Simian Immunodeficiency Virus SIVagm.sab Infection of Caribbean African Green Monkeys: a New Model for the Study of SIV Pathogenesis in Natural Hosts

Size: px
Start display at page:

Download "Simian Immunodeficiency Virus SIVagm.sab Infection of Caribbean African Green Monkeys: a New Model for the Study of SIV Pathogenesis in Natural Hosts"

Transcription

1 JOURNAL OF VIROLOGY, May 2006, p Vol. 80, No X/06/$ doi: /jvi Copyright 2006, American Society for Microbiology. All Rights Reserved. Simian Immunodeficiency Virus SIVagm.sab Infection of Caribbean African Green Monkeys: a New Model for the Study of SIV Pathogenesis in Natural Hosts Ivona Pandrea, 1 * Cristian Apetrei, 1 Jason Dufour, 1 Nora Dillon, 1 Joseph Barbercheck, 1 Michael Metzger, 1 Béatrice Jacquelin, 2 Rudolf Bohm, 1 Preston A. Marx, 1 Françoise Barre-Sinoussi, 2 Vanessa M. Hirsch, 3 Michaela C. Müller-Trutwin, 2 Andrew A. Lackner, 1 and Ronald S. Veazey 1 Tulane National Primate Research Center, Covington, Louisiana 70433; Tulane University Health Science Center, New Orleans, Louisiana ; Unite de Régulation des Infections Rétrovirales, Institut Pasteur, Paris, France 2 ; and Laboratory of Molecular Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland Received 16 December 2005/Accepted 3 March 2006 Caribbean-born African green monkeys (AGMs) were classified as Chlorocebus sabaeus by cytochrome b sequencing. Guided by these phylogenetic analyses, we developed a new model for the study of simian immunodeficiency virus (SIV) infection in natural hosts by inoculating Caribbean AGMs with their speciesspecific SIVagm.sab. SIVagm.sab replicated efficiently in Caribbean AGM peripheral blood mononuclear cells in vitro. During SIVagm.sab primary infection of six Caribbean AGMs, the virus replicated at high levels, with peak viral loads (VLs) of 10 7 to 10 8 copies/ml occurring by day 8 to 10 postinfection (p.i.). Set-point values of up to copies/ml were reached by day 42 p.i. and maintained throughout follow-up (through day 450 p.i.). CD4 T-cell counts in the blood showed a transient depletion at the peak of VL, and then returned to near preinfection values by day 28 p.i. and remained relatively stable during the chronic infection. Preservation of CD4 T cells was also found in lymph nodes (LNs) of chronic SIVagm.sab-infected Caribbean AGMs. No activation of CD4 T cells was detected in the periphery in SIV-infected Caribbean AGMs. These virological and immunological profiles from peripheral blood and LNs were identical to those previously reported in African-born AGMs infected with the same viral strain (SIVagm.sab92018). Due to these similarities, we conclude that Caribbean AGMs are a useful alternative to AGMs of African origin as a model for the study of SIV infection in natural African hosts. Simian immunodeficiency virus (SIV) infection in macaques, compared to SIV infection of African nonhuman primates (NHPs) such as African green monkeys (AGMs), sooty mangabeys (SMs), mandrills, and chimpanzees, differs in one fundamental aspect: clinical outcome. While macaques infected with SIVmac progress relatively rapidly to AIDS, African NHPs naturally or experimentally infected with their species-specific SIV rarely develop disease (4 6, 12, 26, 39, 42, 43, 47, 49, 50). Only a few cases of immunodeficiency have been reported to date in African NHPs (3, 29, 31, 44, 55). Numerous investigations have tried to explain this paradox, but no generally accepted explanation presently exists. In the past several years three experimental nonpathogenic models have been developed: infection of (African-origin) AGM species with their respective variants of SIVagm (9, 26, 42), infection of sooty mangabeys with SIVsm or SIVmac (25, 49), and infection of mandrills with SIVmnd-1 and SIVmnd-2 (39, 40). The study of these models showed that in naturally and experimentally SIV-infected African monkeys, the dynamics of plasma viral load (VL) is surprisingly similar to human immunodeficiency virus-infected humans and SIVmac-infected rhesus macaques (Rh), with peaks of viral replication ranging * Corresponding author. Mailing address: Division of Comparative Pathology, Tulane National Primate Research Center, Three Rivers Road, Covington, LA Phone: (985) Fax: (985) ipandrea@tulane.edu. between and and chronic VL levels of to (5, 9, 12, 13, 25, 26, 39, 40, 42, 43, 49, 50). High levels of viral replication in the absence of clinical progression were reported for SIVagm-infected