Distribution, persistence, and efficacy of adoptively transferred central and effector memory-derived autologous simian immunodeficiency virus-specific CD8+ T cell clones in rhesus macaques during acute infection - PubMed (original) (raw)

. 2010 Jan 1;184(1):315-26.

doi: 10.4049/jimmunol.0902410. Epub 2009 Nov 30.

Matthew T Trivett, Diane L Bolton, Charles M Trubey, Jacob D Estes, Yuan Li, Jeremy Smedley, Rhonda Pung, Margherita Rosati, Rashmi Jalah, George N Pavlakis, Barbara K Felber, Michael Piatak Jr, Mario Roederer, Jeffrey D Lifson, David E Ott, Claes Ohlen

Affiliations

Distribution, persistence, and efficacy of adoptively transferred central and effector memory-derived autologous simian immunodeficiency virus-specific CD8+ T cell clones in rhesus macaques during acute infection

Jacob T Minang et al. J Immunol. 2010.

Abstract

Plasma viremia decreases coincident with the appearance of virus-specific CD8(+) T cells during acute HIV or SIV infection. This finding, along with demonstrations of viral mutational escape from CD8(+) T cell responses and transient increase in plasma viremia after depletion of CD8(+) T cells in SIV-infected monkeys strongly suggest a role for CD8(+) T cells in controlling HIV/SIV. However, direct quantitative or qualitative correlates between CD8(+) T cell activity and virus control have not been established. To directly assess the impact of large numbers of virus-specific CD8(+) T cells present at time of SIV infection, we transferred in vitro expanded autologous central and effector memory-derived Gag CM9-, Nef YY9-, and Vif WY8-specific CD8(+) T cell clones to acutely infected rhesus macaques. The cells persisted in PBMCs between 4 and 9 d, but were not detected in gut-associated lymphoid tissue or lymph nodes. Interestingly, a high frequency of the infused cells localized to the lungs, where they persisted at high frequency for >6 wk. Although persisting cells in the lungs were Ag reactive, there was no measurable effect on virus load. Sequencing of virus from the animal receiving Nef YY9-specific CD8(+) T cells demonstrated an escape mutation in this epitope <3 wk postinfection, consistent with immune selection pressure by the infused cells. These studies establish methods for adoptive transfer of autologous SIV-specific CD8(+) T cells for evaluating immune control during acute infection and demonstrate that infused cells retain function and persist for at least 2 mo in specific tissues.

PubMed Disclaimer

Figures

Figure 1

Figure 1

Gating strategy and flow cytometry sorting analysis of central (TCM) and effector (TEM) memory CD8+ T-cell populations from a rhesus macaque (DBN2) immunized with a DNA vaccine construct containing full length SIVmac239 Gag, Pol, Vif, Tat, and Nef sequences. CD8+ lymphocytes were singlet-gated and defined as memory cells based on surface expression of CD95. TCM and TEM fractions were further defined and sorted based on CD28 and CCR7 expression.

Figure 2

Figure 2

TCM- and TEM-derived CD8+ T-cell clones exhibit similar functional reactivity and both display an effector memory surface phenotype after culture in vitro. TCM- and TEM-derived SIV-specific CD8+ T-cell clones from two rhesus macaques, AZ15 and DBN2, were stimulated with PBMC pulsed with the appropriate peptides (A) or autologous SIV-infected CD4+ T-cell clones (B). IFN-γ-expression by the TCM- and TEM-derived clones was measured by ICS and flow cytometry. The surface phenotype of TCM- and TEM-derived clones was determined using flourochrome conjugated mAbs to CD28 and CD95 followed by flow cytometric analyzes (C).

Figure 3

Figure 3

Intravenously infused ex vivo expanded virus-specific TCM- and TEM-derived CD8+ T-cell clones show similar distribution and persistence in vivo. Half of the clonal population of TCM- and TEM-derived virus-specific CD8+ T-cell clones from two rhesus macaques, DBN2 and AZ15, were stained with PKH26 and CFSE, respectively, and the pool of stained and unstained cells adoptively transferred to the monkeys. The distribution and persistence of the infused cells was analyzed by flow cytometry by gating for the CD3+CD8+ T-cell fraction of the “live gate” of the forward and side scatter plot (A). PBMC (B), LN and GALT (C) samples were analyzed at indicated time points post infusion.

Figure 4

Figure 4

Adoptive transfer of ex vivo expanded virus-specific CD8+ T-cell clones show no measurable effect on the virus load or the number of circulating CD4+ T cells in peripheral blood. Virus load in two animals infused with virus-specific CD8+ T cells, DBN2 and AZ15, as well as a control animal, DBK1, was determined on day 0, 3 (day of infusion), 4, 5 and 7 post challenge and once a week thereafter, by analyzing cell-free plasma viral RNA (A) or cell-associated viral DNA (B) levels by QPCR. The frequency of circulating CD4+ T cells in PBMC was determined using the BD Tru Count™ kit (C).

