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.

  • Review Article
  • Published:

The molecular basis of the memory T cell response: differential gene expression and its epigenetic regulation

Key Points

  • Memory T cells, which are derived from naive T cells and carry the 'memory' of a previous exposure to an antigen, are long lived, have enhanced functional capacity compared with naive T cells and serve as the cellular basis of immunological memory.

  • Memory T cells are heterogeneous populations and can be divided into two main subsets: central memory T cells (which have stem cell-like properties and circulate in lymphoid organs) and effector memory T cells (which circulate in non-lymphoid tissues and perform rapid effector functions following activation).

  • Compared with naive T cells, memory T cells highly express a few hundred genes that have an array of functions and serve as the transcriptional basis for the unique function of memory T cells.

  • The highly expressed genes in memory T cells can be further divided into two main classes based on the pattern of their expression before and after antigen-mediated T cell activation. The first group includes genes that are highly expressed by resting memory T cells but not by resting naive T cells, whereas the second group includes genes that are expressed more highly by activated memory T cells than by activated naive T cells. Genes that are expressed at similar levels in resting naive and memory T cells but are more highly expressed by activated memory T cells than by activated naive T cells are called 'poised' genes.

  • Chromatin states (open or closed) are regulated by chemical modifications of DNA and histones; such modifications are considered to be epigenetic changes. The particular patterns of DNA methylation and histone modification are closely associated with the expression status of memory T cell highly expressed genes and may have a key role in the regulation of the differential gene expression and function of memory T cells.

  • A better understanding of the functions and epigenetic regulation of highly expressed genes in memory T cells will enable us to elucidate how memory T cell formation, maintenance and function, as well as the consequences of dysregulation of epigenetic changes, contribute to the altered function of the immune system in degenerative processes, such as autoimmunity and ageing.

Abstract

How the immune system remembers a previous encounter with a pathogen and responds more efficiently to a subsequent encounter has been one of the central enigmas for immunologists for over a century. The identification of pathogen-specific memory lymphocytes that arise after an infection provided a cellular basis for immunological memory. But the molecular mechanisms of immunological memory remain only partially understood. The emerging evidence suggests that epigenetic changes have a key role in controlling the distinct transcriptional profiles of memory lymphocytes and thus in shaping their function. In this Review, we summarize the recent progress that has been made in assessing the differential gene expression and chromatin modifications in memory CD4+ and CD8+ T cells, and we present our current understanding of the molecular basis of memory T cell function.

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

Figure 1: Comparison of overall gene expression in naive and memory T cells.
Figure 2: Types of genes that are differentially expressed in memory T cells based on their expression kinetics before and after T cell activation.
Figure 3: The chromatin basis for differential gene expression in memory T cells involves histone methylation.

Similar content being viewed by others

References

  1. Bevan, M. J. Understand memory, design better vaccines. Nature Immunol. 12, 463–465 (2011).

    Article  CAS  Google Scholar 

  2. Pulendran, B. & Ahmed, R. Immunological mechanisms of vaccination. Nature Immunol. 12, 509–517 (2011).

    Article  CAS  Google Scholar 

  3. Sallusto, F., Lenig, D., Forster, R., Lipp, M. & Lanzavecchia, A. Two subsets of memory T lymphocytes with distinct homing potentials and effector functions. Nature 401, 708–712 (1999).

    Article  CAS  PubMed  Google Scholar 

  4. Pepper, M. & Jenkins, M. K. Origins of CD4+ effector and central memory T cells. Nature Immunol. 12, 467–471 (2011).

    Article  CAS  Google Scholar 

  5. Hyatt, G. et al. Gene expression microarrays: glimpses of the immunological genome. Nature Immunol. 7, 686–691 (2006).

    Article  CAS  Google Scholar 

  6. Haining, W. N. & Wherry, E. J. Integrating genomic signatures for immunologic discovery. Immunity 32, 152–161 (2010).

    Article  CAS  PubMed  Google Scholar 

  7. Kaech, S. M., Hemby, S., Kersh, E. & Ahmed, R. Molecular and functional profiling of memory CD8 T cell differentiation. Cell 111, 837–851 (2002). This pioneering study showed the changes in global gene expression that occur as CD8+ naive T cells differentiate into effector and then memory T cells after viral infection and suggested that antigen-specific CD8+ T cells progressively differentiate into memory T cells following viral infection.