AGMs (5, 12, 13, 26, 42) as well as for other African NHP hosts of SIV, such as SMs (6, 25, 30, 47, 49, 50) and mandrills (39, 40, 43, 44). However, although these models provided key new insights into SIV biology, they suffer from significant practical limitations, such as availability of experimental animals due to difficulties in their importation, risk of transfer of various pathogenic agents from Africa, and endangered species limitations. Therefore, development of an alternative animal model for natural SIV infection would be a significant achievement in the field. Given the endangered nature of other African NHP species currently used in AIDS research, AGMs would be the primate model of choice for investigation of viral replication and immune responses in the tissues in natural hosts infected with SIV: they are abundant and widely spread throughout sub- Saharan Africa, and they have a high prevalence of SIVagm infection in the wild (24, 45). Therefore, AGMs represent the largest reservoir of SIV. Different species of AGMs (vervet [Chlorocebus pygerythrus], grivet [C. aethiops], tantalus [C. tantalus], and sabaeus [C. sabaeus]) each carry their own SIVagm subtypes, named SIVagm.ver, SIVagm.gri, SIVagm.tan, and SIVagm.sab, respectively (1, 10, 11, 18, 23, 33). Since AGMs from Africa are currently difficult to import to the United 4858

2 VOL. 80, 2006 CARIBBEAN AGMs AS A MODEL FOR SIV INFECTION 4859 States and may carry various pathogenic agents, a possible alternative to the AGM model based on animals from Africa exists in the colonies of AGMs established approximately 300 years ago on several Caribbean islands by the transfer of AGMs from West Africa. In contrast to their African relatives, in which SIV prevalence is up to 60 to 70% in the adult population (24, 45), Caribbean AGMs are negative for SIV (7, 16). This led to the suggestion that Caribbean AGMs might be resistant to SIV infection (20). A more plausible explanation could be that the founder animals had been captured at a young age, when they were still uninfected (36). A second difference between the African-born and Caribbeanborn AGMs seems to lie in the CD4 and CD8 expression. It is widely accepted that CD4 T lymphocytes from most AGMs from Africa coexpress CD8 (12, 20, 32, 34, 37). However, this has been demonstrated to be due mainly to expression of the CD8 on AGM CD4 T cells, rather than to CD8 expression (20). The same study reported that Caribbean AGMs differ from their African counterparts in that they lack the double CD4 CD8 T-cell population. It was suggested that the lack of a double-positive phenotype might be considered a reversion to normal of a species that had not encountered SIVagm since separation from their African counterparts (20). The latter raised the question of whether the susceptibility of Caribbean AGMs to AIDS has changed (20). To determine whether Caribbean AGMs represent a suitable alternative to AGMs from Africa for SIV pathogenesis studies, we investigated the dynamics of VLs and changes in the major cell populations in peripheral blood and lymph nodes (LNs) of SIVagm-infected Caribbean AGMs and compared them with previous results from SIVagm-infected AGMs from Africa. We show here that Caribbean AGMs support productive and nonpathogenic SIV infection for up to 420 days. In SIVagm.sab92018-infected Caribbean AGMs, VLs and immunophenotypic changes are similar to those in African AGMs. No CD4 T-cell activation was detected in this new model during SIVagm infection. Thus, Caribbean AGMs infected with SIVagm.sab are appropriate models for studying the pathogenesis of SIV infection of natural hosts. These monkeys are plentiful and available; their use in this line of research will alleviate the previously mentioned restrictions. MATERIALS AND METHODS Animals. Twelve healthy, uninfected Caribbean AGMs originating from St. Kitts island were tested for CD4, CD8, and CD8 expression. The SIVagm.sab92018 infection study group comprised six female Caribbean AGMs (Chlorocebus sabaeus). The animals were adults (mean age, 8 years). All animals were negative for simian T-cell lymphotropic virus (Vironostika human T-cell lymphotropic virus-i/ii enzyme-linked immunosorbent assay [ELISA]; biomérieux, Durham, NC) and SIV by both ELISA and Western blot assay using SIVmac strips (Zeptometrix, Buffalo, N.Y.). Animals were housed at the Tulane National Primate Research Center (TNPRC), an AAALAC International-accredited