940 DOI: 10.1038/s41467-025-56560-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 01/21/2025] [Indexed: 02/16/2025] Open
Abstract
Mapping cellular organization in the developing brain presents significant challenges due to the multidimensional nature of the data, characterized by complex spatial patterns that are difficult to interpret without high-throughput tools. Here, we present DeepCellMap, a deep-learning-assisted tool that integrates multi-scale image processing with advanced spatial and clustering statistics. This pipeline is designed to map microglial organization during normal and pathological brain development and has the potential to be adapted to any cell type. Using DeepCellMap, we capture the morphological diversity of microglia, identify strong coupling between proliferative and phagocytic phenotypes, and show that distinct spatial clusters rarely overlap as human brain development progresses. Additionally, we uncover an association between microglia and blood vessels in fetal brains exposed to maternal SARS-CoV-2. These findings offer insights into whether various microglial phenotypes form networks in the developing brain to occupy space, and in conditions involving haemorrhages, whether microglia respond to, or influence changes in blood vessel integrity. DeepCellMap is available as an open-source software and is a powerful tool for extracting spatial statistics and analyzing cellular organization in large tissue sections, accommodating various imaging modalities. This platform opens new avenues for studying brain development and related pathologies.
Collapse
Affiliation(s)
- Theo Perochon
- Group of Data Modeling and Computational Biology, IBENS, École Normale Supérieure, Paris, France
| | - Zeljka Krsnik
- Croatian Institute for Brain Research, University of Zagreb, Zagreb, Croatia
| | - Marco Massimo
- Centre for Developmental Neurobiology, MRC Centre for Neurodevelopmental Disorders, King's College London, London, UK
| | - Yana Ruchiy
- Department of Women's and Children's Health, Karolinska Institutet, Solna, Sweden
| | | | - Elyas Mohammadi
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Solna, Sweden
| | - Xiaofei Li
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Solna, Sweden
| | - Katherine R Long
- Centre for Developmental Neurobiology, MRC Centre for Neurodevelopmental Disorders, King's College London, London, UK
| | - Laura Parkkinen
- Department of Neuropathology and The Queen's College, University of Oxford, Oxford, United Kingdom
| | - Klas Blomgren
- Department of Women's and Children's Health, Karolinska Institutet, Solna, Sweden
| | - Thibault Lagache
- BioImage Analysis Unit, CNRS UMR3691, Institut Pasteur, Université Paris Cité, Paris, France.
| | - David A Menassa
- Department of Women's and Children's Health, Karolinska Institutet, Solna, Sweden.
- Department of Neuropathology and The Queen's College, University of Oxford, Oxford, United Kingdom.
| | - David Holcman
- Group of Data Modeling and Computational Biology, IBENS, École Normale Supérieure, Paris, France.
- DAMPT, University of Cambridge, DAMPT and Churchill College, Cambridge, United Kingdom.
| |
Collapse
|
4
|
Gui G, Bingham MA, Herzog JR, Wong-Rolle A, Dillon LW, Goswami M, Martin E, Reeves J, Kim S, Bahrami A, Degenhardt H, Zaki G, Divakar P, Schrom EC, Calvo K, Hourigan CS, Hansen K, Zhao C. Single Cell Spatial Transcriptomics Reveals Immunotherapy-Driven Bone Marrow Niche Remodeling in AML. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.24.634753. [PMID: 39975227 PMCID: PMC11838223 DOI: 10.1101/2025.01.24.634753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Given the successful graft-versus-leukemia cell treatment effect observed with allogeneic hematopoietic stem cell transplant for patients with refractory or relapsed acute myeloid leukemia, immunotherapies have also been investigated in the nontransplant setting. Here, we use a multi-omic approach to investigate spatiotemporal interactions in the bone marrow niche between leukemia cells and immune cells in patients with refractory or relapsed acute myeloid leukemia treated with a combination of the immune checkpoint inhibitor pembrolizumab and hypomethylating agent decitabine. We derived precise segmentation data by extensively training nuclear and membrane cell segmentation models, which enabled accurate transcript assignment and deep learning-feature-based image analysis. To overcome read-depth limitations, we integrated the single-cell RNA sequencing data with single-cell-resolution spatial transcriptomic data from the same sample. Quantifying cell-cell distances between cell edges rather than cell centroids allowed us to conduct a more accurate downstream analysis of the tumor microenvironment, revealing that multiple cell types of interest had global enrichment or local enrichment proximal to leukemia cells after pembrolizumab treatment, which could be associated with their clinical responses. Furthermore, ligand-receptor analysis indicated a potential increase in TWEAK signaling between leukemia cells and immune cells after pembrolizumab treatment.
