17
www.sciencemag.org/content/344/6190/1401/suppl/DC1 Supplementary Materials for HIV-1–induced AIDS in monkeys Theodora Hatziioannou,* Gregory Q. Del Prete, Brandon F. Keele, Jacob D. Estes, Matthew W. McNatt, Julia Bitzegeio, Alice Raymond, Anthony Rodriguez, Fabian Schmidt, C. Mac Trubey, Jeremy Smedley, Michael Piatak Jr., Vineet N. KewalRamani,* Jeffrey D. Lifson,* Paul D. Bieniasz* *Corresponding author. E-mail: [email protected] (T.H.); [email protected] (V.N.K.); [email protected] (J.D.L.); [email protected] (P.D.B.) Published 20 June 2014, Science 344, 1401 (2014) DOI: 10.1126/science.1250761 This PDF file includes: Materials and Methods Figs. S1 to S8 Full Reference List

Supplementary Materials for - Science · 2014. 6. 18. · weeks 12 and 13 or 11 and 12, for P3-A and P3-B, respectively, and at 50 mg/kg of body weight at week 27 or 26, for P3-A

  • Upload
    others

  • View
    2

  • Download
    0

Embed Size (px)

Citation preview

  • www.sciencemag.org/content/344/6190/1401/suppl/DC1

    Supplementary Materials for

    HIV-1–induced AIDS in monkeys

    Theodora Hatziioannou,* Gregory Q. Del Prete, Brandon F. Keele, Jacob D. Estes,

    Matthew W. McNatt, Julia Bitzegeio, Alice Raymond, Anthony Rodriguez, Fabian

    Schmidt, C. Mac Trubey, Jeremy Smedley, Michael Piatak Jr., Vineet N. KewalRamani,*

    Jeffrey D. Lifson,* Paul D. Bieniasz*

    *Corresponding author. E-mail: [email protected] (T.H.); [email protected] (V.N.K.);

    [email protected] (J.D.L.); [email protected] (P.D.B.)

    Published 20 June 2014, Science 344, 1401 (2014)

    DOI: 10.1126/science.1250761

    This PDF file includes:

    Materials and Methods

    Figs. S1 to S8

    Full Reference List

  • Materials and Methods

    HIV-1 strains.

    A proviral plasmid clone of HIV-1 encoding SIVmac Vif and the macaque adapted

    envelope from SHIV/KB9 has previously been described(11). Derivatives of this proviral

    plasmid were constructed by replacing Env-encoding sequences with the corresponding

    sequences from previously described NL4-3-derived proviral plasmids(26). In all these

    chimeric Env proteins, the signal peptide is derived from NL4-3 Env whereas the

    remaining gp120 encoding sequences were derived from YU2, BaL, AD8 or

    KB9(V3ADA) strains. At the C-terminal end of the introduced R5-tropic Env sequences,

    the junction between YU2, BaL or KB9(V3ADA) and NL4-3 Env sequences is at amino

    acid 753 (within the Env cytoplasmic tail) while the junction between AD8 and NL4-3 is

    at amino acid 590 (in the gp41 ectodomain). The KB9(V3ADA) Env sequence was

    constructed using overlapping PCR primers to replace 35 amino acids of the V3 loop in

    the KB9 envelope by the corresponding sequence from ADA, which is identical to the

    HIV-1 subtype B consensus. We confirmed that all of the aforementioned constructs

    generated viruses that replicated in vitro in MT2 cell lines engineered to express CCR5.

    To generate HIV-1KB9 Vpu 15/21, Vpu-coding sequences were amplified from a selected

    amplicon obtained from the plasma of P3-A and used to replace the corresponding Vpu

    sequences in HIVKB9 using overlapping PCR and the EcoRI-KpnI sites in HIV-1NL4-3. A

    Vpu-defective HIV-1NL4-3 construct has been previously described(21).

    HIV-1 adaptation to pigtailed macaques

    Pigtailed macaques (Macacca nemestrina) were housed and cared for in accordance with

    American Association for Accreditation of Laboratory Animal Care (AAALAC)

    standards in an AAALAC-accredited facility and all animal procedures were performed

    according to a protocol approved by the Institutional Animal Care and Use Committee of

    the National Cancer Institute. At the start of the study, all animals were free of

    cercopithicine herpesvirus 1, simian immunodeficiency virus (SIV), simian type-D

    retrovirus, and simian T-lymphotropic virus type 1. Plasma for viral RNA (vRNA)

    quantification, sequencing analysis and Western blots, and peripheral blood mononuclear

    cells (PBMCs) for flow cytometry assays were isolated from whole blood collected in

    EDTA-anticoagulated Vacutainer tubes (BD) at the time points indicated. Plasma was

    separated from the blood by centrifugation and was frozen at -80°C in aliquots before

    analysis for the presence of vRNA or antibodies. PBMCs were isolated from whole

    EDTA blood by Ficoll-Paque Plus (GE Healthcare) gradient centrifugation.

