Skip to main content

This is a preprint.

It has not yet been peer reviewed by a journal.

The National Library of Medicine is running a pilot to include preprints that result from research funded by NIH in PMC and PubMed.

bioRxiv logoLink to bioRxiv
[Preprint]. 2023 Dec 15:2023.12.14.571745. [Version 1] doi: 10.1101/2023.12.14.571745

50-color phenotyping of the human immune system with in-depth assessment of T cells and dendritic cells

Andrew J Konecny 1,2,#, Peter Mage 3,#, Aaron J Tyznik 4,*, Martin Prlic 1,2,*, Florian Mair 1,5,*
PMCID: PMC10760076  PMID: 38168221

Abstract

We report the development of an optimized 50-color spectral flow cytometry panel designed for the in-depth analysis of the immune system in human blood and tissues, with the goal of maximizing the amount of information that can be collected using currently available flow cytometry platforms. We established and tested this panel using peripheral blood mononuclear cells (PBMCs), but included CD45 to enable its use for the analysis of human tissue samples. The panel contains lineage markers for all major immune cell subsets, and an extensive set of phenotyping markers focused on the activation and differentiation status of the T cell and dendritic cell (DC) compartment.

We outline the biological insight that can be gained from the simultaneous measurement of such a large number of proteins and propose that this approach provides a unique opportunity for the comprehensive exploration of the immune status in tissue biopsies and other human samples with a limited number of cells. Of note, we tested the panel to be compatible with cell sorting for further downstream applications. Furthermore, to facilitate the wide-spread implementation of such a panel across different cohorts and samples, we established a trimmed-down 45-color version which can be used with different spectral cytometry platforms.

Finally, to generate this panel, we utilized not only existing panel design guidelines, but also developed new metrics to systematically identify the optimal combination of 50 fluorochromes and evaluate fluorochrome-specific resolution in the context of a 50-color unmixing matrix.

Keywords: high-dimensional, spectral cytometry, immunophenotyping, human PBMCs, BD FACSDiscover S8, Sony ID7000

Background:

The immune system serves as the body's defense system against pathogens and is also essential for maintaining steady-state homeostasis in tissues (1) and preventing the development of malignant tumors (2). The composition and activation status of immune cells in the periphery and in tissues can be used to extrapolate immune cell differentiation and function. To facilitate data interpretation, an immune cell population is ideally analyzed in the context of other immune cell populations. Thus, to comprehensively study the functional state of the immune system, it is highly beneficial to capture as much information from as many different cell types as feasible. This is particularly relevant for assessing immune cell function in situ, for example in human tissue samples (3). Moreover, these human tissues are often limited in size and availability, e.g. tissue biopsies (4) or resected pieces of tumor tissue (5), which precludes parallel analysis with multiple panels or multiple applications. The development of an analysis approach that can provide broad and, for some cell subsets, in depth-phenotyping, paired with the ability to preserve cell populations of interest for downstream applications such as single-cell RNA-sequencing, is hence of importance.

The interactions between professional antigen-presenting cells (APCs) and different T cell subsets (6) are of particular interest in the context of studying anti-tumor immune responses (7). Dendritic cells (DCs) are highly specialized APCs, and generally divided into cross-presenting cDC1s and cDC2s (8), with CD163+ cDC3s being a more recently described subset during inflammatory conditions (9). Each of these DC populations appears to have a distinct function for steering an adaptive immune response. T cells consist of conventional CD4+ T cells, regulatory CD4+ T cells (Tregs), CD8+ T cells, γδ T cells and subsets of T cells with semi-invariant T cell receptors including mucosal-associated invariant T cells (MAIT cells) and invariant NK-T cells (10).

The panel presented here (Figure 1A) was designed to comprehensively capture the differentiation and activation status of T cells and APCs, while also measuring B cell, NK cell and innate lymphoid cells (ILCs) phenotypes (list of markers depicted in Figure 1B and Table 2). Optimization was done on human cryopreserved PBMCs, but the panel includes CD45 as a pan-hematopoietic marker and has been tested on human tissue-derived leukocytes (data not shown).

Figure 1: Overview gating of the 50-color panel on cryopreserved PBMCs.