    Article  CAS  PubMed  Google Scholar 

  8. Liu, K. et al. Augmentation in expression of activation-induced genes differentiates memory from naive CD4+ T cells and is a molecular mechanism for enhanced cellular response of memory CD4+ T cells. J. Immunol. 166, 7335–7344 (2001).

    Article  CAS  PubMed  Google Scholar 

  9. Holmes, S., He, M., Xu, T. & Lee, P. P. Memory T cells have gene expression patterns intermediate between naive and effector. Proc. Natl Acad. Sci. USA 102, 5519–5523 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Willinger, T., Freeman, T., Hasegawa, H., McMichael, A. J. & Callan, M. F. Molecular signatures distinguish human central memory from effector memory CD8 T cell subsets. J. Immunol. 175, 5895–5903 (2005).

    Article  CAS  PubMed  Google Scholar 

  11. Luckey, C. J. et al. Memory T and memory B cells share a transcriptional program of self-renewal with long-term hematopoietic stem cells. Proc. Natl Acad. Sci. USA 103, 3304–3309 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Haining, W. N. et al. Identification of an evolutionarily conserved transcriptional signature of CD8 memory differentiation that is shared by T and B cells. J. Immunol. 181, 1859–1868 (2008).

    Article  CAS  PubMed  Google Scholar 

  13. Araki, Y. et al. Genome-wide analysis of histone methylation reveals chromatin state-based regulation of gene transcription and function of memory CD8+ T cells. Immunity 30, 912–925 (2009). This study identifies four different modes of association between histone methylation and differential gene expression in CD8+ memory T cells, providing a chromatin basis for the differential gene expression and function of memory CD8+ T cells.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Wirth, T. C. et al. Repetitive antigen stimulation induces stepwise transcriptome diversification but preserves a core signature of memory CD8+ T cell differentiation. Immunity 33, 128–140 (2010). This study reveals signature genes of CD8+ memory T cells and their complex regulation following repeated antigenic challenges.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Chevalier, N. et al. CXCR5 expressing human central memory CD4 T cells and their relevance for humoral immune responses. J. Immunol. 186, 5556–5568 (2011).

    Article  CAS  PubMed  Google Scholar 

  16. Novershtern, N. et al. Densely interconnected transcriptional circuits control cell states in human hematopoiesis. Cell 144, 296–309 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Marshall, H. D. et al. Differential expression of Ly6C and T-bet distinguish effector and memory Th1 CD4+ cell properties during viral infection. Immunity 35, 633–646 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Rossi, R. L. et al. Distinct microRNA signatures in human lymphocyte subsets and enforcement of the naive state in CD4+ T cells by the microRNA miR-125b. Nature Immunol. 12, 796–803 (2011).

    Article  CAS  Google Scholar 

  19. Hertoghs, K. M. et al. Molecular profiling of cytomegalovirus-induced human CD8+ T cell differentiation. J. Clin. Invest. 120, 4077–4090 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Kato, K. et al. Identification of stem cell transcriptional programs normally expressed in embryonic and neural stem cells in alloreactive CD8+ T cells mediating graft-versus-host disease. Biol. Blood Marrow Transplant. 16, 751–771 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Turtle, C. J. et al. Innate signals overcome acquired TCR signaling pathway regulation and govern the fate of human CD161hi CD8α+ semi-invariant T cells. Blood 118, 2752–2762 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Lai, W. et al. Transcriptional control of rapid recall by memory CD4 T cells. J. Immunol. 187, 133–140 (2011).

    Article  CAS  PubMed  Google Scholar 

  23. Bonasio, R., Tu, S. & Reinberg, D. Molecular signals of epigenetic states. Science 330, 612–616 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Berger, S. L. The complex language of chromatin regulation during transcription. Nature 447, 407–412 (2007).

