OMIP-080: 29-Color flow cytometry panel for comprehensive evaluation of NK and T cells reconstitution after hematopoietic stem cells transplantation

Sarka Vanikova, Abhishek Koladiya, Jan Musil, Sarka Vanikova, Abhishek Koladiya, Jan Musil

Abstract

This 29-color panel was developed and optimized for the monitoring of NK cell and T cell reconstitution in peripheral blood of patients after HSCT. We considered major post-HSCT complications during the design, such as relapses, viral infections, and GvHD and identification of lymphocyte populations relevant to their resolution. The panel includes markers for all major NK cell and T cell subsets and analysis of their development and qualitative properties. In the NK cell compartment, we focus mainly on CD57 + NKG2C+ cells and the expression of activating (NKG2D, DNAM-1) and inhibitory receptors (NKG2A, TIGIT). Another priority is the characterization of T cell reconstitution; therefore, we included detection of CD4+ RTEs based on CD45RA, CD62L, CD95, and CD31 as a marker of thymus function. Besides that, we also analyze the emergence and properties of major T cell populations with a particular interest in CD8, Th1, ThCTL, and Treg subsets. Overall, the panel allows for comprehensive analysis of the reconstituting immune system and identification of potential markers of immune cell dysfunction.

Conflict of interest statement

The authors have no conflict of interest to declare.

© 2021 The Authors. Cytometry Part A published by Wiley Periodicals LLC on behalf of International Society for Advancement of Cytometry.

Figures

FIGURE 1
FIGURE 1
Manual gating strategy and algorithmic analysis. (A) Before the identification of individual subsets, debris, doublets and dead cells were removed. (B) Cell lineages were identified based on the expression of the following phenotypes: CD56+ (NK cells), CD56−CD3+ γδTCR+ (γδT cells), CD3+γδTCR‐CD56+ T cells, CD3+γδTCR−CD56−CD4+ (T helper cells) and CD3+γδTCR−CD56−CD8+ (T cytotoxic cells). (C) CD4+ T cells were delineated into CD25+FoxP3+ Tregs and non‐Treg cells. Activation of Tregs was evaluated based on the expression of CD39. Furthermore, expression of PD1, TIGIT, and Tim3 identifying highly suppressive Tregs was evaluated. Cells in the non‐Treg gate were divided into individual T helper subsets based on the combination of chemokine receptors. These cells were first gated for perforin+CD27−CTL cells and perforin− cells. The perforin− cells were gated for CCR10+ CCR4+ CCR6+ Th22 cells, CCR10+ CCR4+CCR6− ThGM‐CSF, CCR10−CCR4+CCR6− Th2 cells, CCR10−CCR4+CCR6+ Th17 cells, CCR10‐CCR4‐CCR6+CXCR3− Th9 cells and CCR10−CCR4−CCR6+/−CXCR3+ Th1 cells. Thymic function was evaluated on CD4+ gated cell via identification of CD45RA+CD62L+CD95−CD31+ cells. (D) Gated CD8+ cells were subtyped into individual memory subsets based on CD45RA and CD62L and expression of inhibitory receptors, activation markers and perforin was evaluated. Inhibiting receptors are only weakly expressed on T cells from healthy donors, and therefore we show a comparison between healthy and patients after HSCT. (E) Gating of γδT cells based on CD57−NKG2A+ cytokine‐producing cells, CD57+NKG2A− terminally differentiated cytotoxic cells and CD57−NKG2A− with intermediate phenotype. Expression of perforin and TIGIT on these cells is depicted using a histogram. (F) NK cells were subtyped based on the expression of CD56 and CD16, and a comparison of a healthy donor and post‐HSCT patient is shown. CD56hiCD16‐ cells represent less differentiated cytokine‐producing cells characterized by low expression perforin and high expression of CD27, whereas CD56loCD16+ cells express high levels of perforin and lack NKG2A and CD27. These cells include a subset of CD57+NKG2C+ memory‐like NK cells. (G) CD56+ T cells can be divided into three main lineages based on CD4 and CD8. The CD4+ cells are enriched with cytotoxic perforin expressing cells mostly terminally differentiated, as evident by expression of CD57. The CD8+ cell can be split into two subsets based on the expression of NKG2C. These subsets show distinct differentiation status, as evident by the expression patterns of CD45RA and CD62L. (H) Identification of individual cell types was performed on gated live single cells using the Embedding Guided by Self‐Organizing Map (EmbedSOM) algorithm [20]. Expression pattern of markers on individual cell subsets is show in Figure S8. FSC, forward‐scatter; SSC, side‐scatter [Color figure can be viewed at wileyonlinelibrary.com]