facility. Housing and handling of animals were in accordance with the Guide for the Care and Use of Laboratory Animals (U.S. Public Health Service) (35) and the Animal Welfare Act. All protocols and procedures were reviewed and approved by the Tulane University Institutional Animal Care and Use Committee. Genetic characterization of Caribbean AGMs by cytochrome b sequencing. In order to confirm the species of Caribbean AGMs, DNA was extracted from 300 l of whole blood from 12 monkeys originating from St. Kitts, 5 monkeys originating from Barbados, 7 sabaeus monkeys from Senegal, 4 vervet monkeys from Tanzania, and 3 tantalus monkeys from the Central African Republic. A QIAamp blood kit (QIAGEN, Valencia, CA) was used to extract the DNA, which was eluted into 60 l ofh 2 O and stored at 20 C. A 267-base-pair (bp) region of cytochrome b, representing bases to of the human mitochondrial genome (2), was amplified using primers L14841 and H15149 (21). One microgram of host DNA was used in a 100- l PCR using a PerkinElmer model PE 9700 thermocycler. The PCR conditions were as follows: 2 min at 94 C, followed by 40 cycles of 30 s at 94 C, 30 s at 55 C, and 30 s at 72 C, with a final extension of 5 min at 72 C. Following purification with a QIAquick PCR purification kit (QIAGEN), the PCR products were cycle sequenced and analyzed on an ABI-377 automated DNA sequencer. Phylogenetic trees. Cytochrome b sequences from different species of AGMs were aligned to that from Chlorocebus aethiops (GenBank accession no. AY863426) and to sequences from other African NHPs retrieved from GenBank or generated in our laboratory. Sequences were aligned using Clustalw version 1.8. Unrooted maximum parsimony trees were inferred using PAUP version 4.0b8 (54) with uniformly weighted unordered characters. Distance matrix-based trees were constructed with the neighbor-joining method (48) using the HKY85 model of nucleotide substitution (15). The reliability of the branching order was estimated by performing 1,000 bootstrap replicates. In vitro infection of Caribbean PBMCs with SIVagm.sab. To verify the capacity of SIVagm.sab to infect peripheral blood mononuclear cells (PBMCs) from Caribbean AGMs in vitro, cells from four different animals were used. In order to test the virus in both CD4 phenotypes described in Caribbean AGMs, two animals with the double-positive CD4 CD8 T-cell population and two animals lacking this phenotype were examined. Cells were first stimulated with phytohemagglutinin (PHA; 1 ng/ml) for 3 days at 37 C in a 5% CO 2 incubator. Cells were then washed in 10% complete RPMI, counted, resuspended at cells/ml in complete medium containing 10% human interleukin-2 (IL-2), and cultured for an additional 3 days in Transwell plates (Costar, Corning, NY). In order to verify whether there was any modification in CD4 expression before and after stimulation of AGM PBMCs, CD4 expression was tested by flow cytometry at each of these steps. Before infection, the concentration of cells was again adjusted to /ml and 1 to 5 ml of virus stock (60 50% tissue culture infective doses of SIVagm.sab 2) was added to the culture for 24 h. The virus inoculum was washed out after 1 day, and the cell culture was maintained for 14 days. Viability of the cells was monitored daily, and fresh medium and IL-2 were added in order to maintain the culture at cells/ml. Every 3 days, 1 ml of tissue culture supernatant was removed from the culture and clarified by centrifugation (1,500 rpm for 10 min). Supernatants were tested for the presence of the virus using the SIV core P27 antigen capture assay (Zeptometrix, Buffalo, NY). As a control, SIVmac251 was grown on Rh PBMCs in the same conditions. SIVagm.sab in vivo infection. To avoid selection of viral variants in vitro, inocula used in this study consisted of plasma obtained from an experimentally SIVagm.sab92018-infected sabaeus monkey from Senegal during primary infection, and stocks were prepared and titers were determined on SupT1 as described elsewhere (9). All six AGMs were inoculated with plasma equivalent to % tissue culture infective doses of SIVagm.sab9018. Three AGMs (EI45, EI52, and EI53) received a viral stock that was prepared in a sabaeus monkey from Senegal. The same stock had been used in our previous studies for infection of African AGMs (26, 42). The other three AGMs (EI42, EI44, and EI51) were inoculated with a stock obtained in a Caribbean AGM (EI45) experimentally infected with the virus