Antiretroviral Therapy in Simian Immunodeficiency Virus-Infected Sooty Mangabeys: Implications for AIDS Pathogenesis

Similar documents
CTLA-4-expressing CD4 T cells are critical contributors to SIV viral persistence

Combined IL-21 and IFNα treatment limits inflammation and delay viral rebound in ART-treated, SIV-infected macaques

Interleukin-21 combined with ART reduces inflammation and viral reservoir in SIV-infected macaques

Novel Approaches for the Assessment of the In Vivo Residual Virus Pool and Viral Eradication Strategies in SIV-infected Rhesus Macaques

Severe Depletion of Mucosal CD4 + T Cells in AIDS-Free Simian Immunodeficiency Virus-Infected Sooty Mangabeys

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

Analysis of the In Vivo Turnover of CD4+T-Cell Subsets in Chronically SIV-Infected Sooty Mangabeys

Availability of activated CD4+ T cells dictates the level of viremia in naturally SIV-infected sooty mangabeys

Establishment and Targeting of the Viral Reservoir in Rhesus Monkeys

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

Copyright information:

Nature Medicine: doi: /nm.2109

Over thousands of years, species-specific

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

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

Mother-to-Infant Transmission of Simian Immunodeficiency Virus Is Rare in Sooty Mangabeys and Is Associated with Low Viremia

Supplementary Figure 1

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

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

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

JAK3 inhibitor administration in vivo in chronically SIV Infected Rhesus Macaques

Supporting Information

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

Experimental SIV infection of rhesus macaques (RMs),

Viral Reservoirs and anti-latency interventions in nonhuman primate models of SIV/SHIV infection

With over 20 drugs and several viable regimens, the mo6vated pa6ent with life- long access to therapy can control HIV indefinitely, elimina6ng the

HIV 101: Fundamentals of HIV Infection

Received 4 December 2001/Accepted 29 April 2002

Supplementary Materials

Target Cell Availability, Rather than Breast Milk Factors, Dictates Mother-to-Infant Transmission of SIV in Sooty Mangabeys and Rhesus Macaques

Therapeutic strategies for immune reconstitution in acquired immunodeficiency syndrome

Supplementary Materials for

Supplementary Figures. Supplementary Figure 1. Treatment schematic of SIV infection and ARV and PP therapies.

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

CROI 2016 Review: Immunology and Vaccines

PD-1 blockade during chronic SIV infection reduces hyperimmune activation and microbial translocation in rhesus macaques

CD4 Depletion in SIV-Infected Macaques Results in Macrophage and Microglia Infection with Rapid Turnover of Infected Cells

cure research HIV & AIDS

HIV Basics: Pathogenesis

Chronic HIV-1 Infection Frequently Fails to Protect against Superinfection

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

Where the Wild Things Are: Pathogenesis of SIV Infection in African Nonhuman Primate Hosts

IAS 2015 Towards an HIV Cure symposium Vancouver. Distinct HIV Genetic Populations in Effector Memory T Cells after Prolonged Therapy

VIRAL TITER COUNTS. The best methods of measuring infectious lentiviral titer

SUPPLEMENTARY FIGURES

Guidelines for the Use of Antiretroviral Agents in HIV-1-Infected Adults and Adolescents

Rapid Seeding of the Viral Reservoir Prior to SIV Viremia in Rhesus Monkeys

HIV 101: Overview of the Physiologic Impact of HIV and Its Diagnosis Part 2: Immunologic Impact of HIV and its Effects on the Body

HIV-1 Viral Load Real Time (RG)

A VACCINE FOR HIV BIOE 301 LECTURE 10 MITALI BANERJEE HAART

Received 7 November 2007/Accepted 15 January 2008

January 25, 2017 Scientific Research Process Name of Journal: ESPS Manuscript NO: Manuscript Type: Title: Authors: Correspondence to

HLA-DR CD38 CD4 T Lymphocytes Have Elevated CCR5 Expression and Produce the Majority of R5-Tropic HIV-1 RNA In Vivo

Supporting Information

Can HIV be cured? (how about long term Drug free remission?)

Središnja medicinska knjižnica

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

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

Decay characteristics of HIV-1- infected compartments during combination therapy

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

Blocking antibodies and peptides. Rat anti-mouse PD-1 (29F.1A12, rat IgG2a, k), PD-

A Highly Intensified ART Regimen Induces Long-Term Viral Suppression and Restriction of the Viral Reservoir in a Simian AIDS Model

HIV Reservoirs in Developing Countries: Implication for HIV CURE Strategies

Supplementary Information

MID 36. Cell. HIV Life Cycle. HIV Diagnosis and Pathogenesis. HIV-1 Virion HIV Entry. Life Cycle of HIV HIV Entry. Scott M. Hammer, M.D.

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

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

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

Journal of Infectious Diseases Advance Access published July 11, Effect of Antiretroviral Therapy on HIV Reservoirs in Elite Controllers

CD4 T Cells, Including Th17 and Cycling Subsets, Are Intact in the Gut Mucosa of HIV-1-Infected Long-Term Nonprogressors

Alexander O. Pasternak, Mirte Scherpenisse, Ben Berkhout

York criteria, 6 RA patients and 10 age- and gender-matched healthy controls (HCs).

Dr Anna Herasimtschuk

Micro 301 HIV/AIDS. Since its discovery 31 years ago 12/3/ Acquired Immunodeficiency Syndrome (AIDS) has killed >32 million people

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

Complete Transcriptome Analysis of Latently Infected CD4 + T Cells

Supplemental Materials and Methods Plasmids and viruses Quantitative Reverse Transcription PCR Generation of molecular standard for quantitative PCR

Clinical Development of ABX464, drug candidate for HIV Functional Cure. Chief Medical Officer ABIVAX

Molecular Diagnosis Future Directions

Application of μmacs Streptavidin MicroBeads for the analysis of HIV-1 directly from patient plasma

Professor Jonathan Weber

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

Hepatitis B Antiviral Drug Development Multi-Marker Screening Assay

Loss of Effector and Anti-Inflammatory Natural Killer T Lymphocyte Function in Pathogenic Simian Immunodeficiency Virus Infection

BRIEF REPORT METHODS. Twenty-seven HIV-infected individuals receiving ART for a median of 2 years were included in this study (Table 1).

In vitro human regulatory T cell suppression assay

Management of patients with antiretroviral treatment failure: guidelines comparison

Diversity and Tropism of HIV-1 Plasma Rebound Virus after Treatment Discontinuation

In vitro human regulatory T cell expansion

Supplementary Table e-1. Flow cytometry reagents and staining combinations

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

Animal models in HIV cure research.

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

staining and flow cytometry

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)

X/01/$ DOI: /JVI Copyright 2001, American Society for Microbiology. All Rights Reserved.

