Progressive Telomere Shortening of Epstein-Barr Virus Specific Memory T Cells during HIV Infection: Contributor to Exhaustion?

Similar documents
EBV Infection and Immunity. Andrew Hislop Institute for Cancer Studies University of Birmingham

Detailed analysis of Epstein Barr virus-specific CD4 + and CD8 + T cell responses during infectious mononucleosis

Cytotoxicity assays. Rory D. de Vries, PhD 1. Viroscience lab, Erasmus MC, Rotterdam, the Netherlands

Commercially available HLA Class II tetramers (Beckman Coulter) conjugated to

CD27 and CD57 Expression Reveals Atypical Differentiation of Human Immunodeficiency Virus Type 1-Specific Memory CD8 T Cells

Supplementary Figure 1. Enhanced detection of CTLA-4 on the surface of HIV-specific

HD1 (FLU) HD2 (EBV) HD2 (FLU)

Ex vivo Human Antigen-specific T Cell Proliferation and Degranulation Willemijn Hobo 1, Wieger Norde 1 and Harry Dolstra 2*

staining and flow cytometry

Dynamics of Cytomegalovirus (CMV) Specific T Cells in HIV-1 Infected Individuals Progressing to AIDS with CMV End-Organ Disease

Detailed step-by-step operating procedures for NK cell and CTL degranulation assays

Decreased EBNA-1-specific CD8 T cells in patients with Epstein Barr virus-associated

Subset analysis of the EBV-specific CD4+ T cell response in primary. and persistent infection. and

Chapter 8. Slower CD4 T cell decline in Ethiopian versus Dutch HIV 1 infected individuals is due to lower T cell proliferation rates

Cover Page. The handle holds various files of this Leiden University dissertation.

Rapid antigen-specific T cell enrichment (Rapid ARTE)

Viral Persistence Alters CD8 T-Cell Immunodominance and Tissue Distribution and Results in Distinct Stages of Functional Impairment

Direct ex vivo characterization of human antigen-specific CD154 + CD4 + T cells Rapid antigen-reactive T cell enrichment (Rapid ARTE)

PBMC from each patient were suspended in AIM V medium (Invitrogen) with 5% human

Human Immunodeficiency Virus Type-1 Myeloid Derived Suppressor Cells Inhibit Cytomegalovirus Inflammation through Interleukin-27 and B7-H4

Innate and Cellular Immunology Control of Infection by Cell-mediated Immunity

Supplementary Data 1. Alanine substitutions and position variants of APNCYGNIPL. Applied in

Nature Biotechnology: doi: /nbt Supplementary Figure 1. Binding capacity of DNA-barcoded MHC multimers and recovery of antigen specificity

7-AAD/CFSE Cell-Mediated Cytotoxicity Assay Kit

CD8 T cells play a critical role in the control of viral infections

Supplemental Methods. CD107a assay

Cellular Immunity in Aging and HIV: Correlates of Protection. Immune Senescence

x Lymphocyte count /µl CD8+ count/µl 800 Calculated

Supplementary Data. Treg phenotype

Tonsillar homing of Epstein-Barr virus specific CD8 + T cells and the virus-host balance

Institut für Medizinische Immunologie, Berliner Hochschulmedizin Charite, Charité Campus Mitte, Berlin, Germany

SUPPLEMENTARY INFORMATION

Supplementary Figures

Supporting Online Material for

MHC MULTIMER PROFICIENCY PANEL 2015

Supplementary Fig. 1: Ex vivo tetramer enrichment with anti-c-myc beads

Low Avidity CMV + T Cells accumulate in Old Humans

MHC Tetramers and Monomers for Immuno-Oncology and Autoimmunity Drug Discovery

Naive, memory and regulatory T lymphocytes populations analysis

MHC MULTIMER PROFICIENCY PANEL 2017

Epstein-Barr Virus Infects B and Non-B Lymphocytes in HIV-1 Infected Children and Adolescents

Critical Role for Alpha/Beta and Gamma Interferons in Persistence of Lymphocytic Choriomeningitis Virus by Clonal Exhaustion of Cytotoxic T Cells

MATERIALS AND METHODS. Neutralizing antibodies specific to mouse Dll1, Dll4, J1 and J2 were prepared as described. 1,2 All

CD25-PE (BD Biosciences) and labeled with anti-pe-microbeads (Miltenyi Biotec) for depletion of CD25 +