Collapse
Affiliation(s)
- Gege Gui
- Fralin Biomedical Research Institute, Virginia Tech FBRI Cancer Research Center, Washington, DC
- Laboratory of Myeloid Malignancies, Hematology Branch, NHLBI, Bethesda, MD, USA
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Molly A. Bingham
- Thoracic and GI Malignancies Branch, CCR, NCI, Bethesda, MD, USA
| | - Julius R. Herzog
- Thoracic and GI Malignancies Branch, CCR, NCI, Bethesda, MD, USA
| | | | - Laura W. Dillon
- Laboratory of Myeloid Malignancies, Hematology Branch, NHLBI, Bethesda, MD, USA
| | - Meghali Goswami
- Laboratory of Myeloid Malignancies, Hematology Branch, NHLBI, Bethesda, MD, USA
| | - Eddie Martin
- Department of Laboratory Medicine, NIH Clinical Center, Bethesda, MD, USA
| | | | - Sean Kim
- NanoString Technologies Inc., Seattle, WA, USA
| | | | - Hermann Degenhardt
- Optical Microscopy and Analysis Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - George Zaki
- Biomedical Informatics and Data Science Directorate, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | | | - Edward C Schrom
- Lymphocyte Biology Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD
| | - Katherine Calvo
- Department of Laboratory Medicine, NIH Clinical Center, Bethesda, MD, USA
| | - Christopher S. Hourigan
- Fralin Biomedical Research Institute, Virginia Tech FBRI Cancer Research Center, Washington, DC
- Laboratory of Myeloid Malignancies, Hematology Branch, NHLBI, Bethesda, MD, USA
- Department of Internal Medicine, Virginia Tech Carilion School of Medicine
| | - Kasper Hansen
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Chen Zhao
- Thoracic and GI Malignancies Branch, CCR, NCI, Bethesda, MD, USA
| |
Collapse
|
5
|
Taube JM, Sunshine JC, Angelo M, Akturk G, Eminizer M, Engle LL, Ferreira CS, Gnjatic S, Green B, Greenbaum S, Greenwald NF, Hedvat CV, Hollmann TJ, Jiménez-Sánchez D, Korski K, Lako A, Parra ER, Rebelatto MC, Rimm DL, Rodig SJ, Rodriguez-Canales J, Roskes JS, Schalper KA, Schenck E, Steele KE, Surace MJ, Szalay AS, Tetzlaff MT, Wistuba II, Yearley JH, Bifulco CB. Society for Immunotherapy of Cancer: updates and best practices for multiplex immunohistochemistry (IHC) and immunofluorescence (IF) image analysis and data sharing. J Immunother Cancer 2025; 13:e008875. [PMID: 39779210 PMCID: PMC11749220 DOI: 10.1136/jitc-2024-008875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 11/12/2024] [Indexed: 01/11/2025] Open
Abstract
OBJECTIVES Multiplex immunohistochemistry and immunofluorescence (mIHC/IF) are emerging technologies that can be used to help define complex immunophenotypes in tissue, quantify immune cell subsets, and assess the spatial arrangement of marker expression. mIHC/IF assays require concerted efforts to optimize and validate the multiplex staining protocols prior to their application on slides. The best practice guidelines for staining and validation of mIHC/IF assays across platforms were previously published by this task force. The current effort represents a complementary manuscript for mIHC/IF analysis focused on the associated image analysis and data management. METHODS The Society for Immunotherapy of Cancer convened a task force of pathologists and laboratory leaders from academic centers as well as experts from pharmaceutical and diagnostic companies to develop best practice guidelines for the quantitative image analysis of mIHC/IF output and data management considerations. RESULTS Best-practice approaches for image acquisition, color deconvolution and spectral unmixing, tissue and cell segmentation, phenotyping, and algorithm verification are reviewed. Additional quality control (QC) measures such as batch-to-batch correction and QC for assembled images are also discussed. Recommendations for sharing raw outputs, processed results, key analysis programs and source code, and representative photomicrographs from mIHC/IF assays are included. Lastly, multi-institutional harmonization efforts are described. CONCLUSIONS mIHC/IF technologies are maturing and are routinely included in research studies and moving towards clinical use. Guidelines for how to perform and standardize image analysis on mIHC/IF-stained slides will likely contribute to more comparable results across laboratories and pave the way for clinical implementation. A checklist encompassing these two-part guidelines for the generation of robust data from quantitative mIHC/IF assays will be provided in a third publication from this task force. While the current effort is mainly focused on best practices for characterizing the tumor microenvironment, these principles are broadly applicable to any mIHC/IF assay and associated image analysis.