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

When to start: guidelines comparison

Transcription:

Antiretroviral Therapy in Simian Immunodeficiency Virus-Infected Sooty Mangabeys: Implications for AIDS Pathogenesis Francesca Calascibetta, Emory University Luca Micci, Emory University Diane Carnathan, Emory University Benton Lawson, Emory University Thomas Howerton Vanderford, Emory University Steven Bosinger, Emory University Kirk Easley, Emory University Ann Chahroudi, Emory University Joseph Mackel, Emory University Brandon F. Keele, Leidos Biomedical Research, Inc. Only first 10 authors above; see publication for full author list. Journal Title: Journal of Virology Volume: Volume 90, Number 16 Publisher: American Society of Microbiology 2016-08-01, Pages 7541-7551 Type of Work: Article Final Publisher PDF Publisher DOI: 10.1128/JVI.00598-16 Permanent URL: https://pid.emory.edu/ark:/25593/rwnrj Final published version: http://dx.doi.org/10.1128/jvi.00598-16 Copyright information: 2016, American Society for Microbiology. All Rights Reserved. Accessed May 7, 2018 6:59 PM EDT

Calascibetta et al. portant successes in HIV/AIDS research, significantly reducing the mortality and morbidity of HIV infection. Importantly, ART results in a marked improvement of the complex immunological abnormalities that are associated with HIV infection, including a restoration of peripheral CD4 T cell counts, the attenuation of HIV-associated generalized immune activation, and an improvement of the immune response against many opportunistic pathogens (16 18). While current antiretroviral drugs were designed, identified, and developed based on their ability to effectively inhibit specific enzymatic activities of HIV-1 gene products (i.e., reverse transcriptase [RT], protease, and integrase), most of these compounds are also able to effectively suppress the same enzymatic activities of SIV (19 22). In this regard, a number of studies involving ART administration in experimentally SIV- or simianhuman immunodeficiency virus (SHIV)-infected rhesus macaques (RMs) were recently conducted by several groups, including ours (19, 22 25). In the setting of SIV infection of SMs, we previously conducted a study in which six animals were treated with short-term, two-drug ART (tenofovir and emtricitabine) and the decay of viremia was examined to estimate the average in vivo life span of productively SIV-infected cells, which was found to be similar to that described for HIV-infected humans (26). However, no studies have been performed involving longer, multidrug ART administration in SIV-infected SMs. To directly assess the role of SIV replication in determining the main characteristics of chronic SIV infection of SMs, we treated a cohort of animals with a potent ART regimen consisting of tenofovir, emtricitabine, raltegravir, and darunavir for periods ranging from 2 to 12 months and conducted a number of virological and immunological analyses before, during, and after ART interruption. We observed that ART effectively suppressed viremia in 10 of 12 SIV-infected SMs and that these animals experienced (i) an increase of the percentage of circulating CD4 T CM cells, (ii) an increase in the level of total CD4 T cells in the rectal mucosa, and (iii) significant declines in the levels of activated HLA-DR CD8 T cells in the blood and rectum. In addition, we observed that ART interruption resulted in rapid viral rebound in all treated animals, indicating that, in SIV-infected SMs, the virus reservoir persists for 1 year under ART despite relatively low levels of infection in CD4 T CM and T SCM cells. Based on these data, we concluded that suppressive ART reverts some of the key immunological abnormalities observed during nonpathogenic SIV infection of SMs, thereby helping to define the direct effects of virus replication in SIV-infected natural hosts. MATERIALS AND METHODS Ethics statement. All animals were housed at the Yerkes National Primate Research Center (YNPRC), Atlanta, GA, which is accredited by the Association for Assessment and Accreditation of Laboratory Animal Care. SMs were maintained according to the Animal Welfare Act and NIH guidelines for housing and care of laboratory animals. Experimentation was performed in accordance with the institutional regulations and after approval by the Institutional Animal Care and Use Committee (IACUC) at the YNPRC (IACUC protocol YER-2002673-032017GA, entitled Studies of the Natural SIV Infection of Sooty Mangabeys ). Commercial dry food supplemented with fruit was provided by the veterinary personnel, and water was available ad libitum. Adjustments were made as necessary depending on sex, age, and weight. Room temperature was maintained at 21 C, with a relative air humidity of 50% and a 12-h light-dark cycle. Appropriate procedures were performed to ensure limited distress, pain, discomfort, and/or injury. The sedatives ketamine and Telazol were applied by intramuscular injection at doses of 10 mg/kg of body weight and 4 to 5 mg/kg, respectively, for blood and tissue collections. Animals and ART regimen. Twelve chronically SIV-infected SMs (3 females and 9 males; ranging in age from 14 to 25 years), not homozygous for CCR5-null alleles and with viral loads of 1,000 copies/ml, were included in this study. Starting at day 30, all animals were treated with a four-drug ART regimen comprising two nucleoside reverse transcriptase (RT) inhibitors (tenofovir [PMPA] and emtricitabine [FTC]), one integrase inhibitor (raltegravir [RAL]), and one protease inhibitor (darunavir [DRV]). PMPA and FTC were administered subcutaneously (s.c.) once a day at dosages of 20 mg/kg and 30 mg/kg, respectively. RAL and DRV were administered orally (mixed with food) twice a day at the following dosages: 150 mg twice a day (b.i.d.) and 400 mg b.i.d., respectively. The 12 SMs were divided into four groups of three animals each, and each group received ART for 2, 6, 9, or 12 months. ART was interrupted at day 60, 184, 274, or 365, and all animals were then monitored for an additional period of at least 2 months and up to 6 months. ART was administered to all 12 animals, but only 11 SMs completed treatment, as one animal was sacrificed due to complications unrelated to either SIV infection or ART. Sample collection and processing. Peripheral blood (PB) and colorectal mucosa biopsy (RB) specimens were collected longitudinally at various experimental points prior to, during, and after ART administration. All animals were first sedated by intramuscular injection before any access. Blood collections of up to 40 ml and RB were performed every 6 weeks, while monitoring of viral load and immunophenotypic analysis were performed every 2 or 4 weeks on smaller blood volumes (3 to 5 ml). Collections and processing of PB and RB were performed as previously described (7, 27 29). Briefly, blood samples were used for complete blood counts and routine chemical analysis, and plasma was separated by centrifugation within1hofphlebotomy. Peripheral blood mononuclear cells (PBMCs) were isolated by density gradient centrifugation. For rectal biopsies, fecal material was removed from the rectum, and up to 10 colorectal pinches were collected with biopsy forceps under visual control via the use of an anoscope. RB-derived lymphocytes were isolated by digestion with 0.5 to 2 U/mg collagenase type II plus 10 U/ l DNase I recombinant, RNase-free solution for 2 h at 37 C and then passed through a 70- m cell strainer to remove residual tissue fragments. All samples were processed to get a final concentration of 1 10 6 cells/ml in complete RPMI 1640 medium and then were stained and fixed (1% paraformaldehyde). Flow cytometry analyses were performed within 24 to 48 h of collection. Flow cytometric analysis. Fourteen-parameter flow cytometric analysis was performed on cells from PB and RB specimens by using panels of monoclonal antibodies that are cross-reactive to SMs (4, 5, 7, 27 30). Predetermined optimal concentrations of the following antibodies were used to determine variations in the percentages of CD4 T cells and specific CD4 T cell subsets: anti-cd3 allophycocyanin (APC) Cy7 (clone SP34-2), anti-cd95 phycoerythrin (PE) Cy5 (clone DX2), anti- CD62L PE (clone SK11), anti-ccr5 APC (clone 3A9), anti-cd45ra fluorescein isothiocyanate (FITC) (clone L48), and anti-ccr7 PE Cy7 (clone 3D12), all from BD Biosciences, San Jose, CA; anti-cd28 PE Texas Red (clone CD28.2), from Beckman Coulter, Brea, CA; and anti-cd8 BV711 (clone RPA-T8), anti-cd4 BV650 (clone OKT4), anti-pd1 BV421 (clone EH12.2H7), and anti-cd27 BV605 (clone O323), from BioLegend Inc., San Diego, CA. Activation and proliferation markers on CD4 and CD8 T cell populations were assessed using anti-hla-dr peridinin chlorophyll protein (PerCP) Cy5.5 (clone G46-6) and anti-ki-67 Alexa 700 (clone B56) (both from BD Biosciences, San Jose, CA). The key CD4 T cell subsets were defined as follows: naive (T N ), CD45RA CCR7 CD27 CD28 CD95 ; stem cell memory (T SCM ), CD45RA CCR7 CD27 CD28 CD95 ; central memory (T CM ), CD45RA CD28 CD95 CCR7 CD62L ; transitional memory (T TM ), CD45RA CD28 CD95 CCR7 CD62L ; and effector memory (T EM ), CD45RA CD28 CD95 CCR7 CD62L. Flow cytometric acquisitions were performed on an LSRII flow cytometer 7542 jvi.asm.org Journal of Virology August 2016 Volume 90 Number 16

