Skip to main page content
U.S. flag

An official website of the United States government

Dot gov

The .gov means it’s official.
Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

Https

The site is secure.
The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
. 2011 Jun;178(6):2740-51.
doi: 10.1016/j.ajpath.2011.02.022.

Aberrant CD8+ T-cell responses and memory differentiation upon viral infection of an ataxia-telangiectasia mouse model driven by hyper-activated Akt and mTORC1 signaling

Affiliations

Aberrant CD8+ T-cell responses and memory differentiation upon viral infection of an ataxia-telangiectasia mouse model driven by hyper-activated Akt and mTORC1 signaling

Anthony D D'Souza et al. Am J Pathol. 2011 Jun.

Abstract

Immune system-related pathology is common in ataxia-telangiectasia (A-T) patients and mice that lack the protein kinase, A-T mutated (ATM). However, it has not been studied how ATM influences immune responses to a viral infection. Using the lymphocytic choriomeningitis virus (LCMV) infection model, we show that ATM(-/-) mice, despite having fewer naïve CD8⁺ T cells, effectively clear the virus. However, aberrant CD8⁺ T-cell responses are observed, including defective expansion and contraction, effector-to-memory differentiation, and a switch in viral-epitope immunodominance. T-cell receptor-activated, but not naïve, ATM(-/-) splenic CD8⁺ T cells have increased ribosomal protein S6 and Akt phosphorylation and do not proliferate well in response to IL-15, a cytokine important for memory T-cell development. Accordingly, pharmacological Akt or mammalian target of rapamycin complex 1 (mTORC1) inhibition during T-cell receptor activation alone rescues the IL-15 proliferation defect. Finally, rapamycin treatment during LCMV infection in vivo increases the number of memory T cells in ATM(-/-) mice. Altogether, these results show that CD8⁺T cells lacking ATM have hyperactive Akt and mTORC1 signaling in response to T-cell receptor activation, which results in aberrant cytokine responses and memory T-cell development. We speculate that similar signaling defects contribute to the immune system pathology of A-T, and that inhibition of Akt and/or mTORC1 may be of therapeutic value.