References

    1. Wall SA, Devine S, Vasu S. The who, how and why: allogeneic transplant for acute myeloid leukemia in patients older than 60 years. Blood Rev. 2017;31:362–9.
    1. Bartenstein M, Deeg HJ. Hematopoietic stem cell transplantation for MDS. Hematol Oncol Clin North Am. 2010;24:407–22.
    1. Naik S, Martinez CA, Omer B, Sasa G, Yassine K, Allen CE, et al. Allogeneic hematopoietic stem cell transplant for relapsed and refractory non‐Hodgkin lymphoma in pediatric patients. Blood Adv. 2019;3:2689–95.
    1. Palumbo A, Cavallo F, Gay F, di Raimondo F, Ben Yehuda D, Petrucci MT, et al. Autologous transplantation and maintenance therapy in multiple myeloma. New Engl J Med. 2014;371:895–905.
    1. Pai S‐Y, Logan BR, Griffith LM, Buckley RH, Parrott RE, Dvorak CC, et al. Transplantation outcomes for severe combined immunodeficiency, 2000–2009. New Engl J Med. 2014;371:434–46.
    1. Gratwohl A, Brand R, Frassoni F, Rocha V, Niederwieser D, Reusser P, et al. Cause of death after allogeneic haematopoietic stem cell transplantation (HSCT) in early leukaemias: an EBMT analysis of lethal infectious complications and changes over calendar time. Bone Marrow Transplant. 2005;36:757–69.
    1. Kaeuferle T, Krauss R, Blaeschke F, Willier S, Feuchtinger T. Strategies of adoptive T‐cell transfer to treat refractory viral infections post allogeneic stem cell transplantation. J Hematol Oncol. 2019;12:13.
    1. Cichocki F, Verneris MR, Cooley S, Bachanova V, Brunstein CG, Blazar BR, et al. The past, present, and future of NK cells in hematopoietic cell transplantation and adoptive transfer. Curr Top Microbiol Immunol. 2016;395:225–43.
    1. Shaffer BC, Hsu KC. How important is NK alloreactivity and KIR in allogeneic transplantation? Best Pract Res Clin Haematol. 2016;29:351–8.
    1. Storek J, Geddes M, Khan F, Huard B, Helg C, Chalandon Y, et al. Reconstitution of the immune system after hematopoietic stem cell transplantation in humans. Semin Immunopathol. 2008;30:425–37.
    1. Ullah MA, Hill GR, Tey SK. Functional reconstitution of natural killer cells in allogeneic hematopoietic stem cell transplantation. Front Immunol. 2016;7:144.
    1. Cooper MA, Fehniger TA, Turner SC, Chen KS, Ghaheri BA, Ghayur T, et al. Human natural killer cells: a unique innate immunoregulatory role for the CD56bright subset. Blood. 2001;97:3146–51.
    1. Grzywacz B, Kataria N, Verneris MR. CD56dimCD16+ NK cells downregulate CD16 following target cell induced activation of matrix metalloproteinases [4]. Leukemia. 2007;21:356–9.
    1. Amand M, Iserentant G, Poli A, Sleiman M, Fievez V, Sanchez IP, et al. Human CD56dimCD16dimCells as an individualized natural killer cell subset. Front Immunol. 2017;8:699.
    1. Simonetta F, Pradier A, Bosshard C, Masouridi‐Levrat S, Chalandon Y, Roosnek E. NK cell functional impairment after allogeneic hematopoietic stem cell transplantation is associated with reduced levels of T‐bet and Eomesodermin. J Immunol. 2015;195:4712–20.
    1. Hilpert J, Grosse‐Hovest L, Grünebach F, Buechele C, Nuebling T, Raum T, et al. Comprehensive analysis of NKG2D ligand expression and release in leukemia: implications for NKG2D‐mediated NK cell responses. J Immunol. 2012;189:1360–71.
    1. Carlsten M, Baumann BC, Simonsson M, Jädersten M, Forsblom AM, Hammarstedt C, et al. Reduced DNAM‐1 expression on bone marrow NK cells associated with impaired killing of CD34+ blasts in myelodysplastic syndrome. Leukemia. 2010;24:1607–16.
    1. Stringaris K, Sekine T, Khoder A, Alsuliman A, Razzaghi B, Sargeant R, et al. Leukemia‐induced phenotypic and functional defects in natural killer cells predict failure to achieve remission in acute myeloid leukemia. Haematologica. 2014;99:836–47.
    1. Bosch M, Khan FM, Storek J. Immune reconstitution after hematopoietic cell transplantation. Curr Opin Hematol. 2012;19:324–55.
    1. Kratochvíl M, Koladiya A, Vondrášek J. Generalized EmbedSOM on quadtree‐structured self‐organizing maps. F1000Res. 2020;8:2120.
    1. Simões AE, di Lorenzo B, Silva‐Santos B. Molecular determinants of target cell recognition by human γδ T cells. Front Immunol. 2018;9:929.
    1. Pauza CD, Liou ML, Lahusen T, Xiao L, Lapidus RG, Cairo C, et al. Gamma delta T cell therapy for cancer: it is good to be local. Front Immunol. 2018;9:1305.
