Thymic output generates a new and diverse TCR repertoire after autologous stem cell transplantation in multiple sclerosis patients

Paolo A Muraro, Daniel C Douek, Amy Packer, Katherine Chung, Francisco J Guenaga, Riccardo Cassiani-Ingoni, Catherine Campbell, Sarfraz Memon, James W Nagle, Frances T Hakim, Ronald E Gress, Henry F McFarland, Richard K Burt, Roland Martin, Paolo A Muraro, Daniel C Douek, Amy Packer, Katherine Chung, Francisco J Guenaga, Riccardo Cassiani-Ingoni, Catherine Campbell, Sarfraz Memon, James W Nagle, Frances T Hakim, Ronald E Gress, Henry F McFarland, Richard K Burt, Roland Martin

Abstract

Clinical trials have indicated that autologous hematopoietic stem cell transplantation (HSCT) can persistently suppress inflammatory disease activity in a subset of patients with severe multiple sclerosis (MS), but the mechanism has remained unclear. To understand whether the beneficial effects on the course of disease are mediated by lympho-depletive effects alone or are sustained by a regeneration of the immune repertoire, we examined the long-term immune reconstitution in patients with MS who received HSCT. After numeric recovery of leukocytes, at 2-yr follow-up there was on average a doubling of the frequency of naive CD4(+) T cells at the expense of memory T cells. Phenotypic and T cell receptor excision circle (TREC) analysis confirmed a recent thymic origin of the expanded naive T cell subset. Analysis of the T cell receptor repertoire showed the reconstitution of an overall broader clonal diversity and an extensive renewal of clonal specificities compared with pretherapy. These data are the first to demonstrate that long-term suppression of inflammatory activity in MS patients who received HSCT does not depend on persisting lymphopenia and is associated with profound qualitative immunological changes that demonstrate a de novo regeneration of the T cell compartment.

Figures

Figure 1.
Figure 1.
Resolution of inflammatory disease activity after HSCT. Representative contrast-enhanced MRI axial scans of the brain of patient 2 at pretransplant evaluation (top) and at 2-yr follow-up (bottom) illustrate a long-lasting resolution of inflammatory disease in this patient with high pretransplant activity (arrows point at 3 out of 17 total enhancing lesions present in the brain pretherapy; no enhancing lesions were detected after therapy).
Figure 2.
Figure 2.
Phenotypic analysis of naive and memory T cell populations. All values are the percent expression of the indicated T cell subpopulation at pretherapy and follow-up time points. (left) Subsets of CD4+ T cells and (right) subsets of CD8+ T cells, each categorized as Tnaive, naive T cells; TCM, central–memory T cells; and TEM, effector–memory T cells. The boundaries of the boxes indicate the 25th and 75th percentiles, the lines within the boxes indicate the median, and the whiskers mark the 10th and the 90th percentiles. We observed a statistically significant (greater than twofold) increase of CD4 Tnaive and a decrease of CD4 TCM.
Figure 3.
Figure 3.
Measures of thymic output after HSCT. (A) Phenotypic identification of RTEs in the CD4+ subset showed a significant increase of CD4+ RTEs at 2-yr follow-up. (B) TCR excision circle (TREC) analysis showed full reconstitution and increases of CD4 RTE output at 2-yr follow-up. (C) Due to extensive peripheral cell division, CD8 TREC levels only recovered up to near-pretherapy levels. Age–related interindividual variance and the small number of patients precluded a statistical evaluation of TREC data.
Figure 4.
Figure 4.
Analysis of TCR diversity. High-resolution TCRBV CDR3 spectratyping analysis identified three basic patterns of evolution of repertoire diversity: (A) recovery of diversity from a restricted repertoire, (B) reconstitution of diversity from a normally diverse repertoire; (C) and total or partial persistence (or reemergence) of repertoire skewing. Different numbers of examples are shown to convey a correct impression of the observed frequency of each pattern. Patterns A and B were largely predominant except for patient 5, who had recurrent infections. Vertical lines are reference DNA size markers to provide the exact correspondence of CDR3 peaks across different samples. n.d., not determined (due to unavailability of 2-yr follow-up for patient 7).
Figure 5.
Figure 5.
Extensive clonotypic renewal within the T cell repertoire. We cloned and sequenced 90 clonal TCRs from sample pairs for each of the three patterns of evolution of CDR3 length diversity previously identified by spectratyping. One representative example is shown for each pattern. Colored pie slices represent individual expanded T clones. Color and size of slices allow identification of the expanded clones and show their relative frequency, respectively. The group of nonexpanded clones (unique sequences, not shown individually for their large numbers) is shown in light gray in pretransplant samples and in white in posttransplant samples and, within each one, the proportion of single clones persisting posttransplantation is shown in dark gray. For the predominant patterns in A and B, we found a prominent increase of unique sequences, denoting increased clonal diversity also within the same CDR3 length. Pre-existing expanded clones were either undetectable or reduced after therapy. In the less frequent pattern in C, preexisting or new expanded clones constituted the majority of the examined T repertoire after therapy.
Figure 6.
Figure 6.
Phenotypic changes of the peripheral T cell repertoire. (A) Transient increase of CD95/Fas+ in the CD4+ and CD8+ T cell subset in the early (peripheral) phases of immune reconstitution, suggesting an increased susceptibility to Fas/FasL-mediated apoptosis. Representative histograms from an individual patient are shown (left) as is a summary of data from all patients (right). (B) Persistent increase of CD8+/CD28−/CD57+ T cells during follow-up. (left) An individual patient's example of CD8+ gated analysis with the top histograms showing CD57 expression, and dot plots (bottom) showing CD57 (x axis) versus CD28 (y axis) expression. (right) The box plot summarizes the data from all patients. CD8+/CD28−/CD57+ have been characterized as a terminally differentiated subpopulation with short telomeres, defective cytolytic functions, and increased susceptibility to apoptosis.
Figure 7.
Figure 7.
Characterization of oligoclonally expanded CD8+ populations. (A) In the example shown, TCR Vβ back-gating in combination with CDR3 spectratyping of CD8+ T cells (top inlays: black box, preHSCT; red box, 6-mo follow-up) demonstrates that oligoclonal expanded cells, representing up to 50% of all CD8+ cells (B), are CD57+/CD45RA+/CD45RO+/CD27−/CD95(Fas)+ (C), consistent with the phenotype of atypical effector–memory cells that reached replicative senescence after extensive division in vivo.

