Deep sequencing and flow cytometric characterization of expanded effector memory CD8+CD57+ T cells frequently reveals T-cell receptor Vβ oligoclonality and CDR3 homology in acquired aplastic anemia

Valentina Giudice, Xingmin Feng, Zenghua Lin, Wei Hu, Fanmao Zhang, Wangmin Qiao, Maria Del Pilar Fernandez Ibanez, Olga Rios, Neal S Young, Valentina Giudice, Xingmin Feng, Zenghua Lin, Wei Hu, Fanmao Zhang, Wangmin Qiao, Maria Del Pilar Fernandez Ibanez, Olga Rios, Neal S Young

Abstract

Oligoclonal expansion of CD8+ CD28- lymphocytes has been considered indirect evidence for a pathogenic immune response in acquired aplastic anemia. A subset of CD8+ CD28- cells with CD57 expression, termed effector memory cells, is expanded in several immune-mediated diseases and may have a role in immune surveillance. We hypothesized that effector memory CD8+CD28-CD57+ cells may drive aberrant oligoclonal expansion in aplastic anemia. We found CD8+CD57+ cells frequently expanded in the blood of aplastic anemia patients, with oligoclonal characteristics by flow cytometric Vβ usage analysis: skewing in 1-5 Vβ families and frequencies of immunodominant clones ranging from 1.98% to 66.5%. Oligoclonal characteristics were also observed in total CD8+ cells from aplastic anemia patients with CD8+CD57+ cell expansion by T-cell receptor deep sequencing, as well as the presence of 1-3 immunodominant clones. Oligoclonality was confirmed by T-cell receptor repertoire deep sequencing of enriched CD8+CD57+ cells, which also showed decreased diversity compared to total CD4+ and CD8+ cell pools. From analysis of complementarity-determining region 3 sequences in the CD8+ cell pool, a total of 29 sequences were shared between patients and controls, but these sequences were highly expressed in aplastic anemia subjects and also present in their immunodominant clones. In summary, expansion of effector memory CD8+ T cells is frequent in aplastic anemia and mirrors Vβ oligoclonal expansion. Flow cytometric Vβ usage analysis combined with deep sequencing technologies allows high resolution characterization of the T-cell receptor repertoire, and might represent a useful tool in the diagnosis and periodic evaluation of aplastic anemia patients. (Registered at clinicaltrials.gov identifiers: 00001620, 01623167, 00001397, 00071045, 00081523, 00961064).

Trial registration: ClinicalTrials.gov NCT00001620 NCT01623167 NCT00001397 NCT00071045 NCT00081523 NCT00961064.

Copyright © 2018 Ferrata Storti Foundation.

