Circulating Tfh1 (cTfh1) cell numbers and PD1 expression are elevated in low-grade B-cell non-Hodgkin's lymphoma and cTfh gene expression is perturbed in marginal zone lymphoma

Elliot T Byford, Matthew Carr, Eleni Ladikou, Matthew J Ahearne, Simon D Wagner, Elliot T Byford, Matthew Carr, Eleni Ladikou, Matthew J Ahearne, Simon D Wagner

Abstract

CD4+ T-cell subsets are found in the tumour microenvironment (TME) of low-grade B-cell non-Hodgkin's lymphomas such as marginal zone lymphoma (MZL) or follicular lymphoma (FL). Both numbers and architecture of activating follicular helper T-cells (Tfh) and suppressive Treg in the TME of FL are associated with clinical outcomes. There has been almost no previous work on CD4+ T-cells in MZL. It is now recognised that circulating CD4+CXCR5+ T-cells are the memory compartment of Tfh cells. We determined differences in number of circulating Tfh (cTfh) cells and cTfh subsets between normal subjects and patients with FL or MZL. Lymphoma patients showed increased numbers of cTfh1 and reduced cTfh17 cells due to decreased expression of the subset-defining marker CCR6 in patients. PD1, a surface marker associated with Tfh cells, showed increased expression on cTfh subsets in patients. Focusing on MZL we determined expression of 96 T-cell associated genes by microfluidic qRT-PCR. Analysis of differentially expressed genes showed significant differences between normal subjects and patients both for bulk cTfh (CCL4) and the cTfh1 subset (JAK3). While our findings require confirmation in larger studies we suggest that analysis of number and gene expression of circulating T-cells might be a source of clinically useful information as is the case for T-cells within lymphoma lymph nodes.

Conflict of interest statement

Competing Interests: The authors have declared that no competing interests exist.