stock mentioned above. The virus stock was named SIVagm.sab92018 (EI45). The SIVagm.sab92018 (EI45) inoculum was adjusted to a viral RNA load equivalent to the initial stock, both determined by real-time (rt)-pcr. Specimen collection. Five milliliters of EDTA blood was collected from the femoral vein of each monkey two times before infection (days 15 and 0), twice weekly throughout the first 3 weeks postinfection (p.i.), weekly thereafter until the set point (days 28 and 35 p.i.), and finally, every month during chronic infection (between 2 and 18 months p.i.). Whole blood was used for flow cytometry within 1 hour after collection. Plasma was aliquoted and stored at 80 C prior to the VL quantification. Excisional biopsy specimens of axillary LNs were collected from all the animals as previously described (57). LNs were sampled before infection and at selected time points (days 0, 7, 10, 14, 28, 200, and 350 p.i.) during the acute and chronic phases of SIVagm infection. Isolation of lymphocytes. Lymphocytes were isolated from the axillary LNs by gently mincing and pressing tissues through nylon mesh screens and stained for flow cytometry as previously described (57). Flow cytometry. Whole blood was stained using a lysis technique as previously described (56). Blood cells and cell suspensions from LNs were stained for flow cytometric analysis using four-color staining combinations with monoclonal antibodies CD20-fluorescein isothiocyanate (FITC), CD3-phycoerythrin, CD8 -

3 4860 PANDREA ET AL. J. VIROL. peridinin chlorophyll A protein, CD4-allophycocyanin, and HLA-DR (BD Biosciences Pharmingen, San Diego, CA). Cells were incubated with an excess of monoclonal antibodies at 4 C for 30 min, followed by a phosphate-buffered saline wash (400 g, 7 min) and fixation in 2% paraformaldehyde. Data were acquired with a FACSCalibur flow cytometer and analyzed with CellQuest software (BD Bioscience Immunocytometry Systems). CD4 and CD8 expression on T-cell subsets was determined by gating on lymphocytes and then on CD3 T cells. To reflect the relative proportion of CD4 and CD8 T cells present in the LNs, we calculated an index of these cell populations. This index is the product of the percentage of CD3 T cells (gated on lymphocytes) multiplied by the percentage of CD4 or CD8 T cells, divided by 100. This index provides a more accurate reflection of target cells, since it is a function of both total CD3 T cells and CD4 or CD8 T cells. Antibody detection. Anti-SIVagm.sab92018 antibody dynamics were monitored by SIVagm.sab-specific enzyme immunoassay (primate immunodeficiency virus enzyme immunoassay) based on peptides mapping the conserved Gp41 immunodominant region and the highly variable V3 loop of SIVagm.sab2 (51). Serological reactivities were confirmed by Western blotting for all the monkeys included in this study (Zeptometrix Corp., Buffalo, NY). Quantification of viral P27 antigenemia. Plasma samples were assayed for P27 antigen by ELISA with standards provided in the SIV core p27 antigen capture assay kit (Zeptometrix Corp., Buffalo, NY). Quantification was done according to the manufacturer s instructions. This ELISA kit was initially designed to detect P27 of SIVmac but has also been shown to detect Gag antigens of SIVagm (27). In addition, in previous studies we confirmed its cross-reactivity for SIVagm.sab Gag (9, 26, 42). Viral RNA quantification. Viral RNA was extracted from 540 l of plasma using the QIAamp viral RNA extraction kit (QIAGEN, Valencia, CA). Plasma RNA quantification was done using rt-pcr assays specific for SIVagm.sab as previously described (26, 42). Briefly, total RNA was retrotranscribed into cdna using the TaqMan Gold rt-pcr kit and random hexamers (PE Applied Biosystems, Foster City, CA). PCRs were carried out in a spectrofluorometric thermal cycler (ABI PRISM 7700; PerkinElmer). The quantification was based on the amplification of a 180-bp segment located in the long terminal repeat region. This region is highly conserved within different SIVagm strains. The primers and probe were specifically designed for SIVagm.sab (J15S, 5 J15S, and J15P) as previously described (9). Absolute viral RNA copy numbers were deduced by comparing the relative signal strength to corresponding values obtained for five 10-fold dilutions of an SIVagm.sab long terminal repeat RNA that was reverse transcribed and amplified in parallel. The detection limit of the SIVagm.sab quantification assays was RNA copies/0.5 