Collapse
Affiliation(s)
- Janis M Taube
- Mark Foundation Center for Advanced Genomics and Imaging, Johns Hopkins University SOM, Baltimore, Maryland, USA
- Bloomberg~Kimmel Institute of Cancer Immunotherapy, Johns Hopkins University SOM, Baltimore, Maryland, USA
| | - Joel C Sunshine
- Bloomberg~Kimmel Institute of Cancer Immunotherapy, Johns Hopkins University SOM, Baltimore, Maryland, USA
| | - Michael Angelo
- Department of Pathology, Stanford University School of Medicine, Palo Alto, California, USA
| | | | - Margaret Eminizer
- Johns Hopkins University, Baltimore, Maryland, USA
- Department of Astronomy and Physics, Johns Hopkins University, Baltimore, Maryland, USA
| | - Logan L Engle
- Bloomberg~Kimmel Institute of Cancer Immunotherapy, Johns Hopkins University SOM, Baltimore, Maryland, USA
| | - Cláudia S Ferreira
- Pharma Research and Early Development (pRED), Roche Innovation Center Munich, Penzberg, Germany
| | - Sacha Gnjatic
- Icahn School of Medicine at Mount Sinai Tisch Cancer Institute, New York, New York, USA
| | - Benjamin Green
- Bloomberg~Kimmel Institute of Cancer Immunotherapy, Johns Hopkins University SOM, Baltimore, Maryland, USA
| | - Shirley Greenbaum
- Department of Pathology, Stanford University School of Medicine, Palo Alto, California, USA
| | - Noah F Greenwald
- Department of Pathology, Stanford University School of Medicine, Palo Alto, California, USA
| | - Cyrus V Hedvat
- Department of Pathology, Molecular and Cell-based Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Travis J Hollmann
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Daniel Jiménez-Sánchez
- Bloomberg~Kimmel Institute of Cancer Immunotherapy, Johns Hopkins University SOM, Baltimore, Maryland, USA
| | - Konstanty Korski
- Department of Personalized Healthcare, Data, Analytics and Imaging Group, F Hoffmann-La Roche AG, Basel, Switzerland
| | - Ana Lako
- Dana Farber/Brigham and Women's Cancer Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Edwin R Parra
- Department of Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | | | - David L Rimm
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Scott J Rodig
- Dana Farber/Brigham and Women's Cancer Center, Harvard Medical School, Boston, Massachusetts, USA
| | | | - Jeffrey S Roskes
- Bloomberg~Kimmel Institute of Cancer Immunotherapy, Johns Hopkins University SOM, Baltimore, Maryland, USA
- Department of Astronomy and Physics, Johns Hopkins University, Baltimore, Maryland, USA
| | - Kurt A Schalper
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut, USA
| | | | | | | | - Alexander S Szalay
- Mark Foundation Center for Advanced Genomics and Imaging, Johns Hopkins University SOM, Baltimore, Maryland, USA
- Bloomberg~Kimmel Institute of Cancer Immunotherapy, Johns Hopkins University SOM, Baltimore, Maryland, USA
- Department of Astronomy and Physics, Johns Hopkins University, Baltimore, Maryland, USA
| | - Michael T Tetzlaff
- Departments of Pathology and Dermatology, University of California San Francisco, San Francisco, California, USA
| | - Ignacio I Wistuba
- Department of Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | | | | |
Collapse
|
6
|
Chen H, Murphy RF. CytoSpatio: Learning cell type spatial relationships using multirange, multitype point process models. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.31.621408. [PMID: 39553984 PMCID: PMC11565948 DOI: 10.1101/2024.10.31.621408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
Recent advances in multiplexed fluorescence imaging have provided new opportunities for deciphering the complex spatial relationships among various cell types across diverse tissues. We introduce CytoSpatio, open-source software that constructs generative, multirange, and multitype point process models that capture interactions among multiple cell types at various distances simultaneously. On analyzing five cell types across five tissues, our software showed consistent spatial relationships within the same tissue type, with certain cell types like proliferating T cells consistently clustering across tissue types. It also revealed that the attraction-repulsion relationships between cell types like B cells and CD4-positive T cells vary with tissue type. CytoSpatio can also generate synthetic tissue structures that preserve the spatial relationships seen in training images, a capability not provided by previous descriptive, motif-based approaches. This potentially allows spatially realistic simulations of how cell relationships affect tissue biochemistry.
