Treatment with IL-7 Prevents the Decline of Circulating CD4 + T Cells during the Acute Phase of SIV Infection in Rhesus Macaques

Similar documents
Supporting Information

SUPPLEMENTARY INFORMATION

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

NIH Public Access Author Manuscript Nature. Author manuscript; available in PMC 2009 July 1.

Table S1. Viral load and CD4 count of HIV-infected patient population

The CAPRI-T was one of two (along with the CAPRI-NK, see reference. [29]) basic immunological studies nested within the CAMELIA trial.

Antibody-Dependent Cell-Mediated Cytotoxicity in Simian Immunodeficiency Virus-Infected Rhesus Monkeys

IMMUNOLOGICAL MEMORY. CD4 T Follicular Helper Cells. Memory CD8 T Cell Differentiation

Nature Medicine: doi: /nm.2109

Supplementary Figure 1

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

Supplementary Materials

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

IgG3 regulates tissue-like memory B cells in HIV-infected individuals

Th17 and Th17/Treg ratio at early HIV infection associate with protective HIV-specific CD8 + T-cell responses and disease progression

Long-term persistence of T cell memory in Italian vaccinees

On an individual level. Time since infection. NEJM, April HIV-1 evolution in response to immune selection pressures

Figure S1. Alignment of predicted amino acid sequences of KIR3DH alleles identified in 8

DNA and Protein Vaccination Confers Protection Upon Mucosal Challenge with Heterologous SIVsmE660 (OA 10.04)

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

Virus-specific CD8 cytotoxic T lymphocytes (CTLs) are major

Abstract. Introduction. Results

Received 4 December 2001/Accepted 29 April 2002

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

Supplemental materials

ndln NK Cells (x10 3 ) Days post-infection (A/PR/8) *** *** *** Liver NK Cells (x10 4 ) Days post-infection (MCMV)

White Blood Cells (WBCs)

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

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

HIV Anti-HIV Neutralizing Antibodies

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

Antigen Presentation and T Lymphocyte Activation. Abul K. Abbas UCSF. FOCiS

Antibody Dependent Cellular Cytotxic activity: Past and Future. Guido Ferrari, M.D. Duke University Medical Center

NIH Public Access Author Manuscript Nat Med. Author manuscript; available in PMC 2010 September 1.

Supplemental Figure 1

Are we targeting the right HIV determinants?

Supplementary Figure 1 Cytokine receptors on developing thymocytes that can potentially signal Runx3d expression.

Cellular Immune response. Jianzhong Chen, Ph.D Institute of immunology, ZJU

Is an HIV Vaccine Possible?

Nature Neuroscience: doi: /nn Supplementary Figure 1. Behavioral training.

Fayth K. Yoshimura, Ph.D. September 7, of 7 HIV - BASIC PROPERTIES

Supplementary Materials for

Therapeutic DNA Vaccine Induces Broad T Cell Responses in the Gut and Sustained Protection from Viral Rebound and AIDS in SIV-Infected Rhesus Macaques

Received 19 September 2007/Accepted 21 November 2007

Human Immunodeficiency Virus

Supplementary Figure 1. Gating strategy and quantification of integrated HIV DNA in sorted CD4 + T-cell subsets.

Spleen. mlns. E Spleen 4.1. mlns. Spleen. mlns. Mock 17. Mock CD8 HIV-1 CD38 HLA-DR. Ki67. Spleen. Spleen. mlns. Cheng et al. Fig.

Supporting Information

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

An Evolutionary Story about HIV

David Verhoeven, Sumathi Sankaran, Melanie Silvey, and Satya Dandekar*

Simian-Human Immunodeficiency Infection Is the Course Set in the Acute Phase?

Supplementary Figure 1. IL-12 serum levels and frequency of subsets in FL patients. (A) IL-12

Chapter 1. Chapter 1 Concepts. MCMP422 Immunology and Biologics Immunology is important personally and professionally!

Question 1. Kupffer cells, microglial cells and osteoclasts are all examples of what type of immune system cell?