Figure 1:

PBMCs were obtained from commercial vendors, stained as described in the online section of the manuscript and acquired on a BD FACSDiscover S8. The optical configuration of the instrument is described in Online Figures 1-2. Additional gating and staining controls are shown in Online Figures 9-11. Pre-gating of plots is annotated in the figure or indicated by dotted black arrows. For some plots different donors are shown for clarity. The gating strategy has been devised in such a way that the staining pattern for every marker in the panel can be shown at least once on a single A4 page. The raw data has been deposited on Flowrepository with the identifier FR-FCM-Z73V. Abbreviations: BUV: Brilliant Ultraviolet; BV: Brilliant Violet; BB: Brilliant Blue; RB: RealBlue; AF: AlexaFluor; PE: Phycoerythrin; APC: Allophycocyanin; Qdot: Quantum Dot; NIR: Near-Infrared;

(A) Gating strategy for CD45+ live cells, monocytes, B cells and γδ and αβ T cells.

(B) Overview of the 50 targets analyzed with this experiment. Some of the markers can be used for phenotyping multiple immune cell lineages.

(C) Representative plots for the main phenotyping markers in the B cell lineage (IgG, IgD, IgM and CD24).

(D) Gating strategy to delineate invariant NKT cells, MAIT cells, CD4+ and CD8+ T cells, as well as CD4+ regulatory T cells (Tregs).

(E) Representative plots for CD69, CD103, CD57 and PD-1 expression on non-naïve CD8+ cytotoxic T cells.

(F) Expression pattern for CD39, CXCR3, CCR4, CD45RO and ICOS on the CD4+ Treg population.

(G) Histogram overlays for the expression pattern of BTLA, CD27, CD28, CD38, TIGIT and KLRG1 on NK cells (grey), MAIT cells (orange), CD4+ Tregs (red), CD4+ non Tregs (purple), CD8+ naïve T cells (green) and CD8+ non-naïve T cells (blue). Dotted red lines indicate positivity cut-offs.

(H) Gating strategy for NK cells and NK cell subsets based on CD56, CD161, CD16 and Nkp46.

(I) Gating strategy for Basophils (CD123+ FcER1+ HLA-DR), plasmacytoid DCs (CD303+ HLA-DR+), pan conventional DCs (CD11c+ HLA-DR+), and the cDC1 (CD141+) and cDC2 (FcER1+) subsets.

(J) Histogram overlays for the expression pattern of CD86, CD40, CD11b, CD1c and CD163 on B cells (grey), CD8+ naïve T cells (green, negative control), CD14+ monocytes (purple), CD16+ monocytes (yellow), CD141+ cDC1s (red) and FcER1+ cDC2s (blue). Dotted red lines indicate positivity cut-offs.

(K) Gating strategy for Lin CD2+ CD127+ innate lymphoid cells (ILCs).

(L) A selection of fluorescence-minus-one (FMO) controls for the indicated markers: PD-1 and BTLA on CD8+ T cells, ICOS and CCR4 on Tregs, and CD86 and CD1c on Lin-cells as indicated. Dotted red lines indicate positivity cut-offs. Note that there is no or only negligible spreading error (SE) present. Additional FMO and gating controls are shown in Online Figure 9.

Table 2.