Introduction. Materials and methods

Peptide stimulation and Intracellular Cytokine Staining

Technical Resources. BD Immunocytometry Systems. FastImmune Intracellular Cytokine Staining Procedures

Low immune activation despite high levels of pathogenic HIV-1 results in long-term asymptomatic disease

Interferon γ regulates idiopathic pneumonia syndrome, a. Th17 + CD4 + T-cell-mediated GvH disease

Supplementary Figure 1. Using DNA barcode-labeled MHC multimers to generate TCR fingerprints

Rapid perforin upregulation directly ex vivo by CD8 + T cells is a defining characteristic of HIV elite controllers

CHAPTER 3 LABORATORY PROCEDURES

Superior Fluorescent Labeling Dyes Spanning the Full Visible Spectrum...1. Trademarks: HiLyte Fluor (AnaSpec, Inc.)

Gladstone Institutes, University of California (UCSF), San Francisco, USA

Therapeutic PD L1 and LAG 3 blockade rapidly clears established blood stage Plasmodium infection

ASSESSING THE FUNCTION OF EBV-SPECIFIC CD4 + T cells

MATERIALS. Peptide stimulation and Intracellular Cytokine Staining- EFFECTOR 45RA PANEL

Total genomic DNA was extracted from either 6 DBS punches (3mm), or 0.1ml of peripheral or

GM-CSF and IL-2 induce specific cellular immunity and provide protection against Epstein-Barr virus lymphoproliferative disorder

Supplementary webappendix

Gastric Carcinoma with Lymphoid Stroma: Association with Epstein Virus Genome demonstrated by PCR

Supplementary Figure 1. BMS enhances human T cell activation in vitro in a

Product Datasheet. HLA ABC Antibody (W6/32) NB Unit Size: 0.25 mg. Store at -20C. Avoid freeze-thaw cycles. Reviews: 1 Publications: 22

attomol HLA-B*27-Realtime LT 2 Assay for the detection of the human HLA-B*27-locus using LightCycler (Do not use for tissue typing!

Optimizing Intracellular Flow Cytometry

90 min 18 min. 45 min. 14 d

SUPPLEMENTARY INFORMATION. Involvement of IL-21 in the epidermal hyperplasia of psoriasis

Loss of CD96 Expression as a New Biomarker for T-cell Senescence in HIV-1 Infection

Lecture 6. Burr BIO 4353/6345 HIV/AIDS. Tetramer staining of T cells (CTL s) Andrew McMichael seminar: Background

Immune surveillance: The immune system can recognize and destroy nascent malignant cells

Translating EBV Immunology from Bench to Bedside: Somnium Animadverto A Dream Realized

MAJOR ARTICLE. (See the editorial commentary by Bitan on pages )

Application Information Bulletin: Human NK Cells Phenotypic characterizing of human Natural Killer (NK) cell populations in peripheral blood

Estimating primate effector T cell responses to DNA vaccination

Supplementalgfigureg1gSchematicgdiagramgofgtumor1modellingg

Alternate Antibody-Based Therapeutic Strategies To Purge the HIV Cell Reservoir

Nature Medicine: doi: /nm.2109

In vitro human regulatory T cell expansion

Fluorochrome Panel 1 Panel 2 Panel 3 Panel 4 Panel 5 CTLA-4 CTLA-4 CD15 CD3 FITC. Bio) PD-1 (MIH4, BD) ICOS (C398.4A, Biolegend) PD-L1 (MIH1, BD)

Supporting Information

CD14 + S100A9 + Monocytic Myeloid-Derived Suppressor Cells and Their Clinical Relevance in Non-Small Cell Lung Cancer

Mass Cytometry Applications for Immunology Research. Pub Note PN 13 01_150505

Shenghua Zhou, Rong Ou, Lei Huang, and Demetrius Moskophidis*

EBV infection B cells and lymphomagenesis. Sridhar Chaganti

Cover Page. The handle holds various files of this Leiden University dissertation

Effector cell mediated cytotoxicity measured by intracellular Granzyme B release in HIV infected subjects

Supplementary Figure 1. Example of gating strategy

The Journal of Experimental Medicine

Concentration Estimation from Flow Cytometry Exosome Data Protocol

CONTRACTING ORGANIZATION: Johns Hopkins University School of Medicine Baltimore, MD 21205

Principle of the FluoroSpot assay. Anti-tag mab-green. Streptavidin-Red. Detection mab-tag. Detection mab-biotin. Analyte. Analyte.