    1. Lawand M, Déchanet‐Merville J, Dieu‐Nosjean MC. Key features of gamma‐delta T‐cell subsets in human diseases and their immunotherapeutic implications. Front Immunol. 2017;8:761.
    1. Torina A, Guggino G, la Manna MP, Sireci G. The janus face of NKT cell function in autoimmunity and infectious diseases. Int J Mol Sci. 2018;19(2):440.
    1. Pita‐López ML, Pera A, Solana R. Adaptive memory of human NK‐like CD8+ T‐cells to aging, and viral and tumor antigens. Front Immunol. 2016;7:616.
    1. Godfrey DI, Koay HF, McCluskey J, Gherardin NA. The biology and functional importance of MAIT cells. Nat Immunol. 2019;20:1110–28.
    1. Seggewiss R, Einsele H. Immune reconstitution after allogeneic transplantation and expanding options for immunomodulation: an update. Blood. 2010;115:3861–8.
    1. Ogonek J, Juric MK, Ghimire S, Varanasi PR, Holler E, Greinix H, et al. Immune reconstitution after allogeneic hematopoietic stem cell transplantation. Front Immunol. 2016;7:507.
    1. Ravkov E, Slev P, Heikal N. Thymic output: assessment of CD4+ recent thymic emigrants and T‐cell receptor excision circles in infants. Cytometry. Part B. Clin Cytom. 2017;92:249–57.
    1. Schnorfeil FM, Lichtenegger FS, Emmerig K, Schlueter M, Neitz JS, Draenert R, et al. T cells are functionally not impaired in AML: increased PD‐1 expression is only seen at time of relapse and correlates with a shift towards the memory T cell compartment. J Hematol Oncol. 2015;8:93.
    1. Kong Y, Zhang J, Claxton DF, Ehmann WC, Rybka WB, Zhu L, et al. PD‐1hiTIM‐3+ T cells associate with and predict leukemia relapse in AML patients post allogeneic stem cell transplantation. Blood Cancer J. 2015;5:e330.
    1. Kong Y, Zhu L, Schell TD, Zhang J, Claxton DF, Ehmann WC, et al. T‐cell immunoglobulin and ITIM domain (TIGIT) associates with CD8+ T‐cell exhaustion and poor clinical outcome in AML patients. Clin Cancer Res. 2016;22:3057–66.
    1. Shenghui Z, Yixiang H, Jianbo W, Kang Y, Laixi B, Yan Z, et al. Elevated frequencies of CD4+ CD25+ CD127lo regulatory T cells is associated to poor prognosis in patients with acute myeloid leukemia. Int J Cancer. 2011;129:1373–81.
    1. Bansal AK, Sharawat SK, Gupta R, Vishnubhatla S, Dhawan D, Bakhshi S. Regulatory T cells in pediatric AML are associated with disease load and their serial assessment suggests role in leukemogenesis. Am J Blood Res. 2020;10:90–6.
    1. Elias S, Rudensky AY. Therapeutic use of regulatory T cells for graft‐versus‐host disease. Br J Haematol. 2019;187:25–38.
    1. Duhen T, Duhen R, Lanzavecchia A, Sallusto F, Campbell DJ. Functionally distinct subsets of human FOXP3 + Treg cells that phenotypically mirror effector Th cells. Blood. 2012;119:4430–40.
    1. Gu J, Ni X, Pan X, Lu H, Lu Y, Zhao J, et al. Human CD39hi regulatory T cells present stronger stability and function under inflammatory conditions. Cell Mol Immunol. 2017;14:521–8.
    1. Anderson AC, Joller N, Kuchroo VK. Lag‐3, Tim‐3, and TIGIT: co‐inhibitory receptors with specialised functions in immune regulation. Immunity. 2016;44:989–1004.
    1. Lim EY, Jackson SE, Wills MR. The CD4+ T cell response to human cytomegalovirus in healthy and Immunocompromised people. Front Cell Infect Microbiol. 2020;10:202.
    1. van Balen P, van Bergen CAM, van Luxemburg‐Heijs SAP, de Klerk W, van Egmond EHM, Veld SAJ, et al. CD4 donor lymphocyte infusion can cause conversion of chimerism without GVHD by inducing immune responses targeting minor histocompatibility antigens in HLA class II. Front Immunol. 2018;9:3016.
    1. Wu W, Huang J, Duan B, Traficante DC, Hong H, Risech M, et al. Th17‐stimulating protein vaccines confer protection against Pseudomonas aeruginosa pneumonia. Am J Respir Crit Care Med. 2012;186:420–7.
    1. Marqués JM, Rial A, Muñoz N, Pellay FX, van Maele L, Léger H, et al. Protection against Streptococcus pneumoniae serotype 1 acute infection shows a signature of Th17‐ and IFN‐γ‐mediated immunity. Immunobiology. 2012;217:420–9.
    1. Hoving JC, Kolls JK. New advances in understanding the host immune response to pneumocystis. Curr Opin Microbiol. 2017;40:65–71.

Source: PubMed

3
Sottoscrivi