References

    1. Hafler, D.A. 2004. Multiple sclerosis. J. Clin. Invest. 113:788–794.
    1. Muraro, P.A., R. Cassiani Ingoni, and R. Martin. 2003. Hematopoietic stem cell transplantation for multiple sclerosis: current status and future challenges. Curr. Opin. Neurol. 16:299–305.
    1. Fassas, A., and V.K. Kimiskidis. 2003. Stem cell transplantation for multiple sclerosis: what is the evidence? Blood Rev. 17:233–240.
    1. Burt, R.K., S. Slavin, W.H. Burns, and A.M. Marmont. 2002. Induction of tolerance in autoimmune diseases by hematopoietic stem cell transplantation: getting closer to a cure? Blood. 99:768–784.
    1. Fassas, A., A. Anagnostopoulos, A. Kazis, K. Kapinas, I. Sakellari, V. Kimiskidis, and A. Tsompanakou. 1997. Peripheral blood stem cell transplantation in the treatment of progressive multiple sclerosis: first results of a pilot study. Bone Marrow Transplant. 20:631–638.
    1. Burt, R.K., A.E. Traynor, B. Cohen, K.H. Karlin, F.A. Davis, D. Stefoski, C. Terry, L. Lobeck, E.J. Russell, C. Goolsby, et al. 1998. T cell-depleted autologous hematopoietic stem cell transplantation for multiple sclerosis: report on the first three patients. Bone Marrow Transplant. 21:537–541.
    1. Openshaw, H., B.T. Lund, A. Kashyap, R. Atkinson, I. Sniecinski, L.P. Weiner, and S. Forman. 2000. Peripheral blood stem cell transplantation in multiple sclerosis with busulfan and cyclophosphamide conditioning: report of toxicity and immunological monitoring. Biol. Blood Marrow Transplant. 6:563–575.
    1. Sun, W., U. Popat, G. Hutton, Y.C. Zang, R. Krance, G. Carrum, G.A. Land, H. Heslop, M. Brenner, and J.Z. Zhang. 2004. Characteristics of T-cell receptor repertoire and myelin-reactive T cells reconstituted from autologous haematopoietic stem-cell grafts in multiple sclerosis. Brain. 127:996–1008.
    1. Reimer, P., V. Kunzmann, M. Wilhelm, B. Weissbrich, D. Kraemer, H. Berghammer, and F. Weissinger. 2003. Cellular and humoral immune reconstitution after autologous peripheral blood stem cell transplantation (PBSCT). Ann. Hematol. 82:263–270.
    1. Koehne, G., W. Zeller, M. Stockschlaeder, and A.R. Zander. 1997. Phenotype of lymphocyte subsets after autologous peripheral blood stem cell transplantation. Bone Marrow Transplant. 19:149–156.
    1. Berzins, S.P., A.P. Uldrich, J.S. Sutherland, J. Gill, J.F. Miller, D.I. Godfrey, and R.L. Boyd. 2002. Thymic regeneration: teaching an old immune system new tricks. Trends Mol. Med. 8:469–476.
    1. Kivisakk, P., D.J. Mahad, M.K. Callahan, C. Trebst, B. Tucky, T. Wei, L. Wu, E.S. Baekkevold, H. Lassmann, S.M. Staugaitis, et al. 2003. Human cerebrospinal fluid central memory CD4+ T cells: evidence for trafficking through choroid plexus and meninges via P-selectin. Proc. Natl. Acad. Sci. USA. 100:8389–8394.
    1. Jameson, S.C. 2002. Maintaining the norm: T-cell homeostasis. Nat. Rev. Immunol. 2:547–556.
    1. Murali-Krishna, K., and R. Ahmed. 2000. Cutting edge: naive T cells masquerading as memory cells. J. Immunol. 165:1733–1737.