    To initiate adaptation of HIV-1 to macaques, P1-A and P1-B were intravenously (IV)

    inoculated (saphenous vein) with a cocktail of cell culture supernatants from transfected

    293T cells that contained 5x105 i.u. of each HIV-1 variant. During subsequent serial

    passages, some of the animals, as noted below, received subcutaneous administration

    (s.c.) of either a chimeric ‘humanized’ anti-CD8 monoclonal antibody (MAb), cM-T807,

    or a chimeric ‘rhesusized’ anti-CD8 MAb, M-T807R1 (both obtained through the NCRR

    non-human primate reagent program, from Keith Reimann, Harvard/Beth Israel

    Deaconess Medical Center), at the dosages indicated. For serial passage of blood from

    infected to naïve macaques, whole blood from donor animals was drawn into acid citrate

    dextrose (ACD) Vacutainer tubes immediately prior to i.v. infusion into recipients. At

  • weeks 32 and 33 after infection, P1-A received cM-T807 at 25 mg/kg of body weight.

    Thereafter, 10ml of blood withdrawn from P1-A at week 34 after infection was used to

    inoculate P2-A. P2-A received s.c. injection of cM-T807 at 25 mg/kg of body weight at

    the time of infection and at one week postinfection (pi). Blood (10ml) obtained from P2-

    A at week 23 pi, immediately prior to administration of M-T807R1 at 50 mg/kg, was

    used to inoculate animal P3-A and blood (10ml) obtained from P2-A at week 24 pi, one

    week after M-T807R1 administration, was used to inoculate P3-B. Both P3-A and P3-B

    received M-T807R1 at 25 mg/kg of body weight at the time of inoculation and one week

    later. Thereafter, P3-A and P3-B received M-T807R1 at 25 mg/kg of body weight at

    weeks 12 and 13 or 11 and 12, for P3-A and P3-B, respectively, and at 50 mg/kg of body

    weight at week 27 or 26, for P3-A and P3-B, respectively. Passage 4 was initiated using

    blood obtained from P3-A and P3-B at weeks 47 and 46 after infection, respectively, one

    week after administration of M-T807R1 at 50 mg/kg. P4-A was inoculated with 10ml

    blood obtained from P3-A, P4-B was inoculated with blood from both P3-A and P3-B

    (5ml each) and P4-C was inoculated with 10ml blood from P3-B. M-T807R1 was

    administered at 25 mg/kg to P4-A, P4-B and P4-C at the time of inoculation and at one

    week post inoculation. For passage 5, blood obtained from animal P4-C at week 28 after

    infection (necropsy) was used to inoculate four animals (10ml blood per animal). P5-A

    and P5-B were not subjected to CD8+ T-cell depletion, while P5-C and P5-D were

    injected with M-T807R1 at the time of inoculation and one week thereafter. M-T807R1

    was subsequently administered to P5-A and P5-B at weeks 23 and 24 pi. Blood (10ml)

    obtained from P5-C at week 39 after infection was used to inoculate P6-A, that was not

    subjected to CD8+ T-cell depletion, and P6-B that was injected with M-T807R1 at the

    time of inoculation and one week thereafter.

    Viral load measurements

    Virions were pelleted from plasma and virion-associated RNA extracted as described

    previously(11). Plasma viral load was quantified using real time qRT-PCR, based on

    amplification of an HIV-1NL4-3-derived sequence located in the Gag coding region.

    Reverse transcription was performed with random hexamers and SuperScript II reverse

    transcriptase (Invitrogen). For the real time PCR amplification, the forward primer was 5’

    CTA GAA CGA TTC GCA GTT AAT CCT 3’ the reverse primer was 5’ CTA TCC TTT

    GAT GCA CAC AAT AGA G 3’ and the FRET probe was 5’ FAM

    TCCCAGTATTTGTCTACAGCCTTCTGATG-BHQ 3’. Each PCR mix contained

    cDNA templates, 1x PCR II buffer (ABI), 4.5 mM MgCl2, 0.6 μM primers, 0.1 μM

    probe, and 1.25 units AmpliTaq Gold DNA polymerase (ABI). PCR reactions were

    performed on an ABI 7500 Sequence Detection System, (1 cycle of 95 °C for 10 min

    followed by 45 cycles of 95 °C for 15 seconds and 60 °C for 1 min). Fluorescent signal-

    based quantification of vRNA copy numbers in test samples were determined by ABI

    7500 System SDS software, using a standard curve constructed from serial dilutions of an

    appropriate RNA control template.

    Flow cytometry

    Antibodies and reagents were obtained from BD Biosciences, unless indicated otherwise

    and data analysis was performed using FCS Express (De Novo Software). Sample

    preparation absolute cell counting and lymphocyte immunophenotyping methods were

  • performed as previously described(11, 27, 28) Briefly, absolute cell counts were

    performed on EDTA-anti-coagulated whole blood using the following surface antigen

    staining panel: CD45 FITC (DO58-1283), CD3 PE (SP34-2), CD4 APC (L200), CD14

    APC-Cy7 (M5E2; BioLegend), CD8α PE-Cy7 (SK1), and CD20 Pacific Blue (2H7;

    BioLegend). FACS Lysing solution (BD Biosciences) was then added and approximately

    50,000 CD45+CD3+ cells were acquired for each sample, using a BD FACSVerse flow

    cytometer equipped with a volumetric flow sensor. Lymphocyte immunophenotyping

    was performed on freshly isolated mononuclear cells using the following antibodies: CD4

    Pacific Blue (OKT4; BioLegend), CCR5 PE (3A9), CD28 ECD (CD28.2; Beckman

    Coulter), CD95 PE-Cy5 (DX2), CD8 PE-Cy7 (SK1), CD38 APC (OK10; NIH

    Nonhuman Primate Resource), CD3 APC-Cy7 (SP34-2), and Ki67 FITC (B56). Surface

    and intracellular staining was performed using BD Cytofix/Cytoperm reagents and

    protocol. Approximately 200,000 CD3+ T-cells were acquired for each sample using a

    BD LSR-II flow cytometer.

