CD4+ T Cell-Receptor Repertoire Diversity is Compromised in the Spleen but Not in the Bone Marrow of Aged Mice Due to Private and Sporadic Clonal Expansions

Reduction in T cell receptor (TCR) diversity in old age is considered as a major cause for immune complications in the elderly population. Here, we explored the consequences of aging on the TCR repertoire in mice using high-throughput sequencing (TCR-seq). We mapped the TCRβ repertoire of CD4+ T cells isolated from bone marrow (BM) and spleen of young and old mice. We found that TCRβ diversity is reduced in spleens of aged mice but not in their BM. Splenic CD4+ T cells were also skewed toward an effector memory phenotype in old mice, while BM cells preserved their memory phenotype with age. Analysis of Vβ and Jβ gene usage across samples, as well as comparison of CDR3 length distributions, showed no significant age dependent changes. However, comparison of the frequencies of amino-acid (AA) TCRβ sequences between samples revealed repertoire changes that occurred at a more refined scale. The BM-derived TCRβ repertoire was found to be similar among individual mice regardless of their age. In contrast, the splenic repertoire of old mice was not similar to those of young mice, but showed an increased similarity with the BM repertoire. Each old-mouse had a private set of expanded TCRβ sequences. Interestingly, a fraction of these sequences was found also in the BM of the same individual, sharing the same nucleotide sequence. Together, these findings show that the composition and phenotype of the CD4+ T cell BM repertoire are relatively stable with age, while diversity of the splenic repertoire is severely reduced. This reduction is caused by idiosyncratic expansions of tens to hundreds of T cell clonotypes, which dominate the repertoire of each individual. We suggest that these private and abundant clonotypes are generated by sporadic clonal expansions, some of which correspond to pre-existing BM clonotypes. These organ- and age-specific changes of the TCRβ repertoire have implications for understanding and manipulating age-associated immune decline.

[1]  M. Blackman,et al.  The narrowing of the CD8 T cell repertoire in old age. , 2011, Current opinion in immunology.

[2]  F. Miedema,et al.  Maintenance of peripheral naive T cells is sustained by thymus output in mice but not humans. , 2012, Immunity.

[3]  Daniel C. Douek,et al.  Convergent recombination shapes the clonotypic landscape of the naïve T-cell repertoire , 2010, Proceedings of the National Academy of Sciences.

[4]  Rustom Antia,et al.  Peripheral selection rather than thymic involution explains sudden contraction in naive CD4 T-cell diversity with age , 2012, Proceedings of the National Academy of Sciences.

[5]  F. Greenway,et al.  Obesity Increases the Production of Proinflammatory Mediators from Adipose Tissue T Cells and Compromises TCR Repertoire Diversity: Implications for Systemic Inflammation and Insulin Resistance , 2010, The Journal of Immunology.

[6]  C. Weyand,et al.  Understanding immunosenescence to improve responses to vaccines , 2013, Nature Immunology.

[7]  Katherine Kedzierska,et al.  Methods for comparing the diversity of samples of the T cell receptor repertoire. , 2007, Journal of immunological methods.

[8]  Peter Dalgaard,et al.  R Development Core Team (2010): R: A language and environment for statistical computing , 2010 .

[9]  C. Weyand,et al.  Aging and T-cell diversity , 2007, Experimental Gerontology.

[10]  R. Miller,et al.  Idiosyncratic alterations of TCR size distributions affecting both CD4 and CD8 T cell subsets in aging mice. , 1998, Cellular immunology.

[11]  N. Friedman,et al.  CNS-specific immunity at the choroid plexus shifts toward destructive Th2 inflammation in brain aging , 2013, Proceedings of the National Academy of Sciences.

[12]  R. Pabst,et al.  The bone marrow: a nest for migratory memory T cells. , 2005, Trends in immunology.

[13]  Robin Callard,et al.  Quantifying Thymic Export: Combining Models of Naive T Cell Proliferation and TCR Excision Circle Dynamics Gives an Explicit Measure of Thymic Output1 , 2009, The Journal of Immunology.

[14]  Wilfred Ndifon,et al.  Chromatin conformation governs T-cell receptor Jβ gene segment usage , 2012, Proceedings of the National Academy of Sciences.

[15]  J. Nikolich-Žugich Ageing and life-long maintenance of T-cell subsets in the face of latent persistent infections , 2008, Nature Reviews Immunology.

