1
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      OMIP 076: High‐dimensional immunophenotyping of murine T‐cell, B‐cell, and antibody secreting cell subsets

      research-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          1 PURPOSE AND APPROPRIATE SAMPLE TYPES This 19‐parameter, 18‐color flow cytometry panel was designed and optimized to enable the comprehensive and simultaneous immunophenotyping of distinct T‐cell, B‐cell, and antibody secreting cell (ASC) subsets within murine tissues (Table 1). Cellular populations identified by using this OMIP include two major subsets of B‐cells (memory and activated), two ASC subsets (plasma cells and plasmablasts), and seven major subsets of CD4+ T‐cells (naïve, central memory, effector memory, helper, regulatory, follicular helper, and follicular regulatory). Staining was performed on freshly isolated splenocytes from 21‐day‐old BALB/c mice, however, due to the omission of mouse strain‐specific markers, this OMIP can be implemented across a range of murine models where in‐depth immunophenotyping of the diverse repertoire of T‐cell, B‐cell, and ASC populations is required. TABLE 1 Summary table Purpose Comprehensive immunophenotyping of T‐cell, B‐cell, and ASC subsets Species Mouse Cell types Murine tissues containing lymphocyte populations Cross‐reference OMIP‐031, OMIP‐032, OMIP‐054, OMIP‐061 2 BACKGROUND There is now considerable evidence demonstrating that both prenatal and postnatal exposure to particular classes of microbial stimuli can provide beneficial signals during early life immune development, resulting in the protection against future inflammatory disease [1, 2, 3]. The principal target of this beneficial immunostimulation appears to be the innate immune system [4, 5], and the mechanisms driving protection underlay the paradigm of innate immune training, whereby certain classes of microbial stimuli can alter the functional state of innate immune cells, leading to the optimization of immunocompetence [6]. Immune training focuses on the phenotypic and transcriptional profiles of several prototypical innate populations [6, 7], however, the characterization of downstream adaptive responses associated with protection via innate immune training are of critical importance for understanding disease pathogenesis, and the potential for therapeutic mitigation. Due to this gap in our current understanding, the broader protective mechanisms remain incompletely understood. To address this requirement, we have developed and optimized a novel 19‐parameter flow cytometry panel to comprehensively and simultaneously characterize distinct T‐cell, B‐cell, and ASC subsets localized within tissues of BALB/c mice in response to immune training during early life. The developmental phase of this flow cytometry panel involved the prioritization of T‐cell, B‐cell, and ASC subsets central to the maintenance of immunological homeostasis, as based on the current literature and forerunner studies. As such, a degree of emphasis was placed on effector, regulatory, and memory subsets within T‐cell and B‐cell populations. In regard to T‐cells, the conversion of peripheral naïve CD4+ T‐cells to effector T (Teff) cells is denoted by upregulation of the activation marker CD25, while concomitant upregulation of both CD25 and intracellular Foxp3 expression is essential for the peripheral induction of regulatory T‐cells (Treg) [8], a process previously recognized in the protection against allergic airways inflammation following microbial‐derived immunomodulation [9, 10]. Furthermore, the expression of CD44 on Treg has been implicated in promoting enhanced function [11, 12], while inducible costimulator (ICOS)+ Tregs are recognized to have superior suppressive capacity and interleukin (IL)‐10 production compared to ICOS− Tregs [13, 14]. Following activation and contraction, CD4+ T‐cells transition toward a memory phenotype via the gradual upregulation of CD44 expression in parallel with transient expression of CD62L, driving the establishment of a dynamic repository of central memory (TCM) and effector memory (TEM) T‐cells [15, 16, 17]. In