Reagents used for OMIP-XX

Specificity Alternative Name Clone Fluorochrome Purpose
CD45RA Isoform of CD45 HI100 Spark UV 387 Phenotyping of T cells, naive vs memory
CD40 TNFRSF5 5C3 BUV395 Phenotyping of B cells and DCs, activation
CD3 Part of the TCR complex UCHT1 BUV496 Lineage marker of pan T cells
CD56 Neural cell adhesion molecule 1, NCAM1 NCAM16.1 BUV563 Lineage marker of pan NK cells
CD2 LFA-2 S5.5 Qdot 605 Identification of ILCs, NK cell phenotyping
CD141 BDCA-3, or Thrombomodulin 1A4 BUV615 Lineage marker of cDC1s
Nkp46 CD335, Natural cytotoxicity triggering 9E2 Qdot 625 Phenotyping of NK cells
CD303 Clec4c V24-785 BUV661 Lineage marker of pDCs
CD86 B7-2 FUN-1 BUV737 Phenotyping of DCs and monocytes, activation
CD45 Protein tyrosine phosphatase, receptor HI30 BUV805 Pan-Haematopoietic marker
CD161 Killer cell lectin-like receptor subfamily B 1, DX12 BV421 Phenotyping of T cells and NK cells, MAIT marker
IgM Immunoglobulin M SA-DA4 Super Bright 436 Phenotyping of B cells and DCs
CD1c N.A L161 Pacific Blue Phenotyping of DCs and monocytes
CD28 N.A CD28.2 BV480 Phenotyping of T cells
CD19 N.A SJ25C1 AmCyan Lineage marker of B cells
CD11c Integrin alpha X, ITGAX S-HCL-3 BV510 Lineage marker for conventional DCs
CD45RO Isoform of CD45 UCHL1 BV570 Phenotyping of T cells, naive vs memory
CD197 CCR7, chemokine receptor 7 G043H7 BV605 Phenotyping of T cells, naive vs memory
CD69 N.A FN50 BV650 Phenotyping of T cells, tissue residency marker
FcER1 high-affinity IgE receptor AER-37 BV711 Lineage marker for cDC2s and basophils
CD103 Integrin alpha E, ITGAE Ber-ACT8 BV750 Phenotyping of T cells, tissue residency marker
CD194 CCR4, chemokine receptor 4 1G1 BV786 Phenotyping of T cells, migration
CD25 Interleukin-2 receptor alpha chain, IL2RA BC96 BB515 Phenotyping of T cells, marker for Tregs
CD14 Lipopolysaccharide receptor M5E2 RB545 Lineage marker for monocytes
CD8 N.A OKT8 NovaFluor Blue 555 Lineage marker for CD8+ T cells
CD4 N.A SK3 NovaFluor Blue 585 Lineage marker for CD4+ T cells
CD272 B- and T-lymphocyte attenuator, BTLA J168-540 RB613 Phenotyping of T cells, activation
CD27 TNFRSF7 M-T271 BB660 Phenotyping of T cells, activation
CD11b Integrin alpha M, ITGAM M1/70 PerCP Phenotyping of APCs
IgG Immunoglobulin G G18-145 BB700 Phenotyping of B cells, differentiation
TCRgd Gamma delta T cell receptor B1 RB705 Lineage marker of gd T cells
CD127 Interleukin-7 receptor subunit alpha, IL7RA HIL7R- RB744 Phenotyping of T cells, Treg identification
TIGIT T cell immunoreceptor with Ig and ITIM TgMab-2 RB780 Phenotyping of T cells and NK cells
IgD Immunoglobulin D W18340F PerCP-Fire 806 Phenotyping of B cells, differentiation
CD57 Human natural killer-1, HNK-1 NK-1 PE Phenotyping of T cells and NK cells
CD20 N.A 2H7 Spark YG 593 Lineage marker of B cells
CD24 Signal transducer CD24 SN3 PE-Alexa Fluor 610 Phenotyping of B cells, differentiation
CD183 CXCR3, CX chemokine receptor 3 G025H7 PE-Fire 640 Phenotyping of T cells, migration
CD123 Interleukin-3 receptor, IL3RA 9F5 PE-Cy5 Lineage marker of Basophils and pDCs
CD278 Inducible T-cell costimulator, ICOS ISA-3 PE-Cy5.5 Phenotyping of T cells, activation
CD16 Fc gamma receptor, FcγRIII 3G8 PE-Alexa Fluor 700 Lineage marker of monocytes, phenotyping of NK
CD279 PD-1, Programmed Death 1 EH12.1 PE-Cy7 Phenotyping of T cells, activation and exhaustion
HLA-DR MHC class II L243 PE-Fire 810 MHC class II
Va24-JA18 V alpha 24 – J alpha 18 TCR chain 6B11 APC Marker for invariant NKT cells
Va7.2 V alpha 7.2 TCR chain 3C10 Alexa Fluor 647 Marker for MAIT cells
KLRG1 Killer cell lectin-like receptor subfamily G SA231A2 Spark NIR 685 Phenotyping of T cells and NK cells, activation
CD39 Ectonucleoside triphosphate A1 R718 Phenotyping of T cells and NK cells, activation
Live/Dead N.A Amine Zombie-NIR Live/Dead cell discrimination
CD163 Scavenger receptor for hemoglobin GHI/61 APC-Cy7 DC phenotyping marker, marker for DC3s
CD38 cyclic ADP ribose hydrolase HB-7 APC-Fire 810 Phenotyping of T cells and NK cells, activation

B cells are identified by the lineage markers CD19 and CD20 (Figure 1A). Basic B cell differentiation status can be assessed using expression of the immunoglobulin subclasses IgM, IgD, IgG and the sialoglycoprotein CD24 (Figure 1C), as well as CD27 and CD38 (11).