[16]  I. Kang,et al.  Age-Associated Change in the Frequency of Memory CD4+ T Cells Impairs Long Term CD4+ T Cell Responses to Influenza Vaccine1 , 2004, The Journal of Immunology.

[17]  Janko Nikolich- Zcaron,et al.  Ageing and life-long maintenance of T-cell subsets in the face of latent persistent infections. , 2008 .

[18]  P. Linton,et al.  Age-related changes in lymphocyte development and function , 2004, Nature Immunology.

[19]  E. Sercarz,et al.  Limitations in plasticity of the T-cell receptor repertoire. , 1991, Proceedings of the National Academy of Sciences of the United States of America.

[20]  A. Lustig,et al.  T Cell Aging: A Review of the Transcriptional Changes Determined from Genome-Wide Analysis , 2013, Front. Immunol..

[21]  Charles P. Lin,et al.  In vivo imaging of Treg cells providing immune privilege to the haematopoietic stem-cell niche , 2011, Nature.

[22]  Andreas Radbruch,et al.  Professional memory CD4+ T lymphocytes preferentially reside and rest in the bone marrow. , 2009, Immunity.

[23]  L. Haynes,et al.  Effects of aging on T cell function , 2009, Current Opinion in Immunology.

[24]  Ramit Mehr,et al.  Models and methods for analysis of lymphocyte repertoire generation, development, selection and evolution. , 2012, Immunology letters.

[25]  R. Ahmed,et al.  Bone marrow contains virus-specific cytotoxic T lymphocytes. , 1997, Blood.

[26]  M. Blackman,et al.  Clonal Expansions and Loss of Receptor Diversity in the Naive CD8 T Cell Repertoire of Aged Mice1 , 2009, The Journal of Immunology.

[27]  F. Alt,et al.  The Mechanism and Regulation of Chromosomal V(D)J Recombination , 2002, Cell.

[28]  A. Vallejo,et al.  The Influence of Age on T Cell Generation and TCR Diversity1 , 2005, The Journal of Immunology.

[29]  David Kipling,et al.  Age-Related Changes in Human Peripheral Blood IGH Repertoire Following Vaccination , 2012, Front. Immun..

[30]  R Core Team,et al.  R: A language and environment for statistical computing. , 2014 .

[31]  P. Marrack,et al.  Unexpected expansions of CD8-bearing cells in old mice. , 1993, Journal of immunology.

[32]  Jérôme Lane,et al.  IMGT®, the international ImMunoGeneTics information system® , 2004, Nucleic Acids Res..

[33]  P. Kourilsky,et al.  Expanded CD4+ and CD8+ T cell clones in elderly humans. , 1997, Journal of immunology.

[34]  Mushtaq Ahmed,et al.  Age-associated decline in T cell repertoire diversity leads to holes in the repertoire and impaired immunity to influenza virus , 2008, The Journal of experimental medicine.

[35]  P. Doherty,et al.  Ecological analysis of antigen-specific CTL repertoires defines the relationship between naïve and immune T-cell populations , 2013, Proceedings of the National Academy of Sciences.

[36]  R. Würzner,et al.  Long-Term Cytomegalovirus Infection Leads to Significant Changes in the Composition of the CD8+ T-Cell Repertoire, Which May Be the Basis for an Imbalance in the Cytokine Production Profile in Elderly Persons , 2005, Journal of Virology.

[37]  Robert Gentleman,et al.  ShortRead: a bioconductor package for input, quality assessment and exploration of high-throughput sequence data , 2009, Bioinform..

[38]  Achim Zeileis,et al.  Measuring Inequality, Concentration, and Poverty , 2014 .

[39]  G. Bastert,et al.  Therapy of human tumors in NOD/SCID mice with patient-derived reactivated memory T cells from bone marrow , 2001, Nature Medicine.

[40]  M. Davenport,et al.  Nonrandom attrition of the naive CD8+ T-cell pool with aging governed by T-cell receptor:pMHC interactions , 2011, Proceedings of the National Academy of Sciences.

[41]  D. Netanely,et al.  Age-dependent spatial memory loss can be partially restored by immune activation. , 2008, Rejuvenation research.

[42]  Utpal Banerjee,et al.  The hematopoietic stem cell and its niche: a comparative view. , 2007, Genes & development.

[43]  Hadley Wickham,et al.  ggplot2 - Elegant Graphics for Data Analysis (2nd Edition) , 2017 .