addition to establishing peripheral memory, activated CD4+ T‐cells have the capacity to upregulate extracellular expression of CXCR5, ICOS, and programmed cell death protein 1 (PD‐1) [18, 19], resulting in the generation of a highly specialized population of T follicular helper (TFH) cells required for the formation of germinal centers within secondary lymphoid organs, while also providing crucial survival signals to support high‐affinity B‐cells during affinity maturation and proliferation [20, 21]. A separate subset of thymic‐derived cells that share homology with the TFH phenotype in addition to Foxp3 and bimodal CD25 expression, termed follicular regulatory T (TFR) cells, have also been identified, however, this subset has been attributed to the inhibition of TFH activity and subsequent generation of humoral immunity [22, 23]. The immunophenotypic characterization of B‐cell and ASC subsets for this OMIP was centered around the classic expression of CD19 and B220. To maximize the capacity of a 5‐laser BD LSRFortessa™, CD19 (B‐cell and ASC subsets) and CD4 (T‐cell) antibodies were conjugated to the same fluorochrome, since co‐expression is essentially absent in single‐cell analysis. Within secondary lymphoid tissues, the antigen‐specific activation of B‐cells involves the constitutive upregulation of major histocompatibility complex class‐II (MHC class II; mouse I‐A/I‐E) and CD80 expression, in conjunction with the membrane‐bound expression of both immunoglobulin (Ig) M and IgD [24, 25, 26]. Following antigen‐specific activation, B‐cells upregulate Synd‐1 expression and differentiate into the two major classes of ASC; the rapidly produced and short‐lived plasmablasts and the short‐lived peripheral plasma cells, both of which have the capacity to secrete IgM [27, 28, 29, 30]. A major difference between these two antibody‐secreting subsets, however, is the absence of classic mature B‐cell markers CD19, B220, and MHC‐II on plasma cells [28, 31]. The eventual transition of B‐cells toward a memory phenotype results in the loss of Synd‐1 expression with parallel upregulation of programmed cell death protein 1 ligand 2 (PD‐L2), generating a long‐lived secondary lymphoid population expressing IgM +/− IgD that can rapidly differentiate into ASC upon re‐stimulation [32, 33, 34, 35, 36]. Panel optimization was performed on a BD LSRFortessa™, with all fluorochrome‐conjugated antibodies (Table 2) titrated during the optimization phase (Figure S1). Prior to multicolor extracellular staining, splenocytes were incubated in Fc Block™ (Purified recombinant CD16/32) to inhibit non‐antigen‐specific binding of fluorochrome‐conjugated antibodies to the nonpolymorphic epitope of FcγIII (CD16) and FcγII (CD32) receptors expressed on multiple myeloid populations and B‐cells. A representative gating strategy to delineate the T‐cell, B‐cell, and ASC subsets described above is detailed in Figure 1. Briefly, splenocytes were first gated on side‐scatter (SSC) and forward‐scatter (FSC) parameters (Figure 1A) to remove sample debris, followed by single‐cell gating (Figure 1B) to remove doublets. Gating was then performed on viable CD45+ cells (Figure 1C) to remove dead/dying cells and stromal cells from the analysis. The primary T‐cell/B‐cell/ASC separation involved delineation of TCRβ and CD4/CD19 expression (Figure 1D). Double positive cells were classified as CD4+ T‐cells, as CD19+ B‐cells and ASC subsets will be present within the TCRβ− population (Figure 1D) due to the absence of TCRβ/CD19 co‐expression (Figure S2A). An additional TCRβ−CD4/CD19− gate was included to enable the characterization of B220−Synd‐1+MHC class II−IgM+ plasma cells (PC; Figure 1E). CD19+ B‐cells and ASC subsets were then defined as B220lo/+Synd‐1+MHC class II+IgM+ plasmablasts (PB; Figure 1F), B220+Synd‐1−CD80+PD‐L2−MHC class II+IgM+IgD+ activated B‐cells (Figure 1G) and B220+Synd‐1−CD80+PD‐L2+IgM+IgD+/− memory B‐cells (Figure 1H). CD4+ T‐cells were defined as CD62L+CD44lo/− naïve T‐cells (Figure 1I), CD62L+CD44hi TCM (Figure 1I), CD62L−CD44hi TEM (Figure 1I), CD25+Foxp3− Teff (Figure 1J), CD25+Foxp3+ Treg (Figure 1J) ICOS+CD44+ Treg (Figure 1K), CXCR5+ICOS+PD‐1+ TFH (Figure 1L), and CXCR5+ICOS+PD‐1+CD25+/‐Foxp3+ TFR (Figure 1M). TABLE 2 Reagents used for OMIP Specificity Fluorochrome Clone Purpose PD‐L2 (CD273) BUV395 TY25 Memory B‐cells IgD BUV496 AMS 9.1 Activated/memory B‐cells CD44 BUV737 IM7 T‐cell subsets ICOS (CD278) BV421 7E.17G9 T Follicular helper/Treg PD‐1 (CD279) BV480 J43 T Follicular helper cells Live/Dead FVS575 N/A Viable cells CD80 BV650 16‐10A1 Activated B‐cells IgM BV711 R6.60.2 B‐cell/ASC subsets CD4 BV786 RM4‐5 CD4+ T‐cells CD19 BV786 1D3 B‐cell subsets Synd‐1 (CD138) BB515 281‐2 Plasmablasts/Plasma cells TCRβ BB700 H57‐597 Pan T‐cells Foxp3 PE FJK‐16s Regulatory T‐cells B220 (CD45R) PE‐CF594 RA3‐6B2 B‐cell subsets CD25 PE‐Cy5 PC61 Activated T‐cells CXCR5 (CD185) PE‐Cy7 2G8 T Follicular helper cells MHC class II (I‐A/I‐E) AF647 M5/114.15.2 B‐cell subsets CD62L APC‐R700 MEL‐14 T‐cell subsets CD45 APC‐Cy7 30‐F11 Pan leukocyte FIGURE 1 Overview of 19‐parameter gating strategy developed for the characterization of T‐cell, B‐cell, and ASC subsets within freshly isolated splenocytes from 21‐day‐old BALB/c mice. 1 × 106 splenocytes were incubated in Fc Block™, followed by fixable viability stain (FVS) and a 17‐parameter extracellular antibody cocktail containing 10% brilliant stain buffer plus (BD biosciences). Intracellular staining was performed following fixation‐permeabilization of extracellular stained splenocytes. Data were acquired on a BD LSRFortessa™ (BD Biosciences). (A–C) Removal of cellular debris, doublets, nonviable cells and stromal cells. (D) Primary delineation of TCRβ−CD19+, TCRβ+CD4+, and TCRβ−CD4/CD19− cells. (E–M) Characterization of (E) plasma cells, (F) plasmablasts, (G) activated B‐cells, (H) memory B‐cells, (I) naïve, effector memory and central memory T‐cells, (J) effector and regulatory T‐cells, (K) ICOS+CD44+ Treg, (L) T follicular helper cells, and (M) follicular regulatory T‐cells. All plots are representative of individual samples. Manual gating was determined using fluorescence minus one (FMO) controls where necessary (Figure S4) [Color figure can be viewed at wileyonlinelibrary.com] To perform high‐dimensional analysis on 21‐day‐old naïve splenocytes, viable CD45+ cells (Figure 1C) underwent high‐resolution FlowSOM clustering to define cell populations, followed by metaclustering for visualization with Uniform Manifold Approximation and Projection (UMAP) [37] using the Cytometry Data Analysis Tool (CATALYST) pipeline [38, 39]. Primary unsupervised analysis was performed to identify CD4+ T‐cell and B‐cell/ASC clusters based on extracellular receptor co‐expression (Figure S3A). CD4+ T‐cell (Figure S3B), and B‐cell/ASC (Figure S3C) clusters were then isolated for secondary subset analysis. 3 SIMILARITIES TO OTHER OMIPS The OMIP described here shares a small degree of marker similarity (TCRβ, CD4, CD44, CD62L, PD‐1, CD19, B220) with OMIP‐031 [40], OMIP‐032 [41], and OMIP‐061 [42], which are focused on immunologic checkpoint expression on murine T‐cell subsets, the characterization of innate and adaptive populations within the murine mammary gland and murine antigen‐presenting cells, respectively. While both OMIP‐031 and OMIP‐032 characterize TCRβ+CD4+ effector and memory T‐cell subsets based on a combination of CD44 and/or CD62L expression, OMIP‐032 employs an additional CD19+ gate to delineate B‐cells. OMIP‐061 utilized B220 to identify B‐cells. A distinct difference between these OMIPs and the OMIP described here is that our panel was developed for the sole purpose of comprehensively immunophenotyping T‐cell, B‐cell, and ASC subsets simultaneously, and we therefore include an additional 12 markers to allow the characterization of two major B‐cell, two ASC and seven major T‐cell populations within a single sample. The OMIP described here also exhibits minor overlap with OMIP‐054 [43], however, our panel was developed to maximize the potential of a 5‐laser BD LSRFortessa™ in facilities without the capacity to perform mass cytometry. AUTHOR CONTRIBUTIONS Kyle Mincham: Conceptualization; data curation; formal analysis; funding