Targeting α4β7 integrin reduces mucosal transmission of simian immunodeficiency virus and protects gut-associated lymphoid tissue from infection

Siddappa N Byrareddy, Brianne Kallam, James Arthos, Claudia Cicala, Fatima Nawaz, Joseph Hiatt, Ellen N Kersh, Janet M McNicholl, Debra Hanson, Keith A Reimann, Markus Brameier, Lutz Walter, Kenneth Rogers, Ann E Mayne, Paul Dunbar, Tara Villinger, Dawn Little, Tristram G Parslow, Philip J Santangelo, Francois Villinger, Anthony S Fauci, Aftab A Ansari, Siddappa N Byrareddy, Brianne Kallam, James Arthos, Claudia Cicala, Fatima Nawaz, Joseph Hiatt, Ellen N Kersh, Janet M McNicholl, Debra Hanson, Keith A Reimann, Markus Brameier, Lutz Walter, Kenneth Rogers, Ann E Mayne, Paul Dunbar, Tara Villinger, Dawn Little, Tristram G Parslow, Philip J Santangelo, Francois Villinger, Anthony S Fauci, Aftab A Ansari

Abstract

α4β7 integrin-expressing CD4(+) T cells preferentially traffic to gut-associated lymphoid tissue (GALT) and have a key role in HIV and simian immunodeficiency virus (SIV) pathogenesis. We show here that the administration of an anti-α4β7 monoclonal antibody just prior to and during acute infection protects rhesus macaques from transmission following repeated low-dose intravaginal challenges with SIVmac251. In treated animals that became infected, the GALT was significantly protected from infection and CD4(+) T cell numbers were maintained in both the blood and the GALT. Thus, targeting α4β7 reduces mucosal transmission of SIV in macaques.

Conflict of interest statement

COMPETING FINANCIAL INTERESTS

The authors declare no competing financial interests. The findings in this report are those of the authors and do not necessarily reflect the views of the Centers for Disease Control and Prevention.

Figures

Fig. 1. Kinetics of plasma viral load…
Fig. 1. Kinetics of plasma viral load and tissue and organ specific pro-viral DNA loads
Plasma viral loads from two groups of macaques (n=12 for each group) that received 50 mg/kg of either aα4β7-mAb (a) or a rhesus IgG (b) I.V. at 3 week intervals following the initiation of six, once-weekly, low-dose intra-vaginal challenges with SIVmac251 (arrows). Kaplan-Meier curves based on plasma viral RNA (n=12 each group; log-rank test; p=0.002) (c).Pro-viral DNA load (# of copies/ng DNA) in rectal biopsies from α4β7-mAb (d) or IgG control macaques (e) (n=6 for α4β7-mAb, n=10 for IgG control).Median values of data in d and e (random mixed effects (RME) model; p=0.006) (f). Two infected macaques from each group (α4β7-mAb in blue (T), control IgG in red (C)) were sacrificed at 2 weeks p.i. (filled circles) and two sacrificed at 16–18 weeks p.i. (open circles). Pro-viral DNA load (# copies/ng DNA) in jejunal, ileum and colon tissue samples from both groups (n=4; Mann-Whitney U-test; P<0.01) (g), spleen, and internal iliac, colonic, inguinal, axillary, mesenteric lymph-nodes (n=4; Mann-Whitney U-test; P<0.01) from both groups (h). Pro-viral DNA load (# copies/ng DNA) from cervical tissue samples from 3 macaques from both groups (i). Macaque I.D.’s in panels a, b, d and e are specified.
Fig. 2. Frequency of lymphocyte subsets from…
Fig. 2. Frequency of lymphocyte subsets from infected macaque PBMCs and inhibition of MAdCAM or SIVmac251 gp120 by α4β7-mAb
Changes in the absolute numbers of CD4+ T cells (top left), CD8+ T cells (top right), NK cells (bottom left) and B cells (bottom right) in PBMC from infected α4β7-mAb (blue) or IgG treated (red) and uninfected (purple) macaques (n=6 for α4β7-mAb infected, n=10 for IgG infected, n=8 for uninfected); median values are given (RME model) (a).Frequencies of CD4+CD45+ T cells in cytobrush samples (n=6 for α4β7-mAb, n=9 for IgG; RME model; p=0.052) (b), and gut biopsy(c) (n=6 for α4β7 mAb and n=9 for IgG; RME model; p<0.0001).Flow-cytometric analysis of CD4+α4β7+ T cells stained with PE labeled MAdCAM-Ig in the presence of IgG (d) or α4β7-mAb (e) in the presence of MgCl2. MAdCAM-Ig-reactive cells are included within the blue box and the percent reactive cells are indicated. Binding of neutravidin-PE alone (lane 1), increasing amounts of PE labeled SIVMac251 gp120 (lanes 2–4), 2μg PE labeled SIVMac251 gp120 in the presence of an unlabeled α4 mAb (lane 5), and 2μg PE labeled SIVMac251 gp120 in the absence of divalent cations (lane 6) as controls, to CD4+α4β7+ T cells (f). Flow-cytometric profile of CD4+α4β7+ T cells stained with neutravidin PE alone (grey) or PE labeled SIVmac251 gp120 in the absence (blue) or presence (red) of unlabeled α4β7-mAb (g). CD4+α4β7+ T cells stained with neutravidin-PE alone (grey) or labeled SIVmac251 gp120 in the absence (blue) or presence (red) of unlabeled MAdCAM-Ig (h). Experiments shown in panels f, g and h were performed in presence of an anti-CD4 mAb.