Collapse
Affiliation(s)
- Haoran Chen
- Computational Biology Department, School of Computer Science, Carnegie Mellon University
| | - Robert F. Murphy
- Computational Biology Department, School of Computer Science, Carnegie Mellon University
| |
Collapse
|
7
|
Azimi M, Cho S, Bozkurt E, McDonough E, Kisakol B, Matveeva A, Salvucci M, Dussmann H, McDade S, Firat C, Urganci N, Shia J, Longley DB, Ginty F, Prehn JH. Spatial effects of infiltrating T cells on neighbouring cancer cells and prognosis in stage III CRC patients. J Pathol 2024; 264:148-159. [PMID: 39092716 DOI: 10.1002/path.6327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 05/03/2024] [Accepted: 06/03/2024] [Indexed: 08/04/2024]
Abstract
Colorectal cancer (CRC) is one of the most frequently occurring cancers, but prognostic biomarkers identifying patients at risk of recurrence are still lacking. In this study, we aimed to investigate in more detail the spatial relationship between intratumoural T cells, cancer cells, and cancer cell hallmarks as prognostic biomarkers in stage III colorectal cancer patients. We conducted multiplexed imaging of 56 protein markers at single-cell resolution on resected fixed tissue from stage III CRC patients who received adjuvant 5-fluorouracil (5FU)-based chemotherapy. Images underwent segmentation for tumour, stroma, and immune cells, and cancer cell 'state' protein marker expression was quantified at a cellular level. We developed a Python package for estimation of spatial proximity, nearest neighbour analysis focusing on cancer cell-T-cell interactions at single-cell level. In our discovery cohort (Memorial Sloan Kettering samples), we processed 462 core samples (total number of cells: 1,669,228) from 221 adjuvant 5FU-treated stage III patients. The validation cohort (Huntsville Clearview Cancer Center samples) consisted of 272 samples (total number of cells: 853,398) from 98 stage III CRC patients. While there were trends for an association between the percentage of cytotoxic T cells (across the whole cancer core), it did not reach significance (discovery cohort: p = 0.07; validation cohort: p = 0.19). We next utilised our region-based nearest neighbour approach to determine the spatial relationships between cytotoxic T cells, helper T cells, and cancer cell clusters. In both cohorts, we found that shorter distance between cytotoxic T cells, T helper cells, and cancer cells was significantly associated with increased disease-free survival. An unsupervised trained model that clustered patients based on the median distance between immune cells and cancer cells, as well as protein expression profiles, successfully classified patients into low-risk and high-risk groups (discovery cohort: p = 0.01; validation cohort: p = 0.003). © 2024 The Author(s). The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Mohammadreza Azimi
- Department of Physiology and Medical Physics, RCSI Centre for Systems Medicine, Royal College of Surgeons in Ireland University of Medicine and Health Sciences, Dublin, Ireland
| | - Sanghee Cho
- GE HealthCare Technology and Innovation Center (formerly GE Research Center), Niskayuna, NY, USA
| | - Emir Bozkurt
- Department of Physiology and Medical Physics, RCSI Centre for Systems Medicine, Royal College of Surgeons in Ireland University of Medicine and Health Sciences, Dublin, Ireland
| | - Elizabeth McDonough
- GE HealthCare Technology and Innovation Center (formerly GE Research Center), Niskayuna, NY, USA
| | - Batuhan Kisakol
- Department of Physiology and Medical Physics, RCSI Centre for Systems Medicine, Royal College of Surgeons in Ireland University of Medicine and Health Sciences, Dublin, Ireland
| | - Anna Matveeva
- Department of Physiology and Medical Physics, RCSI Centre for Systems Medicine, Royal College of Surgeons in Ireland University of Medicine and Health Sciences, Dublin, Ireland
| | - Manuela Salvucci
- Department of Physiology and Medical Physics, RCSI Centre for Systems Medicine, Royal College of Surgeons in Ireland University of Medicine and Health Sciences, Dublin, Ireland
| | - Heiko Dussmann
- Department of Physiology and Medical Physics, RCSI Centre for Systems Medicine, Royal College of Surgeons in Ireland University of Medicine and Health Sciences, Dublin, Ireland
| | - Simon McDade
- School of Medicine, Dentistry and Biomedical Sciences, Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, UK
| | - Canan Firat
- Memorial Sloan Kettering Cancer Centre, New York, NY, USA
| | - Nil Urganci
- Memorial Sloan Kettering Cancer Centre, New York, NY, USA
| | - Jinru Shia
- Memorial Sloan Kettering Cancer Centre, New York, NY, USA
| | - Daniel B Longley
- School of Medicine, Dentistry and Biomedical Sciences, Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, UK
| | - Fiona Ginty
- GE HealthCare Technology and Innovation Center (formerly GE Research Center), Niskayuna, NY, USA
| | - Jochen Hm Prehn
- Department of Physiology and Medical Physics, RCSI Centre for Systems Medicine, Royal College of Surgeons in Ireland University of Medicine and Health Sciences, Dublin, Ireland
| |
Collapse
|
8
|
Williams HL, Frei AL, Koessler T, Berger MD, Dawson H, Michielin O, Zlobec I. The current landscape of spatial biomarkers for prediction of response to immune checkpoint inhibition. NPJ Precis Oncol 2024; 8:178. [PMID: 39138341 PMCID: PMC11322473 DOI: 10.1038/s41698-024-00671-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 08/05/2024] [Indexed: 08/15/2024] Open
Abstract
Enabling the examination of cell-cell relationships in tissue, spatially resolved omics technologies have revolutionised our perspectives on cancer biology. Clinically, the development of immune checkpoint inhibitors (ICI) has advanced cancer therapeutics. However, a major challenge of effective implementation is the identification of predictive biomarkers of response. In this review we examine the potential added predictive value of spatial biomarkers of response to ICI beyond current clinical benchmarks.
