1
|
Liechti T, Lelios I, Schroeder A, Decman V, Gonneau C, Groves C, Green C, Alcaide EG. Potential and challenges of clinical high-dimensional flow cytometry: A call to action. Cytometry A 2024; 105:829-837. [PMID: 39444224 DOI: 10.1002/cyto.a.24902] [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: 06/21/2024] [Revised: 10/02/2024] [Accepted: 10/06/2024] [Indexed: 10/25/2024]
Abstract
Clinical biomarker strategies increasingly integrate translational research to gain new insights into disease mechanisms or to define better biomarkers in clinical trials. High-dimensional flow cytometry (HDFCM) holds the promise to enhance the exploratory potential beyond traditional, targeted biomarker strategies. However, the increased complexity of HDFCM poses several challenges, which need to be addressed in order to fully leverage its potential and to align with current regulatory requirements in clinical flow cytometry. These challenges include among others extended timelines for assay development and validation, the necessity for extensive knowledge in HDFCM, and sophisticated data analysis strategies. However, no guidelines exist on how to manage such challenges in adopting clinical HDFCM. Our CYTO 2024 workshop "Potential and challenges of clinical high-dimensional flow cytometry" aimed to find consensus across the pharmaceutical industry and broader scientific community on the overall benefits and most urgent challenges of HDFCM in clinical trials. Here, we summarize the insights we gained from our workshop. While this report does not provide a blueprint, it is a first step in defining and summarizing the most pressing challenges in implementing HDFCM in clinical trials. Furthermore, we compile current efforts with the goal to overcome some of these challenges. As such we bring the scientific community and health authorities together to build solutions, which will accelerate and simplify the full adoption of HDFCM in clinical trials.
Collapse
Affiliation(s)
- Thomas Liechti
- Translational Medicine, Genentech, San Francisco, California, USA
| | - Iva Lelios
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland
| | - Aaron Schroeder
- Translational Medicine, Genentech, San Francisco, California, USA
| | - Vilma Decman
- Cellular Biomarkers, GSK, Collegeville, Pennsylvania, USA
| | | | - Christopher Groves
- Translational Science and Innovation Laboratory, Q2 Solutions, Durham, North Carolina, USA
| | - Cherie Green
- Translational Science, Ozette, Seattle, Washington, USA
| | - Enrique Gomez Alcaide
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland
| |
Collapse
|
2
|
Coppard V, Szep G, Georgieva Z, Howlett SK, Jarvis LB, Rainbow DB, Suchanek O, Needham EJ, Mousa HS, Menon DK, Feyertag F, Mahbubani KT, Saeb-Parsy K, Jones JL. FlowAtlas: an interactive tool for high-dimensional immunophenotyping analysis bridging FlowJo with computational tools in Julia. Front Immunol 2024; 15:1425488. [PMID: 39086484 PMCID: PMC11288863 DOI: 10.3389/fimmu.2024.1425488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 07/02/2024] [Indexed: 08/02/2024] Open
Abstract
As the dimensionality, throughput and complexity of cytometry data increases, so does the demand for user-friendly, interactive analysis tools that leverage high-performance machine learning frameworks. Here we introduce FlowAtlas: an interactive web application that enables dimensionality reduction of cytometry data without down-sampling and that is compatible with datasets stained with non-identical panels. FlowAtlas bridges the user-friendly environment of FlowJo and computational tools in Julia developed by the scientific machine learning community, eliminating the need for coding and bioinformatics expertise. New population discovery and detection of rare populations in FlowAtlas is intuitive and rapid. We demonstrate the capabilities of FlowAtlas using a human multi-tissue, multi-donor immune cell dataset, highlighting key immunological findings. FlowAtlas is available at https://github.com/gszep/FlowAtlas.jl.git.
Collapse
Affiliation(s)
- Valerie Coppard
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
| | - Grisha Szep
- Randall Centre for Cell & Molecular Biophysics, King’s College London, London, United Kingdom
| | - Zoya Georgieva
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
| | - Sarah K. Howlett
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
| | - Lorna B. Jarvis
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
| | - Daniel B. Rainbow
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
| | - Ondrej Suchanek
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Edward J. Needham
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
| | - Hani S. Mousa
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
| | - David K. Menon
- Department of Anaesthesia, University of Cambridge, Cambridge, United Kingdom
| | | | - Krishnaa T. Mahbubani
- Department of Surgery, University of Cambridge, Cambridge, United Kingdom
- Collaborative Biorepository for Translational Medicine (CBTM), Cambridge NIHR Biomedical Research Centre, Cambridge, United Kingdom
| | - Kourosh Saeb-Parsy
- Department of Surgery, University of Cambridge, Cambridge, United Kingdom
- Collaborative Biorepository for Translational Medicine (CBTM), Cambridge NIHR Biomedical Research Centre, Cambridge, United Kingdom
| | - Joanne L. Jones
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
3
|
Zheng Y, Caron DP, Kim JY, Jun SH, Tian Y, Florian M, Stuart KD, Sims PA, Gottardo R. ADTnorm: Robust Integration of Single-cell Protein Measurement across CITE-seq Datasets. RESEARCH SQUARE 2024:rs.3.rs-4572811. [PMID: 39041028 PMCID: PMC11261982 DOI: 10.21203/rs.3.rs-4572811/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/24/2024]
Abstract
CITE-seq enables paired measurement of surface protein and mRNA expression in single cells using antibodies conjugated to oligonucleotide tags. Due to the high copy number of surface protein molecules, sequencing antibody-derived tags (ADTs) allows for robust protein detection, improving cell-type identification. However, variability in antibody staining leads to batch effects in the ADT expression, obscuring biological variation, reducing interpretability, and obstructing cross-study analyses. Here, we present ADTnorm (https://github.com/yezhengSTAT/ADTnorm), a normalization and integration method designed explicitly for ADT abundance. Benchmarking against 14 existing scaling and normalization methods, we show that ADTnorm accurately aligns populations with negative- and positive-expression of surface protein markers across 13 public datasets, effectively removing technical variation across batches and improving cell-type separation. ADTnorm enables efficient integration of public CITE-seq datasets, each with unique experimental designs, paving the way for atlas-level analyses. Beyond normalization, ADTnorm includes built-in utilities to aid in automated threshold-gating as well as assessment of antibody staining quality for titration optimization and antibody panel selection. Applying ADTnorm to a published COVID-19 CITE-seq dataset allowed for identifying previously undetected disease-associated markers, illustrating a broad utility in biological applications.
Collapse
Affiliation(s)
- Ye Zheng
- Basic Science Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| | - Daniel P. Caron
- Department of Microbiology and Immunology, Columbia University, New York, NY 10032, USA
| | - Ju Yeong Kim
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| | - Seong-Hwan Jun
- Department of Biostatistics and Computational Biology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Yuan Tian
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| | - Mair Florian
- Department of Biology, ETH Zürich, Zürich 8093, Switzerland
| | - Kenneth D. Stuart
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA, United States
| | - Peter A. Sims
- Department of Systems Biology, Columbia University, New York, NY 10032, USA
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY 10032, USA
| | - Raphael Gottardo
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
- Biomedical Data Science Center, Lausanne University Hospital and University of Lausanne, Lausanne 1005, Switzerland
| |
Collapse
|
4
|
Dinalankara W, Ng DP, Marchionni L, Simonson PD. Comparison of three machine learning algorithms for classification of B-cell neoplasms using clinical flow cytometry data. CYTOMETRY. PART B, CLINICAL CYTOMETRY 2024; 106:282-293. [PMID: 38721890 DOI: 10.1002/cyto.b.22177] [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: 11/29/2023] [Revised: 03/22/2024] [Accepted: 04/12/2024] [Indexed: 05/18/2024]
Abstract
Multiparameter flow cytometry data is visually inspected by expert personnel as part of standard clinical disease diagnosis practice. This is a demanding and costly process, and recent research has demonstrated that it is possible to utilize artificial intelligence (AI) algorithms to assist in the interpretive process. Here we report our examination of three previously published machine learning methods for classification of flow cytometry data and apply these to a B-cell neoplasm dataset to obtain predicted disease subtypes. Each of the examined methods classifies samples according to specific disease categories using ungated flow cytometry data. We compare and contrast the three algorithms with respect to their architectures, and we report the multiclass classification accuracies and relative required computation times. Despite different architectures, two of the methods, flowCat and EnsembleCNN, had similarly good accuracies with relatively fast computational times. We note a speed advantage for EnsembleCNN, particularly in the case of addition of training data and retraining of the classifier.
Collapse
Affiliation(s)
- Wikum Dinalankara
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, New York, USA
| | - David P Ng
- Department of Pathology, University of Utah, Salt Lake City, Utah, USA
| | - Luigi Marchionni
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, New York, USA
| | - Paul D Simonson
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, New York, USA
| |
Collapse
|
5
|
Caron DP, Specht WL, Chen D, Wells SB, Szabo PA, Jensen IJ, Farber DL, Sims PA. Multimodal hierarchical classification of CITE-seq data delineates immune cell states across lineages and tissues. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.07.06.547944. [PMID: 37461466 PMCID: PMC10350048 DOI: 10.1101/2023.07.06.547944] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/27/2023]
Abstract
Single-cell RNA sequencing (scRNA-seq) is invaluable for profiling cellular heterogeneity and dissecting transcriptional states, but transcriptomic profiles do not always delineate subsets defined by surface proteins, as in cells of the immune system. Cellular Indexing of Transcriptomes and Epitopes (CITE-seq) enables simultaneous profiling of single-cell transcriptomes and surface proteomes; however, accurate cell type annotation requires a classifier that integrates multimodal data. Here, we describe MultiModal Classifier Hierarchy (MMoCHi), a marker-based approach for classification, reconciling gene and protein expression without reliance on reference atlases. We benchmark MMoCHi using sorted T lymphocyte subsets and annotate a cross-tissue human immune cell dataset. MMoCHi outperforms leading transcriptome-based classifiers and multimodal unsupervised clustering in its ability to identify immune cell subsets that are not readily resolved and to reveal novel subset markers. MMoCHi is designed for adaptability and can integrate annotation of cell types and developmental states across diverse lineages, samples, or modalities.
Collapse
Affiliation(s)
- Daniel P. Caron
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY, USA
| | - William L. Specht
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY, USA
| | - David Chen
- Department of Systems Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Steven B. Wells
- Department of Systems Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Peter A. Szabo
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY, USA
| | - Isaac J. Jensen
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY, USA
| | - Donna L. Farber
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY, USA
- Department of Surgery, Columbia University Irving Medical Center, New York, NY, USA
| | - Peter A. Sims
- Department of Systems Biology, Columbia University Irving Medical Center, New York, NY, USA
- Department of Biochemistry and Molecular Biophysics, Columbia University Irving Medical Center, New York, NY, USA
| |
Collapse
|
6
|
Giovenzana A, Bezzecchi E, Bichisecchi A, Cardellini S, Ragogna F, Pedica F, Invernizzi F, Di Filippo L, Tomajer V, Aleotti F, Scotti GM, Socci C, Cesana G, Olmi S, Morelli MJ, Falconi M, Giustina A, Bonini C, Piemonti L, Ruggiero E, Petrelli A. Fat-to-blood recirculation of partially dysfunctional PD-1 +CD4 Tconv cells is associated with dysglycemia in human obesity. iScience 2024; 27:109032. [PMID: 38380252 PMCID: PMC10877684 DOI: 10.1016/j.isci.2024.109032] [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: 08/09/2023] [Revised: 01/03/2024] [Accepted: 01/23/2024] [Indexed: 02/22/2024] Open
Abstract
Obesity is characterized by the accumulation of T cells in insulin-sensitive tissues, including the visceral adipose tissue (VAT), that can interfere with the insulin signaling pathway eventually leading to insulin resistance (IR) and type 2 diabetes. Here, we found that PD-1+CD4 conventional T (Tconv) cells, endowed with a transcriptomic and functional profile of partially dysfunctional cells, are diminished in VAT of obese patients with dysglycemia (OB-Dys), without a concomitant increase in apoptosis. These cells showed enhanced capacity to recirculate into the bloodstream and had a non-restricted TCRβ repertoire divergent from that of normoglycemic obese and lean individuals. PD-1+CD4 Tconv were reduced in the circulation of OB-Dys, exhibited an altered migration potential, and were detected in the liver of patients with non-alcoholic steatohepatitis. The findings suggest a potential role for partially dysfunctional PD-1+CD4 Tconv cells as inter-organ mediators of IR in obese patients with dysglycemic.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | - Stefano Olmi
- San Marco Hospital GSD, Zingonia, Bergamo, Italy
- Università Vita-Salute San Raffaele, Milan, Italy
| | | | - Massimo Falconi
- IRCCS Ospedale San Raffaele, Milan, Italy
- Università Vita-Salute San Raffaele, Milan, Italy
| | - Andrea Giustina
- IRCCS Ospedale San Raffaele, Milan, Italy
- Università Vita-Salute San Raffaele, Milan, Italy
| | - Chiara Bonini
- IRCCS Ospedale San Raffaele, Milan, Italy
- Università Vita-Salute San Raffaele, Milan, Italy
| | - Lorenzo Piemonti
- IRCCS Ospedale San Raffaele, Milan, Italy
- Università Vita-Salute San Raffaele, Milan, Italy
| | | | | |
Collapse
|
7
|
Perik-Zavodskaia O, Perik-Zavodskii R, Nazarov K, Volynets M, Alrhmoun S, Shevchenko J, Sennikov S. Murine Bone Marrow Erythroid Cells Have Two Branches of Differentiation Defined by the Presence of CD45 and a Different Immune Transcriptome Than Fetal Liver Erythroid Cells. Int J Mol Sci 2023; 24:15752. [PMID: 37958735 PMCID: PMC10650492 DOI: 10.3390/ijms242115752] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Revised: 10/18/2023] [Accepted: 10/29/2023] [Indexed: 11/15/2023] Open
Abstract
Mouse erythropoiesis is a multifaceted process involving the intricate interplay of proliferation, differentiation, and maturation of erythroid cells, leading to significant changes in their transcriptomic and proteomic profiles. While the immunoregulatory role of murine erythroid cells has been recognized historically, modern investigative techniques have been sparingly applied to decipher their functions. To address this gap, our study sought to comprehensively characterize mouse erythroid cells through contemporary transcriptomic and proteomic approaches. By evaluating CD71 and Ter-119 as sorting markers for murine erythroid cells and employing bulk NanoString transcriptomics, we discerned distinctive gene expression profiles between bone marrow and fetal liver-derived erythroid cells. Additionally, leveraging flow cytometry, we assessed the surface expression of CD44, CD45, CD71, and Ter-119 on normal and phenylhydrazine-induced hemolytic anemia mouse bone marrow and splenic erythroid cells. Key findings emerged: firstly, the utilization of CD71 for cell sorting yielded comparatively impure erythroid cell populations compared to Ter-119; secondly, discernible differences in immunoregulatory molecule expression were evident between erythroid cells from mouse bone marrow and fetal liver; thirdly, two discrete branches of mouse erythropoiesis were identified based on CD45 expression: CD45-negative and CD45-positive, which had been altered differently in response to phenylhydrazine. Our deductions underscore (1) Ter-119's superiority over CD71 as a murine erythroid cell sorting marker, (2) the potential of erythroid cells in murine antimicrobial immunity, and (3) the importance of investigating CD45-positive and CD45-negative murine erythroid cells separately and in further detail in future studies.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Sergey Sennikov
- Laboratory of Molecular Immunology, Federal State Budgetary Scientific Institution Research Institute of Fundamental and Clinical Immunology, 630099 Novosibirsk, Russia; (O.P.-Z.); (R.P.-Z.); (K.N.); (M.V.); (S.A.)
| |
Collapse
|
8
|
Régnier P, Le Joncour A, Maciejewski-Duval A, Darrasse-Jèze G, Dolladille C, Meijers WC, Bastarache L, Fouret P, Bruneval P, Arbaretaz F, Sayetta C, Márquez A, Rosenzwajg M, Klatzmann D, Cacoub P, Moslehi JJ, Salem JE, Saadoun D. CTLA-4 Pathway Is Instrumental in Giant Cell Arteritis. Circ Res 2023; 133:298-312. [PMID: 37435729 DOI: 10.1161/circresaha.122.322330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Accepted: 06/28/2023] [Indexed: 07/13/2023]
Abstract
BACKGROUND Giant cell arteritis (GCA) causes severe inflammation of the aorta and its branches and is characterized by intense effector T-cell infiltration. The roles that immune checkpoints play in the pathogenesis of GCA are still unclear. Our aim was to study the immune checkpoint interplay in GCA. METHODS First, we used VigiBase, the World Health Organization international pharmacovigilance database, to evaluate the relationship between GCA occurrence and immune checkpoint inhibitors treatments. We then further dissected the role of immune checkpoint inhibitors in the pathogenesis of GCA, using immunohistochemistry, immunofluorescence, transcriptomics, and flow cytometry on peripheral blood mononuclear cells and aortic tissues of GCA patients and appropriated controls. RESULTS Using VigiBase, we identified GCA as a significant immune-related adverse event associated with anti-CTLA-4 (cytotoxic T-lymphocyte-associated protein-4) but not anti-PD-1 (anti-programmed death-1) nor anti-PD-L1 (anti-programmed death-ligand 1) treatment. We further dissected a critical role for the CTLA-4 pathway in GCA by identification of the dysregulation of CTLA-4-derived gene pathways and proteins in CD4+ (cluster of differentiation 4) T cells (and specifically regulatory T cells) present in blood and aorta of GCA patients versus controls. While regulatory T cells were less abundant and activated/suppressive in blood and aorta of GCA versus controls, they still specifically upregulated CTLA-4. Activated and proliferating CTLA-4+ Ki-67+ regulatory T cells from GCA were more sensitive to anti-CTLA-4 (ipilimumab)-mediated in vitro depletion versus controls. CONCLUSIONS We highlighted the instrumental role of CTLA-4 immune checkpoint in GCA, which provides a strong rationale for targeting this pathway.