PubMed Disclaimer

Figures

Figure 1
Figure 1
Fewer splenic T cells and altered immunodominance hierarchy in CD8+ T cells in ataxia-telangiectasia mutated (ATM)−/− mice in response to lymphocytic choriomeningitis virus (LCMV) infection. A: Total splenocytes in wild-type ATM+/+ (black circles in AD) and ATM−/− (grey squares in AD) mice at day 0 (d0), day 8 (d8), and day 41 (d41) post-LCMV infection. B: Total splenic CD8+ T cells at d0, d8, and d41 after post-LCMV infection ATM+/+ and ATM−/− mice. C: Total splenic CD4 T cells at d0, d8, and d41 post-LCMV infection of ATM+/+ and ATM−/− mice. D: Total DbNP396-CD8+ and DbGP33-CD8+ T cells at d8 and d41 post-LCMV infection in ATM+/+ and ATM−/− mice, respectively. E: Total CD8+ T cells in the blood (expressed as the number in 1 × 106 peripheral blood mononuclear cells (PBMCs) in wild-type and ATM−/− mice at d0, d8, day 15, day 20, day 30, and d41 post-LCMV infection. F: Percentage of DbNP396-CD8+ and DbGP33-CD8+ T cells at d0, d8, day 15, day 20, day 30, and d41 post-LCMV infection in wild-type and ATM−/− mice, respectively. A–C: 5 ATM+/+ and 5 ATM−/− mice were analyzed in two separate experiments on d0; the combined data from two separate experiments is shown representing a total of 5 ATM+/+ and 5 ATM−/− mice analyzed on d8 postinfection and a total of 7 ATM+/+ and 8 ATM−/− mice analyzed on day 41 postinfection from two separate experiments. For blood analyses in data from E and F, 10 ATM+/+ and 9 ATM−/− mice were analyzed in two independent experiments. Statistically significant differences are indicated directly or by asterisks, which represent P values from an unpaired t-test of <0.05.
Figure 2
Figure 2
Defective memory CD8+ T-cell differentiation ataxia-telangiectasia mutated (ATM)−/− mice post-LCMV infection. A: Number of viral specific DbNP396-CD8+, DbGP33-CD8+ and DbGP61-CD4+ memory T cells in spleens from ATM+/+ and ATM−/− mice at day 41 after lymphocytic choriomeningitis virus (LCMV) infection. B: Comparison of median fluorescence intensities (MFI) obtained from fluorescence-activated cell sorting (FACS) analyses of intracellular staining of interferon-γ, tumor necrosis factor-α, and IL-2 of DbGP33-CD8 T cells from ATM+/+ and ATM−/− mice at day 41 postinfection. C: Relative intracellular expression of granzyme B (GzmB) in DbGP33-CD8+ T cells from in ATM+/+ and ATM−/− mice at days 8 and 41 postinfection. The MFI from wild-type cells was given a value of 1.0 for normalization of the results. D: Top row, percent KLRG1hi, CD127hi, and CD62Lhi DbGP33-CD8+ T cells at days 8, 15, 20, 30, and 41 in the blood of LCMV-infected ATM+/+ (bold black line) and ATM−/− (dashed gray line) mice. Bottom row, percent KLRG1hi, CD127hi, and CD62Lhi DbGP33-CD8+ T cells at days 8 and 41 post-LCMV infection for ATM+/+ (black circles) and ATM−/− (grey squares) mice are shown. Whereas for the blood, the combined data from two separate experiments representing 7 ATM+/+ and 9 ATM−/− mice are shown, and for the spleen, the combined data from separate experiments representing 5 pairs of wild-type and ATM−/− mice on day 8 and 7 wild-type and 9 ATM−/− mice on day 41 are shown. Statistically significant differences are indicated directly or by asterisks, which represent P values from an unpaired t-test of <0.05.
Figure 3
Figure 3
T-cell receptor (TCR)-activated ataxia-telangiectasia mutated (ATM)−/− CD8+ lymphocytes sustain heightened Akt phosphorylation and exhibit a proliferation defect in response to IL-15 treatment that is rescued by the Akt inhibitor triciribine. A: Experimental scheme used to test the response of ATM−/− lymphocytes to IL-2 and IL-15, which after TCR activation (using anti-CD3 and anti-CD28 antibodies) skews the cell populations toward effector and memory phenotypes in vitro. B: Number of CD8+ T cells after IL-15 (right) or IL-2 (left) treatment of ATM+/+ and ATM−/− splenocytes, according to the scheme in A. For culturing in IL-2 and IL-15, the starting CD8+ T-cell number was 2.5 × 104 and 1 × 105, respectively. The P value from a two-tailed unpaired t-test is indicated for the IL-15 experiment, and no difference was observed in the IL-2 experiment. C: Western blot analyses of phosphorylated Akt (S473, T308), PTEN (S380/T382/383), S6 (S235/236), AMPK (T172), and ACC (S79) and the corresponding unphosphorylated proteins in splenic CD8+ T lymphocytes from wild-type and ATM−/− mice treated with IL-15 according to the scheme in A. Actin was used as the loading control. Similar results were obtained 3 times from different sets of mice. D: Total CD8+ T cells from ATM+/+ (open bars) or ATM−/− (filled bars) mice after IL-15 treatment according to the scheme, (A) with (+) or without (−) 10 nmol/L triciribine added at the stage indicated by T (ie, during TCR activation alone or during both TCR activation and IL-15 treatment). (E) Total percent CD8+ T-cell death evaluated by propidium iodide (PI)-positive cells after IL-15 treatment according to the scheme (A), with (+) or without (−)10 nmol/L triciribine added at the stage indicated by T. Analysis of cells from ATM+/+ (open bars) and ATM−/− (filled bars) mice are shown. D and E: The P value from a two-tailed unpaired t-test for the comparison of ATM−/− to ATM+/+ without drug treatment is shown above the brackets, and those for comparisons of drug-treated to untreated within in each group/genotype are as indicated.
Figure 4
Figure 4
Rapamycin treatment during T-cell receptor (TCR) activation rescues the IL-15 proliferation defect in ataxia-telangiectasia mutated (ATM)−/− splenic CD8+ T lymphocytes. A: Western blot analyses (as described in Figure 3C), except the ATM+/+ and ATM−/− cells were collected before TCR stimulation (pre-TCR) and 72 hours after TCR activation (post-TCR) (as described in Figure 3A). B: Graphed are the median fluorescence intensities (MFI) from intracellular staining for phosphorylated ribosomal protein S6 (phospho-S6; S235/236) calculated from FACS plots of ATM+/+ (white bars) and ATM−/− (gray bars) CD8+ T cells before (at 0 hours), and at 24, 48, and 72 hours of TCR activation in vitro. C: Same as B, except the analysis of CD8+ T cells on day 8 post-LCMV infection in vivo is shown. The data from 5 ATM+/+ and 5 ATM−/− mice were analyzed. D: Total CD8+ T cells from ATM+/+ (open bars) or ATM−/− (filled bars) after IL-15 treatment, according to the scheme (A), with (+) or without (−) 10 nmol/L rapamycin added at the stage indicated by “R” (ie, during TCR alone or during both TCR activation and IL-15 treatment). E: Total percent CD8+ T-cell death evaluated by propidium iodide (PI)-positive cells after IL-15 treatment, according to the scheme (A), with (+) or without (−) 10 nmol/L rapamycin added at the stage indicated by “R”. Analysis of cells from ATM+/+ (open bars) and ATM−/− (filled bars) mice are shown. D and E: The P value from a two-tailed unpaired t-test for the comparison of ATM−/− to ATM+/+ without drug treatment is shown above the brackets, and those for comparisons of drug treated to untreated within each group/genotype are as indicated.
Figure 5
Figure 5
Rapamycin treatment during lymphocytic choriomeningitis virus infection improves memory CD8+ T-cell development in ataxia-telangiectasia mutated (ATM)−/− mice. A: Percent of KLRG1hi, CD127hi, and CD62Lhi DbGP33-CD8+ T cells in ATM+/+ and ATM−/− mice at days 8, 15, and 30 post-LCMV infection with (dashed gray) or without (black line) rapamycin treatment between day −1 to day +8 (indicated by the gray shading). B: Same as in A, except total DbGP33+ and DbNP396+ CD8+ T lymphocytes were analyzed. Statistically significant differences are indicated by asterisks, which represent P values from an unpaired t-test of <0.05. PBMCs, peripheral blood mononuclear cells. X-axes denotes days after LCMV infection.