SUPPLEMENTARY INFORMATION

Immune response to infection

Supplemental Figure 1. Gating strategies for flow cytometry and intracellular cytokinestaining

T Memory Stem Cells: A Long-term Reservoir for HIV-1

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

Fluid movement in capillaries. Not all fluid is reclaimed at the venous end of the capillaries; that is the job of the lymphatic system

Cytotoxic T Cells in HIV and Other Retroviral Infections

Medical Virology Immunology. Dr. Sameer Naji, MB, BCh, PhD (UK) Head of Basic Medical Sciences Dept. Faculty of Medicine The Hashemite University

CLINICAL. Immune Restoration in the Context of HAART

Chapter 35 Active Reading Guide The Immune System

A VACCINE FOR HIV BIOE 301 LECTURE 10 MITALI BANERJEE HAART

Massive infection and loss of memory CD4 + T cells in multiple tissues during acute SIV infection

B220 CD4 CD8. Figure 1. Confocal Image of Sensitized HLN. Representative image of a sensitized HLN

Defining kinetic properties of HIV-specific CD8 + T-cell responses in acute infection

The encephalitogenicity of TH17 cells is dependent on IL-1- and IL-23- induced production of the cytokine GM-CSF

Principles of Adaptive Immunity

The Journal of Experimental Medicine

SYSTEMS BIOLOGY APPROACHES TO IDENTIFY MECHANISMS OF IMMUNE MEDIATED PROTECTION TRANSLATING RESEARCH INTO HEALTH

<10. IL-1β IL-6 TNF + _ TGF-β + IL-23

Impaired T Lymphocyte Responses in Older Macaques: Possible Implications for Lentiviral Disease Progression

DEBATE ON HIV ENVELOPE AS A T CELL IMMUNOGEN HAS BEEN GAG-GED

Approaching a Cure Daniel R. Kuritzkes, MD

Rapid Influx and Death of Plasmacytoid Dendritic Cells in Lymph Nodes Mediate Depletion in Acute Simian Immunodeficiency Virus Infection

Gp96-Ig-SIV VACCINES INDUCE PREDOMINANT IMMUNE RESPONSES AT MUCOSAL SITES

Increased Loss of CCR5 CD45RA CD4 T Cells in CD8 Lymphocyte-Depleted Simian Immunodeficiency Virus-Infected Rhesus Monkeys

MHC class I MHC class II Structure of MHC antigens:

JAK3 inhibitor administration in vivo in chronically SIV Infected Rhesus Macaques

Mon, Wed, Fri 11:00 AM-12:00 PM. Owen, Judy, Jenni Punt, and Sharon Stranford Kuby-Immunology, 7th. Edition. W.H. Freeman and Co., New York.

Effector memory T cell responses are associated with protection of rhesus monkeys from mucosal simian immunodeficiency virus challenge

Nature Immunology: doi: /ni Supplementary Figure 1. Transcriptional program of the TE and MP CD8 + T cell subsets.

Scott Abrams, Ph.D. Professor of Oncology, x4375 Kuby Immunology SEVENTH EDITION

Immune correlates of protection from congenital cytomegalovirus after primary infection in pregnancy

Supplementary Information. Tissue-wide immunity against Leishmania. through collective production of nitric oxide

ILC1 and ILC3 isolation and culture Following cell sorting, we confirmed that the recovered cells belonged to the ILC1, ILC2 and

(ii) The effective population size may be lower than expected due to variability between individuals in infectiousness.

SUPPLEMENTARY INFORMATION. Divergent TLR7/9 signaling and type I interferon production distinguish

JVI Accepts, published online ahead of print on 7 March 2007 J. Virol. doi: /jvi

Heterosubtypic immunity. Professor Ajit Lalvani FMedSci Chair of Infectious Diseases 14/07/2014

The Role of B Cell Follicles in HIV Replication and Persistence

ABSTRACT. HIV-induced immunodeficiency may be mediated by the activation of

NK mediated Antibody Dependent Cellular Cytotoxicity in HIV infections

Alexander O. Pasternak, Mirte Scherpenisse, Ben Berkhout

Helminth worm, Schistosomiasis Trypanosomes, sleeping sickness Pneumocystis carinii. Ringworm fungus HIV Influenza

La risposta immune all infezione da virus ebola. Chiara Agrati, PhD

Transcription:

SUPPORTING INFORMATION FOR: Treatment with IL-7 Prevents the Decline of Circulating CD4 + T Cells during the Acute Phase of SIV Infection in Rhesus Macaques Lia Vassena, 1,2 Huiyi Miao, 1 Raffaello Cimbro, 1 Mauro S. Malnati, 2 Giulia Cassina, 2 Michael A. Proschan, 3 Vanessa M. Hirsch, 4 Bernard A. Lafont, 5 Michel Morre, 6 Anthony S. Fauci, 1 and Paolo Lusso 1 1 Laboratory of Immunoregulation, 3 Biostatistics Research Branch and 4 Laboratory of Molecular Medicine, 5 Laboratory of Molecular Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892; 2 Human Virology Unit, DIBIT-HSR, 20132 Milano, Italy; and 6 Cytheris, Issy-les Moulineaux 92370, France This file contains: - Supplementary Methods - Supplementary Data - Supplementary References - Tables S1-S2 - Figures S1-S6