    Western blot analyses

    HIV-1 (AD8) virions, generated by transfection of 293T cells with a proviral plasmid

    were purified by centrifugation through 20% sucrose and resuspended in SDS/PAGE

    loading buffer. Virion proteins were separated on 4 to 12% acrylamide gels and blotted

    onto nitrocellulose membranes, which were then cut into strips. The strips were probed

    with heat-inactivated plasma from infected macaques, diluted 1:200, or serum from an

    HIV-1-infected human as a control, followed by an anti-human IgG-peroxidase

    conjugate. Blots were developed using chemiluminescent detection reagents (Pierce).

    Single genome amplification/sequencing and phylogenetic analysis

    Sequences were obtained from plasma viral RNA and PCR amplified using the single

    genome amplification technique as previously described(18). The entire 3’ half of the

    viral genome (including the entire vif, vpr, vpu, tat, rev env and nef genes) was amplified

    from cDNA generated by reverse transcription of RNA using SuperScript III reverse

    transcriptase according to the manufacturer’s recommendations (Invitrogen). In brief, a

    cDNA reaction of 1× RT buffer, 0.5 mM of each deoxynucleoside triphosphate, 5 mM

    dithiothreitol, 2 U/ml RNaseOUT (RNase inhibitor), 10 U/ml of SuperScript III reverse

    transcriptase, and 0.25 mM antisense primer HIVR3B3.R1 5’-

    ACTACTTGAAGCACTCAAGGCAAGCTTTATTG-3’ was incubated at 50°C for 60

    min, 55°C for 60 min and then heat-inactivated at 70°C for 15 min followed by treatment

    with 1 U of RNase H at 37°C for 20 min. Thereafter, cDNA was amplified via limiting

    dilution PCR where only one amplifiable molecule was present in each reaction using 1×

    PCR buffer, 2mM MgCl2, 0.2mM of each deoxynucleoside triphosphate, 0.2μM of each

    primer, and 0.025 U/μl Platinum Taq polymerase (Invitrogen) in a 20-μl reaction. First

    round PCR was performed with sense primer HIVBK3F1 5’-

    ACAGCAGTACAAATGGCAGTATT-3’ and antisense primer HIVR3B3.R1 under the

    following conditions: 1 cycle of 94°C for 2 min, 35 cycles at 94°C for 15 sec, 55°C for

    30 sec, and 72°C for 4 min, followed by a final extension of 72°C for 10 min. Next, 1μl

    from the first-round PCR product was added to a second-round PCR reaction that

    included the sense primer HIVBK3F2 5’- TGGAAAGGTGAAGGGGCAGT-

    AGTAATAC-3’ and antisense primer HIVR3B6.R2 5’-

  • TGAAGCACTCAAGGCAAGCTTTA-TTGAGGC-3’ performed under the same

    conditions used for first-round PCR, but with a total of 45 cycles. For 5’ genome

    sequences including Gag and Pol, the following primers were used: HIVBK5R1 5’-

    CTTGCCACACAATCATCACCTGCCATCTG-3’, HIVBK5R2 5’-CAATCA-

    TCACCTGCCATCTGTTTTCCATA-3’, HIVU5B1F1 5’-

    CCTTGAGTGCTTCAAGTAGTGT-GTGCCCGTCTGT-3’, and HIVU5B4F2 5’-

    GTAGTGTGTGCCCGTCTGTTGTGTGACTC-3’. Correct sized amplicons were

    identified by agarose gel electrophoresis and directly sequenced with second round PCR

    primers and HIV-1 specific primers using BigDye Terminator technology. Sequences

    were aligned using ClustalW and hand edited using MacClade 4.08 to improve alignment

    quality. Trees were constructed using the neighbor-joining method.

    Immunohistochemistry and In situ hybridization analysis

    Immunohistochemistry (IHC) was performed using a biotin-free polymer approach

    (Golden Bridge International, Inc.) and 5μm tissue sections, mounted on glass slides as

    previously described(27). For CD4+ T cell IHC, heat induced epitope retrieval (HIER)

    was performed by heating sections in 0.01% citraconic anhydride containing 0.05%

    Tween-20 in a pressure cooker (Biocare Medical) set at 122°C for 30 s. Slides were

    incubated with blocking buffer (TBS with 0.05% Tween-20 and 0.25% casein) for 10min

    and then incubated with mouse anti-CD68 (1:400; clone KP1, Dako), mouse anti-CD163

    (1:400; clone 10D6; Novocastra/Leica) and rabbit monoclonal anti-CD4 (1:200; clone