    Article  CAS  PubMed  Google Scholar 

  25. Berger, S. L., Kouzarides, T., Shiekhattar, R. & Shilatifard, A. An operational definition of epigenetics. Genes Dev. 23, 781–783 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Guenther, M. G., Levine, S. S., Boyer, L. A., Jaenisch, R. & Young, R. A. A chromatin landmark and transcription initiation at most promoters in human cells. Cell 130, 77–88 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Barski, A. et al. High-resolution profiling of histone methylations in the human genome. Cell 129, 823–837 (2007).

    CAS  PubMed  Google Scholar 

  28. Zhang, Z. & Pugh, B. F. High-resolution genome-wide mapping of the primary structure of chromatin. Cell 144, 175–186 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Deaton, A. M. & Bird, A. CpG islands and the regulation of transcription. Genes Dev. 25, 1010–1022 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Saxonov, S., Berg, P. & Brutlag, D. L. A genome-wide analysis of CpG dinucleotides in the human genome distinguishes two distinct classes of promoters. Proc. Natl Acad. Sci. USA 103, 1412–1417 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Maunakea, A. K. et al. Conserved role of intragenic DNA methylation in regulating alternative promoters. Nature 466, 253–257 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Avni, O. et al. TH cell differentiation is accompanied by dynamic changes in histone acetylation of cytokine genes. Nature Immunol. 3, 643–651 (2002).

    Article  CAS  Google Scholar 

  33. Chang, S. & Aune, T. M. Dynamic changes in histone-methylation 'marks' across the locus encoding interferon-γ during the differentiation of T helper type 2 cells. Nature Immunol. 8, 723–731 (2007).

    Article  CAS  Google Scholar 

  34. Northrup, D. L. & Zhao, K. Application of ChIP-Seq and related techniques to the study of immune function. Immunity 34, 830–842 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Syrbe, U. et al. Differential regulation of P-selectin ligand expression in naive versus memory CD4+ T cells: evidence for epigenetic regulation of involved glycosyltransferase genes. Blood 104, 3243–3248 (2004).

    Article  CAS  PubMed  Google Scholar 

  36. Schmidl, C. et al. Epigenetic reprogramming of the RORC locus during in vitro expansion is a distinctive feature of human memory but not naive Treg. Eur. J. Immunol. 41, 1491–1498 (2011).

    Article  CAS  PubMed  Google Scholar 

  37. Steinfelder, S. et al. Epigenetic modification of the human CCR6 gene is associated with stable CCR6 expression in T cells. Blood 117, 2839–2846 (2011).

    Article  CAS  PubMed  Google Scholar 

  38. Fitzpatrick, D. R., Shirley, K. M. & Kelso, A. Stable epigenetic inheritance of regional IFN-γ promoter demethylation in CD44highCD8+ T lymphocytes. J. Immunol. 162, 5053–5057 (1999).

    CAS  PubMed  Google Scholar 

  39. Kersh, E. N. et al. Rapid demethylation of the IFN-γ gene occurs in memory but not naive CD8 T cells. J. Immunol. 176, 4083–4093 (2006).

    Article  CAS  PubMed  Google Scholar 

  40. Northrop, J. K., Thomas, R. M., Wells, A. D. & Shen, H. Epigenetic remodeling of the IL-2 and IFN-γ loci in memory CD8 T cells is influenced by CD4 T cells. J. Immunol. 177, 1062–1069 (2006).

    Article  CAS  PubMed  Google Scholar 

  41. Yamashita, M. et al. Bmi1 regulates memory CD4 T cell survival via repression of the Noxa gene. J. Exp. Med. 205, 1109–1120 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Youngblood, B. et al. Chronic virus infection enforces demethylation of the locus that encodes PD-1 in antigen-specific CD8+ T cells. Immunity 35, 400–412 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Deaton, A. M. et al. Cell type-specific DNA methylation at intragenic CpG islands in the immune system. Genome Res. 21, 1074–1086 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Fields, P. E., Kim, S. T. & Flavell, R. A. Changes in histone acetylation at the IL-4 and IFN-γ loci accompany Th1/Th2 differentiation. J. Immunol. 169, 647–650 (2002).

    Article  CAS  PubMed  Google Scholar 

  45. Messi, M. et al. Memory and flexibility of cytokine gene expression as separable properties of human TH1 and TH2 lymphocytes. Nature Immunol. 4, 78–86 (2003).