IFN-γ Secretion Assay Detection Kit (PE) human

Administration of Tumor-Specific Cytotoxic T Lymphocytes Engineered to Resist TGF-ß to Patients with EBV-Associated Lymphomas

10/18/2012. A primer in HLA: The who, what, how and why. What?

Challenges in Development and Validation of an Intracellular Cytokine Staining assay

Nature Immunology: doi: /ni Supplementary Figure 1. Huwe1 has high expression in HSCs and is necessary for quiescence.

Supplementary Table 1. Data collection and refinement statistics (molecular replacement).

Nature Immunology: doi: /ni Supplementary Figure 1. DNA-methylation machinery is essential for silencing of Cd4 in cytotoxic T cells.

Determination of the temporal pattern and importance of BALF1 expression in Epstein-Barr viral infection

a Beckman Coulter Life Sciences: White Paper

Transcription:

BRIEF REPORT Progressive Telomere Shortening of Epstein-Barr Virus Specific Memory T Cells during HIV Infection: Contributor to Exhaustion? Debbie van Baarle, 1 Nening M. Nanlohy, 1 Sigrid Otto, 1 Fiona J. Plunkett, 2 Jean M. Fletcher, 2 and Arne N. Akbar 2 1 Department of Immunology, University Medical Center Utrecht, Utrecht, The Netherlands; 2 Department of Immunology and Molecular Pathology, Royal Free and University College Medical School, London, United Kingdom Individuals infected with human immunodeficiency virus (HIV) have low numbers of functional Epstein-Barr virus (EBV) specific CD8 T cells in the face of a high EBV load, suggesting that these cells have become exhausted. We investigated whether the observed chronic EBV loads during HIV infection could cause exhaustion of EBV-specific T cells by using flow-fish (flow cytometry in combination with fluorescence in situ hybridization) to analyze the telomere length of EBV-specific CD8 T cells. Enhanced telomere shortening of EBV-specific T cells was observed during HIV infection, compared with the decline in telomere length observed in the CD8 T cells of healthy subjects. Thus, chronic exposure to high antigen levels may lead to the progressive shortening of telomeres of antigen-specific T cells, which may impair viral control. Adequate numbers of antiviral T cells are essential to maintain immunological memory. However, excessive numbers of cell divisions may result in the generalized age-related decline in T cell responses [1]. Phenotypic changes have been shown to occur within the memory T cell pool during aging. Furthermore, memory CD8 T cells may lose their replicative capacity as a result of telomere loss, which may restrict their function or persistence during aging [2]. Although telomeres do not shorten Received 8 January 2008; accepted 5 May 2008; electronically published 24 September 2008. Potential conflicts of interest: none reported. Financial support: Netherlands Organization for Scientific Research (talent stipend); European Molecular Biology Organization (travel grant). Reprints or correspondence: Dr. Debbie van Baarle, Dept. of Immunology, University Medical Center Utrecht, hsp KC02.085.2, Lundlaan 6, 3584 EA Utrecht, The Netherlands (d.vanbaarle@umcutrecht.nl). The Journal of Infectious Diseases 2008; 198:1353 7 2008 by the Infectious Diseases Society of America. All rights reserved. 0022-1899/2008/19809-0015$15.00 DOI: 10.1086/592170 during excessive proliferation in acute infections [3, 4] because of up-regulation of the enzyme telomerase, the ability of T cells to up-regulate telomerase activity progressively decreases on repeated stimulation [1], resulting in telomere shortening in virusspecific CD8 T cells after acute infection. Telomere erosion of memory CD8 T cells, especially those specific for persistent antigens, may eventually restrict the capacity of these cells to expand. Telomere erosion may occur in several clinical situations. Patients with X-linked lymphoproliferative syndrome (XLP) experience uncontrolled polyclonal expansion of both T and B cells, usually in response to acute Epstein-Barr virus (EBV) infection. The excessive CD8 T cell proliferation was shown to lead to end-stage differentiation and loss of functional EBV-specific CD8 T cells through replicative senescence [5], which may contribute to defective immunity in patients with XLP. Another situation in which EBV infection is not controlled optimally is during HIV infection. During the course of HIV infection, CD8 T cells increasingly become end-stage effector cells, similar to T cells in elderly persons [6, 7]. Interestingly, HIV-infected individuals have low numbers of functional EBV-specific CD8 T cells [8] in the face of a high EBV load [9], suggesting that these T cells have become exhausted due to chronic exposure to EBV. Therefore, we investigated whether continuous exposure to high levels of EBV during HIV infection may cause increased telomere shortening of EBV-specific T cells. Methods. To study the telomere length of EBV-specific T cells, we selected 4 of 7 HIV-negative blood bank donors (age range, 38 53 years) and 5 of 13 HIV-positive individuals (age range, 36 44 years) from the Amsterdam Cohort Studies of HIV infection, all of whom carried the HLA-B8 allele and had substantial numbers of EBV-specific T cells. Procedures followed were in accordance with the institutional medical ethical committee and with the Helsinki Declaration of 1975, as revised in 1983. Written informed consent was obtained from the subjects at inclusion in the Amsterdam Cohort Studies. The HIVpositive individuals were compared 1 and 8 years (median follow-up, 7.3 years) after HIV seroconversion. After this period, 4 of the individuals progressed to AIDS. Data from the HIVpositive individuals over time were related to data from an additional group of healthy control subjects (n 10; age range, 0 65 years). For determination of EBV load, genomic DNA from 2 10 5 cells was amplified in duplicate using polymerase chain reaction (PCR) primers specific for the nonglycosylated membrane protein BNRF1 p143. Real-time PCR amplification was performed BRIEF REPORT JID 2008:198 (1 November) 1353