    EPR6855; Abcam, Inc.) diluted in blocking buffer overnight at 4oC. Slides were washed

    in 1X TBS with 0.05% Tween-20 and endogenous peroxidases blocked using 1.5% (v/v)

    H2O2 in TBS (pH 7.4) for 10min. Slides were incubated Mouse Polink-1 AP followed by

    Rabbit Polink-1 horseradish peroxidase (HRP, Golden Bridge International, Inc.) for 30

    min each at room temperature. Tissue sections were first incubated with 3,3′-

    diaminobenzidine (Impact™ DAB, Vector Laboratories) to reveal CD4, washed and

    developed with Warp Red (Biocare Medical, Inc.) to mask the low levels of CD4

    expressed on myeloid cells, allowing for specific identification of CD4+ T cells.

    Identification of B-cell lymphomas was performed by performing double IHC for CD3

    (Warp Red) and CD20 (DAB) in a manner analogous to the CD4+ T cell IHC described

    above, but utilizing Mouse Polink-1 HRP followed by Rabbit Polink-1 alakaline

    phosphatase (ALP). Slides were washed in ddH2O, counterstained with hematoxylin,

    mounted in Permount (Fisher Scientific), and scanned at high magnification (x200) using

    the ScanScope CS System (Aperio Technologies) yielding high-resolution images from

    the entire tissue section. Representative regions of interest (500 m2) were identified and

    high-resolution images extracted from the whole-tissue scans. In situ hybridization

    (chromogenic and fluorescent) analysis was performed as previously described using

    HIV-1 clade B lineage specific riboprobes(29, 30). Phenotypic analysis of the cell types

    productively infected with HIV-1 was performed by manually counting the HIV-1

    vRNA+ cells from confocal images taken on an Olympus FV10i confocal microscope

    using a 60X oil-immersion objective (NA 1.35) as previously described(30). Lung tissue

    was stained using the Grocott's methenamine silver stain to identify fungal organisms, in

    particular Pneumocystis which causes Pneumocystis Pneumonia (PCP), a classical AIDS-

    defining illness. Antibodies used for IHC in this study were rabbit monoclonal anti-

    human CD3 (clone SP7; Thermo Scientific), rabbit monoclonal anti-human CD4 (clone

  • EPR6855; Abcam), mouse anti-human CD20 (clone L26; Dako), mouse anti-human

    CD68 (clone KP1; Dako), mouse anti-human CD163 (clone NCL-L-CD163;

    Novocastra/Leica), mouse anti-human collagen I (clone COL-1; Sigma-Aldrich); mouse

    anti-human collagen III (clone FH-7A; Sigma-Aldrich); and rabbit monoclonal anti-

    human Ki67 (clone SP6; Lab Vision/Thermo Fisher Scientific).

    Analysis of adapted envelope function and inhibitor sensitivity

    Env coding sequences, derived from P5-B plasma 3’ viral clones were inserted into

    pcDNA3.1/V5-His-TOPO (Invitrogen). Control plasmids expressing the same cassette

    amplified from the HIV-1AD8 or HIV-1NL4-3 proviral construct were also generated. These

    Env-expressing plasmids were co-transfected with a proviral HIV-1ΔEnv/GFP plasmid in

    293T cells. Clarified and filtered supernatants, harvested 48h post-transfection, were used

    to infect MT2-R5 cells in absence or presence of Maraviroc (8nM) or AMD3100 (1μM).

    Numbers of GFP positive cells obtained in the absence of drugs were set to100% for each

    virus and percentage of infection in the presence of drugs is presented.

    Analysis of Vpu function

    To assess the ability of Vpu to antagonize tetherin, HIV-1 proviral plasmids (HIV-1NL4-3,

    HIV-1NL4-3 delVpu or HIV-1KB9Vpu 15/21) were cotransfected in 293T cells with

    increasing amounts of plasmids expressing human tetherin, or three naturally occurring

    variants of pigtailed macaque tetherin(21). At 48h after transfection, the infectious virion

    yield was determined by applying clarified and filtered cell culture supernatants to TZM-

    bl cells, and measuring β-galactosidase activity in cell lysates, 48h later.

  • Fig. S1.

    Schematic representation of the HIV-1 constructs used in this study. White and black

    regions indicate HIV-1 and SIVMAC239-derived sequences respectively. Env proteins were

    derived from prototype R5-tropic HIV-1 strains (AD8, YU2, BaL) or the macaque-

    adapted KB9 envelope with the V3 loop substituted with that from ADA (KB9V3ADA).

    See methods for details of construction.

  • Fig. S2

    CD8+ T-cell counts in the blood of infected macaques. Absolute numbers of CD8+CD3+

    cells were determined by flow cytometry. Color-coded arrowheads indicate the times at

    which the first of two doses (1 week apart) of a CD8 antibody (either cM-T807 or M-

    T807R1) was administered.

  • Fig. S3

    CD8+ T-cell subset frequencies in GALT of HIV-1 infected macaques. Cell suspensions

    derived from GALT specimens obtained at the indicated times after infection were

    analyzed by flow cytometry using antibodies against CD3, CD4, CD8, CD28 and CD95.