Similar articles

Cited by

References

    1. Savitsky K., Bar-Shira A., Gilad S., Rotman G., Ziv Y., Vanagaite L., Tagle D.A., Smith S., Uziel T., Sfez S., Ashkenazi M., Pecker I., Frydman M., Harnik R., Patanjali S.R., Simmons A., Clines G.A., Sartiel A., Gatti R.A., Chessa L., Sanal O., Lavin M.F., Jaspers N.G., Taylor A.M., Arlett C.F., Miki T., Weissman S.M., Lovett M., Collins F.S., Shiloh Y. A single ataxia telangiectasia gene with a product similar to PI-3 kinase. Science. 1995;268:1749–1753. - PubMed
    1. Lavin M.F., Shiloh Y. The genetic defect in ataxia-telangiectasia. Annu Rev Immunol. 1997;15:177–202. - PubMed
    1. Morrell D., Cromartie E., Swift M. Mortality and cancer incidence in 263 patients with ataxia-telangiectasia. J Natl Cancer Inst. 1986;77:89–92. - PubMed
    1. Barlow C., Hirotsune S., Paylor R., Liyanage M., Eckhaus M., Collins F., Shiloh Y., Crawley J.N., Ried T., Tagle D., Wynshaw-Boris A. Atm-deficient mice: a paradigm of ataxia telangiectasia. Cell. 1996;86:159–171. - PubMed
    1. Elson A., Wang Y., Daugherty C.J., Morton C.C., Zhou F., Campos-Torres J., Leder P. Pleiotropic defects in ataxia-telangiectasia protein-deficient mice. Proc Natl Acad Sci USA. 1996;93:13084–13089. - PMC - PubMed

Publication types

MeSH terms

-