Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

LYMPHOMA

High-dimensional and single-cell transcriptome analysis of the tumor microenvironment in angioimmunoblastic T cell lymphoma (AITL)

Abstract

Angioimmunoblastic T-cell lymphoma (AITL) is an aggressive lymphoid malignancy associated with a poor clinical prognosis. The AITL tumor microenvironment (TME) is unique, featuring a minority population of malignant CD4+ T follicular helper (TFH) cells inter-mixed with a diverse infiltrate of multi-lineage immune cells. While much of the understanding of AITL biology to date has focused on characteristics of the malignant clone, less is known about the many non-malignant populations that comprise the TME. Recently, mutational consistencies have been identified between malignant cells and non-malignant B cells within the AITL TME. As a result, a significant role for non-malignant populations in AITL biology has been increasingly hypothesized. In this study, we have utilized mass cytometry and single-cell transcriptome analysis to identify several expanded populations within the AITL TME. Notably, we find that B cells within the AITL TME feature decreased expression of key markers including CD73 and CXCR5. Furthermore, we describe the expansion of distinct CD8+ T cell populations that feature an exhausted phenotype and an underlying expression profile indicative of dysfunction, impaired cytotoxicity, and upregulation of the chemokines XCL2 and XCL1.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: AITL tumor microenvironment features CD8+ expansion, global shift in B cell phenotypes.
Fig. 2: CD4+ populations in the AITL TME.
Fig. 3: Global shift in AITL B cells marked by loss of CD73, CXCR5.
Fig. 4: Expansion of distinct CD8+ T cell populations within the AITL TME.
Fig. 5: Defining the RNA transcriptome of expanded CD8+ populations in AITL.
Fig. 6: Immunoblasts and NK cell populations in AITL.

Similar content being viewed by others

References

  1. Lunning MA, Vose JM. Angioimmunoblastic T-cell lymphoma: the many-faced lymphoma. Blood. 2017;129:1095–102.

    Article  CAS  PubMed  Google Scholar 

  2. Swerdlow SH, Campo E, Pileri SA, Harris NL, Stein H, Siebert R, et al. The 2016 revision of the World Health Organization classification of lymphoid neoplasms. Blood. 2016;127:2375–90.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Federico M, Rudiger T, Bellei M, Nathwani BN, Luminari S, Coiffier B, et al. Clinicopathologic characteristics of angioimmunoblastic T-Cell lymphoma: analysis of the international peripheral T-cell lymphoma project. J Clin Oncol. 2013;31:240–6.

    Article  CAS  PubMed  Google Scholar 

  4. Mhaidly R, Krug A, Gaulard P, Lemonnier F, Ricci J-E, Verhoeyen E New preclinical models for angioimmunoblastic T-cell lymphoma: filling the GAP. Oncogenesis. 2020; 9. https://doi.org/10.1038/s41389-020-00259-x.

  5. Chiba S, Sakata-Yanagimoto M. Advances in understanding of angioimmunoblastic T-cell lymphoma. Leukemia. 2020;34:2592–606.

    Article  PubMed  PubMed Central  Google Scholar 

  6. Lachenal F, Berger F, Ghesquières H, Biron P, Hot A, Callet-Bauchu E, et al. Angioimmunoblastic T-cell lymphoma: clinical and laboratory features at diagnosis in 77 patients. Medicine. 2007;86:282–92.

    Article  PubMed  Google Scholar 

  7. Gaulard P, de Leval L. The microenvironment in T-cell lymphomas: emerging themes. Semin Cancer Biol. 2014;24:49–60.

    Article  CAS  PubMed  Google Scholar 

  8. Palomero T, Couronné L, Khiabanian H, Kim M-Y, Ambesi-Impiombato A, Perez-Garcia A, et al. Recurrent mutations in epigenetic regulators, RHOA and FYN kinase in peripheral T cell lymphomas. Nat Genet. 2014;46:166–70.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Sakata-Yanagimoto M, Enami T, Yoshida K, Shiraishi Y, Ishii R, Miyake Y, et al. Somatic RHOA mutation in angioimmunoblastic T cell lymphoma. Nat Genet. 2014;46:171–5.

    Article  CAS  PubMed  Google Scholar 

  10. Yoo HY, Sung MK, Lee SH, Kim S, Lee H, Park S, et al. A recurrent inactivating mutation in RHOA GTPase in angioimmunoblastic T cell lymphoma. Nat Genet. 2014;46:371–5.