Figures

Figure 1.
Figure 1.
Immunophenotyping and flow cytometry analysis of Vβ usage in severe aplastic anemia (SAA) patients and healthy subjects. (A) Percentages of CD28+ and CD57+ cells were calculated in both CD4+ and CD8+ compartments for healthy controls and SAA patients. Data are shown as mean±Standard Deviation (SD). Unpaired t-test was performed. *P<0.05; **P<0.01. (B) Vβ usage was studied in T-cell compartments (by row), and percentages of each Vβ family were reported as total CD4+ or CD8+ cell percentage. For Vβ usage in healthy subjects, data are shown as mean+SD, combining the results from all 34 healthy donors. For SAA patients, 2 representative cases are shown.
Figure 2.
Figure 2.
Vβ usage at diagnosis and during treatment. (A) Percentages of Vβ family in CD8+CD57+ cells were calculated on total CD8+ cells, and Vβ skewing in severe aplastic anemia (SAA) patients was defined using the mean+3Standard Deviation (SD) of a given Vβ group in healthy donors. Relative expansion of each Vβ subgroup is shown in the bar graph. Patients were divided based on the absence or presence of expanded CD8+CD57+ cells, using the mean in healthy donors (13.3%). Skewing of one Vβ family is reported as an orange bar. (B) Progression-free survival rate of SAA patients with CD8+CD57+ cells ≤13.3% (n=7) or >13.3% (n=17) prior to treatment. Log-rank (Mantel-Cox) test was performed. (C) Vβ usage was performed in Patient 22 at diagnosis, at 10 days of treatment, and at 6 and 9 months (relapse). Perturbations during treatment and relapse are reported as percentage of positive CD8+CD57+ and Vβ2+ cells (left), or absolute lymphocyte and Vβ2 clone count (right).
Figure 3.
Figure 3.
Characterization of Vβ/Jβ plot, CDR3 size and DJ length profiles in healthy donors by deep sequencing. (A) T-cell receptor β variable (TRBV)/T-cell receptor β joining (TRBJ) plots showed a “citylike” landscape for total CD4+ and CD8+ cell populations in healthy subjects (HC). (B) The size of the complementarity region 3 (CDR3) and DJ length profiles were also defined, showing similar features in CD4+ and CD8+ cells.
Figure 4.
Figure 4.
The TCR repertoire by deep sequencing analysis from total CD8+ cells in severe aplastic anemia (SAA) patients with CD8+CD57+ cell expansion. In contrast to healthy CD8+ profiles, most SAA patients (AA) displayed oligoclonal features in a TRBV/TRBJ rearrangement plot (A) and CDR3 size and DJ length profiles (B). CD4+ and CD8+ profiles are shown for each SAA patient. In AA1 and AA6, only CD8+ cells were sufficient for deep sequencing.
Figure 5.
Figure 5.
The TCR repertoire by deep sequencing of enriched CD8+CD57+ cells in severe aplastic anemia (SAA) patients. (A) The enrichment of the clone in effector memory CD8+ T cells, comparing TRBV/TRBJ rearrangement in total CD8+ cells (left) with those in CD8+CD57+ cells (right) from the same patients. (B) CDR3 size and DJ length profiles from CD8+CD57+ cells also overlapped with those in CD4+ and CD8+ profiles.
Figure 6.
Figure 6.
Homology assessment. (A) CDR3 sequence pools were analyzed among patients (AA) and healthy subjects (HC) for the presence of homology. Shared and immunodominant sequences were reported as a heatmap based on their relative expression: in the same row, from lowest (gray; 5%) values. (B) Structural analysis was performed comparing the sequences for common pattern, using both alignments at the N-terminal of Vβ gene (left) or at the C-terminal of Jβ gene (right).