Pan T cells are identified by expression of CD3 (Figure 1A), followed by subsetting γδ T cells from the larger fraction of αβ T cells. MAIT cells are gated using CD161 and the invariant TCR alpha chain, TCR Vα7.2 (Figure 1D). Invariant NK-T cells can be identified using an antibody against the Vα24 and Jα18 of the TCR alpha locus. For conventional αβ T cells, after gating CD4+ and CD8+ T cells, Tregs can be identified by high expression of the IL-2 receptor alpha chain (CD25) and low expression of the IL-7 receptor (CD127) (Figure 1D). In the CD4+ Th cell and CD8+ cytotoxic T cell fraction, naïve and memory subsets can be identified by the differential expression of CD45RA, CD45RO and the chemokine receptor CCR7 (CD197) (12). Furthermore, tissue-resident memory T cells (TRMs), can be identified by the expression of CD69 and the integrin CD103 (13) (Figure 1E). CD69 also functions as a marker of recent T cell activation.

For detailed assessment of Treg function and phenotype, the ectonucleoside triphosphate diphosphohydrolase CD39 and inducible costimulator of T cells (ICOS) can be used, together with the chemokine receptors CXCR3 and CCR4 (Figure 1F) (14).

Both CD4+ helper T cells and CD8+ cytotoxic T cells can differentiate into different effector and memory lineages, but they can also enter an exhausted state during chronic infections or cancer development (15). In our panel, several phenotyping markers allow us to assess the functional state of T cells in depth: the exhaustion and activation marker Programmed Death 1 (PD-1, CD279), the senescence marker CD57, the co-inhibitory receptor BTLA (CD274), the co-receptors CD27 and CD28, the cyclic ADP ribose hydrolase CD38, TIGIT (T cell immunoreceptor with Ig and ITIM domains) and killer cell lectin-like receptor subfamily G member 1 (KLRG1). Figure 1G depicts well-defined separation for all these markers across multiple T cell subsets, and NK cells as a reference population. Furthermore, for CD4+ Th cells the two main effector lineages can be distinguished using the expression of the chemokine receptors CXCR3 (mostly expressed on Th1 cells) and CCR4 (mostly expressed on Th2 cells). These chemokine receptors can also be used to study different functional capacity and homing properties of CD8+ cytotoxic T cells and Tregs (Figure 1F).

NK cells are gated using the lineage-defining molecules CD56 (NCAM) and CD161 (KLRB1) and can be subsetted by the expression of CD16 (Figure 1H). Nkp46 functions as an additional NK cell marker that is suitable for studying tissue-derived NK cells (16).

In the myeloid cell compartment, CD14 (the LPS receptor) and CD16 (FcγRIII) are used traditionally to distinguish classical (CD14+ CD16+), intermediate (CD14+ CD16dim) and non-classical (CD14CD16+) monocytes (17) (Figure 1A). FcER1 together with the IL-3 receptor alpha chain (CD123) are commonly used markers for basophils. Within the Lin-(CD3CD19CD56CD14CD16) HLA-DR+ compartment, our panel identifies plasmacytoid dendritic cells (pDCs) by the expression of CD303, while classical DCs (cDCs) are marked as CD11c+ HLA-DR+. The cross-presenting cDC1 and the cDC2 subsets are identified by CD141 (also known as BDCA-3) and CD1c (BDCA-1) expression (18), respectively, with FcER1 functioning as an additional and more distinct marker for the cDC2 lineage (9) (Figure 1I). DC activation status can be assessed by the expression of the co-receptors CD40, CD86 and the Integrin alpha M (CD11b). Furthermore, CD163 allows the separation of the recently defined DC3 subset (8,19) and to phenotype monocytes (Figure 1J). Finally, ILCs can be identified by co-expression of CD127 and CD2 (Figure 1K).

A representative gating tree with all the above-mentioned subsets is shown in Figure 1, including some fluorescence-minus-one (FMO) controls: on CD8+ T cells for the molecules PD-1, BTLA; on CD25+ CD127 Tregs for ICOS, CCR4; and on pan CD11c+ MHCII+ cDCs for CD86 and CD1c (Figure 1L). All of these FMOs highlight that by using systematic panel design there is negligible spreading error (SE) (20) for these populations and markers of high interest.

Our panel development strategy was based on established best practices (21-23) and multiple novel approaches. First, we utilized the similarity index (Online Figure 3), the fluorochrome brightness (Online Figure 4) and a newly developed automated algorithm for systematic fluorochrome selection. While previously described metrics such as the complexity index (24) assign an overall “score” to a given set of fluorochromes, our strategy allowed us to identify the best feasible combinations of fluorochromes to move beyond 40 colors. Second, we developed a new metric to evaluate unmixing-dependent spreading error that occurs in highly complex spectral flow cytometry panels and affects all events in the measurement (Online Figure 5, and Mage and Mair, manuscript in preparation). Finally, we utilized the instrument-specific spillover-spreading matrix (SSM) (20) and the total spread matrix (TSM) (Corselli et al, manuscript in preparation) for the optimal assignment of fluorochromes based on the biological co-expression of markers (Online Figure 6).