    The percentage of CD3+ cells that also expressed CD8 and central memory (CD95+,

    CD28+, effector memory (CD95+, CD28-), or naïve markers (CD95-, CD28+) is plotted.

  • Fig. S4

    (A) Immune activation in GALT CD4+ cells. Cell suspensions derived from GALT

    specimens obtained at the indicated times after infection were analyzed by flow

    cytometery using antibodies against CD3, CD4 and Ki67. The percentage of CD3+CD4+

    cells that also expressed Ki67 is plotted. (B) Immmunohistochemical stain for Ki67

    antigen (brown) in lymph node sections taken from P4 animals pre infection or 18

    weeeks after after infection. (C) Immmunohistochemical stain for collagen I, (brown) in

    lymph node sections from the same specimens as in (B). (D) Immmunohistochemical

    stain for collagen III (brown) in lymph node sections from the same specimens as in (B).

  • Fig. S5

    Characterization of AIDS-defining B-cell lymphoma found in P4-C. (A) Hematoxylin

    and eosin stained representative tumor sections from retro-orbital, spinal and renal

    masses. Scale bars = 5mm (top row) 1mm (second row) 400 μm (third row) and 100 μm

    (fourth row). (B) Immunohistochemical staining of CD20+ B-cells (brown) and CD3+ T-

    cells (red) at the three tumor sites. Scale bars = 100μm. (C) Negative control stains

    (brown) for CD3+ T-cells (upper panels) and CD20+ B-cells lower panels from

    unaffected tissues (Kidney) taken from clinically well macaques. Scale bars = 200μm.

  • Fig. S6

    Detection of Pneumocystis organisms in lungs of macaques subjected to Grocott's

    methenamine silver stain. (A) Control sections from clinically well macaques P2-A and

    P3-B (compare with Fig. 2E). (B) Detection of Pnuemocystis organisms in lung sections

    from P5-C (black spots, right panel). A control Pneumocystis-negative lung specimen

    (from P5-A, left panel) is also shown for comparison. Scale bars = 50μm.

  • Fig. S7

    Cell types infected by HIV-1 in macaques. (A) In situ hybridization analysis to detect

    infected HIV-1 RNA-positive cells (brown) in lymph nodes and GALT of an infected

    macaque (P4-C), Scale bars = 100μm. (B) Example of a lymph node section, from P4-C,

    subjected to fluorescent in-situ hybridization (FISH) to detect HIV-1 infected cells (red)

    and immunofluorescence to detect macrophages (CD68+/CD163+; green) and T-cells

    (CD3+, blue). Examples of infected T-cells and macrophages are indicated by black and

    white arrows respectively. Scale bars = 50μm. (C) Phenotypic analysis of the cell types

    (CD3+ T-cells or CD68+/CD163+ macrophages) productively infected by HIV-1 in

    lymph node and GALT sections from infected macaques, at the indicated times after

    infection. The number of infected cells analyzed is given for each animal and time point.

  • Fig. S8

    Antibody responses to HIV-1 in pigtail macaques. Western blot analyses were performed

    using purified HIV-1 virions (encoding AD8 Env) as the antigen and using plasma

    samples recovered from the HIV-1 infected macaques at the indicated number of weeks

    after infection. Serum from an HIV-1-infected human long-term non-progressor (+ve)

    was used as a positive control.

  • References and Notes

    1. D. Blanco-Melo, S. Venkatesh, P. D. Bieniasz, Intrinsic cellular defenses against human

    immunodeficiency viruses. Immunity 37, 399–411 (2012). Medline

    doi:10.1016/j.immuni.2012.08.013

    2. M. Stremlau, C. M. Owens, M. J. Perron, M. Kiessling, P. Autissier, J. Sodroski, The

    cytoplasmic body component TRIM5alpha restricts HIV-1 infection in Old World

    monkeys. Nature 427, 848–853 (2004). Medline doi:10.1038/nature02343

    3. R. Mariani, D. Chen, B. Schröfelbauer, F. Navarro, R. König, B. Bollman, C. Münk, H.

    Nymark-McMahon, N. R. Landau, Species-specific exclusion of APOBEC3G from HIV-

    1 virions by Vif. Cell 114, 21–31 (2003). Medline doi:10.1016/S0092-8674(03)00515-4

    4. Z. Ambrose, V. N. KewalRamani, P. D. Bieniasz, T. Hatziioannou, HIV/AIDS: In search of an

    animal model. Trends Biotechnol. 25, 333–337 (2007). Medline

    doi:10.1016/j.tibtech.2007.05.004

    5. T. Hatziioannou, D. T. Evans, Animal models for HIV/AIDS research. Nat. Rev. Microbiol.

    10, 852–867 (2012). Medline doi:10.1038/nrmicro2911

    6. C. H. Liao, Y. Q. Kuang, H. L. Liu, Y. T. Zheng, B. Su, A novel fusion gene, TRIM5-

    Cyclophilin A in the pig-tailed macaque determines its susceptibility to HIV-1 infection.