using a fluorogenic probe to detect the 74-bp product. As a control for the input DNA, the amount of -albumin DNA was also determined [9]. Major histocompatibility complex (MHC) class I tetramers complexed with EBV peptides were produced. The peptides used (synthesized by solid-phase methods using an automated multiple peptide synthesizer and 9-fluorenylmethoxycarbonyl chemistry) were 2 immunodominant epitopes restricted through HLA-B8: RAKFKQLL (B8-RAK) from the lytic cycle antigen BZLF-1 and FLRGRAYGL (B8-FLR) from the EBV latent antigen EBNA-3A [10]. We included both of these epitopes because they are presented during different phases of the EBV life cycle. Biotinylated class I peptide complexes were tetramerized by addition of cyanine 5 (Cy5) conjugated streptavidin, in addition to the conventional allophycocyanin (APC) or phycoerythrin (PE) conjugated streptavidin. Staining using Cy5- labeled tetramers was compared with that using the APC- or PE-labeled tetramers and was found to give similar results (data not shown). The telomere length of specific CD8 T cells was measured using a 2-color flow-fish (flow cytometry in combination with fluorescence in situ hybridization) assay, as described elsewhere [4]. Briefly, peripheral blood mononuclear cells (PBMCs) were thawed, and 1.5 10 6 cells were stained with MHC class I tetramers containing a Cy5 label, after which cells were washed in PBS and fixed with bis(sulfosuccinimidyl)suberate (BS 3 ). After being washed in hybridization buffer, cells were incubated with the peptide nucleic acid telomeric (C 3 TA 2 ) 3 probe conjugated to fluorescein isothiocyanate. Samples were heated for 10 min at 82 C, rapidly cooled on ice, and hybridized for 1 h atroom temperature in the dark. Cells were washed, and at least 250,000 events were acquired using a FACSCalibur flow cytometer (BD Biosciences). Data were analyzed using CellQuest software (BD Biosciences). Cy5-tetramer staining of CD8 T cells was confirmed using parallel staining with the APC- or PE-labeled tetramers in conjunction with CD8 (BD Biosciences). In vitro T cell proliferation was measured using 5,6- carboxyfluorescein diacetate succinimidyl ester (CFSE; Molecular Probes), in accordance with the manufacturer s protocol. Briefly, PBMCs were thawed, cells were labeled with CFSE for 8 min, and labeling was stopped using human pool serum. Cells were washed, and 2 10 6 cells in 0.5 ml of RPMI 1640 medium were aliquoted in round-bottom tubes. Cells were then stimulated with either RAK or FLR peptide (concentration, 1 g/ml). As a positive control, 0.02 g of anti-cd3 (Sanquin Reagents) and 2 g of anti-cd28 (Sanquin Reagents) was used. After 6 days of incubation at 37 C and 5% CO 2, cells were stained using anti CD8-APC and anti CD3-PerCP (BD Biosciences). The percentage of CD3 CD8 T cells that underwent at least 1 division was calculated by subtracting the proliferation in the absence of stimulus. Stimulation indices (SIs) were calculated by dividing the percentage of proliferation for CD3 CD8 T cells after stimulation by the percentage of proliferation for unstimulated cells. In EBV-positive healthy control individuals, CD8 T cells showed proliferation on EBV peptide stimulation, with a median SI of 4 (data not shown). Results and discussion. To investigate whether the observed chronic exposure to high levels of EBV during HIV infection may cause increased telomere shortening of EBV-specific T cells, we analyzed telomere lengths of both EBV lytic (peptide RAKFKQLL) and EBV latent (peptide FLRGRAYGL) antigen specific CD8 T cells in HLA-B8 positive individuals. To this end, we performed staining of specific T cells with HLA-B8 tetrameric complexes containing the EBV peptides in combination with flow cytometry analysis of telomere length using in situ hybridization with fluorescent telomere probes (flow-fish) [4]. Telomere lengths of EBV-specific T cells in HIV-negative (n 4) and HIV-positive individuals (n 5) were compared 1 and 8 years (median follow-up, 7.3 years) after HIV infection. EBV loads were elevated at these time points during HIV infection, varying from a median of 15,648 copies/1 10 6 PBMCs at the early time point to 5000 EBV copies/1 10 6 PBMCs at the late time point (data not shown). T cells specific for lytic antigen RAK, as identified by HLA-B8 tetrameric complexes labeled with Cy5, were always more abundantly present (range, 0.11% 1.24% tetramer-positive T cells) than those specific for latent antigen FLR (range, 0.15% 0.57% tetramer-positive T cells), as analyzed in a larger group of HIVnegative (n 7) and HIV-positive (n 13) individuals (figure 1A). Subsequently, telomeric probe fluorescence was analyzed within the RAK- and FLR-specific T cell populations in 4 HIVnegative and 5 HIV-positive individuals with substantial numbers of both RAK- and FLR-specific T cells ( 0.4%). Figure 1B shows telomeric probe fluorescence of EBV-specific CD8 T cells in a representative HIV-infected patient at early and late time points during HIV infection. Although CD8 T cells specific for the RAK peptide increased from 0.48% to 0.68% (figure 1B) and those for the FLR peptide increased from 0.40% to 0.63% (figure 1B), telomeric probe median fluorescence intensity (MFI) decreased both for RAK-specific CD8 T cells, from 119.71 to 93.08 (figure 1B), and for FLR-specific CD8 T cells, from 115.48 to 100 (figure 1B). Overall, RAK-specific T cells had shorter telomeres than did FLR-specific T cells (figure 1C), in both HIV-negative (MFI, 89.85 vs. 104.64) and HIV-positive individuals (MFI, 88.19 vs. 101.83 early), a difference that was significant late during HIV infection (MFI, 77.74 vs. 93.06; P.042, Mann-Whitney U test) (figure 1C). This finding suggests that lytic antigen specific T cells underwent more proliferation cycles, and it fits with the described phenotype of EBVspecific T cells showing lytic antigen specific T cells to be more differentiated than latent antigen specific T cells in healthy carriers [10]. Interestingly, further telomere shortening of both lytic and latent antigen specific T cells was observed during HIV infec- 1354 JID 2008:198 (1 November) BRIEF REPORT