Figures

Fig 1. cTfh and cTfh subset proportions…
Fig 1. cTfh and cTfh subset proportions in normal subjects and low-grade B-NHL.
(A) After lymphocyte gating and exclusion of doublets, live and CD45RA- cells were identified for further analysis. The biaxial plot shows CXCR5 and CD4 expression from a representative MZL patient. The gate defines CD4+CXCR5+ (cTfh) cells as being 24.6% of total CD4+ T-cells. (B) cTfh cells as a percentage of CD4+ cells in normal subjects (median±SD, 18.9±5.7) and patients with FL (11.8±7.1) and MZL (10.2±2.7). Horizontal lines represent the median and bars represent inter-quartile range. (C) Biaxial flow cytometry plot showing CXCR3 and CCR6 expression on CD4+CD45RA-CXCR5+ cells. Four populations are identifiable: CXCR3+CCR6- (cTfh1), CXCR3-CCR6- (cTfh2), CXCR3-CCR6+ (cTfh17) and CXCR3+CCR6+ (cTfh1/17). (D) cTfh1 cells as a percentage of total cTfh cells. Medians are significantly (Mann-Whitney U-test) different between normal subjects (n = 12, 20.8±6.7%) and lymphoma patients (MZL, n = 7, 32.1±6.8%, P = 0.013 and FL, n = 9, 35.4±7.6%, P = 0.0056). (E) cTfh2 cells as a percentage of total cTfh cells. There are no significant differences between groups. (F) cTfh17 cells as a percentage of total cTfh cells. Medians are significantly different between normal subjects (37.9±5.9%) and lymphoma patients (MZL 28.5±8.2%, P = 0.045 and FL 22.9±5.7%, P = 0.0006). (G) cTfh1/17 cells as a proportion of total cTfh cells. Medians are significantly different between normal subjects (18.9±5.7%) and lymphoma patients (MZL 10.2±2.7%, P = 0.0008 and FL 11.8±7.1%, P = 0.016).
Fig 2. PD1 expression on cTfh subsets.
Fig 2. PD1 expression on cTfh subsets.
(A) Biaxial flow cytometry plot showing expression of PD1 and CXCR5 on cells gated for CD4+CD45RA-CXCR5+. Gates defining PD1++, PD1+ and PD1- cells were set by employing human tonsillar T-cells as controls. This representative example from a patient with MZL shows PD1- 50.8%, PD1+ 48.1% and PD1++ 0.29%. (B) PD1- cells as a proportion of total cTfh cells. Horizontal lines represent the median and bars represent inter-quartile range. Medians are significantly (Mann-Whitney U-test) different between normal subjects (n = 12, 78.4% (interquartile range 71.2 to 82.1%) and lymphoma patients (MZL n = 7, 44.7% (34.6 to 56.4%), P = 0.0008 and FL n = 9, 54.6% (47.9 to 60%), P = 0.0016). (C) PD1+ cells as a proportion of total cTfh cells. Medians are significantly different between normal subjects (21.5%, interquartile range 17.8 to 28.6%) and lymphoma patients (MZL 54.1% (43.5 to 64.7%), P = 0.0008 and FL 45.2% (39.5 to 50.9%), P = 0.0003). (D) PD1++ cells as a proportion of total cTfh cells. There is no significant difference between normal subjects (median 0.07%, interquartile range 0.03 to 0.09%) and MZL patients (median 0.62%, interquartile range 0.09 to 1.08%, P = 0.05) but there is a significant difference for BNHL (median 0.6%, interquartile range 0.18 to 1.23%, P = 0.003). Distribution of PD1 expression (PD1++, PD1+ or PD1-) within (E) cTfh1 cells, (F) cTfh2 cells, (G) cTfh17 cells and (H) cTfh1/17 cells.
Fig 3. Clustering by ViSNE reveals surface…
Fig 3. Clustering by ViSNE reveals surface marker expression changes in cTfh subsets.
(A) Comparison of VisNE maps and gating from biaxial flow cytometry plots. Areas within blue lines are derived from the ViSNE maps. Cells coloured according to biaxial plots fro CXCR3 and CCR6 are coloured as shown: cTfh1 (blue), cTfh2 (orange), cTfh17 (green), cTfh1/17 (red). (B) The overall ViSNE map generated for each group is coloured according to the fluorescence intensity of the PD1, CXCR3 and CCR6 channels. Cells were pre-gated to cTfh. Level of expression is according to the bar to the right of the figure with red being highest expression.
Fig 4. CCR6 and PD1 expression in…
Fig 4. CCR6 and PD1 expression in cTfh subsets.