Collapse
Affiliation(s)
- Paul Régnier
- Immunology-Immunopathology-Immunotherapy (i3) Laboratory, INSERM UMR-S 959, Sorbonne Université, Paris, France (P.R., A.L.J., A.M.-D., G.D.-J., M.R., D.K., P.C., D.S.), Assistance Publique-Hôpitaux de Paris (AP-HP), France
- Biotherapy Unit (CIC-BTi), Inflammation-Immunopathology-Biotherapy Department (DHU i2B), Groupe Hospitalier Pitié-Salpêtrière (P.R., A.L.J., A.M.-D., M.R., D.K., P.C., D.S.), Assistance Publique-Hôpitaux de Paris (AP-HP), France
| | - Alexandre Le Joncour
- Immunology-Immunopathology-Immunotherapy (i3) Laboratory, INSERM UMR-S 959, Sorbonne Université, Paris, France (P.R., A.L.J., A.M.-D., G.D.-J., M.R., D.K., P.C., D.S.), Assistance Publique-Hôpitaux de Paris (AP-HP), France
- Biotherapy Unit (CIC-BTi), Inflammation-Immunopathology-Biotherapy Department (DHU i2B), Groupe Hospitalier Pitié-Salpêtrière (P.R., A.L.J., A.M.-D., M.R., D.K., P.C., D.S.), Assistance Publique-Hôpitaux de Paris (AP-HP), France
- Département de Médecine Interne et Immunologie Clinique, Sorbonne Université, AP-HP, Groupe Hospitalier Pitié-Salpêtrière, Paris, France (A.L.J., P.C., D.S.)
- Centre National de Référence Maladies Autoimmunes Systémiques Rares, Centre National de Référence Maladies Autoinflammatoires et Amylose Inflammatoire, Inflammation-Immunopathology-Biotherapy Department (DMU 3iD), Paris, France (A.L.J., P.C., D.S.)
| | - Anna Maciejewski-Duval
- Immunology-Immunopathology-Immunotherapy (i3) Laboratory, INSERM UMR-S 959, Sorbonne Université, Paris, France (P.R., A.L.J., A.M.-D., G.D.-J., M.R., D.K., P.C., D.S.), Assistance Publique-Hôpitaux de Paris (AP-HP), France
- Biotherapy Unit (CIC-BTi), Inflammation-Immunopathology-Biotherapy Department (DHU i2B), Groupe Hospitalier Pitié-Salpêtrière (P.R., A.L.J., A.M.-D., M.R., D.K., P.C., D.S.), Assistance Publique-Hôpitaux de Paris (AP-HP), France
| | - Guillaume Darrasse-Jèze
- Immunology-Immunopathology-Immunotherapy (i3) Laboratory, INSERM UMR-S 959, Sorbonne Université, Paris, France (P.R., A.L.J., A.M.-D., G.D.-J., M.R., D.K., P.C., D.S.), Assistance Publique-Hôpitaux de Paris (AP-HP), France
- Faculté de Médecine Paris Descartes (G.D.-J.), Université de Paris, France
| | - Charles Dolladille
- Normandie University, University of Caen Normandy, Centre Hospitalier Universitaire (CHU) de Caen Normandie, PICARO Cardio-Oncology Program, Department of Pharmacology, INSERM ANTICIPE U1086: Unité de Recherche Interdisciplinaire pour la Prévention et le Traitement des Cancers, Centre François Baclesse, France (C.D.)
| | - Wouter C Meijers
- Department of Cardiology, University Medical Center Groningen, University of Groningen, the Netherlands (W.C.M., J.-E.S.)
| | - Lisa Bastarache
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN (L.B.)
| | - Pierre Fouret
- Service d'anatomie et cytologie pathologiques, Groupe Hospitalier Pitié-Salpêtrière (P.F.), Assistance Publique-Hôpitaux de Paris (AP-HP), France
| | - Patrick Bruneval
- Service d'anatomie pathologie, Hôpital Européen Georges Pompidou (P.B.), Assistance Publique-Hôpitaux de Paris (AP-HP), France
| | - Floriane Arbaretaz
- Centre d'Histologie, d'Imagerie et de Cytométrie, Centre de Recherche des Cordeliers, Sorbonne Université, INSERM (F.A.), Université de Paris, France
| | - Célia Sayetta
- ICM Institut du Cerveau, CNRS UMR7225, INSERM U1127, Sorbonne Université, Hôpital de la Pitié-Salpêtrière, Paris, France (C.S.)
| | - Ana Márquez
- Instituto de Parasitología y Biomedicina "López-Neyra," CSIC, PTS Granada, Spain (A.M.)
- Systemic Autoimmune Disease Unit, Instituto de Investigación Biosanitaria de Granada, Spain (A.M.)
| | - Michelle Rosenzwajg
- Immunology-Immunopathology-Immunotherapy (i3) Laboratory, INSERM UMR-S 959, Sorbonne Université, Paris, France (P.R., A.L.J., A.M.-D., G.D.-J., M.R., D.K., P.C., D.S.), Assistance Publique-Hôpitaux de Paris (AP-HP), France
- Biotherapy Unit (CIC-BTi), Inflammation-Immunopathology-Biotherapy Department (DHU i2B), Groupe Hospitalier Pitié-Salpêtrière (P.R., A.L.J., A.M.-D., M.R., D.K., P.C., D.S.), Assistance Publique-Hôpitaux de Paris (AP-HP), France
| | - David Klatzmann
- Immunology-Immunopathology-Immunotherapy (i3) Laboratory, INSERM UMR-S 959, Sorbonne Université, Paris, France (P.R., A.L.J., A.M.-D., G.D.-J., M.R., D.K., P.C., D.S.), Assistance Publique-Hôpitaux de Paris (AP-HP), France
- Biotherapy Unit (CIC-BTi), Inflammation-Immunopathology-Biotherapy Department (DHU i2B), Groupe Hospitalier Pitié-Salpêtrière (P.R., A.L.J., A.M.-D., M.R., D.K., P.C., D.S.), Assistance Publique-Hôpitaux de Paris (AP-HP), France
| | - Patrice Cacoub
- Immunology-Immunopathology-Immunotherapy (i3) Laboratory, INSERM UMR-S 959, Sorbonne Université, Paris, France (P.R., A.L.J., A.M.-D., G.D.-J., M.R., D.K., P.C., D.S.), Assistance Publique-Hôpitaux de Paris (AP-HP), France
- Biotherapy Unit (CIC-BTi), Inflammation-Immunopathology-Biotherapy Department (DHU i2B), Groupe Hospitalier Pitié-Salpêtrière (P.R., A.L.J., A.M.-D., M.R., D.K., P.C., D.S.), Assistance Publique-Hôpitaux de Paris (AP-HP), France
- Département de Médecine Interne et Immunologie Clinique, Sorbonne Université, AP-HP, Groupe Hospitalier Pitié-Salpêtrière, Paris, France (A.L.J., P.C., D.S.)
- Centre National de Référence Maladies Autoimmunes Systémiques Rares, Centre National de Référence Maladies Autoinflammatoires et Amylose Inflammatoire, Inflammation-Immunopathology-Biotherapy Department (DMU 3iD), Paris, France (A.L.J., P.C., D.S.)
| | - Javid J Moslehi
- Section of Cardio-Oncology and Immunology, Division of Cardiology and the Cardiovascular Research Institute, University of California San Francisco (J.J.M.)
| | - Joe-Elie Salem
- Department of Pharmacology, INSERM, CIC-1901, UNICO-GRECO Cardiooncology Program, Sorbonne Université (J.-E.S.), Assistance Publique-Hôpitaux de Paris (AP-HP), France
- Department of Cardiology, University Medical Center Groningen, University of Groningen, the Netherlands (W.C.M., J.-E.S.)
| | - David Saadoun
- Immunology-Immunopathology-Immunotherapy (i3) Laboratory, INSERM UMR-S 959, Sorbonne Université, Paris, France (P.R., A.L.J., A.M.-D., G.D.-J., M.R., D.K., P.C., D.S.), Assistance Publique-Hôpitaux de Paris (AP-HP), France
- Biotherapy Unit (CIC-BTi), Inflammation-Immunopathology-Biotherapy Department (DHU i2B), Groupe Hospitalier Pitié-Salpêtrière (P.R., A.L.J., A.M.-D., M.R., D.K., P.C., D.S.), Assistance Publique-Hôpitaux de Paris (AP-HP), France
- Département de Médecine Interne et Immunologie Clinique, Sorbonne Université, AP-HP, Groupe Hospitalier Pitié-Salpêtrière, Paris, France (A.L.J., P.C., D.S.)
- Centre National de Référence Maladies Autoimmunes Systémiques Rares, Centre National de Référence Maladies Autoinflammatoires et Amylose Inflammatoire, Inflammation-Immunopathology-Biotherapy Department (DMU 3iD), Paris, France (A.L.J., P.C., D.S.)
| |
Collapse
|
9
|
Pura JA, Li X, Chan C, Xie J. TEAM: A MULTIPLE TESTING ALGORITHM ON THE AGGREGATION TREE FOR FLOW CYTOMETRY ANALYSIS. Ann Appl Stat 2023; 17:621-640. [PMID: 38736649 PMCID: PMC11083434 DOI: 10.1214/22-aoas1645] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/14/2024]
Abstract
In immunology studies, flow cytometry is a commonly used multivariate single-cell assay. One key goal in flow cytometry analysis is to detect the immune cells responsive to certain stimuli. Statistically, this problem can be translated into comparing two protein expression probability density functions (pdfs) before and after the stimulus; the goal is to pinpoint the regions where these two pdfs differ. Further screening of these differential regions can be performed to identify enriched sets of responsive cells. In this paper, we model identifying differential density regions as a multiple testing problem. First, we partition the sample space into small bins. In each bin, we form a hypothesis to test the existence of differential pdfs. Second, we develop a novel multiple testing method, called TEAM (Testing on the Aggregation tree Method), to identify those bins that harbor differential pdfs while controlling the false discovery rate (FDR) under the desired level. TEAM embeds the testing procedure into an aggregation tree to test from fine- to coarse-resolution. The procedure achieves the statistical goal of pinpointing density differences to the smallest possible regions. TEAM is computationally efficient, capable of analyzing large flow cytometry data sets in much shorter time compared with competing methods. We applied TEAM and competing methods on a flow cytometry data set to identify T cells responsive to the cytomegalovirus (CMV)-pp65 antigen stimulation. With additional downstream screening, TEAM successfully identified enriched sets containing monofunctional, bifunctional, and polyfunctional T cells. Competing methods either did not finish in a reasonable time frame or provided less interpretable results. Numerical simulations and theoretical justifications demonstrate that TEAM has asymptotically valid, powerful, and robust performance. Overall, TEAM is a computationally efficient and statistically powerful algorithm that can yield meaningful biological insights in flow cytometry studies.
Collapse
Affiliation(s)
- John A Pura
- Center of Innovation to Accelerate Discovery and Practice Transformation, Durham Veterans AfFaIRS MediCal Center, Durham, NC 27701
| | - Xuechan Li
- Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, NC 27705
| | - Cliburn Chan
- Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, NC 27705
| | - Jichun Xie
- Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, NC 27705
| |
Collapse
|
10
|
Ferrer-Font L, Kraker G, Hally KE, Price KM. Ensuring Full Spectrum Flow Cytometry Data Quality for High-Dimensional Data Analysis. Curr Protoc 2023; 3:e657. [PMID: 36744957 DOI: 10.1002/cpz1.657] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Full spectrum flow cytometry (FSFC) allows for the analysis of more than 40 parameters at the single-cell level. Compared to the practice of manual gating, high-dimensional data analysis can be used to fully explore single-cell datasets and reduce analysis time. As panel size and complexity increases so too does the detail and time required to prepare and validate the quality of the resulting data for use in downstream high-dimensional data analyses. To ensure data analysis algorithms can be used efficiently and to avoid artifacts, some important steps should be considered. These include data cleaning (such as eliminating variable signal change over time, removing cell doublets, and antibody aggregates), proper unmixing of full spectrum data, ensuring correct scale transformation, and correcting for batch effects. We have developed a methodical step-by-step protocol to prepare full spectrum high-dimensional data for use with high-dimensional data analyses, with a focus on visualizing the impact of each step of data preparation using dimensionality reduction algorithms. Application of our workflow will aid FSFC users in their efforts to apply quality control methods to their datasets for use in high-dimensional analysis, and help them to obtain valid and reproducible results. © 2023 Wiley Periodicals LLC. Basic Protocol 1: Data cleaning Basic Protocol 2: Validating the quality of unmixing Basic Protocol 3: Data scaling Basic Protocol 4: Batch-to-batch normalization.
Collapse
Affiliation(s)
- Laura Ferrer-Font
- Hugh Green Cytometry Centre, Malaghan Institute of Medical Research, Wellington, New Zealand
| | | | - Kathryn E Hally
- Department of Surgery and Anaesthesia, The University of Otago, Wellington, New Zealand
| | - Kylie M Price
- Hugh Green Cytometry Centre, Malaghan Institute of Medical Research, Wellington, New Zealand
| |
Collapse
|
11
|
Spurgeon BEJ, Frelinger AL. Platelet Phenotyping by Full Spectrum Flow Cytometry. Curr Protoc 2023; 3:e687. [PMID: 36779850 DOI: 10.1002/cpz1.687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/14/2023]
Abstract
Platelets play key roles in hemostasis, immunity, and inflammation, and tests of platelet phenotype and function are useful in studies of disease biology and pathology. Full spectrum flow cytometry offers distinct advantages over standard tests and enables the sensitive and simultaneous detection of many biomarkers. A typical assay provides a wealth of information on platelet biology and allows the assessment of in vivo activation and in vitro reactivity, as well as the discovery of novel phenotypes. Here, we describe the analysis of platelets by full spectrum flow cytometry and discuss a range of controls and methods for interpreting results. © 2023 Wiley Periodicals LLC. Basic Protocol: Platelet phenotyping by full spectrum flow cytometry Support Protocol 1: Spectral unmixing Support Protocol 2: Data preprocessing.