Figure 1. Analysis of telomeric probe fluorescence of Epstein-Barr virus (EBV) specific T cells in HIV-negative and HIV-positive individuals. A, Percentages of lytic antigen RAK and latent antigen FLR specific CD8 T cells in 7 HIV-negative and 13 HIV-positive HLA-B8 positive individuals, as measured by flow cytometry analysis of staining with cyanine 5 (Cy5) labeled HLA-B8 tetrameric complexes containing either the RAK or FLR peptide. B, Flow cytometry analysis of telomere probe fluorescence in tetramer-positive T cells for 1 HIV-infected individual early (left panels) and late (right panels) during HIV infection. RAK-specific (upper panels) and FLR-specific (lower panels) T cells are shown in dot plots, and median fluorescence intensity (MFI) is given as histograms of the tetramer-positive/telomere-positive population (upper right quadrants of dot plots). C, Telomere probe MFI after telomere probe hybridization as a measure of the telomere lengths of lytic antigen (RAK) specific and latent antigen (FLR) specific CD8 T cells in HIV-negative healthy control subjects (n 4) and HIV-positive individuals early (n 4; 1 year after seroconversion) and late (n 5; median follow-up, 7.3 years) during HIV infection. Cells were stained with Cy5-labeled HLA-B8 tetrameric complexes containing 1 of the 2 specific peptides (RAK and FLR) in combination with flow cytometry analysis of telomere length using in situ hybridization with fluorescent (fluorescein isothiocyanate [FITC] labeled) telomere probes (flow-fish), and the MFI of the telomere probes within the HLA-B8-RAK or HLA-B8-FLR specific T cells was determined. P values are indicated. tion. The telomeric probe MFI of RAK-specific T cells decreased from 88.19 to 77.74 (figure 1C). In the case of FLR-specific CD8 T cells, the tendency to have shorter telomeres in late HIV infection (telomere probe MFI, 93.06) than in early infection (MFI, 101.83) was most striking (P.068, Wilcoxon test), suggesting that telomere shortening of EBV-specific T cells can occur BRIEF REPORT JID 2008:198 (1 November) 1355