All steps of this panel design process, including the novel approaches, are described in detail in the online material of this manuscript, including additional staining and gating controls (Online Figure 9). Of note, this panel was developed on two full spectrum cytometers in parallel: a 7-laser instrument with a total of 186 detectors (commercially available from Sony Biotechnology as the Sony ID7000) and a 5-laser instrument with a total of 78 detectors (commercially available from BD Biosciences as the BD FACSDiscover S8). The final and fully optimized panel as shown in Figure 1 was acquired on the BD FACSDiscover S8 (instrument configuration and setup details are listed in Online Figure 1 and 2), together with the FMO control samples. This instrument also allowed cell sorting, highlighting that 50-parameter sorting is feasible to allow very fine-grained isolation of any immune population of interest (sorting strategy and purity of the sorted populations shown in Online Figure 14). Furthermore, we established a trimmed-down panel version of 45 colors that is cross-platform compatible on the BD FACSDiscover S8 and Sony ID7000 (Online Figure 12). To the best of our knowledge, this is the first report of a high-dimensional 40-color+ panel that is usable across multiple independent spectral cytometry platforms.

Manual analysis is critical to assess data quality and separation of markers in any flow cytometry panel (25), but in many cases additional insights can be gleaned from high-dimensional panels by using exploratory computational tools such as dimensionality reduction and clustering (26-28). In Online Figure 15 we show a UMAP plot with overlaid FlowSOM (29) clusters, heatmaps of the annotated clusters and histogram overlays of the raw data for some markers, highlighting the fine-grained cellular heterogeneity that can be revealed using our 50-color panel.

Overall, our data shows that this panel can serve as a widely usable and powerful immunophenotyping resource for comprehensive analysis of human immune cells in peripheral blood and non-lymphoid tissues, including human tissues of small size such as biopsies or some resected tissues (3,5) .

The opportunity to reliably analyze 50 different target molecules (with the option to perform parallel cell sorting) in a high-throughput fashion is likely to enable previously impossible avenues to study the human immune system (30). Finally, development of such a comprehensive panel may enable the consistent use of a single panel suitable for multiple different studies. Together with deposition of these data into publicly accessible databases, such a consistent use would facilitate subsequent cross-study analyses with machine learning approaches such as FAUST (31,32) or other suitable computational techniques.

Similarity to published OMIPs:

The most similar OMIP to our manuscript is OMIP-069 (the first 40-color OMIP to be reported, (24)) and OMIP-044 (the first 28-color OMIP reported (33)). There is some overlap with published 28-color OMIPs focusing on T cell phenotyping (e.g. OMIP-050 and OMIP-058 (34)) and several other lower dimensional OMIPs focused on T cells, but up to date there is no OMIP that reports the use of 50 different fluorochromes allowing such in-depth phenotyping of T cells and antigen-presenting cells (APCs).

Supplementary Material

Supplement 1

Table 1.

Summary table for application of OMIP XX

Purpose 50-color phenotyping of antigen-presenting cells and T cells
Species Human
Cell Types PBMCs and human tissue
Cross-References OMIP-069, OMIP-044, OMIP-050, OMIP-058 and others

Acknowledgements:

This research was supported by the Flow Cytometry Shared Resource (RRID:SCR_022613) of the Fred Hutch/University of Washington/Seattle Children’s Cancer Consortium (P30 CA015704). We would like to thank Michele Black for her state-of-the-art flow cytometry expertise and exemplary leadership in running the Flow Cytometry Shared Resource. We thank Xiaoshan “Shirley” Shi (BD Biosciences) for assistance with equipment, and Jolene Bradford (Thermo Fisher) for support with testing reagents. This work was supported by the Emerson Collective and NIH grants R01 AI123323 and R56 DE032009 (to Martin Prlic). A.J.T. is an ISAC Marylou Scholar. P.M. is an ISAC International Innovator. F.M. is a former ISAC Marylou Scholar.

Footnotes

Conflict of Interest:

Peter Mage and Aaron J. Tyznik are employees of BD Biosciences, the manufacturer of the BD FACSDiscover S8. The other authors declare no conflict of interest.