acquisition; investigation; methodology; validation; visualization; writing‐original draft; writing‐review & editing. Jacob Young: Data curation; formal analysis; investigation; validation; writing‐original draft. Deborah Strickland: Conceptualization; formal analysis; funding acquisition; investigation; methodology; project administration; writing‐original draft; writing‐review & editing. CONFLICT OF INTEREST The authors declare no conflict of interest exists. Supporting information Table S1 Instrument Optical Configuration Table S2. Reagents used in the final OMIP Table S3. Reagents used Table S4. Extracellular multicolor antibody staining cocktail for 1x sample Figure S1. Titrations of each individual component used in the final OMIP. All antibodies were individually titrated on splenocytes from naïve 21‐day‐old BALB/c mice. Data are splenocytes pre‐gated to remove debris (SSC/FSC) and doublets. Figure S2. Absence of TCRβ and CD19 coexpression. (A) TCRβ BB700 and CD19 BV786 staining in the absence of sentinel CD4 BV786 staining (CD4 BV786 FMO), demonstrating the absence of TCRβ and CD19 coexpression. (B) TCRβ BB700 and CD4 BV786 staining in the absence of sentinel CD19 BV786 staining (CD19 BV786 FMO), demonstrating the presence of a minor population of CD4+ non‐T‐cells within 21‐day‐old spleens. Population proportions downstream of TCRβ−CD4+ gate = % of TCRβ−CD4+ cells. Data are splenocytes stained for FVS575 BV605, CD45 APC‐Cy7, TCRβ BB700 and CD19 BV786 or CD4 BV786. Figure S3. High‐dimensional analysis of CD45 + splenocytes. Dimensionality reduction and clustering by UMAP demonstrating (A) distribution of TCRβ, CD4/19 and B220 expression on viable CD45+ splenocytes, (B) CD4+ T‐cell and (C) B‐cell/ASC clusters. Dimensionality reduction and UMAP visualization was performed using 12,000 total splenocytes from 8 individual 21‐day‐old naïve BALB/c mice (1500 cells per sample). Figure S4. Fluorescence Minus One (FMO) controls. Data are splenocytes showing terminal population gates and intermediate gates where required for (A) Synd‐1 BB515, (B‐D) IgM BV711, (E) CD80 BV650, (F) PD‐L2 BUV395 (G) IgM BV711, (H) IgD BUV496, (I) CD44 BUV737, (J) CD62L APC‐R700, (K) CXCR5 PE‐Cy7 and (L) PD‐1 BV480 FMO controls. Figure S5. Titration data for CD3ε PerCP and CD3ε BB700 antibodies. Titrations of (A) CD3ε PerCP and (B) CD3ε BB700 antibodies not used in the final panel. Data are splenocytes pre‐gated to remove debris (SSC/FSC) and doublets. Figure S6. Initial panel staining with CD3ε. Splenocytes were initially stained with CD3ε BB700 at a dilution of 1:200 for the delineation of T‐cells, prior to replacement with TCRβ BB700 in the final iteration of the OMIP. Data are splenocytes from naïve 21‐day‐old BALB/c mice. Figure S7. Titration data for Foxp3 PE clone MF23 antibody. Titration of (A) Foxp3 PE clone MF23 antibody not used in the final panel and (B) Foxp3 PE clone FJK‐16 s antibody used in the final panel. Data are splenocytes pre‐gated to remove debris (SSC/FSC) and doublets. Figure S8. Initial panel staining with CD62L AF700. (A) Titration of CD62L AF700. (B) Poor discrimination of CD45+TCRβ+CD4+CD62L+CD44lo/− naïve, of CD45+TCRβ+CD4+CD62L+CD44hi central memory and of CD45+TCRβ+CD4+CD62L−CD44hi effector memory T‐cell subsets when using the AF700 fluorochrome. Data are splenocytes from naïve 21‐day‐old BALB/c mice. Figure S9. Compensation matrix. Based on data displayed in Figure 1. Acquisition‐defined compensation matrix was manually generated post‐acquisition. Figure S10. Comparison of CXCR5 expression on adolescent and adult splenocytes. Data are representative flow cytometry plots from 21‐day‐old and 20‐week‐old BALB/c mice demonstrating the age‐dependent expression of CXCR5 PE‐Cy7 against PD‐1 BV480 on CD4+ T‐cells. Figure S11. Initial panel staining with IgD BUV496 clone 217–170. (A) Titration of IgD BUV496 clone 217–170. (B) Suboptimal detection of IgD expression on CD45+TCRβ−CD19+B220+Synd‐1−CD80+PD‐L2−MHC class II+IgM+IgD+ activated and CD45+TCRβ−CD19+B220+Synd‐1−CD80+PD‐L2+IgM+IgD− memory B‐cell subsets. Data are splenocytes from naïve 21‐day‐old BALB/c mice. Click here for additional data file.