Collapse
Affiliation(s)
- Benjamin E J Spurgeon
- Center for Platelet Research Studies, Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Harvard Medical School, Boston, Massachusetts
| | - Andrew L Frelinger
- Center for Platelet Research Studies, Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
12
|
Porwit A, Béné MC, Duetz C, Matarraz S, Oelschlaegel U, Westers TM, Wagner-Ballon O, Kordasti S, Valent P, Preijers F, Alhan C, Bellos F, Bettelheim P, Burbury K, Chapuis N, Cremers E, Della Porta MG, Dunlop A, Eidenschink-Brodersen L, Font P, Fontenay M, Hobo W, Ireland R, Johansson U, Loken MR, Ogata K, Orfao A, Psarra K, Saft L, Subira D, Te Marvelde J, Wells DA, van der Velden VHJ, Kern W, van de Loosdrecht AA. Multiparameter flow cytometry in the evaluation of myelodysplasia: Analytical issues: Recommendations from the European LeukemiaNet/International Myelodysplastic Syndrome Flow Cytometry Working Group. CYTOMETRY. PART B, CLINICAL CYTOMETRY 2023; 104:27-50. [PMID: 36537621 PMCID: PMC10107708 DOI: 10.1002/cyto.b.22108] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 11/20/2022] [Accepted: 11/29/2022] [Indexed: 01/18/2023]
Abstract
Multiparameter flow cytometry (MFC) is one of the essential ancillary methods in bone marrow (BM) investigation of patients with cytopenia and suspected myelodysplastic syndrome (MDS). MFC can also be applied in the follow-up of MDS patients undergoing treatment. This document summarizes recommendations from the International/European Leukemia Net Working Group for Flow Cytometry in Myelodysplastic Syndromes (ELN iMDS Flow) on the analytical issues in MFC for the diagnostic work-up of MDS. Recommendations for the analysis of several BM cell subsets such as myeloid precursors, maturing granulocytic and monocytic components and erythropoiesis are given. A core set of 17 markers identified as independently related to a cytomorphologic diagnosis of myelodysplasia is suggested as mandatory for MFC evaluation of BM in a patient with cytopenia. A myeloid precursor cell (CD34+ CD19- ) count >3% should be considered immunophenotypically indicative of myelodysplasia. However, MFC results should always be evaluated as part of an integrated hematopathology work-up. Looking forward, several machine-learning-based analytical tools of interest should be applied in parallel to conventional analytical methods to investigate their usefulness in integrated diagnostics, risk stratification, and potentially even in the evaluation of response to therapy, based on MFC data. In addition, compiling large uniform datasets is desirable, as most of the machine-learning-based methods tend to perform better with larger numbers of investigated samples, especially in such a heterogeneous disease as MDS.
Collapse
Affiliation(s)
- Anna Porwit
- Division of Oncology and Pathology, Department of Clinical Sciences, Faculty of Medicine, Lund University, Lund, Sweden
| | - Marie C Béné
- Hematology Biology, Nantes University Hospital, CRCINA Inserm 1232, Nantes, France
| | - Carolien Duetz
- Department of Hematology, Amsterdam UMC, VU University Medical Center Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Sergio Matarraz
- Cancer Research Center (IBMCC-USAL/CSIC), Department of Medicine and Cytometry Service, Institute for Biomedical Research of Salamanca (IBSAL) and CIBERONC, University of Salamanca, Salamanca, Spain
| | - Uta Oelschlaegel
- Department of Internal Medicine, University Hospital Carl-Gustav-Carus, TU Dresden, Dresden, Germany
| | - Theresia M Westers
- Department of Hematology, Amsterdam UMC, VU University Medical Center Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Orianne Wagner-Ballon
- Department of Hematology and Immunology, Assistance Publique-Hôpitaux de Paris, University Hospital Henri Mondor, Créteil, France
- Inserm U955, Université Paris-Est Créteil, Créteil, France
| | | | - Peter Valent
- Department of Internal Medicine I, Division of Hematology & Hemostaseology and Ludwig Boltzmann Institute for Hematology and Oncology, Medical University of Vienna, Vienna, Austria
| | - Frank Preijers
- Laboratory of Hematology, Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Canan Alhan
- Department of Hematology, Amsterdam UMC, VU University Medical Center Cancer Center Amsterdam, Amsterdam, The Netherlands
| | | | - Peter Bettelheim
- Department of Hematology, Ordensklinikum Linz, Elisabethinen, Linz, Austria
| | - Kate Burbury
- Department of Haematology, Peter MacCallum Cancer Centre, & University of Melbourne, Melbourne, Australia
| | - Nicolas Chapuis
- Laboratory of Hematology, Assistance Publique-Hôpitaux de Paris, Centre-Université de Paris, Cochin Hospital, Paris, France
- Institut Cochin, INSERM U1016, CNRS UMR, Université de Paris, Paris, France
| | - Eline Cremers
- Division of Hematology, Department of Internal Medicine, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Matteo G Della Porta
- IRCCS Humanitas Research Hospital, Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | - Alan Dunlop
- Department of Haemato-Oncology, Royal Marsden Hospital, London, UK
| | | | - Patricia Font
- Department of Hematology, Hospital General Universitario Gregorio Marañon-IiSGM, Madrid, Spain
| | - Michaela Fontenay
- Laboratory of Hematology, Assistance Publique-Hôpitaux de Paris, Centre-Université de Paris, Cochin Hospital, Paris, France
- Institut Cochin, INSERM U1016, CNRS UMR, Université de Paris, Paris, France
| | - Willemijn Hobo
- Department of Internal Medicine I, Division of Hematology & Hemostaseology and Ludwig Boltzmann Institute for Hematology and Oncology, Medical University of Vienna, Vienna, Austria
| | - Robin Ireland
- Department of Haematology and SE-HMDS, King's College Hospital NHS Foundation Trust, London, UK
| | - Ulrika Johansson
- Laboratory Medicine, SI-HMDS, University Hospitals Bristol and Weston NHS Foundation Trust, Bristol, UK
| | | | - Kiyoyuki Ogata
- Metropolitan Research and Treatment Centre for Blood Disorders (MRTC Japan), Tokyo, Japan
| | - Alberto Orfao
- Cancer Research Center (IBMCC-USAL/CSIC), Department of Medicine and Cytometry Service, Institute for Biomedical Research of Salamanca (IBSAL) and CIBERONC, University of Salamanca, Salamanca, Spain
| | - Katherina Psarra
- Department of Immunology - Histocompatibility, Evangelismos Hospital, Athens, Greece
| | - Leonie Saft
- Clinical Pathology and Cancer Diagnostics, Karolinska University Hospital and Institute Solna, Stockholm, Sweden
| | - Dolores Subira
- Department of Hematology, Flow Cytometry Unit, Hospital Universitario de Guadalajara, Guadalajara, Spain
| | - Jeroen Te Marvelde
- Laboratory Medical Immunology, Department of Immunology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | | | - Vincent H J van der Velden
- Laboratory Medical Immunology, Department of Immunology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | | | - Arjan A van de Loosdrecht
- Department of Hematology, Amsterdam UMC, VU University Medical Center Cancer Center Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
13
|
Melsen JE, van Ostaijen-ten Dam MM, Schoorl DJA, Schol PJ, van den Homberg DAL, Lankester AC, Lugthart G, Schilham MW. Single-cell transcriptomics in bone marrow delineates CD56 dimGranzymeK + subset as intermediate stage in NK cell differentiation. Front Immunol 2022; 13:1044398. [PMID: 36505452 PMCID: PMC9730327 DOI: 10.3389/fimmu.2022.1044398] [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: 09/14/2022] [Accepted: 11/03/2022] [Indexed: 11/25/2022] Open
Abstract
Human natural killer (NK) cells in lymphoid tissues can be categorized into three subsets: CD56brightCD16+, CD56dimCD16+ and CD69+CXCR6+ lymphoid tissue-resident (lt)NK cells. How the three subsets are functionally and developmentally related is currently unknown. Therefore, we performed single-cell RNA sequencing combined with oligonucleotide-conjugated antibodies against CD56, CXCR6, CD117 and CD34 on fresh bone marrow NK cells. A minor CD56dimGzmK+ subset was identified that shared features with CD56bright and CD56dimGzmK- NK cells based on transcriptome, phenotype (NKG2AhighCD16lowKLRG1highTIGIThigh) and functional analysis in bone marrow and blood, supportive for an intermediate subset. Pseudotime analysis positioned CD56bright, CD56dimGzmK+ and CD56dimGzmK- cells in one differentiation trajectory, while ltNK cells were developmentally separated. Integrative analysis with bone marrow cells from the Human Cell Atlas did not demonstrate a developmental connection between CD34+ progenitor and NK cells, suggesting absence of early NK cell stages in bone marrow. In conclusion, single-cell transcriptomics provide new insights on development and differentiation of human NK cells.
Collapse
|
14
|
Garg T, Weiss CR, Sheth RA. Techniques for Profiling the Cellular Immune Response and Their Implications for Interventional Oncology. Cancers (Basel) 2022; 14:3628. [PMID: 35892890 PMCID: PMC9332307 DOI: 10.3390/cancers14153628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 07/19/2022] [Accepted: 07/20/2022] [Indexed: 12/07/2022] Open
Abstract
In recent years there has been increased interest in using the immune contexture of the primary tumors to predict the patient's prognosis. The tumor microenvironment of patients with cancers consists of different types of lymphocytes, tumor-infiltrating leukocytes, dendritic cells, and others. Different technologies can be used for the evaluation of the tumor microenvironment, all of which require a tissue or cell sample. Image-guided tissue sampling is a cornerstone in the diagnosis, stratification, and longitudinal evaluation of therapeutic efficacy for cancer patients receiving immunotherapies. Therefore, interventional radiologists (IRs) play an essential role in the evaluation of patients treated with systemically administered immunotherapies. This review provides a detailed description of different technologies used for immune assessment and analysis of the data collected from the use of these technologies. The detailed approach provided herein is intended to provide the reader with the knowledge necessary to not only interpret studies containing such data but also design and apply these tools for clinical practice and future research studies.
Collapse
Affiliation(s)
- Tushar Garg
- Division of Vascular and Interventional Radiology, Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; (T.G.); (C.R.W.)
| | - Clifford R. Weiss
- Division of Vascular and Interventional Radiology, Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; (T.G.); (C.R.W.)
| | - Rahul A. Sheth
- Department of Interventional Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
15
|
Pedersen CB, Dam SH, Barnkob MB, Leipold MD, Purroy N, Rassenti LZ, Kipps TJ, Nguyen J, Lederer JA, Gohil SH, Wu CJ, Olsen LR. cyCombine allows for robust integration of single-cell cytometry datasets within and across technologies. Nat Commun 2022; 13:1698. [PMID: 35361793 PMCID: PMC8971492 DOI: 10.1038/s41467-022-29383-5] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 03/14/2022] [Indexed: 12/21/2022] Open
Abstract
Combining single-cell cytometry datasets increases the analytical flexibility and the statistical power of data analyses. However, in many cases the full potential of co-analyses is not reached due to technical variance between data from different experimental batches. Here, we present cyCombine, a method to robustly integrate cytometry data from different batches, experiments, or even different experimental techniques, such as CITE-seq, flow cytometry, and mass cytometry. We demonstrate that cyCombine maintains the biological variance and the structure of the data, while minimizing the technical variance between datasets. cyCombine does not require technical replicates across datasets, and computation time scales linearly with the number of cells, allowing for integration of massive datasets. Robust, accurate, and scalable integration of cytometry data enables integration of multiple datasets for primary data analyses and the validation of results using public datasets.
Collapse
Affiliation(s)
- Christina Bligaard Pedersen
- Department of Health Technology, Technical University of Denmark, Kongens Lyngby, Denmark
- Center for Genomic Medicine, Rigshospitalet-Copenhagen University Hospital, Copenhagen, Denmark
| | - Søren Helweg Dam
- Department of Health Technology, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Mike Bogetofte Barnkob
- Centre for Cellular Immunotherapy of Haematological Cancer Odense (CITCO), Department of Clinical Immunology, Odense University Hospital, University of Southern Denmark, Odense, Denmark
| | - Michael D Leipold
- Human Immune Monitoring Center, Institute for Immunity, Transplantation, and Infection, Stanford University School of Medicine, Stanford, CA, USA
| | - Noelia Purroy
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- AstraZeneca, Waltham, MA, USA
| | - Laura Z Rassenti
- Division of Hematology-Oncology, Department of Medicine, Moores Cancer Center, University of California, San Diego, La Jolla, CA, USA
| | - Thomas J Kipps
- Division of Hematology-Oncology, Department of Medicine, Moores Cancer Center, University of California, San Diego, La Jolla, CA, USA
| | - Jennifer Nguyen
- Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - James Arthur Lederer
- Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Satyen Harish Gohil
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Academic Haematology, University College London, London, UK
- Department of Haematology, University College London Hospitals NHS Trust, London, UK
| | - Catherine J Wu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Lars Rønn Olsen
- Department of Health Technology, Technical University of Denmark, Kongens Lyngby, Denmark.
| |
Collapse
|
16
|
Dierolf JG, Chadwick K, Brooks CR, Watson AJ, Betts DH. Flow Cytometric Characterization of Pluripotent Cell Protein Markers in Naïve, Formative, and Primed Pluripotent Stem Cells. Methods Mol Biol 2022; 2490:81-92. [PMID: 35486241 DOI: 10.1007/978-1-0716-2281-0_8] [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] [Indexed: 06/14/2023]
Abstract
Here we describe methodologies to characterize, delineate, and quantify pluripotent cells between naïve, formative, and primed pluripotent state mouse embryonic stem cell (mESCs) populations using flow cytometric analysis. This methodology can validate pluripotent states, sort individual cells of interest, and determine the efficiency of transitioning naïve mESCs to a primed-like state as mouse epiblast-like cells (mEpiLCs) and onto fully primed mouse epiblast stem cells (mEpiSCs). Quantification of the cell surface markers; SSEA1(CD15) and CD24 introduces an effective method of distinguishing individual cells from a population by their respective positioning in the pluripotent spectrum. Additionally, this protocol can be used to demarcate and sort cells via fluorescently activated cell sorting for downstream applications. Flow cytometric analysis within mESCs, mEpiLCs, and mEpiSCs can be efficiently completed using these optimized protocols.
Collapse
Affiliation(s)
- Joshua G Dierolf
- Department of Physiology and Pharmacology, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, ON, Canada
| | - Kristin Chadwick
- Robarts Research Institute, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, ON, Canada
| | - Courtney R Brooks
- Department of Physiology and Pharmacology, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, ON, Canada
| | - Andrew J Watson
- Department of Physiology and Pharmacology, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, ON, Canada
- Department of Obstetrics and Gynecology, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, ON, Canada
- The Children's Health Research Institute (CHRI), Lawson Health Research Institute, London, ON, Canada
| | - Dean H Betts
- Department of Physiology and Pharmacology, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, ON, Canada.
- Department of Obstetrics and Gynecology, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, ON, Canada.
- The Children's Health Research Institute (CHRI), Lawson Health Research Institute, London, ON, Canada.
| |
Collapse
|
17
|
Burton RJ, Ahmed R, Cuff SM, Baker S, Artemiou A, Eberl M. CytoPy: An autonomous cytometry analysis framework. PLoS Comput Biol 2021; 17:e1009071. [PMID: 34101722 PMCID: PMC8213167 DOI: 10.1371/journal.pcbi.1009071] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Revised: 06/18/2021] [Accepted: 05/12/2021] [Indexed: 12/24/2022] Open
Abstract
Cytometry analysis has seen a considerable expansion in recent years in the maximum number of parameters that can be acquired in a single experiment. In response to this technological advance there has been an increased effort to develop new computational methodologies for handling high-dimensional single cell data acquired by flow or mass cytometry. Despite the success of numerous algorithms and published packages to replicate and outperform traditional manual analysis, widespread adoption of these techniques has yet to be realised in the field of immunology. Here we present CytoPy, a Python framework for automated analysis of cytometry data that integrates a document-based database for a data-centric and iterative analytical environment. In addition, our algorithm-agnostic design provides a platform for open-source cytometry bioinformatics in the Python ecosystem. We demonstrate the ability of CytoPy to phenotype T cell subsets in whole blood samples even in the presence of significant batch effects due to technical and user variation. The complete analytical pipeline was then used to immunophenotype the local inflammatory infiltrate in individuals with and without acute bacterial infection. CytoPy is open-source and licensed under the MIT license. CytoPy is available at https://github.com/burtonrj/CytoPy, with notebooks accompanying this manuscript (https://github.com/burtonrj/CytoPyManuscript) and software documentation at https://cytopy.readthedocs.io/.