ml of plasma. Statistical analyses. Flow cytometry data were analyzed for significant differences by the Wilcoxon test using StatView software. Nucleotide sequence accession no. The nucleotide sequences of the cytochrome b genes from different species of AGMs were deposited in GenBank (accession no., DQ through DQ451245). RESULTS CD4, CD8, and CD8 expression in Caribbean AGMs. It was reported that Caribbean AGMs have a different CD4 T-cell phenotype than their African counterparts in that they lack the double-positive CD4 CD8 T-cell population (20). We have analyzed the blood of 12 Caribbean AGMs originating from St. Kitts and showed that, similar to AGMs from Africa, including sabaeus monkeys originating from Senegal (20, 26), Caribbean sabaeus monkeys are actually heterogeneous in terms of CD4 and CD8 coexpression (Fig. 1a and b). Furthermore, Caribbean sabaeus monkeys showed the same pattern of CD8 expression on CD4 T cells as AGMs from Africa (Fig. 1c and d). This indicates that there is no major difference between Caribbean monkeys and their African counterparts regarding the presence of double-positive CD4 CD8 T cells. Genetic classification of AGMs. A cytochrome b phylogenetic tree was constructed to determine to which species of AGMs Caribbean monkeys belonged. Data were also used to determine if there were any genetic differences between the FIG. 1. CD4 and CD8 expression in blood of Caribbean AGMs. Caribbean (St. Kitts) AGMs display two different phenotypes of CD4 T cells; a representative example of each is shown here. In some animals, such as AGM EI49, very few CD4 CD8 T cells are observed (a); AGM EI53 is representative of animals the majority of whose CD4 T cells have a low expression of CD8 (b). When CD8 T cells are defined with a CD8 monoclonal antibody, a similar pattern of staining is observed for all Caribbean AGMs, due to few CD4 CD8 T cells (c and d). monkeys that show the double CD4 CD8 phenotype and the animals which lack this phenotype. The phylogenetic analysis confirms that the monkeys originating from St. Kitts are AGMs (Fig. 2). These animals tightly clustered with AGMs originating from Barbados, a different Caribbean island. All Caribbean AGMs were different from vervet monkeys originating from Tanzania, tantalus monkeys from Central Africa, and grivet monkeys (sequence retrieved from GenBank) but formed a cluster with sabaeus monkeys originating from Senegal. The 12 AGMs from our study group, originating from St. Kitts, formed two closely related groups, differentiated by one nucleotide in the studied region (Fig. 2). No correlation between the CD4 CD8 phenotype or CD4 CD8 neg phenotype and the two subgroups of Caribbean AGMs was found. Monkeys with both phenotypes clustered in both subgroups (data not shown). Minimal CD4 down-regulation occurs after in vitro stimulation of AGM PBMCs. In order to study the susceptibility of Caribbean AGMs to in vitro infection by SIVagm, we first evaluated the levels of CD4 expression on activated CD4 T cells. Early studies reported a marked down-regulation of the CD4 expression in AGM PBMCs upon stimulation and suggested that this may explain the lack of pathogenicity of SIVagm in its natural host (34). Therefore, CD4 expression was examined in unstimulated and stimulated PBMCs from four AGMs (two representative of the CD4 CD8 phenotype and two that lacked CD8 on their CD4 T cells) in order to exclude down-regulation of CD4 as a potential cause of the failure of SIVagm.sab to grow in Caribbean AGM PBMCs. The mean percentage of CD4 cells normalized to CD3 after PBMC separation was 9.7% 1.3% in the four

4 VOL. 80, 2006 CARIBBEAN AGMs AS A MODEL FOR SIV INFECTION 4861 FIG. 2. Caribbean AGM speciation based on mitochondrial DNA characterization. Cytochrome b sequences (267 bp) were amplified from Caribbean AGMs and compared with those of different species of AGMs from Africa. Sequences originating from different species of AGMs of African origin are color coded: sabaeus in red, tantalus in blue, grivet in orange, and vervet in green. Sequences from Caribbean AGMs are shown in black. Monkeys coded EI are from St. Kitts island; monkeys coded A and B are from Barbados. Cytochrome b sequences from other species of African monkeys are shown in gray. The phylogenetic trees were estimated by the maximum parsimony method. The reliability was estimated from 1,000 bootstrap replicates; only bootstrap values relevant for lineage definition are shown.