(A) Concatenated FACS histogram showing superimposed PD1 expression in CD4+CXCR5+ cells from normal subjects (n = 12) and patients with MZL (n = 7) and BNHL (n = 9). The right hand column chart demonstrates PD1 expression (geometric mean fluorescence intensity) for each group (mean±SEM). There are significant increases in PD1 expression in MZL (Mann-Whitney U-test; P = 0.0008) and BNHL (P = 0.0005). (B) FACS histograms showing PD1 expression in CD4+CXCR5+CCR6+ cells from normal subjects and patients with MZL and BNHL. The right hand column chart demonstrates PD1 expression (geometric mean fluorescence intensity) for each group (mean±SEM). There are significant increases in PD1 expression in MZL (Mann-Whitney U-test; P = 0.0052) and BNHL (P = 0.0033). (C) FACS histograms showing CCR6 expression in CD4+CXCR5+CCR6+ cells from normal subjects and patients with MZL and BNHL. The right hand column chart demonstrates PD1 expression (geometric mean fluorescence intensity) for each group (mean±SEM). There are significant decreases in CCR6 expression in MZL (Mann-Whitney U-test; P = 0.0005) and BNHL (P = 0.0035).
Fig 5. Suppressive Tregs and Tfr in…
Fig 5. Suppressive Tregs and Tfr in normal subjects and low-grade lymphoma.
(A) Biaxial flow cytometry plot showing expression of CD4 and FOXP3 on cells gated for CD4+CD45RA-. The highest 5% of cells from 11 concatenated NS were employed for the FOXP3hi gate. This representative example from a patient with lymphoma shows Treg 5.9% of CD4+CD45RA- cells. (B) Percentage of Treg within total CD4+ T-cells in normal subjects (n = 11), MZL (N = 4) and BNHL (n = 9). Horizontal lines represent the median and bars represent inter-quartile range. No significant differences were found between groups (Mann-Whitney U test). (C) Biaxial flow cytometry plot showing expression of CD4 and CXCR5 on cells gated for CD4+CD45RA-FOXP3hi. This representative example from a patient with lymphoma shows cTfr 21.4% of CD4+CD45RA-FOXP3hi cells. (D) cTfr as a proportion of total CD4+ T-cells. Horizontal lines represent the median and bars represent inter-quartile range. There are no significant differences between normal subjects (n = 11) and patient with MZL (n = 4) or BNHL (n = 9). (E) Biaxial flow cytometry plot showing expression of CD4 and FOXP3 on cells gated for CD4+CD45RA-CXCR5+. Gates were set by employing human tonsillar T-cells as controls. This representative example from a patient with lymphoma shows cTfr (FOXP3hi) 7.3% and cTfh (FOXP3-/lo) 92.7% of CD4+CD45RA-CXCR5+ cells. (F) cTfr:cTfh ratio. Horizontal lines represent the median and bars represent inter-quartile range. BNHL patients (n = 9) showed a significant difference (Mann-Whitney U-test, P = 0.038) from normal subjects (n = 11) but MZL patients (n = 4) did not.
Fig 6. Gene expression changes between normal…
Fig 6. Gene expression changes between normal subjects and MZL patients.
(A) Listing of genes showing the greatest relative change of expression level comparing cTfh PD1+ cells from normal subjects (NS) and patients with MZL. Genes showing an increase in rank in MZL i.e. increased expression are shaded red and those showing a decrease in rank are shaded blue. (B) Box and whisker plot (median, interquartile ranges and 10th and 90th percentile) showing expression levels of CCL4, which differed significantly between cTfh cells of normal subjects and MZL patients (Mann-Whitney U-test, P = 0.03). Examples of non-significant expression level changes are shown for GZMB, MAF, STAT5B, CD27 and PTEN. (C) Relative change in gene expression comparing cTfh1 PD1+ cells from normal subjects (NS) and patients with MZL. Genes showing an increase in rank in MZL i.e. increased expression are shaded red and those showing a decrease in rank are shaded blue. (D) Expression levels of JAK3 differed significantly (Mann-Whitney U-test, P = 0.01) between cTfh1 cells of normal subjects and MZL patients. Examples of non-significant expression level changes are shown for PTEN, IL12RB1, LEF1, GATA3 and TNFAIP8.