Antiretroviral Therapy of Nonpathogenic SIV Infection driven by the FACS DiVa software package (BD Biosciences), and data were analyzed with FlowJo (Tree Star Inc., Ashland, OR). Detection of plasma SIV RNA by standard RT-PCR assay. Plasma SIV loads were determined by standard quantitative RT-PCR as previously described (limit of detection, 60 copies/ml) (31). Briefly, 150 lof plasma was used to extract viral SIV smm RNA by using a QIAamp viral RNA minikit (Qiagen, Valencia, CA). Ten microliters of extracted RNA was subjected to RT-PCR by using random hexamers to prime reverse transcription (Invitrogen, Carlsbad, CA). Primer and probe sequences were targeted to the 5=-untranslated region of the SIV smm genome (forward primer, 5=-GGCAGGAAAATCCCTAGCAG-3=; reverse primer, 5=-GCCCTTACTGCCTTCACTCA-3=; and probe, 5=-AGTCCCTGTTC RGGCGCCAA-3=) (4). The SIV RNA copy number was determined by comparison to an external standard curve created with virion-derived SIV smm RNA. For graphical reasons, samples with SIV RNA levels below the detection limit of the assay were assigned a level of half the lower limit of detection. SIV smm sequence analysis for primer development. In order to generate a specific and sensitive quantitative SIV smm real-time PCR assay, we sequenced the gag genes of the viruses present in all test animals prior to therapeutic intervention. The day 30 pre-art plasma samples were utilized to assess overall sequence diversity in the cohort of naturally infected sooty mangabeys. Viral RNA was extracted from plasma by using a QIAamp viral RNA minikit (Qiagen). RNA was eluted and immediately subjected to cdna synthesis. Reverse transcription of RNA to singlestranded cdna was performed using SuperScript III according to the manufacturer s instructions (Invitrogen). Briefly, a 0.5 mm concentration of each deoxynucleoside triphosphate and 0.25 M antisense primer SIV-Gag-R1 (5=-TGACTACTGGTCTCCTCCAAAGAG-3=) were incubated at 65 C for 5 min and then moved to ice for 1 min. Next, 1 RT buffer, 5 mm dithiothreitol, 2 U/ l RNaseOUT, and 10 U/ l SuperScript III reverse transcriptase were incubated at 50 C for 60 min, followed by an increase in temperature to 55 C for an additional 60 min. SuperScript III was then heat inactivated at 70 C for 15 min and treated with2uofrnase H at 37 C for 20 min. cdna was used immediately for single genome amplification (SGA) PCR. Briefly, 1 High Fidelity Platinum PCR buffer, 2 mm MgSO 4, 0.2 mm (each) deoxynucleoside triphosphates, 0.2 M (each) primers, and 0.025 U/ l Platinum Taq high-fidelity polymerase were combined in a 20- l reaction mixture. cdna was serially diluted until a concentration was found where PCR-positive wells constituted fewer than 30% of the total number of reaction wells. First-round PCR primers included the sense primer SIV-U5-F1 (5=-AGAAGTAAGCTAG TGTGTGTTCCCATCTCT-3=) and the antisense primer SIV-Gag-R1. PCR was performed by denaturation at 94 C for 1 min followed by 35 cycles of 94 C for 20 s, 55 C for 30 s, and 68 C for 1 min, and the reaction was terminated with a single 10-min extension at 68 C. Next, 1 l of each reaction mixture was transferred to a second-round reaction mixture that included the sense primer SIV-U5-F2 (5=-AGCTAGTGTGTGTTCCCAT CTCTCCTA-3=) and the antisense primer SIV-Gag-R2 (5=-CCAAAGAG AGAATTGAGGTGCAG-3=). The second-round PCR was performed under the same PCR conditions, but for 45 cycles. All PCR procedures were performed under PCR clean room conditions and using procedural safeguards against sample contamination, including prealiquoting of all reagents, use of dedicated equipment, and physical separation of sample processing from pre- and post-pcr amplification steps. Correctly sized amplicons, as determined by electrophoresis on an agarose gel, were directly sequenced by cycle sequencing using BigDye Terminator chemistry and protocols recommended by the manufacturer (Applied Biosystems). Individual sequence fragments for each amplicon were assembled and edited using Sequencher (Gene Codes). qpcr assay design and validation. Based on the sequences determined as described above, the quantitative PCR (qpcr) assay primers and probe were designed to accommodate the SIV smm variation identified by this sequence analysis through the introduction of the nonstandard bases P and K (Gen Research) and degenerate bases (IDT). The newly designed assay consisted of the forward primer smgagf (5=-GTCTGCG TCATTTGGTGCATPC-3=), the reverse primer smgagr (5=-CACTAGA TGTCTCTGPACTAT/GTTGTTTTG-3=), and the probe smgagp (5=-6- carboxyfluorescein [FAM]-CTTCPTCAGTKTGTTTCACTTTCTCTTC TGCG-BHQTM1-3=). Assay validation was performed on a mixture of in vitro transcripts generated individually from the 6 dominant variant templates, providing coverage of 96% of the 299 sequences obtained. The assay was optimized for a maximal sensitivity of specific target detection of eight copy equivalents per reaction mix (95% assurance). Quantitative analysis of SIV smm RNA samples by SM-specific RT- PCR assay. SIV smm RNA isolation from cell-free sooty mangabey plasma samples was performed as previously described (32). Quantitative assessment of SIV smm RNA was determined by a quantitative hybrid real-time/ digital RT-PCR approach, which was modified from the plasma viral detection assay as described previously (33). The cdna reaction mixtures were reduced to 15 l, comprising 5 l sample plus 10 l concentrated reaction cocktail, and contained 5 ng/ l random hexamers, 5 U RNaseOUT RNase inhibitor (Invitrogen), and 10 U SuperScript III reverse transcriptase (Invitrogen). Twelve replicate reactions were performed per sample. Quantitative determinations for samples showing amplification in all replicates were derived by interpolation of the averaged threshold cycle (C T ) value for the reactions onto a standard curve of C T versus input template copy number. Quantitative determinations for samples showing fewer than 12 positive amplifications in replicates were derived from the frequency of positive amplifications, corresponding to the presence of at least one target copy in a reaction mixture, according to a Poisson distribution of a given median copy number per reaction mixture. The assay yielded no positive reactions for a total of 72 total reaction mixtures for RNA samples derived from 6 uninfected animals. Memory CD4 T cell subset sorting. Enriched unlabeled CD4 T cells were obtained by magnetic bead isolation and passage through a magnetically activated cell sorting (MACS) selection column placed in a MACS separator (Miltenyi, Auburn, CA). Cells in suspension were stained with the proper antibody mixture and then physically separated by fluorescence-activated cell sorting (FACS). Sorting of CD4 T EM,T CM, T TM, and T SCM cells based on the expression of CD45RA, CCR7, CD28, CD95, and CD62L was performed on a FACS Aria II flow cytometer (BD Biosciences) for samples collected before and during ART. Cells were initially gated on the basis of light scatter, followed by positive staining for CD3 and CD4 and negative staining for the LIVE/DEAD fixable dead cell stain (Invitrogen, Carlsbad, CA). Detection of cell-associated SIV DNA. Sorted CD4 T cell subsets were lysed and homogenized in a highly denaturing guanidine isothiocyanate-containing buffer which inactivates DNases and RNase and disrupts cell membranes. Total DNA was then extracted by use of an AllPrep DNA/ RNA minikit (Qiagen) according to the manufacturer s recommendations. Quantitative assessments of cell-associated total SIV DNA within circulating memory CD4 T cell subsets at day 30 pre-art administration and days 45, 184, 274, and 365 on ART were performed by real-time PCR. Briefly, 45 ng of DNA was loaded into a 50- l reaction mixture with an SIVutr primer/probe set. The same SIV-specific primers and probe were used to amplify and quantify total provirus copies of SIV as described before. Albumin was used as an internal control to quantify the cell number against an external standard curve. An albumin gene-specific probe (5=-VIC-TGACAGAGTCACCAAATGCTGCACAGAA-3=) and flanking primers (5=-TGCATGAGAAAACGCCAGTAA-3= and 5=-ATGGTCGCC TGTTCACCAA-3=) (Applied Biosystems) were used to determine the proportion of SIV DNA cells by dividing the obtained SIV DNA copy number by the albumin gene copy number (5). Statistical analysis. Comparisons between circulating CD4 T cell, CD4 T cell subset, and CD8 T cell frequencies and absolute counts pre-art and during ART were carried out using the nonparametric Wilcoxon matched-pair signed-rank test. The same analysis was carried out for mucosal populations as well. Comparisons between the levels of expression of different markers on CD4 and CD8 lymphocyte popula- August 2016 Volume 90 Number 16 Journal of Virology jvi.asm.org 7543