Collapse
Affiliation(s)
- Hannah L Williams
- Institute for Tissue Medicine and Pathology, University of Bern, Bern, Switzerland.
| | - Ana Leni Frei
- Institute for Tissue Medicine and Pathology, University of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Thibaud Koessler
- Medical Oncology Department, Geneva University Hospitals, 4 rue Gabrielle-Perret-Gentil, 1205, Geneva, Switzerland
- Swiss Cancer Centre Léman, Lausanne, Geneva, Switzerland
- University of Geneva, Faculty of Medicine, Geneva, Switzerland
| | - Martin D Berger
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Heather Dawson
- Institute for Tissue Medicine and Pathology, University of Bern, Bern, Switzerland
| | - Olivier Michielin
- Medical Oncology Department, Geneva University Hospitals, 4 rue Gabrielle-Perret-Gentil, 1205, Geneva, Switzerland
- Swiss Cancer Centre Léman, Lausanne, Geneva, Switzerland
- University of Geneva, Faculty of Medicine, Geneva, Switzerland
| | - Inti Zlobec
- Institute for Tissue Medicine and Pathology, University of Bern, Bern, Switzerland
| |
Collapse
|
9
|
Dezem FS, Arjumand W, DuBose H, Morosini NS, Plummer J. Spatially Resolved Single-Cell Omics: Methods, Challenges, and Future Perspectives. Annu Rev Biomed Data Sci 2024; 7:131-153. [PMID: 38768396 DOI: 10.1146/annurev-biodatasci-102523-103640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Overlaying omics data onto spatial biological dimensions has been a promising technology to provide high-resolution insights into the interactome and cellular heterogeneity relative to the organization of the molecular microenvironment of tissue samples in normal and disease states. Spatial omics can be categorized into three major modalities: (a) next-generation sequencing-based assays, (b) imaging-based spatially resolved transcriptomics approaches including in situ hybridization/in situ sequencing, and (c) imaging-based spatial proteomics. These modalities allow assessment of transcripts and proteins at a cellular level, generating large and computationally challenging datasets. The lack of standardized computational pipelines to analyze and integrate these nonuniform structured data has made it necessary to apply artificial intelligence and machine learning strategies to best visualize and translate their complexity. In this review, we summarize the currently available techniques and computational strategies, highlight their advantages and limitations, and discuss their future prospects in the scientific field.
Collapse
Affiliation(s)
- Felipe Segato Dezem
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
- Center for Spatial Omics, St. Jude Children's Research Hospital, Memphis, Tennessee, USA;
| | - Wani Arjumand
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
- Center for Spatial Omics, St. Jude Children's Research Hospital, Memphis, Tennessee, USA;
| | - Hannah DuBose
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
- Center for Spatial Omics, St. Jude Children's Research Hospital, Memphis, Tennessee, USA;
| | - Natalia Silva Morosini
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
- Center for Spatial Omics, St. Jude Children's Research Hospital, Memphis, Tennessee, USA;
| | - Jasmine Plummer
- Department of Cellular and Molecular Biology and Comprehensive Cancer Center, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
- Center for Spatial Omics, St. Jude Children's Research Hospital, Memphis, Tennessee, USA;
| |
Collapse
|
10
|
Cross A, Issa F. Unraveling Renal Transplant Rejection: How Can We Measure Intragraft Cell-Cell Interactions? Transplantation 2024:00007890-990000000-00824. [PMID: 39044324 DOI: 10.1097/tp.0000000000005136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/25/2024]
Affiliation(s)
- Amy Cross
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, United Kingdom
| | | |
Collapse
|
11
|
Hardwick LJA, Davies BP, Pensa S, Burge-Rogers M, Davies C, Baptista AF, Knott R, S McCrone I, Po E, Strugnell BW, Waine K, Wood P, Khaled WT, Summers HD, Rees P, Wills JW, Hughes K. In the Murine and Bovine Maternal Mammary Gland Signal Transducer and Activator of Transcription 3 is Activated in Clusters of Epithelial Cells around the Day of Birth. J Mammary Gland Biol Neoplasia 2024; 29:10. [PMID: 38722417 PMCID: PMC11081984 DOI: 10.1007/s10911-024-09561-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 03/28/2024] [Indexed: 05/12/2024] Open
Abstract
Signal transducers and activators of transcription (STAT) proteins regulate mammary development. Here we investigate the expression of phosphorylated STAT3 (pSTAT3) in the mouse and cow around the day of birth. We present localised colocation analysis, applicable to other mammary studies requiring identification of spatially congregated events. We demonstrate that pSTAT3-positive events are multifocally clustered in a non-random and statistically significant fashion. Arginase-1 expressing cells, consistent with macrophages, exhibit distinct clustering within the periparturient mammary gland. These findings represent a new facet of mammary STAT3 biology, and point to the presence of mammary sub-microenvironments.