    1. Goldrath, A.W., L.Y. Bogatzki, and M.J. Bevan. 2000. Naive T cells transiently acquire a memory-like phenotype during homeostasis-driven proliferation. J. Exp. Med. 192:557–564.
    1. Kimmig, S., G.K. Przybylski, C.A. Schmidt, K. Laurisch, B. Mowes, A. Radbruch, and A. Thiel. 2002. Two subsets of naive T helper cells with distinct T cell receptor excision circle content in human adult peripheral blood. J. Exp. Med. 195:789–794.
    1. Douek, D.C., R.D. McFarland, P.H. Keiser, E.A. Gage, J.M. Massey, B.F. Haynes, M.A. Polis, A.T. Haase, M.B. Feinberg, J.L. Sullivan, et al. 1998. Changes in thymic function with age and during the treatment of HIV infection. Nature. 396:690–695.
    1. Pannetier, C., M. Cochet, S. Darche, A. Casrouge, M. Zoller, and P. Kourilsky. 1993. The sizes of the CDR3 hypervariable regions of the murine T-cell receptor beta chains vary as a function of the recombined germ-line segments. Proc. Natl. Acad. Sci. USA. 90:4319–4323.
    1. Gorski, J., M. Yassai, X. Zhu, B. Kissela, C. Keever, and N. Flomenberg. 1994. Circulating T cell repertoire complexity in normal individuals and bone marrow recipients analyzed by CDR3 spectratyping: correlation with the immune status. J. Immunol. 152:5109–5119.
    1. Brenchley, J.M., N.J. Karandikar, M.R. Betts, D.R. Ambrozak, B.J. Hill, L.E. Crotty, J.P. Casazza, J. Kuruppu, S.A. Migueles, M. Connors, et al. 2003. Expression of CD57 defines replicative senescence and antigen-induced apoptotic death of CD8+ T cells. Blood. 101:2711–2720.
    1. Mackall, C.L., T.A. Fleisher, M.R. Brown, M.P. Andrich, C.C. Chen, I.M. Feuerstein, M.E. Horowitz, I.T. Magrath, A.T. Shad, S.M. Steinberg, et al. 1995. Age, thymopoiesis, and CD4+ T-lymphocyte regeneration after intensive chemotherapy. N. Engl. J. Med. 332:143–149.
    1. Douek, D.C., R.A. Vescio, M.R. Betts, J.M. Brenchley, B.J. Hill, L. Zhang, J.R. Berenson, R.H. Collins, and R.A. Koup. 2000. Assessment of thymic output in adults after haematopoietic stem-cell transplantation and prediction of T-cell reconstitution. Lancet. 355:1875–1881.
    1. Hug, A., M. Korporal, I. Schroder, J. Haas, K. Glatz, B. Storch-Hagenlocher, and B. Wildemann. 2003. Thymic export function and T cell homeostasis in patients with relapsing remitting multiple sclerosis. J. Immunol. 171:432–437.
    1. Tanchot, C., A. Le Campion, B. Martin, S. Leaument, N. Dautigny, and B. Lucas. 2002. Conversion of naive T cells to a memory-like phenotype in lymphopenic hosts is not related to a homeostatic mechanism that fills the peripheral naive T cell pool. J. Immunol. 168:5042–5046.
    1. Ge, Q., H. Hu, H.N. Eisen, and J. Chen. 2002. Different contributions of thymopoiesis and homeostasis-driven proliferation to the reconstitution of naive and memory T cell compartments. Proc. Natl. Acad. Sci. USA. 99:2989–2994.
    1. Weiner, H.L., and J.A. Cohen. 2002. Treatment of multiple sclerosis with cyclophosphamide: critical review of clinical and immunologic effects. Mult. Scler. 8:142–154.