L. Vassena et al., PLoS Pathogens (Supporting Information) 2 SUPPLEMENTARY METHODS MHC screening and study groups A critical issue that we considered in the study design was the assignment of the 12 animals to the two study groups (i.e., IL-7 treatment and control) in order to avoid potential biases in SIVdisease susceptibility. Since previous work identified MHC class-i alleles with disease/viral load-protective (Mamu-A01, Mamu-A08, Mamu-B08) or -enhancing (Mamu-B01) properties [1-4], we pre-screened the animals for defined Mamu haplotypes. A positive score (+1) was attributed to the presence of Mamu-A01, A08 or B08; a negative score (-1) to the presence of Mamu-B01 (Table S2). The final study groups were balanced considering both MHC scores and baseline counts of peripheral blood CD3 + and CD4 + T lymphocytes (Table S2). Statistical analysis using the O Brien test To compare untreated and IL-7-treated animals with respect to changes from baseline to multiple time points simultaneously we used the O Brien test, which is a natural extension of the Wilcoxon rank sum test to accommodate multiple time points per subject. The analysis was used to compare simultaneously the changes from baseline to each time point during the treatment period (day 7, 14, 21, 28, and 35). First, we ranked changes from baseline to each of the time points separately, and then for each animal we averaged the ranks across the time points. To assess statistical significance, a permutation test was used. Under the null hypothesis of no effect of IL-7 treatment, one should not be able to distinguish treated monkeys from controls; the control monkeys could have been any group of 5 selected from the 11 total (excluding monkey #744 for which several time points were missing). Considering that there are 462 ways to select 5 subjects from 11, for each of these relabelings, we computed the difference between the average for animals labeled as IL-7-treated and the average for animals labeled as untreated. Afterwards, we computed the proportion of relabeled datasets that produced a mean difference at least as extreme as the actual trial data.

L. Vassena et al., PLoS Pathogens (Supporting Information) 3 SUPPLEMENTARY DATA Rapid disease progression in 4 SIV-infected macaques: re-analysis of virological and immunological data after exclusion of rapid progressors During follow-up, two monkeys in each group (H745, H749 in the untreated group; H751, H752 in the IL-7-treated group) developed early signs of SIV-disease progression and were euthanized for humanitarian reasons after a mean of 143.5 ± 20.4 days (range, 115-163). No specific MHC allele was over- or under-represented among these 4 animals. Albeit infrequently, SIV-infected macaques have been shown to undergo a rapid disease course characterized by high-level virus replication in cells of the mononuclear phagocytic lineage rather than in CD4 + T cells [5]. In agreement with these previous observations, our rapid progressors (RP) showed persistently higher levels of SIV plasma viremia (Figure S1A) and antigenemia (Figure S1B), compared to conventional progressors (CP), as well as overall higher numbers of circulating CD4 + T cells, even though the difference reached statistical significance only on day 62 post-infection (Figure S1C). Although it is impossible, without extending the study to a larger group of animals, to establish if the RP course was affected by IL-7 treatment, the fact that two animals in each group progressed rapidly suggests that IL-7 did not influence this unique form of SIV-disease evolution. Since the rapid disease course has been associated with an unusual pathogenesis [5], which is likely influenced by genetic factors and insensitive to IL-7 treatment, we reasoned that the inclusion of RP animals could be a confounding factor in our study, and therefore we reanalyzed all the immunological and virological data after exclusion of these 4 animals. This reanalysis showed no major changes in the statistical comparisons between treated and untreated animals regarding SIV viremia and antigenemia (Figure S2A), CD4 +, CD8 +, naïve, memory and effector T cells in peripheral blood (Figure S2B), ileum and lymph nodes (data not shown). However, the exclusion of RP animals had important effects on the analysis of T-cell apoptosis in lymph nodes, as described in the main text.

L. Vassena et al., PLoS Pathogens (Supporting Information) 4 Changes in T-cell subpopulations in acutely SIV-infected macaques The O Brien permutation test was used to simultaneously compare the changes in naïve, memory and effector CD3 +, CD4 + and CD8 + T cells from baseline to all the time points during the treatment period (day 7-35 post-infection) in IL-7-treated versus untreated animals. As shown in Figure S3A, this analysis demonstrated that the changes in the total numbers of CD3 +, CD4 + and CD8 + T cells were significantly different in IL-7-treated and untreated macaques (Figure S3A). Likewise, significant differences between IL-7-treated and untreated animals were also observed for naïve, memory and effector CD4 + and CD8 + T cells, as well as for naïve and effector CD3 + T cells, but not for memory CD3 + T cells (Figure S3B). However, when the analysis of changes was restricted to day 14 post-infection, the differences between IL-7-treated and untreated animals were significant for all T-cell subpopulations (not shown).