Figure 5

Figure 5

T cell persistence, function and viral load in BAL samples. (A) Virus-specific TCM- and TEM-derived CD8+ T-cell clones from two rhesus macaques, DBN2 and AZ15, half of which were stained with PKH26 and CFSE, respectively, were infused 3 days after challenge with SIVmac239. Infused cells were tracked using flow cytometry at indicated time points (B). Cell-free viral RNA and cell-associated viral DNA and RNA were determined at indicated time-points by QPCR. DBN2 and AZ15 received virus-specific CD8+ T cells. Grey dashed lines show threshold of detection of cell-associated viral DNA and RNA (>10 copies/100,000 cells). (C) Cells from BAL from DBN2 were stimulated with SIV Acc peptide pool or the SIV Gag CM9 peptide as indicated. The frequency of IFN-γ-expressing cells was determined by ICS and flow cytometry.

Figure 6

Figure 6

Adoptive transfer of virus-specific CD8+ T cells does not significantly affect the endogenous SIV-specific T-cell response. (A) PBMC collected at the indicated time points from the two Mamu A*01 positive monkeys, DBN2 and DBK1 were stained with SIV Tat SL8 or Gag CM9 tetramers and CD8 mAb. (B) PBMC from DBN2 and DBK1 collected pre- and post- infusion were stimulated with SIV Tat SL8, Gag CM9 and Acc peptide pool and the percentage of CD8+ IFN-γ-producing T cells determined by flow cytometry, or (C) the number of IFN-γ-producing cells determined by ELISpot. (D) PBMC from AZ15, DBN2 and DBK1 collected on day 39 post infusion were stimulated with SIV Acc, Env, Gag and Pol peptide pools and the number of antigen-induced IFN-γ-producing T cells determined by ELISpot.

Figure 7

Figure 7

Escape mutation in the SIV Nef159-167YY9 epitope detected <3 weeks post infection. Virus isolated from plasma collected from the Mamu A*02-positive monkey AZ15 on days 12 and 20 after challenge with SIVmac239 (days 9 and 17 post T cell infusion) was analyzed by direct sequencing spanning the SIV Nef YY9 epitope. The nucleotide change is highlighted by black circles with the encoded amino acid indicated.

Similar articles

Cited by

References

    1. Hofmann-Lehmann R, Vlasak J, Rasmussen RA, Smith BA, Baba TW, Liska V, Ferrantelli F, Montefiori DC, McClure HM, Anderson DC, Bernacky BJ, Rizvi TA, Schmidt R, Hill LR, Keeling ME, Katinger H, Stiegler G, Cavacini LA, Posner MR, Chou TC, Andersen J, Ruprecht RM. Postnatal passive immunization of neonatal macaques with a triple combination of human monoclonal antibodies against oral simian-human immunodeficiency virus challenge. J. Virol. 2001;75:7470–7480. - PMC - PubMed
    1. Mascola JR, Lewis MG, Stiegler G, Harris D, VanCott TC, Hayes D, Louder MK, Brown CR, Sapan CV, Frankel SS, Lu Y, Robb ML, Katinger H, Birx DL. Protection of Macaques against pathogenic simian/human immunodeficiency virus 89.6PD by passive transfer of neutralizing antibodies. J. Virol. 1999;73:4009–4018. - PMC - PubMed
    1. Veazey RS, Shattock RJ, Pope M, Kirijan JC, Jones J, Hu Q, Ketas T, Marx PA, Klasse PJ, Burton DR, Moore JP. Prevention of virus transmission to macaque monkeys by a vaginally applied monoclonal antibody to HIV-1 gp120. Nat. Med. 2003;9:343–346. - PubMed
    1. Shiver JW, Fu TM, Chen L, Casimiro DR, Davies ME, Evans RK, Zhang ZQ, Simon AJ, Trigona WL, Dubey SA, Huang L, Harris VA, Long RS, Liang X, Handt L, Schleif WA, Zhu L, Freed DC, Persaud NV, Guan L, Punt KS, Tang A, Chen M, Wilson KA, Collins KB, Heidecker GJ, Fernandez VR, Perry HC, Joyce JG, Grimm KM, Cook JC, Keller PM, Kresock DS, Mach H, Troutman RD, Isopi LA, Williams DM, Xu Z, Bohannon KE, Volkin DB, Montefiori DC, Miura A, Krivulka GR, Lifton MA, Kuroda MJ, Schmitz JE, Letvin NL, Caulfield MJ, Bett AJ, Youil R, Kaslow DC, Emini EA. Replication-incompetent adenoviral vaccine vector elicits effective anti-immunodeficiency-virus immunity. Nature. 2002;415:331–335. - PubMed
    1. Rose NF, Marx PA, Luckay A, Nixon DF, Moretto WJ, Donahoe SM, Montefiori D, Roberts A, Buonocore L, Rose JK. An effective AIDS vaccine based on live attenuated vesicular stomatitis virus recombinants. Cell. 2001;106:539–549. - PubMed

Publication types

MeSH terms

Substances

Grants and funding

LinkOut - more resources