Collapse
Affiliation(s)
- Ross J. Burton
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Raya Ahmed
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Simone M. Cuff
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Sarah Baker
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Andreas Artemiou
- School of Mathematics, Cardiff University, Cardiff, United Kingdom
| | - Matthias Eberl
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, United Kingdom
- Systems Immunity Research Institute, Cardiff University, Cardiff, United Kingdom
| |
Collapse
|
18
|
Johansson D, Rauld C, Roux J, Regairaz C, Galli E, Callegari I, Raad L, Waldt A, Cuttat R, Roma G, Diebold M, Becher B, Kuhle J, Derfuss T, Carballido JM, Sanderson NSR. Mass Cytometry of CSF Identifies an MS-Associated B-cell Population. NEUROLOGY-NEUROIMMUNOLOGY & NEUROINFLAMMATION 2021; 8:8/2/e943. [PMID: 33589541 PMCID: PMC8057060 DOI: 10.1212/nxi.0000000000000943] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Accepted: 10/28/2020] [Indexed: 11/15/2022]
Abstract
Objective To identify an MS-specific immune cell population by deep immune phenotyping and relate it to soluble signaling molecules in CSF. Methods We analyzed surface expression of 22 markers in paired blood/CSF samples from 39 patients using mass cytometry (cytometry by time of flight). We also measured the concentrations of 296 signaling molecules in CSF using proximity extension assay. Results were analyzed using highly automated unsupervised algorithmic informatics. Results Mass cytometry objectively identified a B-cell population characterized by the expression of CD49d, CD69, CD27, CXCR3, and human leukocyte antigen (HLA)-DR as clearly associated with MS. Concentrations of the B cell–related factors, notably FCRL2, were increased in MS CSF, especially in early stages of the disease. The B-cell trophic factor B cell activating factor (BAFF) was decreased in MS. Proteins involved in neural plasticity were also reduced in MS. Conclusion When analyzed without a priori assumptions, both the soluble and the cellular compartments of the CSF in MS were characterized by markers related to B cells, and the strongest candidate for an MS-specific cell type has a B-cell phenotype.
Collapse
Affiliation(s)
- David Johansson
- From the Department of Biomedicine (D.J., J.R., E.G., I.C., M.D., J.K., T.D., N.S.R.S.), University Hospital Basel, University of Basel; Novartis Institutes for BioMedical Research (C. Rauld, C. Regairaz, L.R., A.W., R.C., G.R., J.M.C.); Swiss Institute of Bioinformatics (J.R.), Basel; Institute of Experimental Immunology (E.G., B.B.), University of Zurich; and Department of Medicine (E.G., M.D., J.K., T.D.), Neurologic Clinic and Policlinic, University Hospital and University of Basel, Switzerland
| | - Céline Rauld
- From the Department of Biomedicine (D.J., J.R., E.G., I.C., M.D., J.K., T.D., N.S.R.S.), University Hospital Basel, University of Basel; Novartis Institutes for BioMedical Research (C. Rauld, C. Regairaz, L.R., A.W., R.C., G.R., J.M.C.); Swiss Institute of Bioinformatics (J.R.), Basel; Institute of Experimental Immunology (E.G., B.B.), University of Zurich; and Department of Medicine (E.G., M.D., J.K., T.D.), Neurologic Clinic and Policlinic, University Hospital and University of Basel, Switzerland
| | - Julien Roux
- From the Department of Biomedicine (D.J., J.R., E.G., I.C., M.D., J.K., T.D., N.S.R.S.), University Hospital Basel, University of Basel; Novartis Institutes for BioMedical Research (C. Rauld, C. Regairaz, L.R., A.W., R.C., G.R., J.M.C.); Swiss Institute of Bioinformatics (J.R.), Basel; Institute of Experimental Immunology (E.G., B.B.), University of Zurich; and Department of Medicine (E.G., M.D., J.K., T.D.), Neurologic Clinic and Policlinic, University Hospital and University of Basel, Switzerland
| | - Camille Regairaz
- From the Department of Biomedicine (D.J., J.R., E.G., I.C., M.D., J.K., T.D., N.S.R.S.), University Hospital Basel, University of Basel; Novartis Institutes for BioMedical Research (C. Rauld, C. Regairaz, L.R., A.W., R.C., G.R., J.M.C.); Swiss Institute of Bioinformatics (J.R.), Basel; Institute of Experimental Immunology (E.G., B.B.), University of Zurich; and Department of Medicine (E.G., M.D., J.K., T.D.), Neurologic Clinic and Policlinic, University Hospital and University of Basel, Switzerland
| | - Edoardo Galli
- From the Department of Biomedicine (D.J., J.R., E.G., I.C., M.D., J.K., T.D., N.S.R.S.), University Hospital Basel, University of Basel; Novartis Institutes for BioMedical Research (C. Rauld, C. Regairaz, L.R., A.W., R.C., G.R., J.M.C.); Swiss Institute of Bioinformatics (J.R.), Basel; Institute of Experimental Immunology (E.G., B.B.), University of Zurich; and Department of Medicine (E.G., M.D., J.K., T.D.), Neurologic Clinic and Policlinic, University Hospital and University of Basel, Switzerland
| | - Ilaria Callegari
- From the Department of Biomedicine (D.J., J.R., E.G., I.C., M.D., J.K., T.D., N.S.R.S.), University Hospital Basel, University of Basel; Novartis Institutes for BioMedical Research (C. Rauld, C. Regairaz, L.R., A.W., R.C., G.R., J.M.C.); Swiss Institute of Bioinformatics (J.R.), Basel; Institute of Experimental Immunology (E.G., B.B.), University of Zurich; and Department of Medicine (E.G., M.D., J.K., T.D.), Neurologic Clinic and Policlinic, University Hospital and University of Basel, Switzerland
| | - Layla Raad
- From the Department of Biomedicine (D.J., J.R., E.G., I.C., M.D., J.K., T.D., N.S.R.S.), University Hospital Basel, University of Basel; Novartis Institutes for BioMedical Research (C. Rauld, C. Regairaz, L.R., A.W., R.C., G.R., J.M.C.); Swiss Institute of Bioinformatics (J.R.), Basel; Institute of Experimental Immunology (E.G., B.B.), University of Zurich; and Department of Medicine (E.G., M.D., J.K., T.D.), Neurologic Clinic and Policlinic, University Hospital and University of Basel, Switzerland
| | - Annick Waldt
- From the Department of Biomedicine (D.J., J.R., E.G., I.C., M.D., J.K., T.D., N.S.R.S.), University Hospital Basel, University of Basel; Novartis Institutes for BioMedical Research (C. Rauld, C. Regairaz, L.R., A.W., R.C., G.R., J.M.C.); Swiss Institute of Bioinformatics (J.R.), Basel; Institute of Experimental Immunology (E.G., B.B.), University of Zurich; and Department of Medicine (E.G., M.D., J.K., T.D.), Neurologic Clinic and Policlinic, University Hospital and University of Basel, Switzerland
| | - Rachel Cuttat
- From the Department of Biomedicine (D.J., J.R., E.G., I.C., M.D., J.K., T.D., N.S.R.S.), University Hospital Basel, University of Basel; Novartis Institutes for BioMedical Research (C. Rauld, C. Regairaz, L.R., A.W., R.C., G.R., J.M.C.); Swiss Institute of Bioinformatics (J.R.), Basel; Institute of Experimental Immunology (E.G., B.B.), University of Zurich; and Department of Medicine (E.G., M.D., J.K., T.D.), Neurologic Clinic and Policlinic, University Hospital and University of Basel, Switzerland
| | - Guglielmo Roma
- From the Department of Biomedicine (D.J., J.R., E.G., I.C., M.D., J.K., T.D., N.S.R.S.), University Hospital Basel, University of Basel; Novartis Institutes for BioMedical Research (C. Rauld, C. Regairaz, L.R., A.W., R.C., G.R., J.M.C.); Swiss Institute of Bioinformatics (J.R.), Basel; Institute of Experimental Immunology (E.G., B.B.), University of Zurich; and Department of Medicine (E.G., M.D., J.K., T.D.), Neurologic Clinic and Policlinic, University Hospital and University of Basel, Switzerland
| | - Martin Diebold
- From the Department of Biomedicine (D.J., J.R., E.G., I.C., M.D., J.K., T.D., N.S.R.S.), University Hospital Basel, University of Basel; Novartis Institutes for BioMedical Research (C. Rauld, C. Regairaz, L.R., A.W., R.C., G.R., J.M.C.); Swiss Institute of Bioinformatics (J.R.), Basel; Institute of Experimental Immunology (E.G., B.B.), University of Zurich; and Department of Medicine (E.G., M.D., J.K., T.D.), Neurologic Clinic and Policlinic, University Hospital and University of Basel, Switzerland
| | - Burkhard Becher
- From the Department of Biomedicine (D.J., J.R., E.G., I.C., M.D., J.K., T.D., N.S.R.S.), University Hospital Basel, University of Basel; Novartis Institutes for BioMedical Research (C. Rauld, C. Regairaz, L.R., A.W., R.C., G.R., J.M.C.); Swiss Institute of Bioinformatics (J.R.), Basel; Institute of Experimental Immunology (E.G., B.B.), University of Zurich; and Department of Medicine (E.G., M.D., J.K., T.D.), Neurologic Clinic and Policlinic, University Hospital and University of Basel, Switzerland
| | - Jens Kuhle
- From the Department of Biomedicine (D.J., J.R., E.G., I.C., M.D., J.K., T.D., N.S.R.S.), University Hospital Basel, University of Basel; Novartis Institutes for BioMedical Research (C. Rauld, C. Regairaz, L.R., A.W., R.C., G.R., J.M.C.); Swiss Institute of Bioinformatics (J.R.), Basel; Institute of Experimental Immunology (E.G., B.B.), University of Zurich; and Department of Medicine (E.G., M.D., J.K., T.D.), Neurologic Clinic and Policlinic, University Hospital and University of Basel, Switzerland
| | - Tobias Derfuss
- From the Department of Biomedicine (D.J., J.R., E.G., I.C., M.D., J.K., T.D., N.S.R.S.), University Hospital Basel, University of Basel; Novartis Institutes for BioMedical Research (C. Rauld, C. Regairaz, L.R., A.W., R.C., G.R., J.M.C.); Swiss Institute of Bioinformatics (J.R.), Basel; Institute of Experimental Immunology (E.G., B.B.), University of Zurich; and Department of Medicine (E.G., M.D., J.K., T.D.), Neurologic Clinic and Policlinic, University Hospital and University of Basel, Switzerland
| | - José M Carballido
- From the Department of Biomedicine (D.J., J.R., E.G., I.C., M.D., J.K., T.D., N.S.R.S.), University Hospital Basel, University of Basel; Novartis Institutes for BioMedical Research (C. Rauld, C. Regairaz, L.R., A.W., R.C., G.R., J.M.C.); Swiss Institute of Bioinformatics (J.R.), Basel; Institute of Experimental Immunology (E.G., B.B.), University of Zurich; and Department of Medicine (E.G., M.D., J.K., T.D.), Neurologic Clinic and Policlinic, University Hospital and University of Basel, Switzerland
| | - Nicholas S R Sanderson
- From the Department of Biomedicine (D.J., J.R., E.G., I.C., M.D., J.K., T.D., N.S.R.S.), University Hospital Basel, University of Basel; Novartis Institutes for BioMedical Research (C. Rauld, C. Regairaz, L.R., A.W., R.C., G.R., J.M.C.); Swiss Institute of Bioinformatics (J.R.), Basel; Institute of Experimental Immunology (E.G., B.B.), University of Zurich; and Department of Medicine (E.G., M.D., J.K., T.D.), Neurologic Clinic and Policlinic, University Hospital and University of Basel, Switzerland.
| |
Collapse
|
19
|
Abstract
Flow cytometry is an important technology for the study of microbial communities. It grants the ability to rapidly generate phenotypic single-cell data that are both quantitative, multivariate and of high temporal resolution. The complexity and amount of data necessitate an objective and streamlined data processing workflow that extends beyond commercial instrument software. No full overview of the necessary steps regarding the computational analysis of microbial flow cytometry data currently exists. In this review, we provide an overview of the full data analysis pipeline, ranging from measurement to data interpretation, tailored toward studies in microbial ecology. At every step, we highlight computational methods that are potentially useful, for which we provide a short nontechnical description. We place this overview in the context of a number of open challenges to the field and offer further motivation for the use of standardized flow cytometry in microbial ecology research.
Collapse
Affiliation(s)
| | - Ruben Props
- Center for Microbial Ecology & Technology (CMET), Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium
| |
Collapse
|
20
|
Abolhassani H. Specific Immune Response and Cytokine Production in CD70 Deficiency. Front Pediatr 2021; 9:615724. [PMID: 33996677 PMCID: PMC8120026 DOI: 10.3389/fped.2021.615724] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 03/10/2021] [Indexed: 11/13/2022] Open
Abstract
Collective clinical and immunologic findings of defects in the CD27-CD70 axis indicate a primary immunodeficiency associated with terminal B-cell development defect and immune dysregulation leading to autoimmunity, uncontrolled viral infection, and lymphoma. Since the molecular mechanism underlying this entity of primary immunodeficiency has been recently described, more insight regarding the function and profile of immunity is required. Therefore, this study aimed to investigate stimulated antibody production, polyclonal vs. virus-specific T-cell response, and cytokine production of a CD70-deficient patient reported previously with early-onset antibody deficiency suffering from chronic viral infections and B-cell lymphoma. The patient and her family members were subjected to clinical evaluation, immunological assays, and functional analyses. The findings of this study indicate an impaired ability of B cells to produce immunoglobulins, and a poor effector function of T cells was also associated with the severity of clinical phenotype. Reduced proportions of cells expressing the memory marker CD45RO, as well as T-bet and Eomes, were observed in CD70-deficient T cells. The proportion of 2B4+ and PD-1+ virus-specific CD8+ T cells was also reduced in the patient. Although the CD70-mutated individuals presented with early-onset clinical manifestations that were well-controlled by using conventional immunological and anticancer chemotherapies, with better prognosis as compared with CD27-deficient patients, targeted treatment toward specific disturbed immune profile may improve the management and even prevent secondary complications.
Collapse
Affiliation(s)
- Hassan Abolhassani
- Division of Clinical Immunology, Department of Laboratory Medicine, Karolinska Institutet at Karolinska University Hospital Huddinge, Stockholm, Sweden.,Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
21
|
Keyes TJ, Domizi P, Lo YC, Nolan GP, Davis KL. A Cancer Biologist's Primer on Machine Learning Applications in High-Dimensional Cytometry. Cytometry A 2020; 97:782-799. [PMID: 32602650 PMCID: PMC7416435 DOI: 10.1002/cyto.a.24158] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2019] [Revised: 03/10/2020] [Accepted: 05/12/2020] [Indexed: 12/11/2022]
Abstract
The application of machine learning and artificial intelligence to high-dimensional cytometry data sets has increasingly become a staple of bioinformatic data analysis over the past decade. This is especially true in the field of cancer biology, where protocols for collecting multiparameter single-cell data in a high-throughput fashion are rapidly developed. As the use of machine learning methodology in cytometry becomes increasingly common, there is a need for cancer biologists to understand the basic theory and applications of a variety of algorithmic tools for analyzing and interpreting cytometry data. We introduce the reader to several keystone machine learning-based analytic approaches with an emphasis on defining key terms and introducing a conceptual framework for making translational or clinically relevant discoveries. The target audience consists of cancer cell biologists and physician-scientists interested in applying these tools to their own data, but who may have limited training in bioinformatics. © 2020 International Society for Advancement of Cytometry.
Collapse
Affiliation(s)
- Timothy J Keyes
- Medical Scientist Training Program, Stanford University School of Medicine, Stanford, California
- Department of Pediatrics, Stanford University School of Medicine, Stanford, California
| | - Pablo Domizi
- Department of Pediatrics, Stanford University School of Medicine, Stanford, California
| | - Yu-Chen Lo
- Department of Pediatrics, Stanford University School of Medicine, Stanford, California
| | - Garry P Nolan
- Department of Microbiology and Immunology | Baxter Laboratory for Stem Cell Biology, Stanford University School of Medicine, Stanford, California
| | - Kara L Davis
- Department of Pediatrics, Stanford University School of Medicine, Stanford, California
| |
Collapse
|
22
|
Rybakowska P, Alarcón-Riquelme ME, Marañón C. Key steps and methods in the experimental design and data analysis of highly multi-parametric flow and mass cytometry. Comput Struct Biotechnol J 2020; 18:874-886. [PMID: 32322369 PMCID: PMC7163213 DOI: 10.1016/j.csbj.2020.03.024] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Revised: 03/18/2020] [Accepted: 03/25/2020] [Indexed: 01/05/2023] Open
Abstract
High-dimensional, single-cell cell technologies revolutionized the way to study biological systems, and polychromatic flow cytometry (FC) and mass cytometry (MC) are two of the drivers of this revolution. As up to 30-50 dimensions respectively can be measured per single-cell, they allow deep phenotyping combined with cellular functions studies, like cytokine production or protein phosphorylation. In parallel, the bioinformatics field develops algorithms that are able to process incoming data and extract the most useful and meaningful biological information. However, the success of automated analysis tools depends on the generation of high-quality data. In this review we present the most recent FC and MC computational approaches that are used to prepare, process and interpret high-content cytometry data. We also underscore proper experimental design as a key step for obtaining good quality data.