References

    1. Young NS, Calado RT, Scheinberg P. Current concepts in the pathophysiology and treatment of aplastic anemia. Blood. 2006;108(8):2509–2519.
    1. Young NS. Current concepts in the pathophysiology and treatment of aplastic anemia. Hematology Am Soc Hematol Educ Program. 2013;2013:76–81.
    1. Maciejewski JP, Selleri C, Sato T, Anderson S, Young NS. A severe and consistent deficit in marrow and circulating hematopoietic cells (long-term culture-initiating cells) in acquired aplastic anemia. Blood. 1996;88(6):1983–1991.
    1. Maciejewski JP, Kim S, Sloand E, Selleri C, Young NS. Sustained long-term hematologic recovery despite a marked quantitative defect in the stem cell compartment of patients with aplastic anemia after immunosuppressive therapy. Am J Hematol. 2000;65(2):123–131.
    1. Risitano AM, Maciejewski JP, Selleri C, Rotoli B. Function and malfunction of hematopoietic stem cells in primary bone marrow failure syndromes. Curr Stem Cell Res Ther. 2007;2(1):39–52.
    1. Maciejewski JP, O’Keefe C, Gondek L, Tiu R. Immune-mediated bone marrow failure syndromes of progenitor and stem cells: molecular analysis of cytotoxic T cell clones. Folia Histochem Cytobiol. 2007;45(1):5–14.
    1. Feng X, Scheinberg P, Wu CO, et al. Cytokine signature profiles in acquired aplastic anemia and myelodysplastic syndromes. Haematologica. 2011;96(4):602–606.
    1. Risitano AM, Maciejewski JP, Green S, Plasilova M, Zeng W, Young NS. In-vivo dominant immune responses in aplastic anaemia: molecular tracking of putatively pathogenetic T-cell clones by TCR beta-CDR3 sequencing. Lancet. 2004; 364(9431):355–364.
    1. Young NS. Hematopoietic cell destruction by immune mechanisms in aquired aplastic anemia. Semin Hematol. 2000;37(1):3–14.
    1. Sloand EM, Kim S, Maciejewski JP, et al. Intracellular interferon- in circulating and marrow T cells detected by flow cytometry and the response to imunosuppressive therapy in patients with aplastic anemia. Blood. 2002;100:(4)1185–1191.
    1. Maciejewski JP, Selleri C, Anderson S, Young NS. Fas antigen expression on CD34+ human marrow cells is induced by interferon-gamma and tumor necrosis factor-alpha and potentiates cytokine-mediated hematopoietic suppression in vitro. Blood. 1995;85:(11)3183–3190.
    1. Selleri C, Maciejewski JP, Sato T, Young NS. Interferon-γ constitutively expressed in the stromal microenviroment of human marrow cultures mediates potent hematopoietic inhibition. Blood. 1996; 87:(10)4149–4157.
    1. Hirano N, Butler MO, von Bergwelt-Baildon MS, et al. Autoantibodies frequently detected in patients with aplastic anemia. Blood. 2003;102:(13)4567–4575.
    1. Feng X, Chuhjo T, Sugimori C, et al. Diazepam-binding inhibitor-related protein 1: a candidate autoantigen in acquired aplastic anemia patients harboring a minor populatoin of paroxysmal nocturnal hemoglobinuria-type cells. Blood. 2004; 104:(8)2425–2431.
    1. Kook H, Risitano AM, Zeng W, et al. Changes in T-cell receptor VB repertoire in aplastic anemia: effects of different immunosuppressive regimens. Blood. 2002;99(10):3668–3675.
    1. Clemente MJ, Wlodarski MW, Makishima H, et al. Clonal drift demonstrates unexpected dynamics of the T-cell repertoire in T-large granular lymphocyte leukemia. Blood. 2011;118(16):4384–4393.
    1. Clemente MJ, Przychodzen B, Jerez A, et al. Deep sequencing of the T-cell receptor repertoire in CD8+ T-large granular lymphocyte leukemia identifies signature landscapes. Blood. 2013;122(25):4077–4085.
    1. Calis JJ, Rosenberg BR. Characterizing immune repertoires by high throughput sequencing: strategies and applications. Trends Immunol. 2014;35(12):581–590.
    1. Langerak AW, van Den Beemd R, Wolvers-Tettero IL, et al. Molecular and flow cytometric analysis of the Vbeta repertoire for clonality assessment in mature TCRalphabeta T-cell proliferations. Blood. 2001;98(1):165–173.
    1. Wlodarski MW, Gondek LP, Nearman ZP, et al. Molecular strategies for detection and quantitation of clonal cytotoxic T-cell responses in aplastic anemia and myelodysplastic syndrome. Blood. 2006;108(8):2632–2641.
    1. Schuster FR, Hubner B, Führer M, et al. Highly skewed T-cell receptor V-beta chain repertoire in the bone marrow is associated with response to immunosuppressive drug therapy in children with very severe aplastic anemia. Blood Cancer J. 2011;1(3):e8.
    1. Schatz DG, Ji Y. Recombination centres and the orchestration of V(D)J recombination. Nat. Rev. Immunol. 2011;11:(4)251–263.
    1. Gauss GH, Lieber MR. Mechanistic constraints on diversity in human V(D)J recombination. Mol Cell Biol. 1996;16(1):258–269.