Calascibetta et al. FIG 1 Experimental design and baseline characteristics of the SIV-infected SMs included in the study. (A) Experimental design of the study, indicating the days on which ART was administered (day 0; blue arrow) as well as the experimental time points at which blood (PB) and rectal biopsy (RB) specimens were collected (gray dots). ART was interrupted after 60, 184, 274, and 365 days, as indicated by red arrows. The gray boxed area represents the follow-up after structured ART interruption (2 to 6 months). (B) Baseline features of the 12 SIV-infected SMs included in the study, including gender, age, viral load at the chronic stage of SIV infection, CD4 T cell count, years of infection, and CCR5 genotype. tions and their subsets were determined for points during ART versus previous time points and the baseline. Specifically, the unpaired t test, the unpaired t test with Welch s correction, and the nonparametric Wilcoxon matched-pair signed-rank test were carried out. Variations between preand on-art cell-associated virus (SIV DNA) levels in SM blood were monitored over time, and statistical significance was determined using repeated-measure analyses of each CD4 T cell subset (SAS MIXED Procedure, version 9.4). The means standard errors of the means (SEM) were used as descriptive statistics throughout the course of the study after ART initiation. In all cases, significance was attributed for P values of 0.05. All analyses were conducted using GraphPad Prism 5.0d and SAS MIXED Procedure, version 9.4. RESULTS Experimental design and baseline virological and immunological features of chronically SIV-infected SMs. To assess directly the effects of virus replication on the immunological parameters of interest during chronic SIV infection of SMs, we used a potent four-drug ART regimen that was shown to be very effective at suppressing SIV replication in rhesus macaques (19 22). Twelve SIV-infected SMs were included in this study and were divided into four groups, each with three animals receiving ART for 2, 6, 9, or 12 months. On day 0 of the study (Fig. 1A), all animals were treated with PMPA (20 mg/kg/day), FTC (30 mg/kg/day), DRV (400 mg b.i.d.), and RAL (150 mg b.i.d.). Blood and rectal biopsy (RB) specimens were collected longitudinally throughout the course of the study, as shown in Fig. 1A, to monitor a number of virological and immunological parameters prior to and during ART and after ART interruption. After ART interruption, all SIV-infected SMs were monitored for at least 2 months and up to 6 months (Fig. 1A). The 12 SIV-infected SMs included in this study were selected from the colony of animals housed at the YNPRC, and their basic immunological and virological features are shown in Fig. 1B, with viral loads ranging from 1,940 to 261,000 copies/ml of plasma and CD4 T cell counts ranging from 167 to 1,336 cells/mm 3 of blood. Note that, for this study, we selected animals that were not homozygous for the previously described CCR5-null genes (34). ART is well tolerated and induces potent suppression of virus replication in chronically SIV-infected SMs. To investigate the effect of ART on the main virological and immunological pa- 7544 jvi.asm.org Journal of Virology August 2016 Volume 90 Number 16