    Article  CAS  PubMed  Google Scholar 

  11. Lemonnier F, Cairns RA, Inoue S, Li WY, Dupuy A, Broutin S, et al. The IDH2 R172K mutation associated with angioimmunoblastic T-cell lymphoma produces 2HG in T cells and impacts lymphoid development. Proc Natl Acad Sci USA. 2016;113:15084–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Ng SY, Brown L, Stevenson K, deSouza T, Aster JC, Louissaint A, et al. RhoA G17V is sufficient to induce autoimmunity and promotes T-cell lymphomagenesis in mice. Blood. 2018;132:935–47.

    Article  CAS  PubMed  Google Scholar 

  13. Schwartz FH, Cai Q, Fellmann E, Hartmann S, Mäyränpää MI, Karjalainen-Lindsberg M-L, et al. TET2 mutations in B cells of patients affected by angioimmunoblastic T-cell lymphoma. J Pathol. 2017;242:129–33.

    Article  CAS  PubMed  Google Scholar 

  14. Lemonnier F, Mak TW. Angioimmunoblastic T-cell lymphoma: more than a disease of T follicular helper cells. J Pathol. 2017;242:387–90.

    Article  CAS  PubMed  Google Scholar 

  15. Yao W-Q, Wu F, Zhang W, Chuang S-S, Thompson JS, Chen Z, et al. Angioimmunoblastic T-cell lymphoma contains multiple clonal T-cell populations derived from a common TET2 mutant progenitor cell. J Pathol. 2020;250:346–57.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Cader FZ, Schackmann RCJ, Hu X, Wienand K, Redd R, Chapuy B, et al. Mass cytometry of Hodgkin lymphoma reveals a CD4+ regulatory T-cell-rich and exhausted T-effector microenvironment. Blood. 2018;132:825–36.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Neeland MR, Andorf S, Manohar M, Dunham D, Lyu S-C, Dang TD, et al. Mass cytometry reveals cellular fingerprint associated with IgE+ peanut tolerance and allergy in early life. Nat Commun. 2020;11:1091.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Kudryavtsev I, Serebriakova M, Starshinova A, Zinchenko Y, Basantsova N, Malkova A, et al. Imbalance in B cell and T follicular helper cell subsets in pulmonary sarcoidosis. Sci Rep. 2020;10:1059.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Verma K, Ogonek J, Varanasi PR, Luther S, Bünting I, Thomay K, et al. Human CD8+ CD57- TEMRA cells: Too young to be called ‘old’. PloS ONE. 2017;12:e0177405.

    Article  PubMed  PubMed Central  Google Scholar 

  20. Maggi L, Santarlasci V, Capone M, Rossi MC, Querci V, Mazzoni A, et al. Distinctive features of classic and nonclassic (Th17 derived) human Th1 cells. Eur J Immunol. 2012;42:3180–8.

    Article  CAS  PubMed  Google Scholar 

  21. Yang Z-Z, Kim HJ, Villasboas JC, Price-Troska T, Jalali S, Wu H, et al. Mass cytometry analysis reveals that specific intratumoral CD4+ T cell subsets correlate with patient survival in follicular lymphoma. Cell Rep. 2019;26:2178–2193.e3.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Kotecha N, Krutzik PO, Irish JM. Web-based analysis and publication of flow cytometry experiments. Curr Protoc Cytom. 2010;10, https://doi.org/10.1002/0471142956.cy1017s53.

  23. Amir ED, 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;31:545–52.

    Article  CAS  PubMed Central  Google Scholar 

  24. Amir E-AD, Lee B, Badoual P, Gordon M, Guo XV, Merad M, et al. Development of a comprehensive antibody staining database using a standardized analytics pipeline. Front Immunol. 2019;10:1315.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Li H, van der Leun AM, Yofe I, Lubling Y, Gelbard-Solodkin D, van Akkooi ACJ, et al. Dysfunctional CD8 T cells form a proliferative, dynamically regulated compartment within human melanoma. Cell. 2019;176:775–789.e18.

    Article  CAS  PubMed  Google Scholar 

  26. Thommen DS, Koelzer VH, Herzig P, Roller A, Trefny M, Dimeloe S, et al. A transcriptionally and functionally distinct PD-1+ CD8+ T cell pool with predictive potential in non-small-cell lung cancer treated with PD-1 blockade. Nat Med. 2018;24:994–1004.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Jansen CS, Prokhnevska N, Master VA, Sanda MG, Carlisle JW, Bilen MA, et al. An intra-tumoral niche maintains and differentiates stem-like CD8 T cells. Nature. 2019;576:465–70.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Nicolet BP, Guislain A, van Alphen FPJ, Gomez-Eerland R, Schumacher TNM, van den Biggelaar M, et al. CD29 identifies IFN-γ-producing human CD8+ T cells with an increased cytotoxic potential. Proc Natl Acad Sci USA. 2020;117:6686–96.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Fox JC, Nakayama T, Tyler RC, Sander TL, Yoshie O, Volkman BF. Structural and agonist properties of XCL2, the other member of the C-chemokine subfamily. Cytokine. 2015;71:302–11.