L. Vassena et al., PLoS Pathogens (Supporting Information) 5 SUPPLEMENTARY REFERENCES 1. Muhl T, Krawczak M, Ten Haaft P, Hunsmann G, Sauermann U (2002) MHC class I alleles influence set-point viral load and survival time in simian immunodeficiency virus-infected rhesus monkeys. J Immunol 169: 3438-3446. 2. Pal R, Venzon D, Letvin NL, Santra S, Montefiori DC, et al. (2002) ALVAC-SIV-gag-polenv based vaccination and macaque major histocompatibility complex class I (A*01) delay simian immunodeficiency virus SIVmac-induced immunodeficiency. J Virol 76: 292-302. 3. Loffredo JT, Maxwell J, Qi Y, Glidden CE, Borchardt GJ, et al. (2007) Mamu-B*08-positive macaques control simian immunodeficiency virus replication. J Virol 81: 8827-8832. 4. Mothe BR, Weinfurter J, Wang C, Rehrauer W, Wilson N, et al. (2003) Expression of the major histocompatibility complex class I molecule Mamu-A*01 is associated with control of simian immunodeficiency virus SIVmac239 replication. J Virol 77: 2736-2740. 5. Brown CR, Czapiga M, Kabat J, Dang Q, Ourmanov I, et al. (2007) Unique pathology in simian immunodeficiency virus-infected rapid progressor macaques is consistent with a pathogenesis distinct from that of classical AIDS. J Virol 81: 5594-5606.

L. Vassena et al., PLoS Pathogens (Supporting Information) 6 SUPPLEMENTARY TABLES Table S1. P values for the comparison of SIV viremia, antigenemia and DNA load between IL-7-treated and untreated monkeys, obtained by Wilcoxon rank sum test. Statistically significant p values are bolded. Day SIV Viremia SIV DNA SIV Antigenemia Blood GALT Lymph nodes 4 0.0430 0.3095 nt nt nt 7 0.2403 0.3095 nt nt nt 11 0.2403 0.2403 nt nt nt 14 0.0823 0.7922 0.9048 0.4000 nt 18 0.9307 0.9307 nt nt nt 21 0.7922 0.9307 nt nt nt 24 0.4286 0.9307 nt nt 0.4000 28 0.6623 0.6623 nt nt nt 35 0.4286 0.9307 nt nt nt 41 0.7922 0.9307 nt nt nt 48 nt 0.9307 nt nt nt 54 nt 0.9307 nt nt nt 68 0.6623 1.0000 nt nt nt 77 0.7922 1.0000 0.4000 nt nt nt = not tested

L. Vassena et al., PLoS Pathogens (Supporting Information) 7 Table S2. MHC haplotype and baseline peripheral blood CD3 + and CD4 + T-lymphocyte counts in untreated and IL-7-treated macaques. MHC MHC CD3 + CD4 + haplotype score* T cells T cells Untreated: H743 A01-A08 +2 1831 925 H744 B01-1 2471 1774 H745 A08 +1 2878 1879 H747 A08-B01 0 2097 1317 H749 H753 n.d.** B08 0 +1 6771 3194 3656 1996 Mean (±SD): +3 3207±1815 1925±938 IL-7-treated: H746 B08 +1 2604 1338 H748 A01 +1 3167 2211 H750 A08 +1 3972 2367 H751 B01-1 3223 2269 H752 n.d.** 0 2767 1486 H754 A02 0 2233 1351 Mean (±SD): +2 2994±603 1845±483 * The MHC score was calculated by attributing a positive score (+1) to alleles associated with protection from SIV disease/viral load and a negative score (-1) to alleles associated with increased viral load/accelerated disease progression; no score was attributed to alleles with no known effects on SIV disease progression. ** n.d. = none detected. The animals were negative for the MHC alleles tested (A01, A02, A08, A11, B01, B03, B04, B08, B17).

L. Vassena et al., PLoS Pathogens (Supporting Information) 8 SUPPLEMENTARY FIGURES Figure S1. Levels of SIV replication and CD4 + T cells in conventional progressor (CP) versus rapid progressor (RP) SIV-infected macaques. Mean levels (± SEM) of SIV plasma viremia (A) and p27 Gag antigenemia (B) in CP (orange) and RP (green). RP animals showed higher levels of both SIV viremia and antigenemia compared to CP at several time points, starting on day 18 post-infection, as indicated by the asterisks (*p < 0.05, ** p < 0.01, by Wilcoxon rank sum test). (C) Mean absolute numbers (± SEM) of circulating CD4 + T cells in CP (orange) and RP (green). A significant difference was observed on day 62 post-infection, when RP had higher mean levels of circulating CD4 + T cells compared to CP, as indicated by the asterisk (p < 0.042, by Wilcoxon rank sum test).