References

    1. Hacken ten E, Burger JA. Microenvironment dependency in Chronic Lymphocytic Leukemia: The basis for new targeted therapies. Pharmacology and Therapeutics. 2014;144: 338–348. doi:
    1. Burger JA, Gribben JG. The microenvironment in chronic lymphocytic leukemia (CLL) and other B cell malignancies: Insight into disease biology and new targeted therapies. Semin Cancer Biol. Elsevier Ltd; 2013; 1–11. doi:
    1. Lee AM, Clear AJ, Calaminici M, Davies AJ, Jordan S, MacDougall F, et al. Number of CD4+ cells and location of forkhead box protein P3-positive cells in diagnostic follicular lymphoma tissue microarrays correlates with outcome. J Clin Oncol. 2006;24: 5052–5059. doi:
    1. Carreras J, López-Guillermo A, Fox BC, Colomo L, Martinez A, Roncador G, et al. High numbers of tumor-infiltrating FOXP3-positive regulatory T cells are associated with improved overall survival in follicular lymphoma. Blood. American Society of Hematology; 2006;108: 2957–2964. doi:
    1. Carreras J, Lopez-Guillermo A, Roncador G, Villamor N, Colomo L, Martinez A, et al. High numbers of tumor-infiltrating programmed cell death 1-positive regulatory lymphocytesare associated with improved overall survival in follicular lymphoma. J Clin Oncol. 2009;27: 1470–1476. doi:
    1. Amé-Thomas P, Le Priol J, Yssel H, Caron G, Pangault C, Jean R, et al. Characterization of intratumoral follicular helper T cells in follicular lymphoma: role in the survival of malignant B cells. Leukemia. 2012;26: 1053–1063. doi:
    1. Pangault C, Amé-Thomas P, Ruminy P, Rossille D, Caron G, Baia M, et al. Follicular lymphoma cell niche: identification of a preeminent IL-4-dependent TFH–B cell axis. Leukemia. 2010; 1–10.
    1. Ahearne MJ, Willimott S, Piñon L, Kennedy DB, Miall F, Dyer MJS, et al. Enhancement of CD154/IL4 proliferation by the T follicular helper (Tfh) cytokine, IL21 and increased numbers of circulating cells resembling Tfh cells in chronic lymphocytic leukaemia. Br J Haematol. 2013;162: 360–370. doi:
    1. Pascutti MF, Jak M, Tromp JM, Derks IAM, Remmerswaal EBM, Thijssen R, et al. IL-21 and CD40L signals from autologous T cells can induce antigen-independent proliferation of CLL cells. Blood. 2013;122: 3010–3019. doi:
    1. Os A, Bürgler S, Ribes AP, Funderud A, Wang D, Thompson KM, et al. Chronic Lymphocytic Leukemia Cells Are Activated and Proliferate in Response to Specific T Helper Cells. CellReports. The Authors; 2013;4: 566–577. doi:
    1. Arcaini L, Rossi D, Paulli M. Splenic marginal zone lymphoma: from genetics to management. Blood. 2016;127: 2072–2081. doi:
    1. Zucca E, Bertoni F, Vannata B, Cavalli F. Emerging Role of Infectious Etiologies in the Pathogenesis of Marginal Zone B-cell Lymphomas. Clin Cancer Res. 2014;20: 5207–5216. doi:
    1. Vinuesa CG, Linterman MA, Yu D, Maclennan ICM. Follicular Helper T Cells. Annu Rev Immunol. 2016;34: 335–368. doi:
    1. Linterman MA, Pierson W, Lee SK, Kallies A, Kawamoto S, Rayner TF, et al. Foxp3+ follicular regulatory T cells control the germinal center response. Nat Med. Nature Publishing Group; 2011;17: 975–982. doi:
    1. Chung Y, Tanaka S, Chu F, Nurieva RI, Martinez GJ, Rawal S, et al. Follicular regulatory T cells expressing Foxp3 and Bcl-6 suppress germinal center reactions. Nat Med. 2011;17: 983–988. doi:
    1. Förster R, Emrich T, Kremmer E, Lipp M. Expression of the G-protein—coupled receptor BLR1 defines mature, recirculating B cells and a subset of T-helper memory cells. Blood. 1994;84: 830–840.
    1. Morita R, Schmitt N, Bentebibel S-E, Ranganathan R, Bourdery L, Zurawski G, et al. Human Blood CXCR5+CD4+ T Cells Are Counterparts of T Follicular Cells and Contain Specific Subsets that Differentially Support Antibody Secretion. Immunity. 2011;34: 108–121. doi:
    1. He J, Tsai LM, Leong YA, Hu X, Ma CS, Chevalier N, et al. Circulating precursor CCR7(lo)PD-1(hi) CXCR5⁺ CD4⁺ T cells indicate Tfh cell activity and promote antibody responses upon antigen reexposure. Immunity. 2013;39: 770–781. doi:
    1. Sage PT, Alvarez D, Godec J, Andrian von UH, Sharpe AH. Circulating T follicular regulatory and helper cells have memory-like properties. J Clin Invest. 2014;124: 5191–5204. doi:
    1. Schmitt N, Bentebibel SE, Ueno H. Phenotype and functions of memory Tfh cells in human blood. Trends in Immunology. Elsevier Ltd; 2014;35: 436–442. doi:
    1. Sallusto F, Lenig D, Mackay CR, Lanzavecchia A. Flexible programs of chemokine receptor expression on human polarized T helper 1 and 2 lymphocytes. J Exp Med. 1998;187: 875–883.
    1. Acosta-Rodriguez EV, Rivino L, Geginat J, Jarrossay D, Gattorno M, Lanzavecchia A, et al. Surface phenotype and antigenic specificity of human interleukin 17-producing T helper memory cells. Nat Immunol. 2007;8: 639–646. doi:
    1. Bailey SR, Nelson MH, Himes RA, Li Z, Mehrotra S, Paulos CM. Th17 cells in cancer: the ultimate identity crisis. Front Immunol. 2014;5: 276 doi:
    1. Duhen T, Campbell DJ. IL-1β promotes the differentiation of polyfunctional human CCR6+CXCR3+ Th1/17 cells that are specific for pathogenic and commensal microbes. J Immunol. 2014;193: 120–129. doi:
    1. Cha Z, Guo H, Tu X, Zang Y, Gu H, Song H, et al. Alterations of circulating follicular helper T cells and interleukin 21 in diffuse large B-cell lymphoma. Tumour Biol. 2014;35: 7541–7546. doi:
    1. Bentebibel SE, Lopez S, Obermoser G, Schmitt N, Mueller C, Harrod C, et al. Induction of ICOS+CXCR3+CXCR5+ TH cells correlates with antibody responses to influenza vaccination. Science Translational Medicine. 2013;5: 176ra32–176ra32. doi:
    1. Amir E-AD, Davis KL, Tadmor MD, Simonds EF, Levine JH, Bendall SC, et al. viSNE enables visualization of high dimensional single-cell data and reveals phenotypic heterogeneity of leukemia. Nat Biotechnol. 2013;31: 545–552. doi:
    1. Forcade E, Kim HT, Cutler C, Wang K, Alho AC, Nikiforow S, et al. Circulating T follicular helper cells with increased function during chronic graft-versus-host disease. Blood. 2016;127: 2489–2497. doi:
    1. Sage PT, Tan CL, Freeman GJ, Haigis M, Sharpe AH. Defective TFH Cell Function and Increased TFR Cells Contribute to Defective Antibody Production in Aging. CellReports. 2015. doi:
    1. Zhou M, Zou R, Gan H, Liang Z, Li F, Lin T, et al. The effect of aging on the frequency, phenotype and cytokine production of human blood CD4 + CXCR5 + T follicular helper cells: comparison of aged and young subjects. Immun Ageing. BioMed Central; 2014;11: 12 doi:
    1. Le Coz C, Joublin A, Pasquali J-L, Korganow A-S, Dumortier H, Monneaux F. Circulating TFH subset distribution is strongly affected in lupus patients with an active disease. PLoS ONE. 2013;8: e75319 doi:
    1. Herati RS, Reuter MA, Dolfi DV, Mansfield KD, Aung H, Badwan OZ, et al. Circulating CXCR5+PD-1+ response predicts influenza vaccine antibody responses in young adults but not elderly adults. J Immunol. 2014;193: 3528–3537. doi:
    1. Linterman MA. How T follicular helper cells and the germinal centre response change with age. Immunol Cell Biol. 2014;92: 72–79. doi:
    1. Bossaller L, Burger J, Draeger R, Grimbacher B, Knoth R, Plebani A, et al. ICOS deficiency is associated with a severe reduction of CXCR5+CD4 germinal center Th cells. The Journal of Immunology. 2006;177: 4927–4932.
    1. Asrir A, Aloulou M, Gador M, Pérals C, Fazilleau N. Interconnected subsets of memory follicular helper T cells have different effector functions. Nature Communications. Nature Publishing Group; 2017;8: 847 doi:
    1. Ueno H, Banchereau J, Vinuesa CG. Pathophysiology of T follicular helper cells in humans and mice. Nat Immunol. 2015;16: 142–152. doi:
    1. Li X-Y, Wu Z-B, Ding J, Zheng Z-H, Li X-Y, Chen L-N, et al. Role of the frequency of blood CD4+ CXCR5+ CCR6+ T cells in autoimmunity in patients with Sjögren’s syndrome. BIOCHEMICAL AND BIOPHYSICAL RESEARCH COMMUNICATIONS. Elsevier Inc; 2012;422: 238–244. doi:
    1. Romme Christensen J, Börnsen L, Ratzer R, Piehl F, Khademi M, Olsson T, et al. Systemic inflammation in progressive multiple sclerosis involves follicular T-helper, Th17- and activated B-cells and correlates with progression. PLoS ONE. 2013;8: e57820 doi:
    1. Cha Z, Qian G, Zang Y, Gu H, Huang Y, Zhu L, et al. Circulating CXCR5+CD4+ T cells assist in the survival and growth of primary diffuse large B cell lymphoma cells through interleukin 10 pathway. Experimental Cell Research. 2017;350: 154–160. doi:
    1. Martin-Gayo E, Cronin J, Hickman T, Ouyang Z, Lindqvist M, Kolb KE, et al. Circulating CXCR5(+)CXCR3(+)PD-1(lo) Tfh-like cells in HIV-1 controllers with neutralizing antibody breadth. JCI Insight. 2017;2: e89574 doi:
    1. Reinhardt RL, Liang H-E, Locksley RM. Cytokine-secreting follicular T cells shape the antibody repertoire. Nat Immunol. 2009;10: 385–393. doi:
    1. Peuchmaur M, Emilie D, Crevon MC, Brousse N, Gaulard P, D’Agay MF, et al. Interleukin-2 and interferon-gamma production in follicular lymphomas. Am J Clin Pathol. 1991;95: 55–62.
    1. Warzocha K, Ribeiro P, Renard N, Bienvenu J, Charlot C, Coiffier B, et al. Expression of genes coding for the tumor necrosis factor and lymphotoxin ligand-receptor system in non-Hodgkin’s lymphomas. Cancer Immunol Immunother. 2000;49: 469–475.
    1. Vyth-Dreese FA, Boot H, Dellemijn TA, Majoor DM, Oomen LC, Laman JD, et al. Localization in situ of costimulatory molecules and cytokines in B-cell non-Hodgkin’s lymphoma. Immunology. Wiley-Blackwell; 1998;94: 580–586.
    1. Riedel S, Kraft M, Kucharzik T, Pauels HG, Tiemann M, Steinbüchel A, et al. CD4+ Th1-cells predominate in low-grade B-cell lymphoma of gastric mucosa-associated lymphoid tissue (MALT type). Scand J Gastroenterol. 2001;36: 1198–1203.
    1. Nezos A, Gravani F, Tassidou A, Kapsogeorgou EK, Voulgarelis M, Koutsilieris M, et al. Type I and II interferon signatures in Sjogren“s syndrome pathogenesis: Contributions in distinct clinical phenotypes and Sjogren”s related lymphomagenesis. Journal of Autoimmunity. 2015;63: 47–58. doi:
    1. Jackson SW, Jacobs HM, Arkatkar T, Dam EM, Scharping NE, Kolhatkar NS, et al. B cell IFN-γ receptor signaling promotes autoimmune germinal centers via cell-intrinsic induction of BCL-6. J Exp Med. 2016;213: 733–750. doi:
    1. Moore PA, Belvedere O, Orr A, Pieri K, LaFleur DW, Feng P, et al. BLyS: member of the tumor necrosis factor family and B lymphocyte stimulator. Science. 1999;285: 260–263.
    1. Schneider P, MacKay F, Steiner V, Hofmann K, Bodmer JL, Holler N, et al. BAFF, a novel ligand of the tumor necrosis factor family, stimulates B cell growth. J Exp Med. 1999;189: 1747–1756.
    1. Scapini P, Hu Y, Chu C-L, Migone T-S, DeFranco AL, Cassatella MA, et al. Myeloid cells, BAFF, and IFN-γ establish an inflammatory loop that exacerbates autoimmunity in Lyn-deficient mice. J Exp Med. 2010;207: 1757–1773. doi:
    1. Rodig SJ, Shahsafaei A, Li B, Mackay CR, Dorfman DM. BAFF-R, the major B cell-activating factor receptor, is expressed on most mature B cells and B-cell lymphoproliferative disorders. Hum Pathol. 2005;36: 1113–1119. doi:
    1. Novak AJ, Grote DM, Stenson M, Ziesmer SC, Witzig TE, Habermann TM, et al. Expression of BLyS and its receptors in B-cell non-Hodgkin lymphoma: correlation with disease activity and patient outcome. Blood. 2004;104: 2247–2253. doi:
    1. Saberi Hosnijeh F, Krop EJM, Scoccianti C, Krogh V, Palli D, Panico S, et al. Plasma cytokines and future risk of non-Hodgkin lymphoma (NHL): a case-control study nested in the Italian European Prospective Investigation into Cancer and Nutrition. Cancer Epidemiol Biomarkers Prev. 2010;19: 1577–1584. doi:
    1. Shi M, Lin TH, Appell KC, Berg LJ. Janus-kinase-3-dependent signals induce chromatin remodeling at the Ifng locus during T helper 1 cell differentiation. Immunity. 2008;28: 763–773. doi:

Source: PubMed

3
Abonnere