Figure 2. Telomeric probe fluorescence of Epstein-Barr virus specific T cells in HIV-negative vs. HIV-positive individuals over time. The median fluorescence intensity (MFI) related to age (in years) is shown for CD8 T cells from a group of healthy control subjects (black circles) and, longitudinally, for lytic antigen RAK specific T cells (other black symbols) and latent antigen FLR specific T cells (white symbols) from 4 HIV-infected subjects. Three of 4 HIV-infected individuals showed a steeper decline in telomere fluorescence of RAK-specific T cells (upright triangles) or both RAK- and FLR-specific T cells (squares and diamonds). within 7 years of HIV infection (figure 1C). In line with this finding, we did not observe any proliferative capacity of EBVspecific T cells late in HIV infection, as measured using a CFSE dye dilution assay, and we observed it in only 1 of the HIVinfected individuals at the early time point during HIV infection (SI, 6), whereas T cells did proliferate on anti-cd3 stimulation (SI, 5) (data not shown); this individual did not progress to AIDS. To relate these data on telomere length to the normal rate of decline in telomere length with age, we analyzed the telomeric probe fluorescence of CD8 T cells in a group of healthy donors (n 10) with ages ranging from 0 (cord blood) to 80 years. Figure 2 shows that telomeric probe MFI decreases by 7.86 per 10 years. From this curve, the normal decline in MFI was calculated to be 5.7 over a period of 7.3 years (the median follow-up for our HIV-positive individuals) (figure 2). The decline in MFI observed for FLR- and RAK-specific T cells was 2-fold greater in 3 of 4 HIV-infected individuals (figure 2), with median declines of 8.77 and 10.45, respectively. This indicates that telomere shortening of EBV-specific T cells occurs at a faster rate than expected in HIV-infected individuals and may explain the observed functional and phenotypic features of CD8 T cells during HIV infection. HIV-infected individuals (30 40 years of age) resemble elderly individuals ( 80 years of age) with respect to a lack of CD28 and CD27 expression on CD8 T cells, and CD28 T cells from these HIV-positive persons have telomere lengths identical to those of centenarians [1]. This suggests that CD8 T cells during HIV infection have undergone considerable proliferation, comparable to those in elderly persons. Moreover, a longitudinal study in young HIV-infected individuals showed a decrease in telomere length in CD8 T cells over time [11]. Functional defects that have been described in HIV-positive individuals include reduced numbers of cytokine-producing T cells upon antigen stimulation late in infection. Furthermore, only T cells from nonprogressors were shown to maintain high proliferative capacity, which was shown to be coupled with increases in perforin expression [12], suggesting that proliferation is required for obtaining efficient effector T cells. Thus, diminished proliferative capacity leads to a memory T cell population that is less able to control infections because of lower effector functions. EBV-specific T cells have been shown to be important for control of latent EBV infection and to protect against uncontrolled lymphoproliferation of infected B cells and lymphoma development [8, 13]. In both elderly individuals and patients suffering from XLP, EBV-specific T cells show reduced functionality associated with enhanced telomere erosion in the latter group [5]. We now show that EBV-specific T cells during HIV infection also show progressive telomere shortening. Decreased numbers of interferon (IFN) producing EBV-specific T cells upon antigen stimulation in these HIV-infected individuals [8] resemble lower EBV-specific responses (IFN- ) in elderly people. This suggests that, with respect to EBV-specific T cell responses, HIV-positive individuals seem to display a more se- 1356 JID 2008:198 (1 November) BRIEF REPORT