          Related collections

          Most cited references43

          • Record: found
          • Abstract: found
          • Article: not found

          Defining trained immunity and its role in health and disease

          Immune memory is a defining feature of the acquired immune system, but activation of the innate immune system can also result in enhanced responsiveness to subsequent triggers. This process has been termed ‘trained immunity’, a de facto innate immune memory. Research in the past decade has pointed to the broad benefits of trained immunity for host defence but has also suggested potentially detrimental outcomes in immune-mediated and chronic inflammatory diseases. Here we define ‘trained immunity’ as a biological process and discuss the innate stimuli and the epigenetic and metabolic reprogramming events that shape the induction of trained immunity.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Two subsets of memory T lymphocytes with distinct homing potentials and effector functions.

            Naive T lymphocytes travel to T-cell areas of secondary lymphoid organs in search of antigen presented by dendritic cells. Once activated, they proliferate vigorously, generating effector cells that can migrate to B-cell areas or to inflamed tissues. A fraction of primed T lymphocytes persists as circulating memory cells that can confer protection and give, upon secondary challenge, a qualitatively different and quantitatively enhanced response. The nature of the cells that mediate the different facets of immunological memory remains unresolved. Here we show that expression of CCR7, a chemokine receptor that controls homing to secondary lymphoid organs, divides human memory T cells into two functionally distinct subsets. CCR7- memory cells express receptors for migration to inflamed tissues and display immediate effector function. In contrast, CCR7+ memory cells express lymph-node homing receptors and lack immediate effector function, but efficiently stimulate dendritic cells and differentiate into CCR7- effector cells upon secondary stimulation. The CCR7+ and CCR7- T cells, which we have named central memory (TCM) and effector memory (TEM), differentiate in a step-wise fashion from naive T cells, persist for years after immunization and allow a division of labour in the memory response.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Conversion of Peripheral CD4+CD25− Naive T Cells to CD4+CD25+ Regulatory T Cells by TGF-β Induction of Transcription Factor Foxp3