Antiretroviral Therapy of Nonpathogenic SIV Infection FIG 2 Longitudinal assessment of plasma viremia before, during, and after ART administration. (A) Longitudinal viral loads, expressed as numbers of SIV smm RNA copies per milliliter of plasma, for all SIV-infected SMs during ART. Gray shading depicts the period of ART. Significant viral load decreases were observed on days 15, 30, and 45 after ART initiation. (B) Viral load variations after ART interruption. ART (shaded areas) was interrupted on days 60 (upper left; n 2), 184 (upper right; n 3), 274 (bottom left; n 3), and 365 (bottom right; n 3) post-art initiation, and plasma viremia was determined for at least 2 months and up to 6 months after ART cessation. rameters of SIV infection, the 12 SIV-infected SMs received ART for different periods (60, 184, 274, or 365 days). Overall, ART was safe and well tolerated by the majority of SIV-infected SMs that were included in this study. Two animals (FUv and FJy) exhibited a moderate weight loss that was reversed with diet supplementation, with a rapid return to baseline weight after ART interruption. The 21-year-old animal FWo, which was originally included in the 2-month treatment group, was euthanized on day 43 of therapy due to major cardiovascular symptoms that were attributed to aging by the YNPRC veterinary staff. As expected, all animals receiving ART experienced a rapid and significant decline in plasma viral load (Fig. 2A, gray boxed areas of the graphs). As shown in Fig. 2A, 6 of 11 SIV-infected SMs showed viremia suppression to below the level of detection of our standard SIV smm viral load assay (i.e., 60 copies/ml) by day 30 after ART initiation, and the viral loads of four other animals became undetectable by day 45 of treatment. Only one animal (FSs) exhibited relative resistance to ART, with a viral load that declined but remained detectable after 184 days of ART. To further investigate the level of ART-induced virus suppression in SIV-infected SMs that had un- August 2016 Volume 90 Number 16 Journal of Virology jvi.asm.org 7545

Calascibetta et al. FIG 3 Variation in SIV DNA contents of CD4 T cell subsets in response to ART. Analysis of SIV DNA contents in purified CD4 TEM (A), TTM (B), TCM (C), and TSCM (D) cells was performed prior to ART initiation (day 30) and before interruption of ART at days 45, 184, 274, and 365. Results are expressed as numbers of SIV DNA copies per million cells. The total SIV copy number was normalized to the albumin housekeeping gene expression. detectable viral loads with our standard assay, we next measured viremia by using an ultrasensitive SIVsmm-specific RT-PCR assay with a limit of detection of 10 copies/ml. The results of this ultrasensitive assay indicated that low levels of virus production were still present in all ART-treated SIV-infected SMs, though it remains unclear whether this production was related to de novo cycles of virus replication versus persistent virus production in cells that were infected prior to ART (data not shown). ART induces modest changes in the level of SIV DNA in memory CD4ⴙ T cell subsets. In previous studies, we showed that nonpathogenic SIV infection of SMs is associated with lower levels of infection (as measured by total cell-associated SIV DNA) in CD4 TCM and TSCM cells than those seen with pathogenic SIV infection of RMs (4 6). To investigate the effects of ART on the levels of SIV infection in different memory CD4 T cell subsets, we identified and sorted TSCM, TCM, TTM, and TEM cells based on the surface expression of CD45RA, CD62L, CCR7, CD28, and CD95 and quantified the levels of total SIV DNA by RT-PCR for samples collected at various time points during the study (i.e., day 30 pre-art and days 60, 184, 274, and 365 on ART). As shown in Fig. 3A, a significant decrease in SIV DNA content was detected in the TEM CD4 T cell subset (P 0.02) as a result of ART-mediated suppression of virus replication in this subset of short-lived cells. 7546 jvi.asm.org However, we found no statistically significant trends in infection of the other examined CD4 T cell subsets, i.e., TTM, TCM, and TSCM cells (average changes of 0.9-, 1.1-, and 3.7-fold for CD4 TTM, TCM, and TSCM cells, respectively). Note that one SIV-infected SM (FEz) exhibited undetectable levels of SIV gag copies in TSCM and TCM fractions both prior to ART treatment and during treatment. However, the apparent absence of SIV infection in these two longlived memory CD4 T cell compartments was not sufficient to avoid rapid viral rebound once ART was stopped (Fig. 2B). Overall, these data indicate that administration of suppressive ART for 2 to 12 months induced a significant decline in the level of cellassociated SIV DNA in circulating CD4 TEM cells but had little impact on any of the other memory CD4 T cell subsets. ART induces a significant repopulation of peripheral CD4ⴙ TCM cells and total intestinal CD4ⴙ T cells in SIV-infected SMs. In HIV-infected humans, initiation of ART induces a series of immunological changes, including a rapid increase in peripheral CD4 T cell counts as well as a slower and incomplete reconstitution of the mucosal CD4 T cell pool (13, 35 39). To evaluate the impact of ART-induced suppression of virus replication on CD4 T cell reconstitution, we next measured, for our cohort of ARTtreated SIV-infected SMs, the levels of CD4 T cells in peripheral blood (PB) and rectal biopsy (RB) specimens taken at several time Journal of Virology August 2016 Volume 90 Number 16

Antiretroviral Therapy of Nonpathogenic SIV Infection FIG 4 Longitudinal assessment of CD4 T cell levels in peripheral blood and rectal mucosa. (A) Levels of CD4 T cells in blood (expressed as numbers of cells per microliter) and mucosal tissue (RB; expressed as percentages of total CD3 T cells) were calculated for each of the SMs before and during ART (gray shaded areas) and after ART cessation. (B) Levels (means SEM) of naive (yellow), T CM (light blue), T EM (purple), T TM (red), and T SCM (green) CD4 T cell subsets in peripheral blood (PB) of SIV-infected SMs at the different experimental points, divided by group. The x axis shows the time post-art initiation (day 0). In the bottom graphs, mean values, standard deviations, and statistically significant differences are shown (*, P 0.05; **, P 0.01; and ***, P 0.001). Statistical significance was determined by the Wilcoxon matched-pair signed-rank test. (C) Representative flow cytometric plots showing the fraction of CD4 T cells (gated on live CD3 T cells) in RB specimens before, during, and after ART administration for one representative SM. Comparisons of CD4 T cell recovery were performed for different time points during ART (days 92 and 135) and after ART (day 60 post-art). Statistically significant differences are shown (*, P 0.05; **, P 0.01; and ***, P 0.001). Statistical significance was determined by the Wilcoxon matched-pair signed-rank test. points during the study. As shown in Fig. 4A, we found that the number of circulating CD4 T cells remained stable during ART, with most animals experiencing only minor fluctuations. We next investigated how ART influenced the dynamics of the subsets of T N,T SCM,T CM,T TM, and T EM CD4 T cells in peripheral blood. As shown in Fig. 4B, we found that the percentages of CD4 T N, T SCM, and T TM cells remained relatively constant after ART in our cohort of SIV-infected SMs, with only minor fluctuations observed in some animals. In contrast, ART-treated SIV-infected SMs showed a significant expansion of CD4 T CM cells on days 45, 92, and 135 post-art compared to baseline pre-art levels (for day 30 versus day 45, P 0.0039; for day 30 versus day 92, P 0.0039; and for day 30 versus day 135, P 0.0177) that was associated with a concomitant decline of CD4 T EM cells on days 75 and 120 post-art (for day 30 versus day 75, P 0.0039; and for day 30 versus day 120, P 0.0039). Note that the ARTinduced increase in peripheral CD4 T CM cells persisted on day 60 after ART interruption (Fig. 4B, day 30 versus day 60) (P 0.0064). As shown in Fig. 4C, the analysis of CD4 T cells in RB specimens (measured as the fraction of CD3 T cells) from ARTtreated SIV-infected SMs showed a significant increase compared to pre-art levels (for day 30 versus day 92, P 0.0391; and for day 30 versus day 135, P 0.0117) (Fig. 4C). Interestingly, the observed increase of CD4 T cells in RB specimens was not reversed once ART was interrupted, with the percentage of CD4 T cells in the rectal mucosa at day 60 post-art interruption remain- August 2016 Volume 90 Number 16 Journal of Virology jvi.asm.org 7547