5 4862 PANDREA ET AL. J. VIROL. FIG. 3. Expression of CD4 before (a) and after stimulation with PHA (b) and IL-2 (c). An increase of CD4-positive cells was noted after 3 days of PHA stimulation (b). After 5 additional days of treatment with IL-2, a decrease of CD4 T cells was observed (c), but this reduction was insignificant compared with CD4 levels before stimulation (a). animals (Fig. 3a). After 3 days of PHA stimulation, CD4 expression increased slightly (mean, 13.1% 2.4%; P 0.5) (Fig. 3b) and then decreased thereafter (mean, 9.4% 2.6%; P 0.5) after 3 additional days of IL-2 administration (Fig. 3c). The CD4 mean fluorescence intensity followed the same dynamics after the PHA treatment, with an increase to compared to the prestimulation level ( ) (P 0.5). After IL-2 stimulation, the CD4 mean fluorescence intensity remained slightly but not significantly higher than the pretreatment values ( ) (P 0.5). Thus, only minor modifications of CD4 expression could be observed in cell cultures of AGM PB- MCs, and no significant decrease was detected, in contrast with previous reports (34). Caribbean AGM PBMCs are susceptible to SIVagm.sab in vitro and in vivo. To study whether Caribbean AGMs are susceptible to SIVagm.sab, we first verified the susceptibility of AGM PBMCs to infection with the NIH reference strain SIVagm.sab2. We tested this strain for coreceptor usage and demonstrated that it was dual tropic (data not shown), similar to SIVagm.sab92018 (42). Two different phenotypes of AGMs were tested in order to exclude a possible restriction of the virus growth to monkeys with either phenotype: PBMCs from two Caribbean AGMs in which the majority of CD4 T cells were double positive for CD8, and two animals that lacked this particular phenotype. As a positive control, Rh PBMCs were infected with SIVmac251. Our experiment revealed productive replication of SIVagm.sab2 in PBMCs from Caribbean AGMs (mean P27 at day 7 postinoculation 1.73 ng/ml) without regard to CD4 phenotype. No significant differences in the P27 values could be observed between infection with SIVagm in Caribbean AGM PBMCs and SIVmac in Rh PBMCs ( ng/ml versus ng/ml), indicating no restriction of SIVagm.sab in the former. To test the in vivo susceptibility of Caribbean AGMs to SIVagm.sab infection, six animals were inoculated with SIVagm.sab Animals were again selected to display the different phenotypes CD4 CD8 and CD4 CD8 neg (four versus two) in order to determine if these cell subsets play a particular role in SIVagm pathogenesis in Caribbean AGMs, as has been suggested (20). All animals became infected and seroconverted between 21 and 45 days p.i. (Fig. 4a), similar to AGMs from Africa infected with the same SIVagm strain (9). None of the monkeys had clinical signs related to the SIV infection during the follow-up (up to day 420 p.i.). No weight loss, fever, opportunistic infections, or neoplasia was noted. One monkey died at 132 days postinfection due to causes unrelated to SIV infection. The P27 antigenemia levels in SIVagm.sab92018-infected Caribbean AGMs are similar to those observed in their African counterparts. To evaluate systemic viral replication, two viral parameters were measured: P27 antigenemia and plasma RNA VL. P27 antigenemia quantification showed that all animals were positive for P27 during primary infection at day 4 p.i. The P27 values peaked at up to 4.6 to 5.2 ng/ml of plasma between days 8 and 10 p.i. (Fig. 4b). These values are very high, and in the same range as those recorded for SIVagm.sab92018-infected AGMs from Africa (9, 14, 42) and for SIVmac-infected Rh (17, 28, 38, 53, 58). After primary infection, P27 antigenemia became undetectable in SIVagm.sab-infected Caribbean AGMs by day 28 