    AIDS 21 (suppl. 8), S19–S26 (2007). Medline doi:10.1097/01.aids.0000304692.09143.1b

    7. C. A. Virgen, Z. Kratovac, P. D. Bieniasz, T. Hatziioannou, Independent genesis of chimeric

    TRIM5-cyclophilin proteins in two primate species. Proc. Natl. Acad. Sci. U.S.A. 105,

    3563–3568 (2008). Medline doi:10.1073/pnas.0709258105

    8. G. Brennan, Y. Kozyrev, S. L. Hu, TRIMCyp expression in Old World primates Macaca

    nemestrina and Macaca fascicularis. Proc. Natl. Acad. Sci. U.S.A. 105, 3569–3574

    (2008). Medline doi:10.1073/pnas.0709511105

    9. R. M. Newman, L. Hall, A. Kirmaier, L. A. Pozzi, E. Pery, M. Farzan, S. P. O’Neil, W.

    Johnson, Evolution of a TRIM5-CypA splice isoform in old world monkeys. PLOS

    Pathog. 4, e1000003 (2008). Medline doi:10.1371/journal.ppat.1000003

    10. C. A. Virgen, T. Hatziioannou, Antiretroviral activity and Vif sensitivity of rhesus macaque

    APOBEC3 proteins. J. Virol. 81, 13932–13937 (2007). Medline doi:10.1128/JVI.01760-

    07

    11. T. Hatziioannou, Z. Ambrose, N. P. Chung, M. Piatak Jr., F. Yuan, C. M. Trubey, V. Coalter,

    R. Kiser, D. Schneider, J. Smedley, R. Pung, M. Gathuka, J. D. Estes, R. S. Veazey, V.

    N. KewalRamani, J. D. Lifson, P. D. Bieniasz, A macaque model of HIV-1 infection.

    Proc. Natl. Acad. Sci. U.S.A. 106, 4425–4429 (2009). Medline

    doi:10.1073/pnas.0812587106

    12. R. S. Veazey, P. M. Acierno, K. J. McEvers, S. H. Baumeister, G. J. Foster, M. D. Rett, M.

    H. Newberg, M. J. Kuroda, K. Williams, E. Y. Kim, S. M. Wolinsky, E. P. Rieber, M.

    Piatak Jr., J. D. Lifson, D. C. Montefiori, C. R. Brown, V. M. Hirsch, J. E. Schmitz,

    Increased loss of CCR5+ CD45RA

    – CD4

    + T cells in CD8

    + lymphocyte-depleted simian

    immunodeficiency virus-infected rhesus monkeys. J. Virol. 82, 5618–5630 (2008).

    Medline doi:10.1128/JVI.02748-07

    http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=PubMed&list_uids=22999946&dopt=Abstracthttp://dx.doi.org/10.1016/j.immuni.2012.08.013http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=PubMed&list_uids=14985764&dopt=Abstracthttp://dx.doi.org/10.1038/nature02343http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=PubMed&list_uids=12859895&dopt=Abstracthttp://dx.doi.org/10.1016/S0092-8674(03)00515-4http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=PubMed&list_uids=17574286&dopt=Abstracthttp://dx.doi.org/10.1016/j.tibtech.2007.05.004http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=PubMed&list_uids=23154262&dopt=Abstracthttp://dx.doi.org/10.1038/nrmicro2911http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=PubMed&list_uids=18172386&dopt=Abstracthttp://dx.doi.org/10.1097/01.aids.0000304692.09143.1bhttp://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=PubMed&list_uids=18287034&dopt=Abstracthttp://dx.doi.org/10.1073/pnas.0709258105http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=PubMed&list_uids=18287033&dopt=Abstracthttp://dx.doi.org/10.1073/pnas.0709511105http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=PubMed&list_uids=18389077&dopt=Abstracthttp://dx.doi.org/10.1371/journal.ppat.1000003http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=PubMed&list_uids=17942564&dopt=Abstracthttp://dx.doi.org/10.1128/JVI.01760-07http://dx.doi.org/10.1128/JVI.01760-07http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=PubMed&list_uids=19255423&dopt=Abstracthttp://dx.doi.org/10.1073/pnas.0812587106http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=PubMed&list_uids=18367534&dopt=Abstracthttp://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=PubMed&list_uids=18367534&dopt=Abstracthttp://dx.doi.org/10.1128/JVI.02748-07

  • 13. B. U. Orzechowska, M. F. Powers, J. Sprague, H. Li, B. Yen, R. P. Searles, M. K. Axthelm,

    S. W. Wong, Rhesus macaque rhadinovirus-associated non-Hodgkin lymphoma: Animal

    model for KSHV-associated malignancies. Blood 112, 4227–4234 (2008). Medline

    doi:10.1182/blood-2008-04-151498

    14. T. K. Rao, Human immunodeficiency virus (HIV) associated nephropathy. Annu. Rev. Med.

    42, 391–401 (1991). Medline doi:10.1146/annurev.med.42.1.391

    15. E. B. Stephens, C. Tian, Z. Li, O. Narayan, V. H. Gattone II, Rhesus macaques infected with

    macrophage-tropic simian immunodeficiency virus (SIVmacR71/17E) exhibit extensive

    focal segmental and global glomerulosclerosis. J. Virol. 72, 8820–8832 (1998). Medline

    16. P. Simmonds, P. Balfe, C. A. Ludlam, J. O. Bishop, A. J. Brown, Analysis of sequence

    diversity in hypervariable regions of the external glycoprotein of human

    immunodeficiency virus type 1. J. Virol. 64, 5840–5850 (1990). Medline

    17. S. Palmer, M. Kearney, F. Maldarelli, E. K. Halvas, C. J. Bixby, H. Bazmi, D. Rock, J.

    Falloon, R. T. Davey Jr., R. L. Dewar, J. A. Metcalf, S. Hammer, J. W. Mellors, J. M.