nescent T cell phenotype, which may be explained by the persistently high EBV DNA levels found in these individuals [9]. In conclusion, during chronic antigen stimulation, such as during HIV infection, antigen-specific T cells may progressively shorten their telomeres due to frequent activation through increased rates of virus exposure. Although the role played by lytic antigen specific T cells is still unclear, latent antigen specific T cells have been shown to be important for control of EBV infection [14, 15]. This suggests that loss of proliferative potential due to telomere shortening, potentially leading to exhaustion of these cells, may impair EBV control and may predispose these individuals to EBV-related lymphomas. References 1. Effros RB, Cai Z, Linton PJ. CD8 T cells and aging. Crit Rev Immunol 2003; 23:45 64. 2. Akbar AN, Soares MV, Plunkett FJ, Salmon M. Differential regulation of CD8 T cell senescence in mice and men. Mech Ageing Dev 2000; 121: 69 76. 3. Maini MK, Soares MV, Zilch CF, Akbar AN, Beverley PC. Virus-induced CD8 T cell clonal expansion is associated with telomerase upregulation and telomere length preservation: a mechanism for rescue from replicative senescence. J Immunol 1999; 162:4521 6. 4. Plunkett FJ, Soares MV, Annels N, et al. The flow cytometric analysis of telomere length in antigen-specific CD8 T cells during acute Epstein- Barr virus infection. Blood 2001; 97:700 7. 5. Plunkett FJ, Franzese O, Belaramani LL, et al. The impact of telomere erosion on memory CD8 T cells in patients with X-linked lymphoproliferative syndrome. Mech Ageing Dev 2005; 126:855 65. 6. van Baarle D, Tsegaye A, Miedema F, Akbar A. Significance of senescence for virus-specific memory T cell responses: rapid ageing during chronic stimulation of the immune system. Immunol Lett 2005; 97:19 29. 7. Papagno L, Spina CA, Marchant A, et al. Immune activation and CD8 T-cell differentiation towards senescence in HIV-1 infection. PLoS Biol 2004; 2:e20. 8. van Baarle D, Hovenkamp E, Callan MFC, et al. Dysfunctional Epstein- Barr virus (EBV)-specific CD8 T lymphocytes and increased EBV load in HIV-1 infected individuals progressing to AIDS-related non- Hodgkin s lymphoma. Blood 2001; 98:146 55. 9. van Baarle D, Wolthers KC, Hovenkamp E, et al. Absolute level of Epstein-Barr virus DNA in human immunodeficiency virus type 1 infection is not predictive of AIDS-related non-hodgkin lymphoma. J Infect Dis 2002; 186:405 9. 10. Hislop AD, Gudgeon NH, Callan MFC, et al. EBV-Specific CD8 T cell memory: relationships beteween epitope specificity, cell phenotype, and immediate effector function. J Immunol 2001; 167:2019 29. 11. Wolthers KC, Wisman GBA, Otto SA, et al. T-cell telomere length in HIV-1 infection: no evidence for increased CD4 T cell turnover. Science 1996; 274:1543 7. 12. Migueles SA, Laborico AC, Shupert WL, et al. HIV-specific CD8 T cell proliferation is tightly coupled to perforin expression and maintained in patients with immunologic control of virus replication. Nat Immunol 2002; 3:1061 8. 13. Rickinson AB, Moss DJ. Human cytotoxic T lymphocyte responses to Epstein-Barr virus infection. Annu Rev Immunol 1997; 15:405 31. 14. Piriou E, van Dort K, Nanlohy NM, Van Oers MHJ, Miedema F, van Baarle D. Loss of EBNA1-specific memory CD4 and CD8 T cells in HIV-infected patients progressing to AIDS-related non-hodgkin lymphoma. Blood 2005; 106:3166 74. 15. Munz C. Epstein-Barr virus nuclear antigen 1: from immunologically invisible to a promising T cell target. J Exp Med 2004; 199:1301 4. BRIEF REPORT JID 2008:198 (1 November) 1357