References:

  • 1.Meizlish ML, Franklin RA, Zhou X, Medzhitov R. Tissue Homeostasis and Inflammation. Annu Rev Immunol 2021;39:557–581. Available at: https://pubmed.ncbi.nlm.nih.gov/33651964/. [DOI] [PubMed] [Google Scholar]
  • 2.Mellman I, Chen DS, Powles T, Turley SJ. The cancer-immunity cycle: Indication, genotype, and immunotype. Immunity 2023;56:2188–2205. Available at: https://pubmed.ncbi.nlm.nih.gov/37820582/. Accessed November 14, 2023. [DOI] [PubMed] [Google Scholar]
  • 3.Farber DL. Tissues, not blood, are where immune cells function. Nature 2021 593:7860 2021;593:506–509. Available at: https://www.nature.com/articles/d41586-021-01396-y. Accessed September 1, 2023. [DOI] [PubMed] [Google Scholar]
  • 4.Woodward Davis AS, Roozen HN, Dufort MJ, DeBerg HA, Delaney MA, Mair F, Erickson JR, Slichter CK, Berkson JD, Klock AM, Mack M, Lwo Y, Ko A, Brand RM, McGowan I, Linsley PS, Dixon DR, Prlic M. The human tissue-resident CCR5+ T cell compartment maintains protective and functional properties during inflammation. Sci Transl Med 2019;11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Mair F, Erickson JR, Frutoso M, Konecny AJ, Greene E, Voillet V, Maurice NJ, Rongvaux A, Dixon D, Barber B, Gottardo R, Prlic M. Extricating human tumour immune alterations from tissue inflammation. Nature 2022;605:728–735. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Yin X, Chen S, Eisenbarth SC. Dendritic Cell Regulation of T Helper Cells. Annu Rev Immunol 2021;39:759–790. Available at: https://pubmed.ncbi.nlm.nih.gov/33710920/. Accessed September 1, 2023. [DOI] [PubMed] [Google Scholar]
  • 7.Wculek SK, Cueto FJ, Mujal AM, Melero I, Krummel MF, Sancho D. Dendritic cells in cancer immunology and immunotherapy. Nat Rev Immunol 2020;20:7–24. [DOI] [PubMed] [Google Scholar]
  • 8.Cabeza-Cabrerizo M, Cardoso A, Minutti CM, Pereira Da Costa M, Reis E Sousa C. Dendritic Cells Revisited. Annu Rev Immunol 2021;39:131–166. [DOI] [PubMed] [Google Scholar]
  • 9.Dutertre CA, Becht E, Irac SE, Khalilnezhad A, Narang V, Khalilnezhad S, Ng PY, van den Hoogen LL, Leong JY, Lee B, Chevrier M, Zhang XM, Yong PJA, Koh G, Lum J, Howland SW, Mok E, Chen J, Larbi A, Tan HKK, Lim TKH, Karagianni P, Tzioufas AG, Malleret B, Brody J, Albani S, van Roon J, Radstake T, Newell EW, Ginhoux F. Single-Cell Analysis of Human Mononuclear Phagocytes Reveals Subset-Defining Markers and Identifies Circulating Inflammatory Dendritic Cells. Immunity 2019;51:573–589.e8. [DOI] [PubMed] [Google Scholar]
  • 10.Godfrey DI, Uldrich AP, Mccluskey J, Rossjohn J, Moody DB. The burgeoning family of unconventional T cells. Nature Immunology 2015 16:11 2015;16:1114–1123. Available at: https://www.nature.com/articles/ni.3298. Accessed November 14, 2023. [DOI] [PubMed] [Google Scholar]
  • 11.Weisel F, Shlomchik M. Memory B Cells of Mice and Humans. Annu Rev Immunol 2017;35:255–284. Available at: https://pubmed.ncbi.nlm.nih.gov/28142324/. Accessed November 14, 2023. [DOI] [PubMed] [Google Scholar]
  • 12.Sallusto F, Lenig D, Förster R, Lipp M, Lanzavecchia A. Two subsets of memory T lymphocytes with distinct homing potentials and effector functions. Nature 1999;401:708–712. Available at: https://pubmed.ncbi.nlm.nih.gov/10537110/. Accessed November 7, 2023. [DOI] [PubMed] [Google Scholar]