    Coffin, Multiple, linked human immunodeficiency virus type 1 drug resistance mutations

    in treatment-experienced patients are missed by standard genotype analysis. J. Clin.

    Microbiol. 43, 406–413 (2005). Medline doi:10.1128/JCM.43.1.406-413.2005

    18. B. F. Keele, E. E. Giorgi, J. F. Salazar-Gonzalez, J. M. Decker, K. T. Pham, M. G. Salazar,

    C. Sun, T. Grayson, S. Wang, H. Li, X. Wei, C. Jiang, J. L. Kirchherr, F. Gao, J. A.

    Anderson, L. H. Ping, R. Swanstrom, G. D. Tomaras, W. A. Blattner, P. A. Goepfert, J.

    M. Kilby, M. S. Saag, E. L. Delwart, M. P. Busch, M. S. Cohen, D. C. Montefiori, B. F.

    Haynes, B. Gaschen, G. S. Athreya, H. Y. Lee, N. Wood, C. Seoighe, A. S. Perelson, T.

    Bhattacharya, B. T. Korber, B. H. Hahn, G. M. Shaw, Identification and characterization

    of transmitted and early founder virus envelopes in primary HIV-1 infection. Proc. Natl.

    Acad. Sci. U.S.A. 105, 7552–7557 (2008). Medline doi:10.1073/pnas.0802203105

    19. H. Choe, M. Farzan, Y. Sun, N. Sullivan, B. Rollins, P. D. Ponath, L. Wu, C. R. Mackay, G.

    LaRosa, W. Newman, N. Gerard, C. Gerard, J. Sodroski, The β-chemokine receptors

    CCR3 and CCR5 facilitate infection by primary HIV-1 isolates. Cell 85, 1135–1148

    (1996). Medline doi:10.1016/S0092-8674(00)81313-6

    20. S. T. Sina, W. Ren, C. Cheng-Mayer, Coreceptor use in nonhuman primate models of HIV

    infection. J. Transl. Med. 9 (suppl. 1), S7 (2011). Medline doi:10.1186/1479-5876-9-S1-

    S7

    21. M. W. McNatt, T. Zang, T. Hatziioannou, M. Bartlett, I. B. Fofana, W. E. Johnson, S. J. Neil,

    P. D. Bieniasz, Species-specific activity of HIV-1 Vpu and positive selection of tetherin

    transmembrane domain variants. PLOS Pathog. 5, e1000300 (2009). Medline

    doi:10.1371/journal.ppat.1000300

    22. B. Jia, R. Serra-Moreno, W. Neidermyer, A. Rahmberg, J. Mackey, I. B. Fofana, W. E.

    Johnson, S. Westmoreland, D. T. Evans, Species-specific activity of SIV Nef and HIV-1

    Vpu in overcoming restriction by tetherin/BST2. PLOS Pathog. 5, e1000429 (2009).

    Medline doi:10.1371/journal.ppat.1000429

    23. D. Sauter, M. Schindler, A. Specht, W. N. Landford, J. Münch, K. A. Kim, J. Votteler, U.

    Schubert, F. Bibollet-Ruche, B. F. Keele, J. Takehisa, Y. Ogando, C. Ochsenbauer, J. C.

    http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=PubMed&list_uids=18757778&dopt=Abstracthttp://dx.doi.org/10.1182/blood-2008-04-151498http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=PubMed&list_uids=2035984&dopt=Abstracthttp://dx.doi.org/10.1146/annurev.med.42.1.391http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=PubMed&list_uids=9765427&dopt=Abstracthttp://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=PubMed&list_uids=2243378&dopt=Abstracthttp://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=PubMed&list_uids=15635002&dopt=Abstracthttp://dx.doi.org/10.1128/JCM.43.1.406-413.2005http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=PubMed&list_uids=18490657&dopt=Abstracthttp://dx.doi.org/10.1073/pnas.0802203105http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=PubMed&list_uids=8674119&dopt=Abstracthttp://dx.doi.org/10.1016/S0092-8674(00)81313-6http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=PubMed&list_uids=21284906&dopt=Abstracthttp://dx.doi.org/10.1186/1479-5876-9-S1-S7http://dx.doi.org/10.1186/1479-5876-9-S1-S7http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=PubMed&list_uids=19214216&dopt=Abstracthttp://dx.doi.org/10.1371/journal.ppat.1000300http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=PubMed&list_uids=19436700&dopt=Abstracthttp://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=PubMed&list_uids=19436700&dopt=Abstracthttp://dx.doi.org/10.1371/journal.ppat.1000429

  • Kappes, A. Ayouba, M. Peeters, G. H. Learn, G. Shaw, P. M. Sharp, P. Bieniasz, B. H.