    1. Gbadamosi, J., C. Buhmann, W. Tessmer, A. Moench, F. Haag, and C. Heesen. 2003. Effects of mitoxantrone on multiple sclerosis patients' lymphocyte subpopulations and production of immunoglobulin, TNF-alpha and IL-10. Eur. Neurol. 49:137–141.
    1. Peggs, K.S., S. Verfuerth, A. Pizzey, N. Khan, P. Moss, A.H. Goldstone, K. Yong, and S. Mackinnon. 2003. Reconstitution of T-cell repertoire after autologous stem cell transplantation: influence of CD34 selection and cytomegalovirus infection. Biol. Blood Marrow Transplant. 9:198–205.
    1. Gorochov, G., P. Debre, V. Leblond, B. Sadat-Sowti, F. Sigaux, and B. Autran. 1994. Oligoclonal expansion of CD8+ CD57+ T cells with restricted T-cell receptor beta chain variability after bone marrow transplantation. Blood. 83:587–595.
    1. Morley, J.K., F.M. Batliwalla, R. Hingorani, and P.K. Gregersen. 1995. Oligoclonal CD8+ T cells are preferentially expanded in the CD57+ subset. J. Immunol. 154:6182–6190.
    1. Monteiro, J., F. Batliwalla, H. Ostrer, and P.K. Gregersen. 1996. Shortened telomeres in clonally expanded CD28-CD8+ T cells imply a replicative history that is distinct from their CD28+CD8+ counterparts. J. Immunol. 156:3587–3590.
    1. Hakim, F.T., R. Cepeda, S. Kaimei, C.L. Mackall, N. McAtee, J. Zujewski, K. Cowan, and R.E. Gress. 1997. Constraints on CD4 recovery postchemotherapy in adults: thymic insufficiency and apoptotic decline of expanded peripheral CD4 cells. Blood. 90:3789–3798.
    1. Singh, R.K., M.L. Varney, S. Buyukberber, K. Ino, A.G. Ageitos, E. Reed, S. Tarantolo, and J.E. Talmadge. 1999. Fas-FasL-mediated CD4+ T-cell apoptosis following stem cell transplantation. Cancer Res. 59:3107–3111.
    1. Mackall, C.L., T.A. Fleisher, M.R. Brown, M.P. Andrich, C.C. Chen, I.M. Feuerstein, I.T. Magrath, L.H. Wexler, D.S. Dimitrov, and R.E. Gress. 1997. Distinctions between CD8+ and CD4+ T-cell regenerative pathways result in prolonged T-cell subset imbalance after intensive chemotherapy. Blood. 89:3700–3707.
    1. Effros, R.B., and G. Pawelec. 1997. Replicative senescence of T cells: does the Hayflick Limit lead to immune exhaustion? Immunol. Today. 18:450–454.
    1. Selin, L.K., M.Y. Lin, K.A. Kraemer, D.M. Pardoll, J.P. Schneck, S.M. Varga, P.A. Santolucito, A.K. Pinto, and R.M. Welsh. 1999. Attrition of T cell memory: selective loss of LCMV epitope-specific memory CD8 T cells following infections with heterologous viruses. Immunity. 11:733–742.
    1. Autran, B., V. Leblond, B. Sadat-Sowti, E. Lefranc, P. Got, L. Sutton, J.L. Binet, and P. Debre. 1991. A soluble factor released by CD8+ CD57+ lymphocytes from bone marrow transplanted patients inhibits cell-mediated cytolysis. Blood. 77:2237–2241.
    1. Sadat-Sowti, B., P. Debre, L. Mollet, L. Quint, F. Hadida, V. Leblond, G. Bismuth, and B. Autran. 1994. An inhibitor of cytotoxic functions produced by CD8+CD57+ T lymphocytes from patients suffering from AIDS and immunosuppressed bone marrow recipients. Eur. J. Immunol. 24:2882–2888.