p.i., similar to what was described for the asymptomatic phase of chronic SIVmac, HIV-1, SIVagm, SIVsm, and SIVmnd-1/2 infections (9, 17, 39, 40, 42, 43). No differences in P27 antigenemia could be detected between monkeys with high numbers of CD4 CD8 T cells and the animals that lacked this phenotype. These results demonstrate that SIVagm.sab replicates well in Caribbean AGMs. SIVagm.sab92018-infected Caribbean and African AGMs show the same range of plasma RNA VLs. The dynamics of plasma RNA VL of SIVagm.sab in Caribbean AGMs is shown in Fig. 5. Plasma viremia peaked between days 8 and 10 and the values ranged between and copies/ml. This is very similar to what has been described in 11 sabaeus monkeys from Senegal experimentally infected with the same SIVagm. sab92018 strain (42). Thus, plasma RNA quantification confirmed the high antigenemia levels detected in SIVagm.sab-infected Caribbean AGMs. No difference in plasma viremia was observed between animals infected with viral stocks of isolate produced in African or Caribbean AGMs. As reported earlier, these VL values are high and are comparable to levels previously described for pathogenic SIVmac infections (17, 28, 38, 52, 58). The set-point levels in Caribbean AGMs ranged between 10 4 and 10 5 copies/ml (Fig. 5). The plasma VLs during the

6 VOL. 80, 2006 CARIBBEAN AGMs AS A MODEL FOR SIV INFECTION 4863 FIG. 4. Antibody responses (a) and p27 antigenemia (b) in Caribbean AGMs infected with SIVagm.sab. Anti-gp41 antibodies against SIVagm.sab were detected by an in-house peptide ELISA using specific peptides (51); all animals seroconverted between days 21 and 45 p.i. Positive P27 antigenemia was detected only during the acute infection (up to day 28 p.i.). Symbols for animals: no. EI45 (F), no. EI53 (E), no. EI52 ( ), no. EI42 (ƒ), no. EI51 ( ), and no. EI44 ( ). postacute and chronic phases were in the same range as those we and others reported previously in experimentally or naturally SIVagm.sab-infected AGMs from Africa (5, 9, 12, 13, 19, 26, 42). Altogether, our results show that SIVagm.sab replicates at the same level and following the same pattern in Caribbean AGMs as in those from Africa (9, 14, 26, 42). Therefore, we concluded that Caribbean AGMs may be used as an alternative to AGMs from Africa for the study of immunological factors of protection against AIDS. SIVagm.sab-infected Caribbean AGMs showed transient CD4 T-cell declines in peripheral blood and LNs. The dynamics of CD3, CD4, and CD8 T cells and of CD20 B cells were measured in SIVagm.sab92018-infected Caribbean AGMs, in FIG. 5. Viral RNA copy numbers in plasma of SIVagm.sab infected Caribbean AGMs. The detection limit of the assay was 10 3 copies/ml. The range of plasma VLs in Caribbean AGMs is in the same range as values observed in African AGMs infected with the same viral strain (42). Symbols for animals: no. EI45 (F), no. EI53 (E), no. EI52 ( ), no. EI42 (ƒ), no. EI51 ( ), and no. EI44 ( ). blood and LNs (Fig. 6). Caribbean AGMs had a mean of 383 CD4 T cells/ l (range: 292 to 502 cells/ l) prior to SIVagm.sab infection. After inoculation, similar to SIVagm.sab-infected African sabaeus monkeys, Caribbean AGMs demonstrated a significant drop in CD4 T-cell counts, reaching the nadir at day 10 p.i. (P 0.01) (Fig. 6a). After the viral peak, the CD4 T-cell values rebounded close to preinfection values (reached by day 28 for most of the animals) and remained at relatively the same levels during chronic SIV infection. A transient decrease in CD4 T cells was also observed in the LNs that reached a significant degree at day 28 (P 0.03), similar to what has been previously reported in SIVmnd-1-infected mandrills (39). By day 200 the CD4 values were slightly (but not