Collapse
Affiliation(s)
- Paulina Rybakowska
- GENYO, Centre for Genomics and Oncological Research Pfizer/University of Granada/Andalusian Regional Government, PTS Granada, Spain
| | - Marta E. Alarcón-Riquelme
- GENYO, Centre for Genomics and Oncological Research Pfizer/University of Granada/Andalusian Regional Government, PTS Granada, Spain
- Institute for Environmental Medicine, Karolinska Institute, Stockholm, Sweden
| | - Concepción Marañón
- GENYO, Centre for Genomics and Oncological Research Pfizer/University of Granada/Andalusian Regional Government, PTS Granada, Spain
| |
Collapse
|
23
|
Lucchesi S, Furini S, Medaglini D, Ciabattini A. From Bivariate to Multivariate Analysis of Cytometric Data: Overview of Computational Methods and Their Application in Vaccination Studies. Vaccines (Basel) 2020; 8:E138. [PMID: 32244919 PMCID: PMC7157606 DOI: 10.3390/vaccines8010138] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 03/17/2020] [Accepted: 03/18/2020] [Indexed: 12/15/2022] Open
Abstract
Flow and mass cytometry are used to quantify the expression of multiple extracellular or intracellular molecules on single cells, allowing the phenotypic and functional characterization of complex cell populations. Multiparametric flow cytometry is particularly suitable for deep analysis of immune responses after vaccination, as it allows to measure the frequency, the phenotype, and the functional features of antigen-specific cells. When many parameters are investigated simultaneously, it is not feasible to analyze all the possible bi-dimensional combinations of marker expression with classical manual analysis and the adoption of advanced automated tools to process and analyze high-dimensional data sets becomes necessary. In recent years, the development of many tools for the automated analysis of multiparametric cytometry data has been reported, with an increasing record of publications starting from 2014. However, the use of these tools has been preferentially restricted to bioinformaticians, while few of them are routinely employed by the biomedical community. Filling the gap between algorithms developers and final users is fundamental for exploiting the advantages of computational tools in the analysis of cytometry data. The potentialities of automated analyses range from the improvement of the data quality in the pre-processing steps up to the unbiased, data-driven examination of complex datasets using a variety of algorithms based on different approaches. In this review, an overview of the automated analysis pipeline is provided, spanning from the pre-processing phase to the automated population analysis. Analysis based on computational tools might overcame both the subjectivity of manual gating and the operator-biased exploration of expected populations. Examples of applications of automated tools that have successfully improved the characterization of different cell populations in vaccination studies are also presented.
Collapse
Affiliation(s)
- Simone Lucchesi
- Laboratory of Molecular Microbiology and Biotechnology (LA.M.M.B.), Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (S.L.); (D.M.)
| | - Simone Furini
- Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy;
| | - Donata Medaglini
- Laboratory of Molecular Microbiology and Biotechnology (LA.M.M.B.), Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (S.L.); (D.M.)
| | - Annalisa Ciabattini
- Laboratory of Molecular Microbiology and Biotechnology (LA.M.M.B.), Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (S.L.); (D.M.)
| |
Collapse
|
24
|
Van Gassen S, Gaudilliere B, Angst MS, Saeys Y, Aghaeepour N. CytoNorm: A Normalization Algorithm for Cytometry Data. Cytometry A 2019; 97:268-278. [PMID: 31633883 PMCID: PMC7078957 DOI: 10.1002/cyto.a.23904] [Citation(s) in RCA: 108] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 09/12/2019] [Accepted: 09/13/2019] [Indexed: 11/09/2022]
Abstract
High‐dimensional flow cytometry has matured to a level that enables deep phenotyping of cellular systems at a clinical scale. The resulting high‐content data sets allow characterizing the human immune system at unprecedented single cell resolution. However, the results are highly dependent on sample preparation and measurements might drift over time. While various controls exist for assessment and improvement of data quality in a single sample, the challenges of cross‐sample normalization attempts have been limited to aligning marker distributions across subjects. These approaches, inspired by bulk genomics and proteomics assays, ignore the single‐cell nature of the data and risk the removal of biologically relevant signals. This work proposes CytoNorm, a normalization algorithm to ensure internal consistency between clinical samples based on shared controls across various study batches. Data from the shared controls is used to learn the appropriate transformations for each batch (e.g., each analysis day). Importantly, some sources of technical variation are strongly influenced by the amount of protein expressed on specific cell types, requiring several population‐specific transformations to normalize cells from a heterogeneous sample. To address this, our approach first identifies the overall cellular distribution using a clustering step, and calculates subset‐specific transformations on the control samples by computing their quantile distributions and aligning them with splines. These transformations are then applied to all other clinical samples in the batch to remove the batch‐specific variations. We evaluated the algorithm on a customized data set with two shared controls across batches. One control sample was used for calculation of the normalization transformations and the second control was used as a blinded test set and evaluated with Earth Mover's distance. Additional results are provided using two real‐world clinical data sets. Overall, our method compared favorably to standard normalization procedures. The algorithm is implemented in the R package “CytoNorm” and available via the following link: http://www.github.com/saeyslab/CytoNorm © 2019 The Authors. Cytometry Part A published by Wiley Periodicals, Inc. on behalf of International Society for Advancement of Cytometry.
Collapse
Affiliation(s)
- Sofie Van Gassen
- Department of Applied MathematicsComputer Science and Statistics, Ghent UniversityGhentBelgium
- Data Mining and Modeling for BiomedicineVIB Center for Inflammation ResearchGhentBelgium
| | - Brice Gaudilliere
- Department of Anesthesiology, Perioperative and Pain MedicineStanford University School of MedicineStanfordCalifornia
| | - Martin S. Angst
- Department of Anesthesiology, Perioperative and Pain MedicineStanford University School of MedicineStanfordCalifornia
| | - Yvan Saeys
- Department of Applied MathematicsComputer Science and Statistics, Ghent UniversityGhentBelgium
- Data Mining and Modeling for BiomedicineVIB Center for Inflammation ResearchGhentBelgium
| | - Nima Aghaeepour
- Department of Anesthesiology, Perioperative and Pain MedicineStanford University School of MedicineStanfordCalifornia
- Department of Biomedical Data SciencesStanford University School of MedicineStanfordCalifornia
| |
Collapse
|
25
|
Vazquez J, Ong IM, Stanic AK. Single-cell technologies in reproductive immunology. Am J Reprod Immunol 2019; 82:e13157. [PMID: 31206899 PMCID: PMC6697222 DOI: 10.1111/aji.13157] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 06/06/2019] [Accepted: 06/07/2019] [Indexed: 11/29/2022] Open
Abstract
The maternal-fetal interface represents a unique immune privileged site that maintains the ability to defend against pathogens while orchestrating the necessary tissue remodeling required for placentation. The recent discovery of novel cellular families (innate lymphoid cells, tissue-resident NK cells) suggests that our understanding of the decidual immunome is incomplete. To understand this complex milieu, new technological developments allow reproductive immunologists to collect increasingly complex data at a cellular resolution. Polychromatic flow cytometry allows for greater resolution in the identification of novel cell types by surface and intracellular protein. Single-cell RNA-seq coupled with microfluidics allows for efficient cellular transcriptomics. The extreme dimensionality and size of data sets generated, however, requires the application of novel computational approaches for unbiased analysis. There are now multiple dimensionality reduction (tSNE, SPADE) and visualization tools (SPICE) that allow researchers to efficiently analyze flow cytometry data. Development of computational tools has also been extended to RNA-seq data (including scRNA-seq), which requires specific analytical tools. Here, we provide an overview and a brief primer for the reproductive immunology community on data acquisition and computational tools for the analysis of complex flow cytometry and RNA-seq data.
Collapse
Affiliation(s)
- Jessica Vazquez
- Division of Reproductive Sciences, University of Wisconsin-Madison, Madison, WI
| | - Irene M Ong
- Division of Reproductive Sciences, University of Wisconsin-Madison, Madison, WI
- Division of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, WI
| | - Aleksandar K. Stanic
- Division of Reproductive Sciences, University of Wisconsin-Madison, Madison, WI
- Division of Reproductive Endocrinology and Infertility, Departments of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, WI
| |
Collapse
|
26
|
Lux M, Brinkman RR, Chauve C, Laing A, Lorenc A, Abeler-Dörner L, Hammer B. flowLearn: fast and precise identification and quality checking of cell populations in flow cytometry. Bioinformatics 2019; 34:2245-2253. [PMID: 29462241 PMCID: PMC6022609 DOI: 10.1093/bioinformatics/bty082] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Accepted: 02/14/2018] [Indexed: 12/21/2022] Open
Abstract
Motivation Identification of cell populations in flow cytometry is a critical part of the analysis and lays the groundwork for many applications and research discovery. The current paradigm of manual analysis is time consuming and subjective. A common goal of users is to replace manual analysis with automated methods that replicate their results. Supervised tools provide the best performance in such a use case, however they require fine parameterization to obtain the best results. Hence, there is a strong need for methods that are fast to setup, accurate and interpretable. Results flowLearn is a semi-supervised approach for the quality-checked identification of cell populations. Using a very small number of manually gated samples, through density alignments it is able to predict gates on other samples with high accuracy and speed. On two state-of-the-art datasets, our tool achieves median(F1)-measures exceeding 0.99 for 31%, and 0.90 for 80% of all analyzed populations. Furthermore, users can directly interpret and adjust automated gates on new sample files to iteratively improve the initial training. Availability and implementation FlowLearn is available as an R package on https://github.com/mlux86/flowLearn. Evaluation data is publicly available online. Details can be found in the Supplementary Material. Supplementary information Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Markus Lux
- Computational Methods for the Analysis of the Diversity and Dynamics of Genomes, Bielefeld University, Bielefeld, Germany
| | - Ryan Remy Brinkman
- Terry Fox Laboratory, BC Cancer Research Centre, Vancouver, Canada.,Department of Medical Genetics, University of British Columbia, Vancouver, Canada.,Cytapex Bioinformatics Inc., Vancouver, Canada
| | - Cedric Chauve
- Department of Mathematics, Simon Fraser University, Vancouver, Canada
| | - Adam Laing
- Department of Immunobiology, King's College London, London, UK
| | - Anna Lorenc
- Department of Immunobiology, King's College London, London, UK
| | | | | |
Collapse
|
27
|
Characterization of immune cell subtypes in three commonly used mouse strains reveals gender and strain-specific variations. J Transl Med 2019; 99:93-106. [PMID: 30353130 PMCID: PMC6524955 DOI: 10.1038/s41374-018-0137-1] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 08/22/2018] [Accepted: 09/12/2018] [Indexed: 12/21/2022] Open
Abstract
The lack of consensus on bone marrow (BM) and splenic immune cell profiles in preclinical mouse strains complicates comparative analysis across different studies. Although studies have documented relative distribution of immune cells from peripheral blood in mice, similar studies for BM and spleen from naïve mice are lacking. In an effort to establish strain- and gender-specific benchmarks for distribution of various immune cell subtypes in these organs, we performed immunophenotypic analysis of BM cells and splenocytes from both genders of three commonly used murine strains (C57BL/6NCr, 129/SvHsd, and BALB/cAnNCr). Total neutrophils and splenic macrophages were significantly higher in C57BL/6NCr, whereas total B cells were lower. Within C57BL/6NCr female mice, BM B cells were elevated with respect to the males whereas splenic mDCs and splenic neutrophils were reduced. Within BALB/cAnNCr male mice, BM CD4+ Tregs were elevated with respect to the other strains. Furthermore, in male BALB/cAnNCr mice, NK cells were elevated with respect to the other strains in both BM and spleen. Splenic CD4+ Tregs and splenic CD8+ T cells were reduced in male BALB/c mice in comparison to female mice. Bone marrow CD4+ T cells and mDCs were significantly increased in 129/SvHsd whereas splenic CD8+ T cells were reduced. In general, males exhibited higher immature myeloid cells, macrophages, and NK cells. To our knowledge, this study provides a first attempt to systematically establish organ-specific benchmarks on immune cells in studies involving these mouse strains.
Collapse
|
28
|
Abstract
CITRUS is a supervised machine learning algorithm designed to analyze single cell data, identify cell populations, and identify changes in the frequencies or functional marker expression patterns of those populations that are significantly associated with an outcome. The algorithm is a black box that includes steps to cluster cell populations, characterize these populations, and identify the significant characteristics. This chapter describes how to optimize the use of CITRUS by combining it with upstream and downstream data analysis and visualization tools.
Collapse
|
29
|
Abstract
The emergence of flow and mass cytometry technologies capable of generating 40-dimensional data has spurred research into automated methodologies that address bottlenecks across the entire analysis process from quality checking, data transformation, and cell population identification, to biomarker identification and visualizations. We review these approaches in the context of the stepwise progression through the different steps, including normalization, automated gating, outlier detection, and graphical presentation of results.
Collapse
Affiliation(s)
- Sherrie Wang
- Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, BC, Canada
| | - Ryan R Brinkman
- Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, BC, Canada.
| |
Collapse
|
30
|
Olsen LR, Leipold MD, Pedersen CB, Maecker HT. The anatomy of single cell mass cytometry data. Cytometry A 2018; 95:156-172. [DOI: 10.1002/cyto.a.23621] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Revised: 08/28/2018] [Accepted: 09/05/2018] [Indexed: 12/14/2022]
Affiliation(s)
- Lars R. Olsen
- Department of Bio and Health InformaticsTechnical University of Denmark Copenhagen Denmark
- Center for Genomic MedicineCopenhagen University Hospital Copenhagen Denmark
| | - Michael D. Leipold
- Institute for Immunity, Transplantation, and InfectionStanford University School of Medicine Stanford CA
| | - Christina B. Pedersen
- Department of Bio and Health InformaticsTechnical University of Denmark Copenhagen Denmark
- Center for Genomic MedicineCopenhagen University Hospital Copenhagen Denmark
| | - Holden Terry Maecker
- Institute for Immunity, Transplantation, and InfectionStanford University School of Medicine Stanford CA
| |
Collapse
|
31
|
Petkov S, Starodubova E, Latanova A, Kilpeläinen A, Latyshev O, Svirskis S, Wahren B, Chiodi F, Gordeychuk I, Isaguliants M. DNA immunization site determines the level of gene expression and the magnitude, but not the type of the induced immune response. PLoS One 2018; 13:e0197902. [PMID: 29864114 PMCID: PMC5986124 DOI: 10.1371/journal.pone.0197902] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2017] [Accepted: 05/10/2018] [Indexed: 12/19/2022] Open
Abstract
Optimization of DNA vaccine delivery improves the potency of the immune response and is crucial to clinical success. Here, we inquired how such optimization impacts the magnitude of the response, its specificity and type. BALB/c mice were DNA-immunized with two model immunogens, HIV-1 protease and reverse transcriptase by intramuscular or intradermal injections with electroporation. DNA immunogens were co-delivered with DNA encoding luciferase. Delivery and expression were monitored by in vivo bioluminescence imaging (BLI). The endpoint immune responses were assessed by IFN-γ/IL-2 FluoroSpot, multiparametric flow cytometry and antibody ELISA. Expression and immunogenicity were compared in relation to the delivery route. Regardless of the route, protease generated mainly IFN-γ, and reverse transcriptase, IL-2 and antibody response. BLI of mice immunized with protease- or reverse transcriptase/reporter plasmid mixtures, demonstrated significant loss of luminescence over time. The rate of decline of luminescence strongly correlated with the magnitude of immunogen-specific response, and depended on the immunogenicity profile and the immunization route. In vitro and in vivo BLI-based assays demonstrated that intradermal delivery strongly improved the immunogenicity of protease, and to a lesser extent, of reverse transcriptase. Immune response polarization and epitope hierarchy were not affected. Thus, by changing delivery/immunogen expression sites, it is possible to modulate the magnitude, but not the type or fine specificity of the induced immune response.