    Article  CAS  Google Scholar 

  46. Yamashita, M. et al. Interleukin (IL)-4-independent maintenance of histone modification of the IL-4 gene loci in memory Th2 cells. J. Biol. Chem. 279, 39454–39464 (2004).

    Article  CAS  PubMed  Google Scholar 

  47. Fann, M. et al. Histone acetylation is associated with differential gene expression in the rapid and robust memory CD8+ T cell response. Blood 108, 3363–3370 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Araki, Y., Fann, M., Wersto, R. & Weng, N. P. Histone acetylation facilitates rapid and robust memory CD8 T cell response through differential expression of effector molecules (eomesodermin and its targets: perforin and granzyme B). J. Immunol. 180, 8102–8108 (2008).

    Article  CAS  PubMed  Google Scholar 

  49. Northrop, J. K., Wells, A. D. & Shen, H. Chromatin remodeling as a molecular basis for the enhanced functionality of memory CD8 T cells. J. Immunol. 181, 865–868 (2008).

    Article  CAS  PubMed  Google Scholar 

  50. Wei, G. et al. Global mapping of H3K4me3 and H3K27me3 reveals specificity and plasticity in lineage fate determination of differentiating CD4+ T cells. Immunity 30, 155–167 (2009). This study identifies the chromatin states of key transcription factor genes and their target genes in naive CD4+ T cells differentiating into distinct lineages (including T H 1, T H 2, T H 17 and inducible regulatory T cells) and suggests that an epigenetic mechanism underlies the specificity and plasticity of effector and regulatory T cells.

    Article  PubMed  PubMed Central  Google Scholar 

  51. Nakata, Y. et al. c-Myb, Menin, GATA-3, and MLL form a dynamic transcription complex that plays a pivotal role in human T helper type 2 cell development. Blood 116, 1280–1290 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Yamashita, M. et al. Crucial role of MLL for the maintenance of memory T helper type 2 cell responses. Immunity 24, 611–622 (2006). This study demonstrates that MLL regulates histone modifications and expression at the GATA3 locus, indicating that MLL has a crucial role in the maintenance of memory T H 2 cells.

    Article  CAS  PubMed  Google Scholar 

  53. Zediak, V. P., Johnnidis, J. B., Wherry, E. J. & Berger, S. L. Persistently open chromatin at effector gene loci in resting memory CD8+ T cells independent of transcriptional status. J. Immunol. 186, 2705–2709 (2011).

    Article  CAS  PubMed  Google Scholar 

  54. Cannarile, M. A. et al. Transcriptional regulator Id2 mediates CD8+ T cell immunity. Nature Immunol. 7, 1317–1325 (2006).

    Article  CAS  Google Scholar 

  55. Bernstein, B. E. et al. A bivalent chromatin structure marks key developmental genes in embryonic stem cells. Cell 125, 315–326 (2006).

    Article  CAS  PubMed  Google Scholar 

  56. Cui, K. et al. Chromatin signatures in multipotent human hematopoietic stem cells indicate the fate of bivalent genes during differentiation. Cell Stem Cell 4, 80–93 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Seit-Nebi, A., Cheng, W., Xu, H. & Han, J. MLK4 has negative effect on TLR4 signaling. Cell. Mol. Immunol. 9, 27–33 (2012).

    Article  CAS  PubMed  Google Scholar 

  58. Murphy, K. M. & Stockinger, B. Effector T cell plasticity: flexibility in the face of changing circumstances. Nature Immunol. 11, 674–680 (2010).

    CAS  Google Scholar 

  59. Lu, K. T. et al. Functional and epigenetic studies reveal multistep differentiation and plasticity of in vitro-generated and in vivo-derived follicular T helper cells. Immunity 35, 622–632 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Wang, Z. et al. Combinatorial patterns of histone acetylations and methylations in the human genome. Nature Genet. 40, 897–903 (2008).