Collapse
Affiliation(s)
- Laura J A Hardwick
- Department of Veterinary Medicine, University of Cambridge, Madingley Road, Cambridge, CB3 0ES, UK
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Benjamin P Davies
- Department of Veterinary Medicine, University of Cambridge, Madingley Road, Cambridge, CB3 0ES, UK
| | - Sara Pensa
- Department of Pharmacology, University of Cambridge, Cambridge, UK
- Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Maedee Burge-Rogers
- Department of Veterinary Medicine, University of Cambridge, Madingley Road, Cambridge, CB3 0ES, UK
| | - Claire Davies
- The Fold Farm Vets Ltd, Tyne Green, Hexham, Northumberland, UK
| | | | - Robert Knott
- Department of Veterinary Medicine, University of Cambridge, Madingley Road, Cambridge, CB3 0ES, UK
- Bristol Veterinary School, University of Bristol, Langford, UK
| | - Ian S McCrone
- Department of Veterinary Medicine, University of Cambridge, Madingley Road, Cambridge, CB3 0ES, UK
| | - Eleonora Po
- Department of Veterinary Medicine, University of Cambridge, Madingley Road, Cambridge, CB3 0ES, UK
| | | | - Katie Waine
- Farm Post Mortems Ltd, Durham, UK
- Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, T3R 1J3, Canada
| | - Paul Wood
- Department of Veterinary Medicine, University of Cambridge, Madingley Road, Cambridge, CB3 0ES, UK
- SRUC Aberdeen, Craibstone Estate, Bucksburn, Aberdeen, UK
| | - Walid T Khaled
- Department of Pharmacology, University of Cambridge, Cambridge, UK
- Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Huw D Summers
- Department of Biomedical Engineering, Swansea University, Swansea, UK
| | - Paul Rees
- Department of Biomedical Engineering, Swansea University, Swansea, UK
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - John W Wills
- Department of Veterinary Medicine, University of Cambridge, Madingley Road, Cambridge, CB3 0ES, UK.
| | - Katherine Hughes
- Department of Veterinary Medicine, University of Cambridge, Madingley Road, Cambridge, CB3 0ES, UK.
| |
Collapse
|
12
|
Azimi M, Cho S, Bozkurt E, McDonough E, Kisakol B, Matveeva A, Salvucci M, Dussmann H, McDade S, Firat C, Urganci N, Shia J, Longley DB, Ginty F, Prehn JHM. Spatial Effects of Infiltrating T cells on Neighbouring Cancer Cells and Prognosis in Stage III CRC patients. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.30.577720. [PMID: 38352309 PMCID: PMC10862776 DOI: 10.1101/2024.01.30.577720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/22/2024]
Abstract
Colorectal cancer (CRC) is one of the most frequently occurring cancers, but prognostic biomarkers identifying patients at risk of recurrence are still lacking. In this study, we aimed to investigate in more detail the spatial relationship between intratumoural T cells, cancer cells, and cancer cell hallmarks, as prognostic biomarkers in stage III colorectal cancer patients. We conducted multiplexed imaging of 56 protein markers at single cell resolution on resected fixed tissue from stage III CRC patients who received adjuvant 5-fluorouracil-based chemotherapy. Images underwent segmentation for tumour, stroma and immune cells, and cancer cell 'state' protein marker expression was quantified at a cellular level. We developed a Python package for estimation of spatial proximity, nearest neighbour analysis focusing on cancer cell - T cell interactions at single-cell level. In our discovery cohort (MSK), we processed 462 core samples (total number of cells: 1,669,228) from 221 adjuvant 5FU-treated stage III patients. The validation cohort (HV) consisted of 272 samples (total number of cells: 853,398) from 98 stage III CRC patients. While there were trends for an association between percentage of cytotoxic T cells (across the whole cancer core), it did not reach significance (Discovery cohort: p = 0.07, Validation cohort: p = 0.19). We next utilized our region-based nearest neighbourhood approach to determine the spatial relationships between cytotoxic T cells, helper T cells and cancer cell clusters. In the both cohorts, we found that lower distance between cytotoxic T cells, T helper cells and cancer cells was significantly associated with increased disease-free survival. An unsupervised trained model that clustered patients based on the median distance between immune cells and cancer cells, as well as protein expression profiles, successfully classified patients into low-risk and high-risk groups (Discovery cohort: p = 0.01, Validation cohort: p = 0.003).