References

    1. Brenchley JM, Douek DC. The mucosal barrier and immune activation in HIV pathogenesis. Curr Opin HIV AIDS. 2008;3:356–361.
    1. Mehandru S, et al. Primary HIV-1 infection is associated with preferential depletion of CD4+ T lymphocytes from effector sites in the gastrointestinal tract. J Exp Med. 2004;200:761–770.
    1. Veazey RS, et al. Gastrointestinal tract as a major site of CD4+ T cell depletion and viral replication in SIV infection. Science. 1998;280:427–431.
    1. Bargatze RF, Jutila MA, Butcher EC. Distinct roles of L-selectin and integrins alpha 4 beta 7 and LFA-1 in lymphocyte homing to Peyer’s patch-HEV in situ: the multistep model confirmed and refined. Immunity. 1995;3:99–108.
    1. Erle DJ, et al. Expression and function of the MAdCAM-1 receptor, integrin alpha 4 beta 7, on human leukocytes. J Immunol. 1994;153:517–528.
    1. Arthos J, et al. HIV-1 envelope protein binds to and signals through integrin alpha4beta7, the gut mucosal homing receptor for peripheral T cells. Nat Immunol. 2008;9:301–309.
    1. Nakamura GR, Fonseca DP, O’Rourke SM, Vollrath AL, Berman PW. Monoclonal antibodies to the V2 domain of MN-rgp120: fine mapping of epitopes and inhibition of alpha4beta7 binding. PloS one. 2012;7:e39045.
    1. Nawaz F, et al. The Genotype of Early-Transmitting HIV gp120s Promotes alpha(4)beta(7) -Reactivity, Revealing alpha(4)beta(7)/CD4 T cells As Key Targets in Mucosal Transmission. PLoS Pathog. 2011;7:e1001301.
    1. Pereira LE, et al. Preliminary in vivo efficacy studies of a recombinant rhesus anti-alpha(4)beta(7) monoclonal antibody. Cell Immunol. 2009;259:165–176.
    1. Ansari AA, et al. Blocking of alpha4beta7 gut-homing integrin during acute infection leads to decreased plasma and gastrointestinal tissue viral loads in simian immunodeficiency virus-infected rhesus macaques. J Immunol. 2011;186:1044–1059.
    1. Kwa S, et al. Plasmacytoid dendritic cells are recruited to the colorectum and contribute to immune activation during pathogenic SIV infection in rhesus macaques. Blood. 2011;118:2643–2644.
    1. Kelly KA, Rank RG. Identification of homing receptors that mediate the recruitment of CD4 T cells to the genital tract following intravaginal infection with Chlamydia trachomatis. Infection and immunity. 1997;65:5198–5208.
    1. Cicala C, et al. The integrin alpha4beta7 forms a complex with cell-surface CD4 and defines a T-cell subset that is highly susceptible to infection by HIV-1. Proc Natl Acad Sci U S A. 2009;106:20877–20882.
    1. Kader M, et al. Alpha4(+)beta7(hi)CD4(+) memory T cells harbor most Th-17 cells and are preferentially infected during acute SIV infection. Mucosal Immunol. 2009;2:439–449.
    1. Martinelli E, et al. The frequency of alpha4beta7 high memory CD4+ T cells correlates with susceptibility to rectal SIV infection. J Acquir Immune Defic Syndr. 2013;21:21.
    1. McKinnon LR, et al. Characterization of a human cervical CD4+ T cell subset coexpressing multiple markers of HIV susceptibility. J Immunol. 2011;187:6032–6042.
    1. Feagan BG, et al. Vedolizumab as induction and maintenance therapy for ulcerative colitis. The New England journal of medicine. 2013;369:699–710.
    1. Jovani M, Danese S. Vedolizumab for the treatment of IBD: a selective therapeutic approach targeting pathogenic a4b7 cells. Current drug targets. 2013;14:1433–1443.