    Hahn, T. Hatziioannou, F. Kirchhoff, Tetherin-driven adaptation of Vpu and Nef function

    and the evolution of pandemic and nonpandemic HIV-1 strains. Cell Host Microbe 6,

    409–421 (2009). Medline doi:10.1016/j.chom.2009.10.004

    24. V. M. Hirsch, S. Santra, S. Goldstein, R. Plishka, A. Buckler-White, A. Seth, I. Ourmanov,

    C. R. Brown, R. Engle, D. Montefiori, J. Glowczwskie, K. Kunstman, S. Wolinsky, N. L.

    Letvin, Immune failure in the absence of profound CD4+ T-lymphocyte depletion in

    simian immunodeficiency virus-infected rapid progressor macaques. J. Virol. 78, 275–

    284 (2004). Medline doi:10.1128/JVI.78.1.275-284.2004

    25. Single-letter abbreviations for the amino acid residues are as follows: A, Ala; C, Cys; D,

    Asp; E, Glu; F, Phe; G, Gly; H, His; I, Ile; K, Lys; L, Leu; M, Met; N, Asn; P, Pro; Q,

    Gln; R, Arg; S, Ser; T, Thr; V, Val; W, Trp; and Y, Tyr.

    26. Y. J. Zhang, T. Hatziioannou, T. Zang, D. Braaten, J. Luban, S. P. Goff, P. D. Bieniasz,

    Envelope-dependent, cyclophilin-independent effects of glycosaminoglycans on human

    immunodeficiency virus type 1 attachment and infection. J. Virol. 76, 6332–6343 (2002).

    Medline doi:10.1128/JVI.76.12.6332-6343.2002

    27. B. Tabb, D. R. Morcock, C. M. Trubey, O. A. Quiñones, X. P. Hao, J. Smedley, R.

    Macallister, M. Piatak Jr., L. D. Harris, M. Paiardini, G. Silvestri, J. M. Brenchley, W. G.

    Alvord, J. D. Lifson, J. D. Estes, Reduced inflammation and lymphoid tissue

    immunopathology in rhesus macaques receiving anti-tumor necrosis factor treatment

    during primary simian immunodeficiency virus infection. J. Infect. Dis. 207, 880–892

    (2013). Medline doi:10.1093/infdis/jis643

    28. G. Q. Del Prete, M. F. Kearney, J. Spindler, A. Wiegand, E. Chertova, J. D. Roser, J. D.

    Estes, X. P. Hao, C. M. Trubey, A. Lara, K. Lee, C. Chaipan, J. W. Bess Jr., K.

    Nagashima, B. F. Keele, R. Macallister, J. Smedley, V. K. Pathak, V. N. Kewalramani, J.

    M. Coffin, J. D. Lifson, Restricted replication of xenotropic murine leukemia virus-

    related virus in pigtailed macaques. J. Virol. 86, 3152–3166 (2012). Medline

    doi:10.1128/JVI.06886-11

    29. J. M. Brenchley, C. Vinton, B. Tabb, X. P. Hao, E. Connick, M. Paiardini, J. D. Lifson, G.

    Silvestri, J. D. Estes, Differential infection patterns of CD4+ T cells and lymphoid tissue

    viral burden distinguish progressive and nonprogressive lentiviral infections. Blood 120,

    4172–4181 (2012). Medline doi:10.1182/blood-2012-06-437608

    30. A. M. Ortiz, N. R. Klatt, B. Li, Y. Yi, B. Tabb, X. P. Hao, L. Sternberg, B. Lawson, P. M.

    Carnathan, E. M. Cramer, J. C. Engram, D. M. Little, E. Ryzhova, F. Gonzalez-Scarano,

    M. Paiardini, A. A. Ansari, S. Ratcliffe, J. G. Else, J. M. Brenchley, R. G. Collman, J. D.

    Estes, C. A. Derdeyn, G. Silvestri, Depletion of CD4+ T cells abrogates post-peak decline

    of viremia in SIV-infected rhesus macaques. J. Clin. Invest. 121, 4433–4445 (2011).

    Medline doi:10.1172/JCI46023

    http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=PubMed&list_uids=19917496&dopt=Abstracthttp://dx.doi.org/10.1016/j.chom.2009.10.004http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=PubMed&list_uids=14671109&dopt=Abstracthttp://dx.doi.org/10.1128/JVI.78.1.275-284.2004http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=PubMed&list_uids=12021366&dopt=Abstracthttp://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=PubMed&list_uids=12021366&dopt=Abstracthttp://dx.doi.org/10.1128/JVI.76.12.6332-6343.2002http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=PubMed&list_uids=23087435&dopt=Abstracthttp://dx.doi.org/10.1093/infdis/jis643http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=PubMed&list_uids=22238316&dopt=Abstracthttp://dx.doi.org/10.1128/JVI.06886-11http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=PubMed&list_uids=22990012&dopt=Abstracthttp://dx.doi.org/10.1182/blood-2012-06-437608http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=PubMed&list_uids=22005304&dopt=Abstracthttp://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=PubMed&list_uids=22005304&dopt=Abstracthttp://dx.doi.org/10.1172/JCI46023