Collapse
Affiliation(s)
- Mohammadreza Azimi
- Department of Physiology and Medical Physics, RCSI Centre for Systems Medicine, Royal College of Surgeons in Ireland University of Medicine and Health Sciences, Dublin 2, Ireland
| | - Sanghee Cho
- GE HealthCare Technology and Innovation Center, Niskayuna, NY, 12309, USA (formerly GE Research Center)
| | - Emir Bozkurt
- Department of Physiology and Medical Physics, RCSI Centre for Systems Medicine, Royal College of Surgeons in Ireland University of Medicine and Health Sciences, Dublin 2, Ireland
| | - Elizabeth McDonough
- GE HealthCare Technology and Innovation Center, Niskayuna, NY, 12309, USA (formerly GE Research Center)
| | - Batuhan Kisakol
- Department of Physiology and Medical Physics, RCSI Centre for Systems Medicine, Royal College of Surgeons in Ireland University of Medicine and Health Sciences, Dublin 2, Ireland
| | - Anna Matveeva
- Department of Physiology and Medical Physics, RCSI Centre for Systems Medicine, Royal College of Surgeons in Ireland University of Medicine and Health Sciences, Dublin 2, Ireland
| | - Manuela Salvucci
- Department of Physiology and Medical Physics, RCSI Centre for Systems Medicine, Royal College of Surgeons in Ireland University of Medicine and Health Sciences, Dublin 2, Ireland
| | - Heiko Dussmann
- Department of Physiology and Medical Physics, RCSI Centre for Systems Medicine, Royal College of Surgeons in Ireland University of Medicine and Health Sciences, Dublin 2, Ireland
| | - Simon McDade
- School of Medicine, Dentistry and Biomedical Sciences, Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, 97 Lisburn Road, Belfast, BT9 7AE, Northern Ireland, UK
| | | | | | - Jinru Shia
- Memorial Sloan Kettering Cancer Centre, NY
| | - Daniel B Longley
- School of Medicine, Dentistry and Biomedical Sciences, Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, 97 Lisburn Road, Belfast, BT9 7AE, Northern Ireland, UK
| | - Fiona Ginty
- GE HealthCare Technology and Innovation Center, Niskayuna, NY, 12309, USA (formerly GE Research Center)
| | - Jochen H M Prehn
- Department of Physiology and Medical Physics, RCSI Centre for Systems Medicine, Royal College of Surgeons in Ireland University of Medicine and Health Sciences, Dublin 2, Ireland
| |
Collapse
|
13
|
Malaguti M, Lebek T, Blin G, Lowell S. Enabling neighbour labelling: using synthetic biology to explore how cells influence their neighbours. Development 2024; 151:dev201955. [PMID: 38165174 PMCID: PMC10820747 DOI: 10.1242/dev.201955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Accepted: 11/28/2023] [Indexed: 01/03/2024]
Abstract
Cell-cell interactions are central to development, but exploring how a change in any given cell relates to changes in the neighbour of that cell can be technically challenging. Here, we review recent developments in synthetic biology and image analysis that are helping overcome this problem. We highlight the opportunities presented by these advances and discuss opportunities and limitations in applying them to developmental model systems.
Collapse
Affiliation(s)
- Mattias Malaguti
- Centre for Regenerative Medicine, Institute for Stem Cell Research, School of Biological Sciences, University of Edinburgh, 5 Little France Drive, Edinburgh EH16 4UU, UK
| | - Tamina Lebek
- Centre for Regenerative Medicine, Institute for Stem Cell Research, School of Biological Sciences, University of Edinburgh, 5 Little France Drive, Edinburgh EH16 4UU, UK
| | - Guillaume Blin
- Centre for Regenerative Medicine, Institute for Stem Cell Research, School of Biological Sciences, University of Edinburgh, 5 Little France Drive, Edinburgh EH16 4UU, UK
| | - Sally Lowell
- Centre for Regenerative Medicine, Institute for Stem Cell Research, School of Biological Sciences, University of Edinburgh, 5 Little France Drive, Edinburgh EH16 4UU, UK
| |
Collapse
|
14
|
Merighi A, Lossi L. Co-cultures of cerebellar slices from mice with different reelin genetic backgrounds as a model to study cortical lamination. F1000Res 2023; 11:1183. [PMID: 37881513 PMCID: PMC10594056 DOI: 10.12688/f1000research.126787.1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/07/2023] [Indexed: 10/27/2023] Open
Abstract
Background: Reelin has fundamental functions in the developing and mature brain. Its absence gives rise to the Reeler phenotype in mice, the first described cerebellar mutation. In homozygous mutants missing the Reelin gene ( reln -/-), neurons are incapable of correctly positioning themselves in layered brain areas such as the cerebral and cerebellar cortices. We here demonstrate that by employing ex vivo cultured cerebellar slices one can reduce the number of animals and use a non-recovery procedure to analyze the effects of Reelin on the migration of Purkinje neurons (PNs). Methods: We generated mouse hybrids (L7-GFP relnF1/) with green fluorescent protein (GFP)-tagged PNs, directly visible under fluorescence microscopy. We then cultured the slices obtained from mice with different reln genotypes and demonstrated that when the slices from reln -/- mutants were co-cultured with those from reln +/- mice, the Reelin produced by the latter induced migration of the PNs to partially rescue the normal layered cortical histology. We have confirmed this observation with Voronoi tessellation to analyze PN dispersion. Results: In images of the co-cultured slices from reln -/- mice, Voronoi polygons were larger than in single-cultured slices of the same genetic background but smaller than those generated from slices of reln +/- animals. The mean roundness factor, area disorder, and roundness factor homogeneity were different when slices from reln -/- mice were cultivated singularly or co-cultivated, supporting mathematically the transition from the clustered organization of the PNs in the absence of Reelin to a layered structure when the protein is supplied ex vivo. Conclusions: Neurobiologists are the primary target users of this 3Rs approach. They should adopt it for the possibility to study and manipulate ex vivo the activity of a brain-secreted or genetically engineered protein (scientific perspective), the potential reduction (up to 20%) of the animals used, and the total avoidance of severe surgery (3Rs perspective).