    1. Teng G, Papavasiliou FN. Immunoglobulin somatic hypermutation. Annu Rev Genet. 2007;41:107–120.
    1. Matthews AJ, Zheng S, DiMenna LJ, Chaudhuri J. Regulation of immunoglobulin class-switch recombination: choreography of noncoding transcription, targeted DNA deamination, and long-range DNA repair. Adv Immunol. 2014;122:1–57.
    1. Arstila TP, Casrouge A, Baron V, Even J, Kanellopoulos J, Kourilsky P. A direct estimate of the human alphabeta T cell receptor diversity. Science. 1999;286(5441):958–961.
    1. Bretschneider I, Clemente MJ, Meisel C, et al. Discrimination of T-cell subsets and T-cell receptor repertoire distribution. Immunol Res. 2014;58(1):20–27.
    1. Araki K, Youngblood B, Ahmed R. The role of mTOR in memory CD8 T-cell differentiation. Immunol Rev. 2010;235(1):234–243.
    1. Appay V, van Lier RA, Sallusto F, Roederer M. Phenotype and function of human T lymphocyte subsets: consensus and issues. Cytometry A. 2008;73(11):975–983.
    1. Farber DL, Yudanin NA, Restifo NP. Human memory T cells: generation, compartmentalization and homeostasis. Nat Rev Immunol. 2014;14(1):24–35.
    1. World Medical Association. World Medical Association Declaration of Helsinki: ethical principles for medical research involving human subjects. JAMA. 2013;310(20):2191–2194.
    1. International Agranulocytosis and Aplastic Anaemia Study. Incidence of aplastic anemia: the relevance of diagnostic criteria. By the International Agranulocytosis and Aplastic Anemia Study. Blood. 1987;70(6):1718–1721.
    1. Camitta BM, Thomas ED, Nathan DG, et al. Severe aplastic anemia: a prospective study of the effect of early marrow transplantation on acute mortality. Blood. 1976;48(1):63–70.
    1. Bolotin DA, Shugay M, Mamedov IZ, et al. MiTCR: software for T-cell receptor sequencing data analysis. Nat Methods. 2013;10(9):813–814.
    1. Benjamini Y, Yekutieli D. The control of the false discovery rate in multiple testing under dependency. Ann Stat. 2001; 29:(4)1165–1188.
    1. Strioga M, Pasukoniene V, Characiejus D. CD8+ CD28− and CD8+ CD57+ T cells and their role in health and disease. Immunology. 2011;134(1):17–32.
    1. Nickel RS, Horan JT, Fasano RM, et al. Immunophenotypic parameters and RBC alloimmunization in children with sickle cell disease on chronic transfusion. Am J Hematol. 2015;90(12):1135–1141.
    1. Hill MO. Diversity and evenness: a unifying notation and its consequences. Ecology. 1973;54:(2)427–432.
    1. Li H, Ye C, Ji G, Han J. Determinants of public T cell responses. Cell Res. 2012;22(1):33–42.
    1. Gargiulo L, Lastraioli S, Cerruti G, et al. Highly homologous T-cell receptor beta sequences support a common target for autoreactive T cells in most patients with paroxysmal nocturnal hemoglobinuria. Blood. 2007;109(11):5036–5042.
    1. Risitano AM, Kook H, Zeng W, Chen G, Young NS, Maciejewski JP. Oligoclonal and polyclonal CD4 and CD8 lymphocytes in aplastic anemia and paroxysmal nocturnal hemoglobinuria measured by V beta CDR3 spectratyping and flow cytometry. Blood. 2002;100(1):178–183.
    1. Filaci G, Fenoglio D, Fravega M, et al. CD8+ CD28− T regulatory lymphocytes inhibiting T cell proliferative and cytotoxic functions infiltrate human cancers. J Immunol. 2007; 179:(7)4323–4334.
    1. Characiejus D, Pasukoniene V, Jonusauskaite R, et al. Peripheral blood CD8high CD57+ lymphocyte levels may predict outcome in melanoma patients treated with adjuvant interferon-alpha. Anticancer Res. 2008;28:(2B)1139–1142.
    1. Kook H, Zeng W, Guibin C, Kirby M, Young NS, Maciejewski JP. Increased cytotoxic T cells with effector phenotype in aplastic anemia and myelodysplasia. Exp Hematol. 2001;29:(11)1270–1277.
    1. Boucher N, Dufeu-Duchesne T, Vicaut E, Farge D, Effros RB, Schächter F. CD28 expression in T cell aging and human longevity. Exp Gerontol. 1998;33(3):267–282.
    1. Wikby A, Ferguson F, Forsey R, et al. An immune risk phenotype, cognitive impairment, and survival in very late life: impact of allostatic load in Swedish octogenarian and nonagenarian humans. J Gerontol A Biol Sci Med Sci. 2005;60(5):556–565.
    1. Fozza C, Barraqueddu F, Corda G, et al. Study of the T-cell receptor repertoire by CDR3 spectratyping. J Immunol Methods. 2017;440:1–11.
    1. Zeng W, Maciejewski JP, Chen G, Young NS. Limited heterogeneity of T cell receptor BV usage in aplastic anemia. J Clin Invest. 2001;108(5):765–773.
    1. Le Gal FA, Ayyoub M, Dutoit V, et al. Distinct structural TCR repertoires in naturally occurring versus vaccine-induced CD8+ T-cell responses to the tumor-specific antigen NY-ESO-1. J Immunother. 2005;28(3):252–257.

Source: PubMed

3
Subskrybuj