Calascibetta et al. FIG 5 Longitudinal assessment of HLA-DR-expressing CD4 and CD8 T cells in blood and RB specimens. (A) Mean values and standard deviations for percentages of HLA-DR CD4 T cells in PB (red circles) and RB (blue circles) specimens at the different experimental time points are shown for the SM cohorts receiving ART for 2 (n 2),6(n 3),9(n 3), and 12 (n 3) months. Statistically significant differences are shown (*, P 0.05; **, P 0.01; and ***, P 0.001). (B) Longitudinal assessment of CD8 T cells expressing HLA-DR in PB (upper panels) and RB (lower panels) specimens from all SMs. ing higher than that at baseline (i.e., pre-art) (Fig. 4C). Overall, these data indicate that ART induced a significant immune reconstitution in SIV-infected SMs, with increases in both peripheral CD4 T CM and mucosal CD4 T cells. ART reduces the levels of activated CD8 T cells in the peripheral blood and rectal mucosa of SIV-infected SMs. In HIVinfected humans, initiation of ART induces a reduction in the expression of activation and proliferation markers on CD4 and CD8 T cells in peripheral blood as well as on lymphoid and mucosal tissues (13, 35 39). To characterize the impact of ART on the levels of immune activation of SIV-infected SMs, we next longitudinally assessed the expression of HLA-DR, PD-1, and Ki-67, i.e., three key markers of T cell activation (HLA-DR and PD-1) and proliferation (Ki-67), on CD4 and CD8 T cells from peripheral blood and rectal biopsy specimens. As expected based on previous studies, SIV-infected SMs showed relatively low levels of CD4 and CD8 T cell activation before ART. As shown in Fig. 5A, ART did not induce any change in the levels of CD4 HLA- DR T cells in either the peripheral blood or the rectal mucosa. In contrast, we found that the percentage of CD8 HLA-DR T cells declined significantly at day 135 of ART in peripheral blood (for day 30 versus day 135, P 0.0039) and at day 92 of ART in RB specimens (for day 30 versus day 92, P 0.0078) compared to the baseline (pre-art) levels. Interestingly, ART interruption was followed by an increase of the percentage of CD8 HLA-DR T cells in both blood and RB specimens (for PB at day 30 versus day 30, P 0.0163; for PB at day 135 versus day 30, P 0.0039; for PB at day 30 versus day 45, P n.s. [not significant]; for PB at day 135 versus day 45, P 0.0078; for RB specimens at day 30 versus day 106, P n.s.; and for RB specimens at day 92 versus day 106, P 0.0078) (Fig. 5B). No significant changes were found in the level of either PD-1 or Ki-67 T cells (either CD4 or CD8 ) in peripheral blood or rectal mucosa of ART-treated SIV-infected SMs (data not shown). Overall, these data suggest that ART is followed by a decline of CD8 (but not CD4 ) T cell activation that returns to pre-art levels upon ART interruption. ART interruption is followed by rapid virus rebound in SIVinfected SMs. Interruption of ART is followed by a rapid (i.e., within weeks) rebound of viremia in HIV-infected individuals, with numerous studies suggesting that virus persistence under ART is related to the latent infection of long-lived, resting memory CD4 T cells, mainly belonging to the T CM and T SCM subsets (4, 5, 40 43). In our cohort of ART-treated SIV-infected SMs, we interrupted ART at various time points after initiation (see above for details) and measured the level of plasma viremia longitudinally. As shown in Fig. 2B, we found that regardless of the duration 7548 jvi.asm.org Journal of Virology August 2016 Volume 90 Number 16

Antiretroviral Therapy of Nonpathogenic SIV Infection of ART (ranging from 2 to 12 months), all SIV-infected SMs experienced a rapid rebound of virus replication. In most animals, the kinetics of virus rebound after ART interruption showed a peak of viremia at approximately day 30 postinterruption that then stabilized at slightly lower levels (decline of 0.5 to 1 log) in the following weeks (i.e., similar to the pre-art set-point viremia). These results indicate that a reservoir of SIV-infected cells that harbor replication-competent virus clearly remain present in SMs under ART for up to 1 year, despite lower levels of infection in CD4 T CM and T SCM cells. DISCUSSION Natural SIV infection of SMs is typically nonpathogenic despite high levels of virus replication and the short in vivo life span of productively infected cells (26). A series of studies indicated that the main mechanisms of protection from SIV disease progression in these animals are the lack of aberrant immune activation during the chronic phase of the infection, preservation of Th17 cells and mucosal integrity, and the preferential sparing of CD4 T CM and T SCM cells from direct virus infection (4, 5, 41, 42). None of these published studies involved prolonged administration of potent, multidrug antiretroviral regimens, with only one study in which PMPA and FTC were used for a relatively short period as an experimental tool to assess the in vivo life span of productively infected cells (26). The main findings of the current study are that administration of ART in SIV-infected SMs was well tolerated overall and resulted in a rapid decline of viremia to levels below the limit of detection of our standard assay in all but one treated animal. The level of viremia decline observed in the current study is comparable overall to what we observed in similar studies conducted on SIV-infected RMs (19, 44). In addition, we found that ART initiation was followed by an increase in the percentage of CD4 T CM cells in peripheral blood, an increase of CD4 T cells in the rectal mucosa, and a decline in the levels of activated CD8 HLA-DR T cells in both blood and the rectum. Finally, we found that ART interruption resulted in rapid viral rebound in all treated animals. The lower levels of infection in the long-lived CD4 T SCM and T CM cells of SIV-infected SMs suggested that the reservoir is possibly less stable under ART. While the current study does not directly support this hypothesis, there are several arguments against the conclusion that limited CD4 T SCM and T CM cell infection does not affect virus persistence in this model. These arguments include the following: (i) residual viremia was detected in all treated animals (using an ultrasensitive viral load assay that cannot distinguish, however, between rounds of de novo virus replication and residual virus production from cells infected prior to ART), (ii) the reservoir was measured only in peripheral blood, (iii) SIV DNA levels were low but still detectable in CD4 T CM and T SCM cells in the majority of SIV-infected SMs, and (iv) ART was used for a period of 2 to 12 months only. An additional caveat is that this study included several SMs that were infected intravenously with SIV, while most of the naturally SIV-infected animals contracted the infection through sexual intercourse. All these caveats notwithstanding, we found it interesting that virus rebound was also observed in one SIV-infected SM with undetectable SIV DNA in both CD4 T CM and T SCM cells (but still with detectable plasma viremia as measured by the ultrasensitive viral load assay). Future studies involving long-term ART (i.e., 2 years) and in which ART interruption is performed only when the treated SIVinfected SMs show undetectable viremia by the most sensitive assay (i.e., with quasispecies-specific probes and a limit of detection of 3 copies/ml of plasma) will be necessary to ascertain if, under those conditions, the virus reservoir may indeed prove to be less stable as a consequence of the lower infection levels of CD4 T CM and T SCM cells. An interesting and perhaps unexpected finding of this study is that ART had a positive impact on CD4 T cell reconstitution in SIV-infected SMs, as revealed by significant increases of both total CD4 T cells in the rectal mucosa and CD4 T CM cells in peripheral blood. While SIV infection of SMs is typically nonpathogenic, with infected animals experiencing a life span similar to that of uninfected animals (10), the infection is not devoid of some immunological abnormalities. In particular, we have shown that SIV-infected SMs experience a significant decline of CD4 T cells in mucosal tissues (9), even though this decline is not progressive after the acute phase of infection, does not reach the nadirs often observed in pathogenic HIV and SIV infections, and is not associated with a loss of mucosal integrity or detectable microbial translocation (8, 9, 45). In this study, we observed, for the first time, that suppression of virus replication with ART induces a significant increase in the level of CD4 T cells in the rectum and that this increase compared to pre-art values persists even after ART interruption. This observation contrasts with the relatively slow and partial effect of ART in normalizing the levels of mucosal CD4 T cells in humans. It is tempting to speculate that in SIVinfected SMs, the low levels of immune activation as well as the preservation of Th17 cells and mucosal immunity may favor the recovery of mucosal CD4 T cells when virus replication is controlled by ART. The observed impact of ART on the levels of circulating CD4 T CM cells as well as the levels of CD8 T cells expressing the activation marker HLA-DR may also indicate that subtle immunological changes induced by persistent virus replication are present in SIV-infected SMs. The affected processes include CD4 T cell homeostasis, which is largely dependent on the presence of healthy levels of T CM cells in blood and tissues (46), and CD8 T cell activation. The reasons for these changes not translating into a major immunological dysfunction with associated clinical signs of opportunistic infections remain unknown. However, in this regard, the current study opens up new avenues of research aimed at investigating what additional mechanisms may operate in SIVinfected SMs to limit the clinical impact of the immune abnormalities that are revealed and reversed by ART administration. ACKNOWLEDGMENTS We gratefully acknowledge Gilead (Romas Geleziunas), Johnson & Johnson (Guenter Kraus), and Merck (Daria Hazuda) for providing antiretroviral drugs. We also thank Sherrie Jean, Stephanie Ehnert, Christopher Souder, and all the animal care and veterinary staff at the Yerkes National Primate Research Center; Barbara Cervasi and Kiran Gill at the Emory University Flow Cytometry Core; Melon T. Nega at the Emory CFAR Virology & Drug Discovery Core; Ronald G. Collman at the Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA; and Laura Newman (for technical assistance with sequencing). The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health, nor does mention of trade names, commercial products, or organizations imply endorsement by the U.S. Government. August 2016 Volume 90 Number 16 Journal of Virology jvi.asm.org 7549