    1. Mollet, L., B. Sadat-Sowti, J. Duntze, V. Leblond, F. Bergeron, V. Calvez, C. Katlama, P. Debre, and B. Autran. 1998. CD8hi+CD57+ T lymphocytes are enriched in antigen-specific T cells capable of down-modulating cytotoxic activity. Int. Immunol. 10:311–323.
    1. Poser, C.M., D.W. Paty, L.C. Scheinberg, W.I. McDonald, F.A. Davis, G.C. Ebers, K.P. Johnson, K.P. Sibley, D.H. Silberberg, and W.W. Tourtellotte. 1983. New diagnostic criteria for multiple sclerosis: guidelines for research protocols. Ann. Neurol. 13:227–231.
    1. Kurtzke, J.F. 1983. Rating neurologic impairment in multiple sclerosis: an expanded disability status scale. Neurology. 33:1444–1452.
    1. Burt, R.K., B.A. Cohen, E. Russell, K. Spero, A. Joshi, Y. Oyama, W.J. Karpus, K. Luo, B. Jovanovic, A. Traynor, et al. 2003. Hematopoietic stem cell transplantation for progressive multiple sclerosis: failure of a total body irradiation-based conditioning regimen to prevent disease progression in patients with high disability scores. Blood. 102:2373–2378.
    1. Nash, R.A., J.D. Bowen, P.A. McSweeney, S.Z. Pavletic, K.R. Maravilla, M.S. Park, J. Storek, K.M. Sullivan, J. Al-Omaishi, J.R. Corboy, et al. 2003. High-dose immunosuppressive therapy and autologous peripheral blood stem cell transplantation for severe multiple sclerosis. Blood. 102:2364–2372.
    1. Inglese, M., G.L. Mancardi, E. Pagani, M.A. Rocca, A. Murialdo, R. Saccardi, G. Comi, and M. Filippi. 2004. Brain tissue loss occurs after suppression of enhancement in patients with multiple sclerosis treated with autologous haematopoietic stem cell transplantation. J. Neurol. Neurosurg. Psychiatry. 75:643–644.
    1. Mancardi, G.L., R. Saccardi, M. Filippi, F. Gualandi, A. Murialdo, M. Inglese, M.G. Marrosu, G. Meucci, L. Massacesi, A. Lugaresi, et al. 2001. Autologous hematopoietic stem cell transplantation suppresses Gd-enhanced MRI activity in MS. Neurology. 57:62–68.
    1. Carreras, E., A. Saiz, P. Marin, C. Martinez, M. Rovira, N. Villamor, M. Aymerich, M. Lozano, F. Fernandez-Aviles, A. Urbano-Izpizua, et al. 2003. CD34+ selected autologous peripheral blood stem cell transplantation for multiple sclerosis: report of toxicity and treatment results at one year of follow-up in 15 patients. Haematologica. 88:306–314.
    1. Saccardi, R., G.L. Mancardi, A. Solari, A. Bosi, P. Bruzzi, P. Di Bartolomeo, A. Donelli, M. Filippi, A. Guerrasio, F. Gualandi, et al. 2004. Autologous HSCT for severe progressive Multiple Sclerosis in a multicenter trial: impact on disease activity and quality of life. Blood. 10.1182/blood-2004-08-3205.
    1. Muraro, P.A., M. Jacobsen, A. Necker, J.W. Nagle, R. Gaber, N. Sommer, R. Martin, and B. Hemmer. 2000. Rapid identification of local T cell expansion in inflammatory organ diseases by flow cytometric T cell receptor Vb analysis. J. Immunol. Meth. 246:131–143.
    1. Arden, B., S.P. Clark, D. Kabelitz, and T.W. Mak. 1995. Human T-cell receptor variable gene segment families. Immunogenetics. 42:455–500.

Source: PubMed

3
订阅