Collapse
Affiliation(s)
- Stefan Petkov
- Karolinska Institutet, Department of Microbiology, Tumor and Cell Biology, Stockholm, Sweden
| | - Elizaveta Starodubova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
- Chumakov Federal Scientific Center for Research and Development of Immune-and- Biological Products of the Russian Academy of Sciences, Moscow, Russia
| | - Anastasia Latanova
- Karolinska Institutet, Department of Microbiology, Tumor and Cell Biology, Stockholm, Sweden
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
- NF Gamaleja Research Center of Epidemiology and Microbiology, Moscow, Russia
| | - Athina Kilpeläinen
- Karolinska Institutet, Department of Microbiology, Tumor and Cell Biology, Stockholm, Sweden
| | - Oleg Latyshev
- Chumakov Federal Scientific Center for Research and Development of Immune-and- Biological Products of the Russian Academy of Sciences, Moscow, Russia
- NF Gamaleja Research Center of Epidemiology and Microbiology, Moscow, Russia
| | | | - Britta Wahren
- Karolinska Institutet, Department of Microbiology, Tumor and Cell Biology, Stockholm, Sweden
| | - Francesca Chiodi
- Karolinska Institutet, Department of Microbiology, Tumor and Cell Biology, Stockholm, Sweden
| | - Ilya Gordeychuk
- Chumakov Federal Scientific Center for Research and Development of Immune-and- Biological Products of the Russian Academy of Sciences, Moscow, Russia
- Sechenov First Moscow State Medical University, Moscow, Russia
| | - Maria Isaguliants
- Karolinska Institutet, Department of Microbiology, Tumor and Cell Biology, Stockholm, Sweden
- Chumakov Federal Scientific Center for Research and Development of Immune-and- Biological Products of the Russian Academy of Sciences, Moscow, Russia
- NF Gamaleja Research Center of Epidemiology and Microbiology, Moscow, Russia
- Riga Stradins University, Riga, Latvia
| |
Collapse
|
32
|
Lee AJ, Chang I, Burel JG, Lindestam Arlehamn CS, Mandava A, Weiskopf D, Peters B, Sette A, Scheuermann RH, Qian Y. DAFi: A directed recursive data filtering and clustering approach for improving and interpreting data clustering identification of cell populations from polychromatic flow cytometry data. Cytometry A 2018; 93:597-610. [PMID: 29665244 PMCID: PMC6030426 DOI: 10.1002/cyto.a.23371] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Revised: 02/05/2018] [Accepted: 03/15/2018] [Indexed: 11/10/2022]
Abstract
Computational methods for identification of cell populations from polychromatic flow cytometry data are changing the paradigm of cytometry bioinformatics. Data clustering is the most common computational approach to unsupervised identification of cell populations from multidimensional cytometry data. However, interpretation of the identified data clusters is labor-intensive. Certain types of user-defined cell populations are also difficult to identify by fully automated data clustering analysis. Both are roadblocks before a cytometry lab can adopt the data clustering approach for cell population identification in routine use. We found that combining recursive data filtering and clustering with constraints converted from the user manual gating strategy can effectively address these two issues. We named this new approach DAFi: Directed Automated Filtering and Identification of cell populations. Design of DAFi preserves the data-driven characteristics of unsupervised clustering for identifying novel cell subsets, but also makes the results interpretable to experimental scientists through mapping and merging the multidimensional data clusters into the user-defined two-dimensional gating hierarchy. The recursive data filtering process in DAFi helped identify small data clusters which are otherwise difficult to resolve by a single run of the data clustering method due to the statistical interference of the irrelevant major clusters. Our experiment results showed that the proportions of the cell populations identified by DAFi, while being consistent with those by expert centralized manual gating, have smaller technical variances across samples than those from individual manual gating analysis and the nonrecursive data clustering analysis. Compared with manual gating segregation, DAFi-identified cell populations avoided the abrupt cut-offs on the boundaries. DAFi has been implemented to be used with multiple data clustering methods including K-means, FLOCK, FlowSOM, and the ClusterR package. For cell population identification, DAFi supports multiple options including clustering, bisecting, slope-based gating, and reversed filtering to meet various autogating needs from different scientific use cases. © 2018 International Society for Advancement of Cytometry.
Collapse
Affiliation(s)
| | - Ivan Chang
- J. Craig Venter Institute, La Jolla, California
| | - Julie G. Burel
- La Jolla Institute for Allergy and Immunology, La Jolla, California
| | | | | | - Daniela Weiskopf
- La Jolla Institute for Allergy and Immunology, La Jolla, California
| | - Bjoern Peters
- La Jolla Institute for Allergy and Immunology, La Jolla, California
| | - Alessandro Sette
- La Jolla Institute for Allergy and Immunology, La Jolla, California
- Department of Medicine, University of California, San Diego, California
| | - Richard H. Scheuermann
- J. Craig Venter Institute, La Jolla, California
- Department of Pathology, University of California, San Diego, California
| | - Yu Qian
- J. Craig Venter Institute, La Jolla, California
| |
Collapse
|
33
|
Codon optimization and improved delivery/immunization regimen enhance the immune response against wild-type and drug-resistant HIV-1 reverse transcriptase, preserving its Th2-polarity. Sci Rep 2018; 8:8078. [PMID: 29799015 PMCID: PMC5967322 DOI: 10.1038/s41598-018-26281-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Accepted: 05/01/2018] [Indexed: 02/06/2023] Open
Abstract
DNA vaccines require a considerable enhancement of immunogenicity. Here, we optimized a prototype DNA vaccine against drug-resistant HIV-1 based on a weak Th2-immunogen, HIV-1 reverse transcriptase (RT). We designed expression-optimized genes encoding inactivated wild-type and drug-resistant RTs (RT-DNAs) and introduced them into mice by intradermal injections followed by electroporation. RT-DNAs were administered as single or double primes with or without cyclic-di-GMP, or as a prime followed by boost with RT-DNA mixed with a luciferase-encoding plasmid (“surrogate challenge”). Repeated primes improved cellular responses and broadened epitope specificity. Addition of cyclic-di-GMP induced a transient increase in IFN-γ production. The strongest anti-RT immune response was achieved in a prime-boost protocol with electroporation by short 100V pulses done using penetrating electrodes. The RT-specific response, dominated by CD4+ T-cells, targeted epitopes at aa 199–220 and aa 528–543. Drug-resistance mutations disrupted the epitope at aa 205–220, while the CTL epitope at aa 202–210 was not affected. Overall, multiparametric optimization of RT strengthened its Th2- performance. A rapid loss of RT/luciferase-expressing cells in the surrogate challenge experiment revealed a lytic potential of anti-RT response. Such lytic CD4+ response would be beneficial for an HIV vaccine due to its comparative insensitivity to immune escape.
Collapse
|
34
|
Shaham U, Stanton KP, Zhao J, Li H, Raddassi K, Montgomery R, Kluger Y. Removal of batch effects using distribution-matching residual networks. Bioinformatics 2018; 33:2539-2546. [PMID: 28419223 DOI: 10.1093/bioinformatics/btx196] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Accepted: 03/31/2017] [Indexed: 11/12/2022] Open
Abstract
Motivation Sources of variability in experimentally derived data include measurement error in addition to the physical phenomena of interest. This measurement error is a combination of systematic components, originating from the measuring instrument and random measurement errors. Several novel biological technologies, such as mass cytometry and single-cell RNA-seq (scRNA-seq), are plagued with systematic errors that may severely affect statistical analysis if the data are not properly calibrated. Results We propose a novel deep learning approach for removing systematic batch effects. Our method is based on a residual neural network, trained to minimize the Maximum Mean Discrepancy between the multivariate distributions of two replicates, measured in different batches. We apply our method to mass cytometry and scRNA-seq datasets, and demonstrate that it effectively attenuates batch effects. Availability and Implementation our codes and data are publicly available at https://github.com/ushaham/BatchEffectRemoval.git. Contact yuval.kluger@yale.edu. Supplementary information Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Uri Shaham
- Department of Statistics, Yale University, New Haven, CT 06511, USA
| | - Kelly P Stanton
- Department of Pathology, Yale School of Medicine, New Haven, CT 06510, USA.,Program of Computational Biology and Bioinformatics, Yale University, New Haven, CT 06520, USA
| | - Jun Zhao
- Program of Computational Biology and Bioinformatics, Yale University, New Haven, CT 06520, USA
| | - Huamin Li
- Applied Mathematics Program, Yale University, New Haven, CT 06511, USA
| | | | - Ruth Montgomery
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06510, USA
| | - Yuval Kluger
- Department of Pathology, Yale School of Medicine, New Haven, CT 06510, USA.,Program of Computational Biology and Bioinformatics, Yale University, New Haven, CT 06520, USA.,Applied Mathematics Program, Yale University, New Haven, CT 06511, USA
| |
Collapse
|
35
|
Single Cell and Population Level Analysis of HCA Data. Methods Mol Biol 2017. [PMID: 29082497 DOI: 10.1007/978-1-4939-7357-6_15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
High Content Analysis instrumentation has undergone tremendous hardware advances in recent years. It is now possible to obtain images of hundreds of thousands to millions of individual objects, across multiple wells, channels, and plates, in a reasonable amount of time. In addition, it is possible to extract dozens, or hundreds, of features per object using commonly available software tools. Analyzing this data provides new challenges to the scientists. The magnitude of these numbers is reminiscent of flow cytometer, where practitioners have long been taking what effectively amounted to very low resolution, multi-parametric measurements from individual cells for many decades. Flow cytometrists have developed a wide range of tools to effectively analyze and interpret these types of data. This chapter will review the techniques used in flow cytometry and show how they can easily and effectively be applied to High Content Analysis.
Collapse
|
36
|
Baklaushev VP, Kilpeläinen A, Petkov S, Abakumov MA, Grinenko NF, Yusubalieva GM, Latanova AA, Gubskiy IL, Zabozlaev FG, Starodubova ES, Abakumova TO, Isaguliants MG, Chekhonin VP. Luciferase Expression Allows Bioluminescence Imaging But Imposes Limitations on the Orthotopic Mouse (4T1) Model of Breast Cancer. Sci Rep 2017; 7:7715. [PMID: 28798322 PMCID: PMC5552689 DOI: 10.1038/s41598-017-07851-z] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Accepted: 07/04/2017] [Indexed: 01/08/2023] Open
Abstract
Implantation of reporter-labeled tumor cells in an immunocompetent host involves a risk of their immune elimination. We have studied this effect in a mouse model of breast cancer after the orthotopic implantation of mammary gland adenocarcinoma 4T1 cells genetically labelled with luciferase (Luc). Mice were implanted with 4T1 cells and two derivative Luc-expressing clones 4T1luc2 and 4T1luc2D6 exhibiting equal in vitro growth rates. In vivo, the daughter 4T1luc2 clone exhibited nearly the same, and 4T1luc2D6, a lower growth rate than the parental cells. The metastatic potential of 4T1 variants was assessed by magnetic resonance, bioluminescent imaging, micro-computed tomography, and densitometry which detected 100-μm metastases in multiple organs and bones at the early stage of their development. After 3-4 weeks, 4T1 generated 11.4 ± 2.1, 4T1luc2D6, 4.5 ± 0.6; and 4T1luc2, <1 metastases per mouse, locations restricted to lungs and regional lymph nodes. Mice bearing Luc-expressing tumors developed IFN-γ response to the dominant CTL epitope of Luc. Induced by intradermal DNA-immunization, such response protected mice from the establishment of 4T1luc2-tumors. Our data show that natural or induced cellular response against the reporter restricts growth and metastatic activity of the reporter-labelled tumor cells. Such cells represent a powerful instrument for improving immunization technique for cancer vaccine applications.
Collapse
Affiliation(s)
- V P Baklaushev
- Research and Education Center for Medical Nanobiotechnology, Pirogov Russian National Research Medical University, Ministry of Health of the Russian Federation, Moscow, Russia.
- Federal Research and Clinical Center of Specialized Medical Care and Medical Technologies, Federal Biomedical Agency of the Russian Federation, Moscow, Russia.
| | - A Kilpeläinen
- Research and Education Center for Medical Nanobiotechnology, Pirogov Russian National Research Medical University, Ministry of Health of the Russian Federation, Moscow, Russia
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - S Petkov
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - M A Abakumov
- Research and Education Center for Medical Nanobiotechnology, Pirogov Russian National Research Medical University, Ministry of Health of the Russian Federation, Moscow, Russia
| | - N F Grinenko
- Research and Education Center for Medical Nanobiotechnology, Pirogov Russian National Research Medical University, Ministry of Health of the Russian Federation, Moscow, Russia
| | - G M Yusubalieva
- Department of Fundamental and Applied Neurobiology, Serbsky National Research Center for Social and Forensic Psychiatry, Ministry of Health of the Russian Federation, Moscow, Russia
| | - A A Latanova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
- Chumakov Federal Scientific Center for Research and Development of Immunobiological Preparations, Moscow, Russia
| | - I L Gubskiy
- Research and Education Center for Medical Nanobiotechnology, Pirogov Russian National Research Medical University, Ministry of Health of the Russian Federation, Moscow, Russia
| | - F G Zabozlaev
- Federal Research and Clinical Center of Specialized Medical Care and Medical Technologies, Federal Biomedical Agency of the Russian Federation, Moscow, Russia
| | - E S Starodubova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
- Chumakov Federal Scientific Center for Research and Development of Immunobiological Preparations, Moscow, Russia
| | - T O Abakumova
- Department of Fundamental and Applied Neurobiology, Serbsky National Research Center for Social and Forensic Psychiatry, Ministry of Health of the Russian Federation, Moscow, Russia
| | - M G Isaguliants
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden.
- Chumakov Federal Scientific Center for Research and Development of Immunobiological Preparations, Moscow, Russia.
- N.F. Gamaleya Research Center of Epidemiology and Microbiology, Moscow, Russia.
- Riga Stradins University, Riga, Latvia.
| | - V P Chekhonin
- Research and Education Center for Medical Nanobiotechnology, Pirogov Russian National Research Medical University, Ministry of Health of the Russian Federation, Moscow, Russia
- Department of Fundamental and Applied Neurobiology, Serbsky National Research Center for Social and Forensic Psychiatry, Ministry of Health of the Russian Federation, Moscow, Russia
| |
Collapse
|
37
|
Nowicka M, Krieg C, Crowell HL, Weber LM, Hartmann FJ, Guglietta S, Becher B, Levesque MP, Robinson MD. CyTOF workflow: differential discovery in high-throughput high-dimensional cytometry datasets. F1000Res 2017; 6:748. [PMID: 28663787 PMCID: PMC5473464 DOI: 10.12688/f1000research.11622.1] [Citation(s) in RCA: 210] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/24/2017] [Indexed: 01/02/2023] Open
Abstract
High dimensional mass and flow cytometry (HDCyto) experiments have become a method of choice for high throughput interrogation and characterization of cell populations.Here, we present an R-based pipeline for differential analyses of HDCyto data, largely based on Bioconductor packages. We computationally define cell populations using FlowSOM clustering, and facilitate an optional but reproducible strategy for manual merging of algorithm-generated clusters. Our workflow offers different analysis paths, including association of cell type abundance with a phenotype or changes in signaling markers within specific subpopulations, or differential analyses of aggregated signals. Importantly, the differential analyses we show are based on regression frameworks where the HDCyto data is the response; thus, we are able to model arbitrary experimental designs, such as those with batch effects, paired designs and so on. In particular, we apply generalized linear mixed models to analyses of cell population abundance or cell-population-specific analyses of signaling markers, allowing overdispersion in cell count or aggregated signals across samples to be appropriately modeled. To support the formal statistical analyses, we encourage exploratory data analysis at every step, including quality control (e.g. multi-dimensional scaling plots), reporting of clustering results (dimensionality reduction, heatmaps with dendrograms) and differential analyses (e.g. plots of aggregated signals).