Collapse
Affiliation(s)
- Adalberto Merighi
- Department of Veterinary Sciences, University of Turin, Grugliasco, 10095, Italy
| | - Laura Lossi
- Department of Veterinary Sciences, University of Turin, Grugliasco, 10095, Italy
| |
Collapse
|
15
|
Merighi A, Lossi L. Co-cultures of cerebellar slices from mice with different reelin genetic backgrounds as a model to study cortical lamination. F1000Res 2023; 11:1183. [PMID: 37881513 PMCID: PMC10594056 DOI: 10.12688/f1000research.126787.2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/07/2023] [Indexed: 10/27/2023] Open
Abstract
Background: Reelin has fundamental functions in the developing and mature brain. Its absence gives rise to the Reeler phenotype in mice, the first described cerebellar mutation. In homozygous mutants missing the Reelin gene ( reln -/-), neurons are incapable of correctly positioning themselves in layered brain areas such as the cerebral and cerebellar cortices. We here demonstrate that by employing ex vivo cultured cerebellar slices one can reduce the number of animals and use a non-recovery procedure to analyze the effects of Reelin on the migration of Purkinje neurons (PNs). Methods: We generated mouse hybrids (L7-GFP relnF1/) with green fluorescent protein (GFP)-tagged PNs, directly visible under fluorescence microscopy. We then cultured the slices obtained from mice with different reln genotypes and demonstrated that when the slices from reln -/- mutants were co-cultured with those from reln +/- mice, the Reelin produced by the latter induced migration of the PNs to partially rescue the normal layered cortical histology. We have confirmed this observation with Voronoi tessellation to analyze PN dispersion. Results: In images of the co-cultured slices from reln -/- mice, Voronoi polygons were larger than in single-cultured slices of the same genetic background but smaller than those generated from slices of reln +/- animals. The mean roundness factor, area disorder, and roundness factor homogeneity were different when slices from reln -/- mice were cultivated singularly or co-cultivated, supporting mathematically the transition from the clustered organization of the PNs in the absence of Reelin to a layered structure when the protein is supplied ex vivo. Conclusions: Neurobiologists are the primary target users of this 3Rs approach. They should adopt it for the possibility to study and manipulate ex vivo the activity of a brain-secreted or genetically engineered protein (scientific perspective), the potential reduction (up to 20%) of the animals used, and the total avoidance of severe surgery (3Rs perspective).
Collapse
Affiliation(s)
- Adalberto Merighi
- Department of Veterinary Sciences, University of Turin, Grugliasco, 10095, Italy
| | - Laura Lossi
- Department of Veterinary Sciences, University of Turin, Grugliasco, 10095, Italy
| |
Collapse
|
16
|
Abstract
Multiplex imaging has emerged as an invaluable tool for immune-oncologists and translational researchers, enabling them to examine intricate interactions among immune cells, stroma, matrix, and malignant cells within the tumor microenvironment (TME). It holds significant promise in the quest to discover improved biomarkers for treatment stratification and identify novel therapeutic targets. Nonetheless, several challenges exist in the realms of study design, experiment optimization, and data analysis. In this review, our aim is to present an overview of the utilization of multiplex imaging in immuno-oncology studies and inform novice researchers about the fundamental principles at each stage of the imaging and analysis process.
Collapse
Affiliation(s)
- Chen Zhao
- Thoracic and GI Malignancies Branch, CCR, NCI, Bethesda, Maryland, USA
- Lymphocyte Biology Section, Laboratory of Immune System Biology, NIAID, Bethesda, Maryland, USA
| | - Ronald N Germain
- Lymphocyte Biology Section, Laboratory of Immune System Biology, NIAID, Bethesda, Maryland, USA
| |
Collapse
|