  • 13.Gray JI, Farber DL. Tissue-Resident Immune Cells in Humans. 10.1146/annurev-immunol-093019-112809 2022;40:195–220. Available at: https://www.annualreviews.org/doi/abs/10.1146/annurev-immunol-093019-112809. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Sakaguchi S, Mikami N, Wing JB, Tanaka A, Ichiyama K, Ohkura N. Regulatory T Cells and Human Disease. Annu Rev Immunol 2020;38:541–566. Available at: https://pubmed.ncbi.nlm.nih.gov/32017635/. Accessed November 14, 2023. [DOI] [PubMed] [Google Scholar]
  • 15.Blank CU, Haining WN, Held W, Hogan PG, Kallies A, Lugli E, Lynn RC, Philip M, Rao A, Restifo NP, Schietinger A, Schumacher TN, Schwartzberg PL, Sharpe AH, Speiser DE, Wherry EJ, Youngblood BA, Zehn D. Defining ‘T cell exhaustion.’ Nature Reviews Immunology 2019 19:11 2019;19:665–674. Available at: https://www.nature.com/articles/s41577-019-0221-9. Accessed November 14, 2023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Frutoso M, Mair F, Prlic M. OMIP-070: NKp46-Based 27-Color Phenotyping to Define Natural Killer Cells Isolated From Human Tumor Tissues. Cytometry Part A 2020;97:1052–1056. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Mair F, Liechti T. Comprehensive Phenotyping of Human Dendritic Cells and Monocytes. Cytometry Part A 2021;99:231–242. [DOI] [PubMed] [Google Scholar]
  • 18.Dzionek A, Fuchs A, Schmidt P, Cremer S, Zysk M, Miltenyi S, Buck DW, Schmitz J. BDCA-2, BDCA-3, and BDCA-4: three markers for distinct subsets of dendritic cells in human peripheral blood. J Immunol 2000;165:6037–6046. Available at: https://pubmed.ncbi.nlm.nih.gov/11086035/. Accessed November 14, 2023. [DOI] [PubMed] [Google Scholar]
  • 19.Dutertre CA, Becht E, Irac SE, Khalilnezhad A, Narang V, Khalilnezhad S, Ng PY, van den Hoogen LL, Leong JY, Lee B, Chevrier M, Zhang XM, Yong PJA, Koh G, Lum J, Howland SW, Mok E, Chen J, Larbi A, Tan HKK, Lim TKH, Karagianni P, Tzioufas AG, Malleret B, Brody J, Albani S, van Roon J, Radstake T, Newell EW, Ginhoux F. Single-Cell Analysis of Human Mononuclear Phagocytes Reveals Subset-Defining Markers and Identifies Circulating Inflammatory Dendritic Cells. Immunity 2019;51:573–589.e8. [DOI] [PubMed] [Google Scholar]
  • 20.Nguyen R, Perfetto S, Mahnke YD, Chattopadhyay P, Roederer M. Quantifying spillover spreading for comparing instrument performance and aiding in multicolor panel design. Cytometry A 2013;83:306–315. Available at: https://pubmed.ncbi.nlm.nih.gov/23389989/. Accessed November 14, 2023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Mair F, Tyznik AJ. High-Dimensional Immunophenotyping with Fluorescence-Based Cytometry: A Practical Guidebook. Methods Mol Biol 2019;2032:1–29. Available at: https://pubmed.ncbi.nlm.nih.gov/31522410/. Accessed December 4, 2023. [DOI] [PubMed] [Google Scholar]
  • 22.Cossarizza A, Chang HD, Radbruch A, Abrignani S, Addo R, Akdis M, Andrä I, Andreata F, Annunziato F, Arranz E, Bacher P, Bari S, Barnaba V, Barros-Martins J, Baumjohann D, Beccaria CG, Bernardo D, Boardman DA, Borger J, Böttcher C, Brockmann L, Burns M, Busch DH, Cameron G, Cammarata I, Cassotta A, Chang Y, Chirdo FG, Christakou E, Čičin-Šain L, Cook L, Corbett AJ, Cornelis R, Cosmi L, Davey MS, De Biasi S, De Simone G, del Zotto G, Delacher M, Di Rosa F, Di Santo J, Diefenbach A, Dong J, Dörner T, Dress RJ, Dutertre CA, Eckle SBG, Eede P, Evrard M, Falk CS, Feuerer M, Fillatreau S, Fiz-Lopez A, Follo M, Foulds GA, Fröbel J, Gagliani N, Galletti G, Gangaev A, Garbi N, Garrote JA, Geginat J, Gherardin NA, Gibellini L, Ginhoux F, Godfrey DI, Gruarin P, Haftmann C, Hansmann L, Harpur CM, Hayday AC, Heine G, Hernández DC, Herrmann M, Hoelsken O, Huang Q, Huber S, Huber JE, Huehn J, Hundemer M, Hwang WYK, Iannacone M, Ivison SM, Jäck HM, Jani PK, Keller B, Kessler N, Ketelaars S, Knop L, Knopf J, Koay HF, Kobow K, Kriegsmann K, Kristyanto H, Krueger A, Kuehne JF, Kunze-Schumacher H, Kvistborg P, et al. Guidelines for the use of flow cytometry and cell sorting in immunological studies (third edition). Eur J Immunol 2021;51:2708–3145. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Ferrer-Font L, Small SJ, Lewer B, Pilkington KR, Johnston LK, Park LM, Lannigan J, Jaimes MC, Price KM. Panel Optimization for High-Dimensional Immunophenotyping Assays Using Full-Spectrum Flow Cytometry. Curr Protoc 2021;1. Available at: https://pubmed.ncbi.nlm.nih.gov/34492732/. Accessed December 4, 2023. [DOI] [PubMed] [Google Scholar]
  • 24.Park LM, Lannigan J, Jaimes MC. OMIP-069: Forty-Color Full Spectrum Flow Cytometry Panel for Deep Immunophenotyping of Major Cell Subsets in Human Peripheral Blood. Cytometry A 2020;97:1044–1051. Available at: https://pubmed.ncbi.nlm.nih.gov/32830910/. Accessed November 7, 2023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Liechti T, Weber LM, Ashhurst TM, Stanley N, Prlic M, Van Gassen S, Mair F. An updated guide for the perplexed: cytometry in the high-dimensional era. Nat Immunol 2021;22:1190–1197. [DOI] [PubMed] [Google Scholar]
  • 26.Mair F. Gate to the Future: Computational Analysis of Immunophenotyping Data. Cytometry Part A 2019;95:147–149. [DOI] [PubMed] [Google Scholar]
  • 27.Mair F, Hartmann FJ, Mrdjen D, Tosevski V, Krieg C, Becher B. The end of gating? An introduction to automated analysis of high dimensional cytometry data. Eur J Immunol 2016;46:34–43. [DOI] [PubMed] [Google Scholar]
  • 28.Saeys Y, Van Gassen S, Lambrecht BN. Computational flow cytometry: Helping to make sense of high-dimensional immunology data. Nat Rev Immunol 2016;16:449–462. [DOI] [PubMed] [Google Scholar]
  • 29.Van Gassen S, Callebaut B, Van Helden MJ, Lambrecht BN, Demeester P, Dhaene T, Saeys Y. FlowSOM: Using self-organizing maps for visualization and interpretation of cytometry data. Cytometry A 2015;87:636–645. Available at: https://pubmed.ncbi.nlm.nih.gov/25573116/. [DOI] [PubMed] [Google Scholar]
  • 30.Ginhoux F, Yalin A, Dutertre CA, Amit I. Single-cell immunology: Past, present, and future. Immunity 2022;55:393–404. Available at: https://pubmed.ncbi.nlm.nih.gov/35263567/. Accessed September 1, 2023. [DOI] [PubMed] [Google Scholar]
  • 31.Greene E, Finak G, D’Amico LA, Bhardwaj N, Church CD, Morishima C, Ramchurren N, Taube JM, Nghiem PT, Cheever MA, Fling SP, Gottardo R. New interpretable machine-learning method for single-cell data reveals correlates of clinical response to cancer immunotherapy. Patterns (N Y) 2021;2. Available at: https://pubmed.ncbi.nlm.nih.gov/34950900/. Accessed December 13, 2023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Zearfoss R. Tumor biology gets smart. Cell 2022;185:2611–2612. Available at: https://pubmed.ncbi.nlm.nih.gov/35868262/. Accessed December 13, 2023. [DOI] [PubMed] [Google Scholar]
  • 33.Mair F, Prlic M. OMIP-044: 28-color immunophenotyping of the human dendritic cell compartment. Cytometry Part A 2018;93:402–405. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Liechti T, Roederer M. OMIP-058: 30-Parameter Flow Cytometry Panel to Characterize iNKT, NK, Unconventional and Conventional T Cells. Cytometry A 2019;95:946–951. Available at: https://pubmed.ncbi.nlm.nih.gov/31334918/. Accessed November 14, 2023. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Supplement 1

Articles from bioRxiv are provided here courtesy of Cold Spring Harbor Laboratory Preprints

RESOURCES