L. Vassena et al., PLoS Pathogens (Supporting Information) 9 A B Figure S2. Reanalysis of virological and immunological parameters in IL-7-treated and untreated macaques after exclusion of macaques with rapidly progressive (RP) disease course. (A) Mean levels (± SEM) of SIV plasma viremia and p27 Gag antigenemia in untreated (blue) and IL-7-treated (red) animals. (B) Mean absolute numbers (± SEM) of circulating total, naïve, memory and effector CD4 + and CD8 + T cells in untreated (blue) and IL-7-treated (red) animals. The grey shaded area indicates the IL-7-treatment period; blue and red asterisks denote significant differences with baseline values in untreated and IL-7-treated animals, respectively (*p < 0.05, ** p < 0.01, by paired Student s t test).

L. Vassena et al., PLoS Pathogens (Supporting Information) 10 A B Figure S3. Comparison of changes in peripheral blood T-cell subpopulations between IL-7- treated and untreated macaques during the acute phase of SIV infection. Simultaneous comparisons of changes from baseline to all the time points during acute SIV infection as analyzed by the O Brien permutation tests. (A) Analyses for total CD3 +, CD4 + and CD8 + T cells. (B) Analyses for the naïve, memory and effector T-cell subsets. The histograms represent the distribution of all possible values of the difference between IL-7-treated and untreated animals considering all the possible relabelings of the animals in the two groups (by permutation test); dots indicate the values of the differences between IL-7-treated and untreated animals for the actual trial assignments with the relative p values. Significant differences were observed in all T- cell subsets, except in memory CD3 + T cells.

L. Vassena et al., PLoS Pathogens (Supporting Information) 11 Figure S4. Ki67 expression, Annexin-V binding and Bcl-2 expression in two representative animals (one untreated and one treated with IL-7). Histograms show the levels of Ki67 expression (A), Annexin-V binding (B) and Bcl-2 expression (C) at day 14 post-infection in CD4 + and CD8 + T cells from two representative animals: one from the untreated group (#749, shown in blue) and one from the IL-7-treated group (#746, shown in red).

L. Vassena et al., PLoS Pathogens (Supporting Information) 12 A B B Figure S5. SIV Gag-specific T-cell responses in IL-7-treated and untreated macaques. (A) Mean absolute numbers (± SEM) of CD4 + and CD8 + T cells producing one cytokine (singleproducer, SP), two cytokines (double-producer, DP) or all three cytokines (triple-producer, TP) in response to SIV Gag peptide stimulation in untreated (shades of blue) and IL-7-treated (shades of red) animals. (B) Mean absolute numbers of SIV Gag-responding CD4 + and CD8 + T cells in untreated controls (c) and IL-7-treated animals (IL-7). The bars indicate the mean numbers of total responding cells; the colors indicate the mean numbers of IFN-γ, IL-2, or MIP-1β singleproducer cells (shades of blue), IFN-γ/IL-2, IFN-γ/MIP-1β or IL-2/MIP-1β double-producer cells (shades of green), and IFN-γ/IL-2/MIP-1β triple-producer cells (purple). The numbers above each bar indicate the fraction of monkeys that gave a measurable response over background to SIV Gag peptides at the corresponding time point. The grey-shaded areas indicate the IL-7- treatment period.

L. Vassena et al., PLoS Pathogens (Supporting Information) 13 Figure S6. Gating strategy used for the identification of naïve and memory T-cell subsets. Lymphocytes were selected within the lymphocyte gate in a SSC vs FSC dot plot and T cells were further selected for CD3 positivity. Within the CD3 + T-cells gate, CD45RA - cells were selected and further subdivided into central memory (T CM ), transitional memory (T TM ) and effector memory (T EM ) cells by using a combination of the surface markers CCR7 and CD28 as follows: T CM CCR7 + CD28 +, T TM CCR7 - CD28 + and T EM CCR7 - CD28 -. Within the CD45RA + subpopulation, naïve (T N ) and terminal effector (T TE ) cells were identified using the same combination of CCR7 and CD28 markers as double positive and double negative cells, respectively. CD4 + and CD8 + T-cell lineages cells were subsequently identified on the gated subpopulations.