Calascibetta et al. FUNDING INFORMATION This work, including the efforts of Jeffrey Lifson, was funded by HHS NIH National Cancer Institute (NCI) (HHSN261200800001E). This work, including the efforts of Guido Silvestri, was funded by HHS National Institutes of Health (NIH) (R37 AI66998). This work was supported by an R33 NIH grant (AI104278) with principal investigators Guido Silvestri and Mirko Paiardini. Research reported in this publication was also supported by the National Institute of Allergy and Infectious Diseases of the National Institutes of Health under award number P51 OD011132 (to the YNPRC) and also in part by Merck. REFERENCES 1. Chahroudi A, Bosinger SE, Vanderford TH, Paiardini M, Silvestri G. 2012. Natural SIV hosts: showing AIDS the door. Science 335:1188 1193. http://dx.doi.org/10.1126/science.1217550. 2. Silvestri G, Sodora DL, Koup RA, Paiardini M, O Neil SP, McClure HM, Staprans SI, Feinberg MB. 2003. Nonpathogenic SIV infection of sooty mangabeys is characterized by limited bystander immunopathology despite chronic high-level viremia. Immunity 18:441 452. http://dx.doi.org/10.1016/s1074-7613(03)00060-8. 3. Sumpter B, Dunham R, Gordon S, Engram J, Hennessy M, Kinter A, Paiardini M, Cervasi B, Klatt N, McClure H, Milush JM, Staprans S, Sodora DL, Silvestri G. 2007. Correlates of preserved CD4( ) T cell homeostasis during natural, nonpathogenic simian immunodeficiency virus infection of sooty mangabeys: implications for AIDS pathogenesis. J Immunol 178:1680 1691. http://dx.doi.org/10.4049/jimmunol.178.3.1680. 4. Paiardini M, Cervasi B, Reyes-Aviles E, Micci L, Ortiz AM, Chahroudi A, Vinton C, Gordon SN, Bosinger SE, Francella N, Hallberg PL, Cramer E, Schlub T, Chan ML, Riddick NE, Collman RG, Apetrei C, Pandrea I, Else J, Munch J, Kirchhoff F, Davenport MP, Brenchley JM, Silvestri G. 2011. Low levels of SIV infection in sooty mangabey central memory CD(4)( ) T cells are associated with limited CCR5 expression. Nat Med 17:830 836. http://dx.doi.org/10.1038/nm.2395. 5. Cartwright EK, McGary CS, Cervasi B, Micci L, Lawson B, Elliott ST, Collman RG, Bosinger SE, Paiardini M, Vanderford TH, Chahroudi A, Silvestri G. 2014. Divergent CD4 T memory stem cell dynamics in pathogenic and nonpathogenic simian immunodeficiency virus infections. J Immunol 192:4666 4673. http://dx.doi.org/10.4049/jimmunol.1303193. 6. Brenchley JM, Vinton C, Tabb B, Hao XP, Connick E, Paiardini M, Lifson JD, Silvestri G, Estes JD. 2012. Differential infection patterns of CD4 T cells and lymphoid tissue viral burden distinguish progressive and nonprogressive lentiviral infections. Blood 120:4172 4181. http://dx.doi.org/10.1182/blood-2012-06-437608. 7. Brenchley JM, Paiardini M, Knox KS, Asher AI, Cervasi B, Asher TE, Scheinberg P, Price DA, Hage CA, Kholi LM, Khoruts A, Frank I, Else J, Schacker T, Silvestri G, Douek DC. 2008. Differential Th17 CD4 T-cell depletion in pathogenic and nonpathogenic lentiviral infections. Blood 112:2826 2835. http://dx.doi.org/10.1182/blood-2008-05-159301. 8. Brenchley JM, Price DA, Schacker TW, Asher TE, Silvestri G, Rao S, Kazzaz Z, Bornstein E, Lambotte O, Altmann D, Blazar BR, Rodriguez B, Teixeira-Johnson L, Landay A, Martin JN, Hecht FM, Picker LJ, Lederman MM, Deeks SG, Douek DC. 2006. Microbial translocation is a cause of systemic immune activation in chronic HIV infection. Nat Med 12:1365 1371. 9. Gordon SN, Klatt NR, Bosinger SE, Brenchley JM, Milush JM, Engram JC, Dunham RM, Paiardini M, Klucking S, Danesh A, Strobert EA, Apetrei C, Pandrea IV, Kelvin D, Douek DC, Staprans SI, Sodora DL, Silvestri G. 2007. Severe depletion of mucosal CD4 T cells in AIDS-free simian immunodeficiency virus-infected sooty mangabeys. J Immunol 179:3026 3034. http://dx.doi.org/10.4049/jimmunol.179.5.3026. 10. Keele BF, Jones JH, Terio KA, Estes JD, Rudicell RS, Wilson ML, Li Y, Learn GH, Beasley TM, Schumacher-Stankey J, Wroblewski E, Mosser A, Raphael J, Kamenya S, Lonsdorf EV, Travis DA, Mlengeya T, Kinsel MJ, Else JG, Silvestri G, Goodall J, Sharp PM, Shaw GM, Pusey AE, Hahn BH. 2009. Increased mortality and AIDS-like immunopathology in wild chimpanzees infected with SIVcpz. Nature 460:515 519. http://dx.doi.org/10.1038/nature08200. 11. Bosinger SE, Li Q, Gordon SN, Klatt NR, Duan L, Xu L, Francella N, Sidahmed A, Smith AJ, Cramer EM, Zeng M, Masopust D, Carlis JV, Ran L, Vanderford TH, Paiardini M, Isett RB, Baldwin DA, Else JG, Staprans SI, Silvestri G, Haase AT, Kelvin DJ. 2009. Global genomic analysis reveals rapid control of a robust innate response in SIV-infected sooty mangabeys. J Clin Invest 119:3556 3572. http://dx.doi.org/10.1172 /JCI40115. 