              CD4+CD25+ regulatory T cells (Treg) are instrumental in the maintenance of immunological tolerance. One critical question is whether Treg can only be generated in the thymus or can differentiate from peripheral CD4+CD25− naive T cells. In this paper, we present novel evidence that conversion of naive peripheral CD4+CD25− T cells into anergic/suppressor cells that are CD25+, CD45RB−/low and intracellular CTLA-4+ can be achieved through costimulation with T cell receptors (TCRs) and transforming growth factor β (TGF-β). Although transcription factor Foxp3 has been shown recently to be associated with the development of Treg, the physiological inducers for Foxp3 gene expression remain a mystery. TGF-β induced Foxp3 gene expression in TCR-challenged CD4+CD25− naive T cells, which mediated their transition toward a regulatory T cell phenotype with potent immunosuppressive potential. These converted anergic/suppressor cells are not only unresponsive to TCR stimulation and produce neither T helper cell 1 nor T helper cell 2 cytokines but they also express TGF-β and inhibit normal T cell proliferation in vitro. More importantly, in an ovalbumin peptide TCR transgenic adoptive transfer model, TGF-β–converted transgenic CD4+CD25+ suppressor cells proliferated in response to immunization and inhibited antigen-specific naive CD4+ T cell expansion in vivo. Finally, in a murine asthma model, coadministration of these TGF-β–induced suppressor T cells prevented house dust mite–induced allergic pathogenesis in lungs.
                Bookmark

                Author and article information

                Contributors
                deb.strickland@telethonkids.org.au
                Journal
                Cytometry A
                Cytometry A
                10.1002/(ISSN)1552-4930
                CYTO
                Cytometry
                John Wiley & Sons, Inc. (Hoboken, USA )
                1552-4922
                1552-4930
                23 June 2021
                September 2021
                : 99
                : 9 ( doiID: 10.1002/cyto.a.v99.9 )
                : 888-892
                Affiliations
                [ 1 ] Telethon Kids Institute University of Western Australia Nedlands Western Australia Australia
                Author notes
                [*] [* ] Correspondence

                Deborah H. Strickland, Northern Entrance, Perth Children's Hospital, 15 Hospital Avenue, Nedlands, Western Australia, Australia.

                Email: deb.strickland@ 123456telethonkids.org.au

                Author information
                https://orcid.org/0000-0003-4419-7258
                Article
                CYTOA24474
                10.1002/cyto.a.24474
                9546025
                34159723
                b7983131-fc69-4de1-b464-8727ecf7f673
                © 2021 The Authors. Cytometry Part A published by Wiley Periodicals LLC on behalf of International Society for Advancement of Cytometry.

                This is an open access article under the terms of the http://creativecommons.org/licenses/by-nc-nd/4.0/ License, which permits use and distribution in any medium, provided the original work is properly cited, the use is non‐commercial and no modifications or adaptations are made.

                History
                : 14 March 2021
                : 05 August 2020
                : 31 May 2021
                Page count
                Figures: 1, Tables: 2, Pages: 5, Words: 4344
                Funding
                Funded by: Telethon Kids Institute
                Categories
                Omip
                Omip
                Custom metadata
                2.0
                September 2021
                Converter:WILEY_ML3GV2_TO_JATSPMC version:6.2.0 mode:remove_FC converted:07.10.2022

                Cell biology
                antibody secreting cell,b cell,high‐dimensional flow cytometry,lymphocyte,splenocytes,t cell

                Comments

                Comment on this article