    Article  CAS  PubMed  Google Scholar 

  30. Rhodes JW, Tong O, Harman AN, Turville SG. Human dendritic cell subsets, ontogeny, and impact on HIV infection. Front Immunol. 2019;10:1088.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Lei Y, Takahama Y. XCL1 and XCR1 in the immune system. Microbes Infect. 2012;14:262–7.

    Article  CAS  PubMed  Google Scholar 

  32. Kroczek RA, Henn V. The role of XCR1 and its ligand XCL1 in antigen cross-presentation by murine and human dendritic cells. Front Immunol. 2012;3:14.

    Article  PubMed  PubMed Central  Google Scholar 

  33. Ohta T, Sugiyama M, Hemmi H, Yamazaki C, Okura S, Sasaki I, et al. Crucial roles of XCR1-expressing dendritic cells and the XCR1-XCL1 chemokine axis in intestinal immune homeostasis. Sci Rep. 2016;6:23505.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Mizumoto Y, Hemmi H, Katsuda M, Miyazawa M, Kitahata Y, Miyamoto A, et al. Anticancer effects of chemokine-directed antigen delivery to a cross-presenting dendritic cell subset with immune checkpoint blockade. Br J Cancer. 2020;122:1185–93.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Troxell ML, Schwartz EJ, van de Rijn M, Ross DT, Warnke RA, Higgins JP, et al. Follicular dendritic cell immunohistochemical markers in angioimmunoblastic T-cell lymphoma. Appl Immunohistochem Mol Morphol AIMM. 2005;13:297–303.

    Article  CAS  PubMed  Google Scholar 

  36. Aguzzi A, Kranich J, Krautler NJ. Follicular dendritic cells: origin, phenotype, and function in health and disease. Trends Immunol. 2014;35:105–13.

    Article  CAS  PubMed  Google Scholar 

  37. Kim E-S, Ackermann C, Tóth I, Dierks P, Eberhard JM, Wroblewski R, et al. Down-regulation of CD73 on B cells of patients with viremic HIV correlates with B cell activation and disease progression. J Leukoc Biol. 2017;101:1263–71.

    Article  CAS  PubMed  Google Scholar 

  38. Kaku H, Cheng KF, Al-Abed Y, Rothstein TL. A novel mechanism of B cell-mediated immune suppression through CD73 expression and adenosine production. J Immunol Balt Md. 2014;193:5904–13.

    CAS  Google Scholar 

  39. Conter LJ, Song E, Shlomchik MJ, Tomayko MM. CD73 expression is dynamically regulated in the germinal center and bone marrow plasma cells are diminished in its absence. PloS ONE. 2014;9:e92009.

    Article  PubMed  PubMed Central  Google Scholar 

  40. Bowser JL, Blackburn MR, Shipley GL, Molina JG, Dunner K, Broaddus RR. Loss of CD73-mediated actin polymerization promotes endometrial tumor progression. J Clin Investig. 2016;126:220–38.

    Article  PubMed  Google Scholar 

  41. Ohtani H, Komeno T, Agatsuma Y, Kobayashi M, Noguchi M, Nakamura N. Follicular dendritic cell meshwork in angioimmunoblastic T-cell lymphoma is characterized by accumulation of CXCL13(+) cells. J Clin Exp Hematop JCEH. 2015;55:61–69.

    Article  PubMed  Google Scholar 

Download references

Acknowledgements

The authors acknowledge grant funding in support of this work from the Leukemia & Lymphoma Society (LLS), the National Institutes of Health (P50 CA97274), and the Predolin Foundation.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Stephen M. Ansell.

Ethics declarations

Conflict of interest

For JRC research funding provided by NanoString and BMS/Celgene. For SMA, research funding provided by Bristol Myers Squibb, ADC Therapeutics, Seattle Genetics, Regeneron, Affimed, AI Therapeutics, Trillium and Takeda. The remaining authors report nothing to disclose.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Pritchett, J.C., Yang, ZZ., Kim, H.J. et al. High-dimensional and single-cell transcriptome analysis of the tumor microenvironment in angioimmunoblastic T cell lymphoma (AITL). Leukemia 36, 165–176 (2022). https://doi.org/10.1038/s41375-021-01321-2

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41375-021-01321-2

This article is cited by

Search

Quick links