12. Harris LD, Tabb B, Sodora DL, Paiardini M, Klatt NR, Douek DC, Silvestri G, Muller-Trutwin M, Vasile-Pandrea I, Apetrei C, Hirsch V, Lifson J, Brenchley JM, Estes JD. 2010. Downregulation of robust acute type I interferon responses distinguishes nonpathogenic simian immunodeficiency virus (SIV) infection of natural hosts from pathogenic SIV infection of rhesus macaques. J Virol 84:7886 7891. http://dx.doi.org/10.1128/jvi.02612-09. 13. Meythaler M, Wang Z, Martinot A, Pryputniewicz S, Kasheta M, McClure HM, O Neil SP, Kaur A. 2011. Early induction of polyfunctional simian immunodeficiency virus (SIV)-specific T lymphocytes and rapid disappearance of SIV from lymph nodes of sooty mangabeys during primary infection. J Immunol 186:5151 5161. http://dx.doi.org/10.4049 /jimmunol.1004110. 14. Taaffe J, Chahroudi A, Engram J, Sumpter B, Meeker T, Ratcliffe S, Paiardini M, Else J, Silvestri G. 2010. A five-year longitudinal analysis of sooty mangabeys naturally infected with simian immunodeficiency virus reveals a slow but progressive decline in CD4 T-cell count whose magnitude is not predicted by viral load or immune activation. J Virol 84: 5476 5484. http://dx.doi.org/10.1128/jvi.00039-10. 15. Milush JM, Reeves JD, Gordon SN, Zhou D, Muthukumar A, Kosub DA, Chacko E, Giavedoni LD, Ibegbu CC, Cole KS, Miamidian JL, Paiardini M, Barry AP, Staprans SI, Silvestri G, Sodora DL. 2007. Virally induced CD4 T cell depletion is not sufficient to induce AIDS in a natural host. J Immunol 179:3047 3056. http://dx.doi.org/10.4049 /jimmunol.179.5.3047. 16. Palella FJ, Jr, Deloria-Knoll M, Chmiel JS, Moorman AC, Wood KC, Greenberg AE, Holmberg SD, HIV Outpatient Study Investigators. 2003. Survival benefit of initiating antiretroviral therapy in HIV-infected persons in different CD4 cell strata. Ann Intern Med 138:620 626. http: //dx.doi.org/10.7326/0003-4819-138-8-200304150-00007. 17. Muller M, Wandel S, Colebunders R, Attia S, Furrer H, Egger M, IeDEA Southern and Central Africa. 2010. Immune reconstitution inflammatory syndrome in patients starting antiretroviral therapy for HIV infection: a systematic review and meta-analysis. Lancet Infect Dis 10:251 261. http://dx.doi.org/10.1016/s1473-3099(10)70026-8. 18. Greub G, Ledergerber B, Battegay M, Grob P, Perrin L, Furrer H, Burgisser P, Erb P, Boggian K, Piffaretti JC, Hirschel B, Janin P, Francioli P, Flepp M, Telenti A. 2000. Clinical progression, survival, and immune recovery during antiretroviral therapy in patients with HIV-1 and hepatitis C virus coinfection: the Swiss HIV Cohort Study. Lancet 356:1800 1805. http://dx.doi.org/10.1016/s0140-6736(00)03232-3. 19. Micci L, Ryan ES, Fromentin R, Bosinger SE, Harper JL, He T, Paganini S, Easley KA, Chahroudi A, Benne C, Gumber S, McGary CS, Rogers KA, Deleage C, Lucero C, Byrareddy SN, Apetrei C, Estes JD, Lifson JD, Piatak M, Jr, Chomont N, Villinger F, Silvestri G, Brenchley JM, Paiardini M. 2015. Interleukin-21 combined with ART reduces inflammation and viral reservoir in SIV-infected macaques. J Clin Invest 125: 4497 4513. http://dx.doi.org/10.1172/jci81400. 20. Whitney JB, Hill AL, Sanisetty S, Penaloza-MacMaster P, Liu J, Shetty M, Parenteau L, Cabral C, Shields J, Blackmore S, Smith JY, Brinkman AL, Peter LE, Mathew SI, Smith KM, Borducchi EN, Rosenbloom DI, Lewis MG, Hattersley J, Li B, Hesselgesser J, Geleziunas R, Robb ML, Kim JH, Michael NL, Barouch DH. 2014. Rapid seeding of the viral reservoir prior to SIV viraemia in rhesus monkeys. Nature 512:74 77. http://dx.doi.org/10.1038/nature13594. 21. Del Prete GQ, Shoemaker R, Oswald K, Lara A, Trubey CM, Fast R, Schneider DK, Kiser R, Coalter V, Wiles A, Wiles R, Freemire B, Keele BF, Estes JD, Quinones OA, Smedley J, Macallister R, Sanchez RI, Wai JS, Tan CM, Alvord WG, Hazuda DJ, Piatak M, Jr, Lifson JD. 2014. Effect of suberoylanilide hydroxamic acid (SAHA) administration on the residual virus pool in a model of combination antiretroviral therapymediated suppression in SIVmac239-infected Indian rhesus macaques. Antimicrob Agents Chemother 58:6790 6806. http://dx.doi.org/10.1128 /AAC.03746-14. 22. Shytaj IL, Norelli S, Chirullo B, Della Corte A, Collins M, Yalley- Ogunro J, Greenhouse J, Iraci N, Acosta EP, Barreca ML, Lewis MG, Savarino A. 2012. A highly intensified ART regimen induces long-term viral suppression and restriction of the viral reservoir in a simian AIDS 7550 jvi.asm.org Journal of Virology August 2016 Volume 90 Number 16