Collapse
Affiliation(s)
- Malgorzata Nowicka
- Institute for Molecular Life Sciences, University of Zurich, Zurich, 8057, Switzerland
- SIB Swiss Institute of Bioinformatics, University of Zurich, Zurich, 8057, Switzerland
| | - Carsten Krieg
- Institute of Experimental Immunology, University of Zurich, Zurich, 8057, Switzerland
| | - Helena L. Crowell
- Institute for Molecular Life Sciences, University of Zurich, Zurich, 8057, Switzerland
- SIB Swiss Institute of Bioinformatics, University of Zurich, Zurich, 8057, Switzerland
| | - Lukas M. Weber
- Institute for Molecular Life Sciences, University of Zurich, Zurich, 8057, Switzerland
- SIB Swiss Institute of Bioinformatics, University of Zurich, Zurich, 8057, Switzerland
| | - Felix J. Hartmann
- Institute of Experimental Immunology, University of Zurich, Zurich, 8057, Switzerland
| | - Silvia Guglietta
- Department of Experimental Oncology, European Institute of Oncology, Via Adamello 16, Milan, I-20139, Italy
| | - Burkhard Becher
- Institute of Experimental Immunology, University of Zurich, Zurich, 8057, Switzerland
| | - Mitchell P. Levesque
- Department of Dermatology, University Hospital Zurich, Zurich, CH-8091, Switzerland
| | - Mark D. Robinson
- Institute for Molecular Life Sciences, University of Zurich, Zurich, 8057, Switzerland
- SIB Swiss Institute of Bioinformatics, University of Zurich, Zurich, 8057, Switzerland
| |
Collapse
|
38
|
Nowicka M, Krieg C, Crowell HL, Weber LM, Hartmann FJ, Guglietta S, Becher B, Levesque MP, Robinson MD. CyTOF workflow: differential discovery in high-throughput high-dimensional cytometry datasets. F1000Res 2017; 6:748. [PMID: 28663787 PMCID: PMC5473464 DOI: 10.12688/f1000research.11622.3] [Citation(s) in RCA: 184] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/15/2019] [Indexed: 12/15/2022] Open
Abstract
High-dimensional mass and flow cytometry (HDCyto) experiments have become a method of choice for high-throughput interrogation and characterization of cell populations. Here, we present an updated R-based pipeline for differential analyses of HDCyto data, largely based on Bioconductor packages. We computationally define cell populations using FlowSOM clustering, and facilitate an optional but reproducible strategy for manual merging of algorithm-generated clusters. Our workflow offers different analysis paths, including association of cell type abundance with a phenotype or changes in signalling markers within specific subpopulations, or differential analyses of aggregated signals. Importantly, the differential analyses we show are based on regression frameworks where the HDCyto data is the response; thus, we are able to model arbitrary experimental designs, such as those with batch effects, paired designs and so on. In particular, we apply generalized linear mixed models or linear mixed models to analyses of cell population abundance or cell-population-specific analyses of signaling markers, allowing overdispersion in cell count or aggregated signals across samples to be appropriately modeled. To support the formal statistical analyses, we encourage exploratory data analysis at every step, including quality control (e.g., multi-dimensional scaling plots), reporting of clustering results (dimensionality reduction, heatmaps with dendrograms) and differential analyses (e.g., plots of aggregated signals).
Collapse
Affiliation(s)
- Malgorzata Nowicka
- Institute for Molecular Life Sciences, University of Zurich, Zurich, 8057, Switzerland
- SIB Swiss Institute of Bioinformatics, University of Zurich, Zurich, 8057, Switzerland
| | - Carsten Krieg
- Institute of Experimental Immunology, University of Zurich, Zurich, 8057, Switzerland
| | - Helena L. Crowell
- Institute for Molecular Life Sciences, University of Zurich, Zurich, 8057, Switzerland
- SIB Swiss Institute of Bioinformatics, University of Zurich, Zurich, 8057, Switzerland
| | - Lukas M. Weber
- Institute for Molecular Life Sciences, University of Zurich, Zurich, 8057, Switzerland
- SIB Swiss Institute of Bioinformatics, University of Zurich, Zurich, 8057, Switzerland
| | - Felix J. Hartmann
- Institute of Experimental Immunology, University of Zurich, Zurich, 8057, Switzerland
| | - Silvia Guglietta
- Department of Experimental Oncology, European Institute of Oncology, Via Adamello 16, Milan, I-20139, Italy
| | - Burkhard Becher
- Institute of Experimental Immunology, University of Zurich, Zurich, 8057, Switzerland
| | - Mitchell P. Levesque
- Department of Dermatology, University Hospital Zurich, Zurich, CH-8091, Switzerland
| | - Mark D. Robinson
- Institute for Molecular Life Sciences, University of Zurich, Zurich, 8057, Switzerland
- SIB Swiss Institute of Bioinformatics, University of Zurich, Zurich, 8057, Switzerland
| |
Collapse
|
39
|
Nowicka M, Krieg C, Crowell HL, Weber LM, Hartmann FJ, Guglietta S, Becher B, Levesque MP, Robinson MD. CyTOF workflow: differential discovery in high-throughput high-dimensional cytometry datasets. F1000Res 2017; 6:748. [PMID: 28663787 PMCID: PMC5473464 DOI: 10.12688/f1000research.11622.2] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/15/2017] [Indexed: 12/19/2022] Open
Abstract
High dimensional mass and flow cytometry (HDCyto) experiments have become a method of choice for high throughput interrogation and characterization of cell populations.Here, we present an R-based pipeline for differential analyses of HDCyto data, largely based on Bioconductor packages. We computationally define cell populations using FlowSOM clustering, and facilitate an optional but reproducible strategy for manual merging of algorithm-generated clusters. Our workflow offers different analysis paths, including association of cell type abundance with a phenotype or changes in signaling markers within specific subpopulations, or differential analyses of aggregated signals. Importantly, the differential analyses we show are based on regression frameworks where the HDCyto data is the response; thus, we are able to model arbitrary experimental designs, such as those with batch effects, paired designs and so on. In particular, we apply generalized linear mixed models to analyses of cell population abundance or cell-population-specific analyses of signaling markers, allowing overdispersion in cell count or aggregated signals across samples to be appropriately modeled. To support the formal statistical analyses, we encourage exploratory data analysis at every step, including quality control (e.g. multi-dimensional scaling plots), reporting of clustering results (dimensionality reduction, heatmaps with dendrograms) and differential analyses (e.g. plots of aggregated signals).
Collapse
Affiliation(s)
- Malgorzata Nowicka
- Institute for Molecular Life Sciences, University of Zurich, Zurich, 8057, Switzerland
- SIB Swiss Institute of Bioinformatics, University of Zurich, Zurich, 8057, Switzerland
| | - Carsten Krieg
- Institute of Experimental Immunology, University of Zurich, Zurich, 8057, Switzerland
| | - Helena L. Crowell
- Institute for Molecular Life Sciences, University of Zurich, Zurich, 8057, Switzerland
- SIB Swiss Institute of Bioinformatics, University of Zurich, Zurich, 8057, Switzerland
| | - Lukas M. Weber
- Institute for Molecular Life Sciences, University of Zurich, Zurich, 8057, Switzerland
- SIB Swiss Institute of Bioinformatics, University of Zurich, Zurich, 8057, Switzerland
| | - Felix J. Hartmann
- Institute of Experimental Immunology, University of Zurich, Zurich, 8057, Switzerland
| | - Silvia Guglietta
- Department of Experimental Oncology, European Institute of Oncology, Via Adamello 16, Milan, I-20139, Italy
| | - Burkhard Becher
- Institute of Experimental Immunology, University of Zurich, Zurich, 8057, Switzerland
| | - Mitchell P. Levesque
- Department of Dermatology, University Hospital Zurich, Zurich, CH-8091, Switzerland
| | - Mark D. Robinson
- Institute for Molecular Life Sciences, University of Zurich, Zurich, 8057, Switzerland
- SIB Swiss Institute of Bioinformatics, University of Zurich, Zurich, 8057, Switzerland
| |
Collapse
|
40
|
Rubbens P, Props R, Boon N, Waegeman W. Flow Cytometric Single-Cell Identification of Populations in Synthetic Bacterial Communities. PLoS One 2017; 12:e0169754. [PMID: 28122063 PMCID: PMC5266259 DOI: 10.1371/journal.pone.0169754] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Accepted: 12/21/2016] [Indexed: 01/14/2023] Open
Abstract
Bacterial cells can be characterized in terms of their cell properties using flow cytometry. Flow cytometry is able to deliver multiparametric measurements of up to 50,000 cells per second. However, there has not yet been a thorough survey concerning the identification of the population to which bacterial single cells belong based on flow cytometry data. This paper not only aims to assess the quality of flow cytometry data when measuring bacterial populations, but also suggests an alternative approach for analyzing synthetic microbial communities. We created so-called in silico communities, which allow us to explore the possibilities of bacterial flow cytometry data using supervised machine learning techniques. We can identify single cells with an accuracy >90% for more than half of the communities consisting out of two bacterial populations. In order to assess to what extent an in silico community is representative for its synthetic counterpart, we created so-called abundance gradients, a combination of synthetic (i.e., in vitro) communities containing two bacterial populations in varying abundances. By showing that we are able to retrieve an abundance gradient using a combination of in silico communities and supervised machine learning techniques, we argue that in silico communities form a viable representation for synthetic bacterial communities, opening up new opportunities for the analysis of synthetic communities and bacterial flow cytometry data in general.
Collapse
Affiliation(s)
- Peter Rubbens
- KERMIT, Department of Mathematical Modelling, Statistics and Bioinformatics, Ghent University, Ghent, Belgium
- * E-mail:
| | - Ruben Props
- Center for Microbial Technology and Ecology (CMET), Ghent University, Ghent, Belgium
| | - Nico Boon
- Center for Microbial Technology and Ecology (CMET), Ghent University, Ghent, Belgium
| | - Willem Waegeman
- KERMIT, Department of Mathematical Modelling, Statistics and Bioinformatics, Ghent University, Ghent, Belgium
| |
Collapse
|
41
|
CD4+ T cells with an activated and exhausted phenotype distinguish immunodeficiency during aviremic HIV-2 infection. AIDS 2016; 30:2415-2426. [PMID: 27525551 PMCID: PMC5051526 DOI: 10.1097/qad.0000000000001223] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
HIV type 2 (HIV-2) represents an attenuated form of HIV, in which many infected individuals remain ‘aviremic’ without antiretroviral therapy. However, aviremic HIV-2 disease progression exists, and in the current study, we therefore aimed to examine if specific pathological characteristics of CD4+ T cells are linked to such outcome.
Collapse
|
42
|
Azad A, Rajwa B, Pothen A. flowVS: channel-specific variance stabilization in flow cytometry. BMC Bioinformatics 2016; 17:291. [PMID: 27465477 PMCID: PMC4964071 DOI: 10.1186/s12859-016-1083-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Accepted: 05/14/2016] [Indexed: 01/21/2023] Open
Abstract
Background Comparing phenotypes of heterogeneous cell populations from multiple biological conditions is at the heart of scientific discovery based on flow cytometry (FC). When the biological signal is measured by the average expression of a biomarker, standard statistical methods require that variance be approximately stabilized in populations to be compared. Since the mean and variance of a cell population are often correlated in fluorescence-based FC measurements, a preprocessing step is needed to stabilize the within-population variances. Results We present a variance-stabilization algorithm, called flowVS, that removes the mean-variance correlations from cell populations identified in each fluorescence channel. flowVS transforms each channel from all samples of a data set by the inverse hyperbolic sine (asinh) transformation. For each channel, the parameters of the transformation are optimally selected by Bartlett’s likelihood-ratio test so that the populations attain homogeneous variances. The optimum parameters are then used to transform the corresponding channels in every sample. flowVS is therefore an explicit variance-stabilization method that stabilizes within-population variances in each channel by evaluating the homoskedasticity of clusters with a likelihood-ratio test. With two publicly available datasets, we show that flowVS removes the mean-variance dependence from raw FC data and makes the within-population variance relatively homogeneous. We demonstrate that alternative transformation techniques such as flowTrans, flowScape, logicle, and FCSTrans might not stabilize variance. Besides flow cytometry, flowVS can also be applied to stabilize variance in microarray data. With a publicly available data set we demonstrate that flowVS performs as well as the VSN software, a state-of-the-art approach developed for microarrays. Conclusions The homogeneity of variance in cell populations across FC samples is desirable when extracting features uniformly and comparing cell populations with different levels of marker expressions. The newly developed flowVS algorithm solves the variance-stabilization problem in FC and microarrays by optimally transforming data with the help of Bartlett’s likelihood-ratio test. On two publicly available FC datasets, flowVS stabilizes within-population variances more evenly than the available transformation and normalization techniques. flowVS-based variance stabilization can help in performing comparison and alignment of phenotypically identical cell populations across different samples. flowVS and the datasets used in this paper are publicly available in Bioconductor.
Collapse
Affiliation(s)
- Ariful Azad
- Computational Research Division, Lawrence Berkeley National Laboratory, 1 Cyclotron Rd, Berkeley, 94720, CA, USA.
| | - Bartek Rajwa
- Bindley Bioscience Center, Purdue University, West Lafayette, 47907, IN, USA
| | - Alex Pothen
- Department of Computer Science, Purdue University, West Lafayette, 47907, IN, USA
| |
Collapse
|
43
|
Saeys Y, Van Gassen S, Lambrecht BN. Computational flow cytometry: helping to make sense of high-dimensional immunology data. Nat Rev Immunol 2016; 16:449-62. [PMID: 27320317 DOI: 10.1038/nri.2016.56] [Citation(s) in RCA: 309] [Impact Index Per Article: 38.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Recent advances in flow cytometry allow scientists to measure an increasing number of parameters per cell, generating huge and high-dimensional datasets. To analyse, visualize and interpret these data, newly available computational techniques should be adopted, evaluated and improved upon by the immunological community. Computational flow cytometry is emerging as an important new field at the intersection of immunology and computational biology; it allows new biological knowledge to be extracted from high-throughput single-cell data. This Review provides non-experts with a broad and practical overview of the many recent developments in computational flow cytometry.
Collapse
Affiliation(s)
- Yvan Saeys
- VIB Inflammation Research Center, Technologiepark 927, Ghent B-9052, Belgium.,Department of Internal Medicine, Ghent University, De Pintelaan 185, Ghent B-9000, Belgium
| | - Sofie Van Gassen
- VIB Inflammation Research Center, Technologiepark 927, Ghent B-9052, Belgium.,Department of Information Technology, Technologiepark 15, Ghent B-9052, Belgium
| | - Bart N Lambrecht
- VIB Inflammation Research Center, Technologiepark 927, Ghent B-9052, Belgium.,Department of Internal Medicine, Ghent University, De Pintelaan 185, Ghent B-9000, Belgium.,Department of Pulmonary Medicine, Erasmus MC Rotterdam, Dr Molewaterplein 50, Rotterdam 3015 GE, The Netherlands
| |
Collapse
|
44
|
BayesFlow: latent modeling of flow cytometry cell populations. BMC Bioinformatics 2016; 17:25. [PMID: 26755197 PMCID: PMC4709953 DOI: 10.1186/s12859-015-0862-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Accepted: 12/17/2015] [Indexed: 12/23/2022] Open
Abstract
Background Flow cytometry is a widespread single-cell measurement technology with a multitude of clinical and research applications. Interpretation of flow cytometry data is hard; the instrumentation is delicate and can not render absolute measurements, hence samples can only be interpreted in relation to each other while at the same time comparisons are confounded by inter-sample variation. Despite this, most automated flow cytometry data analysis methods either treat samples individually or ignore the variation by for example pooling the data. A key requirement for models that include multiple samples is the ability to visualize and assess inferred variation, since what could be technical variation in one setting would be different phenotypes in another. Results We introduce BayesFlow, a pipeline for latent modeling of flow cytometry cell populations built upon a Bayesian hierarchical model. The model systematizes variation in location as well as shape. Expert knowledge can be incorporated through informative priors and the results can be supervised through compact and comprehensive visualizations. BayesFlow is applied to two synthetic and two real flow cytometry data sets. For the first real data set, taken from the FlowCAP I challenge, BayesFlow does not only give a gating which would place it among the top performers in FlowCAP I for this dataset, it also gives a more consistent treatment of different samples than either manual gating or other automated gating methods. The second real data set contains replicated flow cytometry measurements of samples from healthy individuals. BayesFlow gives here cell populations with clear expression patterns and small technical intra-donor variation as compared to biological inter-donor variation. Conclusions Modeling latent relations between samples through BayesFlow enables a systematic analysis of inter-sample variation. As opposed to other joint gating methods, effort is put at ensuring that the obtained partition of the data corresponds to actual cell populations, and the result is therefore directly biologically interpretable. BayesFlow is freely available at GitHub. Electronic supplementary material The online version of this article (doi:10.1186/s12859-015-0862-z) contains supplementary material, which is available to authorized users.