    1. Sandborn WJ, et al. Vedolizumab as induction and maintenance therapy for Crohn’s disease. The New England journal of medicine. 2013;369:711–721.
    1. Danese S, et al. Biological agents for moderately to severely active ulcerative colitis: a systematic review and network meta-analysis. Annals of internal medicine. 2014;160:704–711.
    1. Moreland AJ, et al. Characterization of killer immunoglobulin-like receptor genetics and comprehensive genotyping by pyrosequencing in rhesus macaques. BMC genomics. 2011;12:295.
    1. O’Leary CE, et al. Identification of novel MHC class I sequences in pig-tailed macaques by amplicon pyrosequencing and full-length cDNA cloning and sequencing. Immunogenetics. 2009;61:689–701.
    1. Lim SY, et al. TRIM5alpha Modulates Immunodeficiency Virus Control in Rhesus Monkeys. PLoS Pathog. 2010;6:e1000738.
    1. Nguyen DC, Scinicariello F, Attanasio R. Characterization and allelic polymorphisms of rhesus macaque (Macaca mulatta) IgG Fc receptor genes. Immunogenetics. 2011;63:351–362.
    1. Butler K, et al. Susceptibility to repeated, low-dose, rectal SHIVSF162P3 challenge is independent of TRIM5 genotype in rhesus macaques. AIDS research and human retroviruses. 2013;29:1091–1094.
    1. Subbarao S, et al. Direct stringency comparison of two macaque models (single-high vs. repeat-low) for mucosal HIV transmission using an identical anti-HIV chemoprophylaxis intervention. Journal of medical primatology. 2007;36:238–243.
    1. Ansari AA, et al. Blocking of alpha4beta7 gut-homing integrin during acute infection leads to decreased plasma and gastrointestinal tissue viral loads in simian immunodeficiency virus-infected rhesus macaques. J Immunol. 2011;186:1044–1059.
    1. Hudgens MG, Gilbert PB. Assessing vaccine effects in repeated low-dose challenge experiments. Biometrics. 2009;65:1223–1232.
    1. Regoes RR, Longini IM, Feinberg MB, Staprans SI. Preclinical assessment of HIV vaccines and microbicides by repeated low-dose virus challenges. PLoS medicine. 2005;2:e249.
    1. Letvin NL, et al. No evidence for consistent virus-specific immunity in simian immunodeficiency virus-exposed, uninfected rhesus monkeys. Journal of virology. 2007;81:12368–12374.
    1. Promadej-Lanier N, et al. Resistance to Simian HIV infection is associated with high plasma interleukin-8, RANTES and Eotaxin in a macaque model of repeated virus challenges. J Acquir Immune Defic Syndr. 2010;53:574–581.
    1. Veazey RS, Shattock RJ, Klasse PJ, Moore JP. Animal models for microbicide studies. Current HIV research. 2012;10:79–87.
    1. Spear G, Rothaeulser K, Fritts L, Gillevet PM, Miller CJ. In captive rhesus macaques, cervicovaginal inflammation is common but not associated with the stable polymicrobial microbiome. PloS one. 2012;7:e52992.
    1. McKinnon LR, et al. Characterization of a human cervical CD4+ T cell subset coexpressing multiple markers of HIV susceptibility. J Immunol. 2011;187:6032–6042.
    1. Pereira LE, et al. Preliminary in vivo efficacy studies of a recombinant rhesus anti-alpha(4)beta(7) monoclonal antibody. Cell Immunol. 2009;259:165–176.
    1. Arthos J, et al. HIV-1 envelope protein binds to and signals through integrin alpha4beta7, the gut mucosal homing receptor for peripheral T cells. Nat Immunol. 2008;9:301–309.

Source: PubMed

3
구독하다