    Article  CAS  PubMed  Google Scholar 

  61. Thomson, J. P. et al. CpG islands influence chromatin structure via the CpG-binding protein Cfp1. Nature 464, 1082–1086 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Wang, Z. et al. Genome-wide mapping of HATs and HDACs reveals distinct functions in active and inactive genes. Cell 138, 1019–1031 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. van der Vlag, J. & Otte, A. P. Transcriptional repression mediated by the human polycomb-group protein EED involves histone deacetylation. Nature Genet. 23, 474–478 (1999).

    Article  CAS  PubMed  Google Scholar 

  64. Zediak, V. P., Wherry, E. J. & Berger, S. L. The contribution of epigenetic memory to immunologic memory. Curr. Opin. Genet. Dev. 21, 154–159 (2011).

    Article  CAS  PubMed  Google Scholar 

  65. Nakaya, H. I. et al. Systems biology of vaccination for seasonal influenza in humans. Nature Immunol. 12, 786–795 (2011).

    Article  CAS  Google Scholar 

  66. Lu, Q. et al. Demethylation of CD40LG on the inactive X in T cells from women with lupus. J. Immunol. 179, 6352–6358 (2007).

    Article  CAS  PubMed  Google Scholar 

  67. Hu, N. et al. Abnormal histone modification patterns in lupus CD4+ T cells. J. Rheumatol. 35, 804–810 (2008).

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank R. Hodes, K. Zhao and the anonymous reviewers for critical reading of the manuscript and helpful suggestions. This research was supported by the Intramural Research Programs of the US National Institute on Aging, National Institutes of Health.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Nan-ping Weng.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Related links

Related links

FURTHER INFORMATION

Gene Expression Omnibus

Glossary

Epigenetic regulation

The modifications on DNA, histones and other targets that collectively determine a stable phenotype without altering the DNA sequence. Epigenetic changes can pass from the parental cells to their offspring and provide a molecular basis for cellular memory.

Chromatin

The combination of DNA, histones and other proteins that comprises eukaryotic chromosomes. The basic repeating unit of chromatin is the nucleosome, which consists of an octamer of histone proteins around which 146 base pairs of DNA is wound.

Central memory T cells

(TCM cells). Antigen-experienced T cells that lack immediate effector function but can mediate rapid recall responses. They also rapidly develop the phenotype and function of effector memory T cells after re-stimulation with antigen. TCM cells retain the migratory properties of naive T cells and therefore circulate through the secondary lymphoid organs.

Effector memory T cells

(TEM cells). Terminally differentiated T cells that lack lymph node-homing receptors but express receptors that enable them to home to inflamed tissues. TEM cells can exert immediate effector functions without the need for further differentiation.

Microarray

A tool for measuring gene transcription. Its use involves the hybridization of fluorescently labelled cDNA prepared from a cell or tissue of interest with thousands of known oligonucleotides or cDNAs dotted on glass slides or other surfaces. The known DNA ideally represents all of the expressed genes in the species.

Heterochromatin

High-density regions in the nucleus that are thought to contain compacted chromatin structures associated with silent genes.

Chromatin immunoprecipitation

A technique that uses antibodies specific for transcription factors or other DNA-binding proteins to precipitate associated DNA sequences from chromatin to study their functional relationship.

Reverse transcription PCR

A type of PCR in which RNA is converted into complementary DNA (cDNA), which is then amplified.

ChIP–seq

A technique in which chromatin immunoprecipitation (ChIP) is followed by high-throughput sequencing to generate a genome-wide distribution map of protein–DNA interactions. This technique can be used to measure transcription factor binding and histone modifications.

Toll-like receptor

(TLR). A member of a family of receptors that are homologous to Drosophila melanogaster Toll. TLRs recognize conserved molecular patterns that are unique to microorganisms. The lipopolysaccharide component of bacterial cell walls is one such ligand. TLRs can also recognize mammalian components and contribute to autoimmunity.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Weng, Np., Araki, Y. & Subedi, K. The molecular basis of the memory T cell response: differential gene expression and its epigenetic regulation. Nat Rev Immunol 12, 306–315 (2012). https://doi.org/10.1038/nri3173

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nri3173

This article is cited by

Search

Quick links

Nature Briefing Microbiology

Sign up for the Nature Briefing: Microbiology newsletter — what matters in microbiology research, free to your inbox weekly.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing: Microbiology