Collapse
|
45
|
Aghaeepour N, Chattopadhyay P, Chikina M, Dhaene T, Van Gassen S, Kursa M, Lambrecht BN, Malek M, Qian Y, Qiu P, Saeys Y, Stanton R, Tong D, Vens C, Walkowiak S, Wang K, Finak G, Gottardo R, Mosmann T, Nolan G, Scheuermann RH, Brinkman RR. A benchmark for evaluation of algorithms for identification of cellular correlates of clinical outcomes. Cytometry A 2016; 89:16-21. [PMID: 26447924 PMCID: PMC4874734 DOI: 10.1002/cyto.a.22732] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Revised: 05/20/2015] [Accepted: 07/16/2015] [Indexed: 11/07/2022]
Abstract
The Flow Cytometry: Critical Assessment of Population Identification Methods (FlowCAP) challenges were established to compare the performance of computational methods for identifying cell populations in multidimensional flow cytometry data. Here we report the results of FlowCAP-IV where algorithms from seven different research groups predicted the time to progression to AIDS among a cohort of 384 HIV+ subjects, using antigen-stimulated peripheral blood mononuclear cell (PBMC) samples analyzed with a 14-color staining panel. Two approaches (FlowReMi.1 and flowDensity-flowType-RchyOptimyx) provided statistically significant predictive value in the blinded test set. Manual validation of submitted results indicated that unbiased analysis of single cell phenotypes could reveal unexpected cell types that correlated with outcomes of interest in high dimensional flow cytometry datasets.
Collapse
Affiliation(s)
- Nima Aghaeepour
- British Columbia Cancer Agency, Vancouver, BC, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
- Baxter Laboratory in Stem Cell Biology, Stanford University, Stanford, CA, USA
| | - Pratip Chattopadhyay
- ImmunoTechnology Section, Vaccine Research Center, National Institutes of Health, Washington, DC, USA
| | - Maria Chikina
- Department Computational and Systems Biology, University of Pittsburgh, Pittsburg, USA
| | - Tom Dhaene
- Department of Information Technology, Ghent University - iMinds, Ghent, Belgium
| | - Sofie Van Gassen
- Department of Information Technology, Ghent University - iMinds, Ghent, Belgium
- Inflammation Research Center, VIB, Ghent, Belgium
- Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium
| | - Miron Kursa
- Interdisciplinary Centre for Mathematical and Computational Modelling, University of Warsaw, Warsaw, Poland
| | - Bart N. Lambrecht
- Inflammation Research Center, VIB, Ghent, Belgium
- Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium
| | | | - Yu Qian
- J. Craig Venter Institute, La Jolla, CA, USA
| | - Peng Qiu
- Department of Biomedical Engineering, Georgia Institute of Technology and Emory University
| | - Yvan Saeys
- Inflammation Research Center, VIB, Ghent, Belgium
- Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium
| | | | - Dong Tong
- The John van Geest Cancer Research Centre, Nottingham Trent University, UK & Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK
| | - Celine Vens
- Inflammation Research Center, VIB, Ghent, Belgium
- Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium
- Department of Public Health and Primary Care, KU Leuven Kulak, Kortrijk, Belgium
| | - Sławomir Walkowiak
- Interdisciplinary Centre for Mathematical and Computational Modelling, University of Warsaw, Warsaw, Poland
| | - Kui Wang
- Department of Mathematics, University of Queensland, St. Lucia, Brisbane, Australia
- School of Medicine, Shihezi University, Xinjiang 832000, China
| | - Greg Finak
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Raphael Gottardo
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Tim Mosmann
- David H. Smith Center for Vaccine Biology and Immunology, University of Rochester Medical Center, Rochester, NY, USA
| | - Garry Nolan
- Baxter Laboratory in Stem Cell Biology, Stanford University, Stanford, CA, USA
| | - Richard H. Scheuermann
- J. Craig Venter Institute, La Jolla, CA, USA
- Department of Pathology, University of California, San Diego, CA, USA
| | - Ryan R. Brinkman
- British Columbia Cancer Agency, Vancouver, BC, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
46
|
Landry BD, Clarke DC, Lee MJ. Studying Cellular Signal Transduction with OMIC Technologies. J Mol Biol 2015; 427:3416-40. [PMID: 26244521 PMCID: PMC4818567 DOI: 10.1016/j.jmb.2015.07.021] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2015] [Revised: 07/25/2015] [Accepted: 07/27/2015] [Indexed: 11/24/2022]
Abstract
In the gulf between genotype and phenotype exists proteins and, in particular, protein signal transduction systems. These systems use a relatively limited parts list to respond to a much longer list of extracellular, environmental, and/or mechanical cues with rapidity and specificity. Most signaling networks function in a highly non-linear and often contextual manner. Furthermore, these processes occur dynamically across space and time. Because of these complexities, systems and "OMIC" approaches are essential for the study of signal transduction. One challenge in using OMIC-scale approaches to study signaling is that the "signal" can take different forms in different situations. Signals are encoded in diverse ways such as protein-protein interactions, enzyme activities, localizations, or post-translational modifications to proteins. Furthermore, in some cases, signals may be encoded only in the dynamics, duration, or rates of change of these features. Accordingly, systems-level analyses of signaling may need to integrate multiple experimental and/or computational approaches. As the field has progressed, the non-triviality of integrating experimental and computational analyses has become apparent. Successful use of OMIC methods to study signaling will require the "right" experiments and the "right" modeling approaches, and it is critical to consider both in the design phase of the project. In this review, we discuss common OMIC and modeling approaches for studying signaling, emphasizing the philosophical and practical considerations for effectively merging these two types of approaches to maximize the probability of obtaining reliable and novel insights into signaling biology.
Collapse
Affiliation(s)
- Benjamin D Landry
- Program in Systems Biology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - David C Clarke
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, V5A 1S6 Canada
| | - Michael J Lee
- Program in Systems Biology, University of Massachusetts Medical School, Worcester, MA 01605, USA; Program in Molecular Medicine, Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01605, USA.
| |
Collapse
|
47
|
Liang M, Schwickart M, Schneider AK, Vainshtein I, Del Nagro C, Standifer N, Roskos LK. Receptor occupancy assessment by flow cytometry as a pharmacodynamic biomarker in biopharmaceutical development. CYTOMETRY PART B-CLINICAL CYTOMETRY 2015; 90:117-27. [PMID: 26054054 PMCID: PMC5042057 DOI: 10.1002/cyto.b.21259] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Revised: 04/20/2015] [Accepted: 05/28/2015] [Indexed: 12/19/2022]
Abstract
Receptor occupancy (RO) assays are designed to quantify the binding of therapeutics to their targets on the cell surface and are frequently used to generate pharmacodynamic (PD) biomarker data in nonclinical and clinical studies of biopharmaceuticals. When combined with the pharmacokinetic (PK) profile, RO data can establish PKPD relationships, which are crucial for informing dose decisions. RO is commonly measured by flow cytometry on fresh blood specimens and is subject to numerous technical and logistical challenges. To ensure that reliable and high quality results are generated from RO assays, careful assay design, key reagent characterization, data normalization/reporting, and thorough planning for implementation are of critical importance during development. In this article, the authors share their experiences and perspectives in these areas and discuss challenges and potential solutions when developing and implementing a flow cytometry‐based RO method in support of biopharmaceutical drug development. © 2015 The Authors Cytometry Part B: Clinical Cytometry Published by Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Meina Liang
- Department of Clinical Pharmacology and DMPK, Medimmune, LLC, Mountain View, California, 94043
| | - Martin Schwickart
- Department of Clinical Pharmacology and DMPK, Medimmune, LLC, Mountain View, California, 94043
| | - Amy K Schneider
- Department of Clinical Pharmacology and DMPK, Medimmune, LLC, Mountain View, California, 94043
| | - Inna Vainshtein
- Department of Clinical Pharmacology and DMPK, Medimmune, LLC, Mountain View, California, 94043
| | - Christopher Del Nagro
- Department of Clinical Pharmacology and DMPK, Medimmune, LLC, Mountain View, California, 94043
| | - Nathan Standifer
- Department of Clinical Pharmacology and DMPK, Medimmune, LLC, Mountain View, California, 94043
| | - Lorin K Roskos
- Department of Clinical Pharmacology and DMPK, Medimmune, LLC, Mountain View, California, 94043
| |
Collapse
|
48
|
Dundar M, Akova F, Yerebakan HZ, Rajwa B. A non-parametric Bayesian model for joint cell clustering and cluster matching: identification of anomalous sample phenotypes with random effects. BMC Bioinformatics 2014; 15:314. [PMID: 25248977 PMCID: PMC4262223 DOI: 10.1186/1471-2105-15-314] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2014] [Accepted: 09/16/2014] [Indexed: 12/13/2022] Open
Abstract
Background Flow cytometry (FC)-based computer-aided diagnostics is an emerging technique utilizing modern multiparametric cytometry systems. The major difficulty in using machine-learning approaches for classification of FC data arises from limited access to a wide variety of anomalous samples for training. In consequence, any learning with an abundance of normal cases and a limited set of specific anomalous cases is biased towards the types of anomalies represented in the training set. Such models do not accurately identify anomalies, whether previously known or unknown, that may exist in future samples tested. Although one-class classifiers trained using only normal cases would avoid such a bias, robust sample characterization is critical for a generalizable model. Owing to sample heterogeneity and instrumental variability, arbitrary characterization of samples usually introduces feature noise that may lead to poor predictive performance. Herein, we present a non-parametric Bayesian algorithm called ASPIRE (anomalous sample phenotype identification with random effects) that identifies phenotypic differences across a batch of samples in the presence of random effects. Our approach involves simultaneous clustering of cellular measurements in individual samples and matching of discovered clusters across all samples in order to recover global clusters using probabilistic sampling techniques in a systematic way. Results We demonstrate the performance of the proposed method in identifying anomalous samples in two different FC data sets, one of which represents a set of samples including acute myeloid leukemia (AML) cases, and the other a generic 5-parameter peripheral-blood immunophenotyping. Results are evaluated in terms of the area under the receiver operating characteristics curve (AUC). ASPIRE achieved AUCs of 0.99 and 1.0 on the AML and generic blood immunophenotyping data sets, respectively. Conclusions These results demonstrate that anomalous samples can be identified by ASPIRE with almost perfect accuracy without a priori access to samples of anomalous subtypes in the training set. The ASPIRE approach is unique in its ability to form generalizations regarding normal and anomalous states given only very weak assumptions regarding sample characteristics and origin. Thus, ASPIRE could become highly instrumental in providing unique insights about observed biological phenomena in the absence of full information about the investigated samples. Electronic supplementary material The online version of this article (doi:10.1186/1471-2105-15-314) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Murat Dundar
- Computer Science Department, IUPUI, 723 W, Michigan St,, 46037 Indianapolis IN, US.
| | | | | | | |
Collapse
|
49
|
Pyne S, Lee SX, Wang K, Irish J, Tamayo P, Nazaire MD, Duong T, Ng SK, Hafler D, Levy R, Nolan GP, Mesirov J, McLachlan GJ. Joint modeling and registration of cell populations in cohorts of high-dimensional flow cytometric data. PLoS One 2014; 9:e100334. [PMID: 24983991 PMCID: PMC4077578 DOI: 10.1371/journal.pone.0100334] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2013] [Accepted: 05/23/2014] [Indexed: 01/20/2023] Open
Abstract
In biomedical applications, an experimenter encounters different potential sources of variation in data such as individual samples, multiple experimental conditions, and multivariate responses of a panel of markers such as from a signaling network. In multiparametric cytometry, which is often used for analyzing patient samples, such issues are critical. While computational methods can identify cell populations in individual samples, without the ability to automatically match them across samples, it is difficult to compare and characterize the populations in typical experiments, such as those responding to various stimulations or distinctive of particular patients or time-points, especially when there are many samples. Joint Clustering and Matching (JCM) is a multi-level framework for simultaneous modeling and registration of populations across a cohort. JCM models every population with a robust multivariate probability distribution. Simultaneously, JCM fits a random-effects model to construct an overall batch template – used for registering populations across samples, and classifying new samples. By tackling systems-level variation, JCM supports practical biomedical applications involving large cohorts. Software for fitting the JCM models have been implemented in an R package EMMIX-JCM, available from http://www.maths.uq.edu.au/~gjm/mix_soft/EMMIX-JCM/.
Collapse
Affiliation(s)
- Saumyadipta Pyne
- CR Rao Advanced Institute of Mathematics, Statistics and Computer Science, Hyderabad, Andhra Pradesh, India
| | - Sharon X. Lee
- Department of Mathematics, University of Queensland, St. Lucia, Queensland, Australia
| | - Kui Wang
- Department of Mathematics, University of Queensland, St. Lucia, Queensland, Australia
| | - Jonathan Irish
- Division of Oncology, Stanford Medical School, Stanford, California, United States of America
- Baxter Laboratory for Stem Cell Biology, Department of Microbiology and Immunology, Stanford School of Medicine, Stanford, California, United States of America
- Department of Cancer Biology, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Pablo Tamayo
- Broad Institute of MIT and Harvard University, Cambridge, Massachusetts, United States of America
| | - Marc-Danie Nazaire
- Broad Institute of MIT and Harvard University, Cambridge, Massachusetts, United States of America
| | - Tarn Duong
- Molecular Mechanisms of Intracellular Transport, Unit Mixte de Recherche 144 Centre National de la Recherche Scientifique/Institut Curie, Paris, France
| | - Shu-Kay Ng
- School of Medicine, Griffith University, Meadowbrook, Queensland, Australia
| | - David Hafler
- Department of Neurology, Yale School of Medicine, New Haven, Connecticut, United States of America
| | - Ronald Levy
- Division of Oncology, Stanford Medical School, Stanford, California, United States of America
| | - Garry P. Nolan
- Baxter Laboratory for Stem Cell Biology, Department of Microbiology and Immunology, Stanford School of Medicine, Stanford, California, United States of America
| | - Jill Mesirov
- Broad Institute of MIT and Harvard University, Cambridge, Massachusetts, United States of America
| | - Geoffrey J. McLachlan
- Department of Mathematics, University of Queensland, St. Lucia, Queensland, Australia
- * E-mail:
| |
Collapse
|
50
|
Mosmann TR, Naim I, Rebhahn J, Datta S, Cavenaugh JS, Weaver JM, Sharma G. SWIFT-scalable clustering for automated identification of rare cell populations in large, high-dimensional flow cytometry datasets, part 2: biological evaluation. Cytometry A 2014; 85:422-33. [PMID: 24532172 PMCID: PMC4238823 DOI: 10.1002/cyto.a.22445] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2013] [Revised: 11/15/2013] [Accepted: 01/02/2014] [Indexed: 01/27/2023]
Abstract
A multistage clustering and data processing method, SWIFT (detailed in a companion manuscript), has been developed to detect rare subpopulations in large, high-dimensional flow cytometry datasets. An iterative sampling procedure initially fits the data to multidimensional Gaussian distributions, then splitting and merging stages use a criterion of unimodality to optimize the detection of rare subpopulations, to converge on a consistent cluster number, and to describe non-Gaussian distributions. Probabilistic assignment of cells to clusters, visualization, and manipulation of clusters by their cluster medians, facilitate application of expert knowledge using standard flow cytometry programs. The dual problems of rigorously comparing similar complex samples, and enumerating absent or very rare cell subpopulations in negative controls, were solved by assigning cells in multiple samples to a cluster template derived from a single or combined sample. Comparison of antigen-stimulated and control human peripheral blood cell samples demonstrated that SWIFT could identify biologically significant subpopulations, such as rare cytokine-producing influenza-specific T cells. A sensitivity of better than one part per million was attained in very large samples. Results were highly consistent on biological replicates, yet the analysis was sensitive enough to show that multiple samples from the same subject were more similar than samples from different subjects. A companion manuscript (Part 1) details the algorithmic development of SWIFT. © 2014 The Authors. Published by Wiley Periodicals Inc.
Collapse
Affiliation(s)
- Tim R Mosmann
- David H. Smith Center for Vaccine Biology and Immunology, University of Rochester Medical Center, University of Rochester, Rochester, New York
| | | | | | | | | | | | | |
Collapse
|