significantly) decreased compared with preinfection values (Fig. 7a). A small but insignificant decrease was also observed in peripheral CD8 T cells at the peak of the VL, but this was readily recovered thereafter (Fig. 6b). No significant changes were noted in the dynamics of CD8 T cells in the LNs of the SIVagm.sab-infected AGMs. Thus, the CD4 and CD8 T-cell dynamics in LNs was similar to those described in AGMs of African origin (9, 26, 42). The numbers of CD20 cells transiently decreased (P 0.05) in peripheral blood during primary infection, with the lowest values occurring at days 7 to 10 p.i (Fig. 6c). At day 21, however, numbers of CD20 cells returned to the preinfection values. During the chronic phase (between days 100 and 175), B lymphocytes transiently increased (P 0.05), and thereafter they returned to normal at around day 200. In the LNs, the percentage of CD20 cells increased temporarily at day 28 (P 0.03) and returned to normal by day 200, concomitant with CD4 T-cell normalization (Fig. 7b). The comparison of CD4 T-cell dynamics in blood and LNs of SIVagm.sab-infected Caribbean and African AGMs clearly shows similarities. In both species, similar to SIVmac251-infected Rh, we observed an early decline in the blood CD4 T cells. However, in contrast to the pathogenic SIV model, there is better preservation of the CD4 T cells in both Caribbean AGMs and those from Africa during the chronic phase of SIV infection.

7 4864 PANDREA ET AL. J. VIROL. CD8 T cells significantly increased at as early as day 3 p.i. and reached a peak at day 7 p.i. (15.19% 4.17%) (Fig. 8). By day 20 p.i., these cells returned to preinoculation values (3.9% 0.67% versus 2.8% 0.5%) (Fig. 8). No significant change was observed in peripheral blood for DR CD4 T cells in SIVinfected Caribbean AGMs (Fig. 8). This pattern of dynamics is very similar to that previously reported in sabaeus monkeys of African origin, in which T-cell activation occurs very early and only transiently during SIVagm.sab infection (26). DISCUSSION FIG. 6. T- and B-cell dynamics in blood of SIVagm.sab-infected Caribbean AGMs. CD4 T cells transiently decreased in the blood (a) of SIVagm.sab-infected AGMs at the peak of VLs. The levels rebounded to levels close to preinfection values during chronic SIVagm.sab infection. No significant modification was observed in the dynamics of CD8 T cells in the peripheral blood. (b) A transient decrease in the numbers of CD20 B cells was present in the blood of SIVagm.sab-infected AGMs (c). Symbols for animals: no. EI45 (F), no. EI53 (E), no. EI52 ( ), no. EI42 (ƒ), no. EI51 ( ), and no. EI44 ( ). Caribbean AGMs and AGMs from Africa respond to SIVsm infection with a similar pattern of T-cell activation. The dynamics of HLA-DR on CD4 and CD8 T cells was determined by flow cytometry in order to determine whether, similarly to AGMs from Africa, Caribbean-born AGMs infected with SIVagm show low levels of T-cell activation. SIVagm.sab92018 infection of Caribbean AGMs was associated with only a transitory increase in the activation status of CD8 T cells. DR In this study we show that AGMs from the Caribbean are C. sabaeus and that they apparently respond identically to SIVagm.sab92018 infection as do African-born AGMs. This is a significant result, since it offers alternatives and expands the avenues for pathogenesis studies in NHP hosts of SIVs. In these monkeys, SIVs generally do not induce disease, in spite of active viral replication (5, 6, 12, 13, 25, 26, 30, 39, 40, 42 44, 47, 49, 50). Therefore, the study of these models allowed the investigation of the reasons for reduced pathogenicity. Such models for pathogenesis studies are needed in order to identify factors associated with protection against lentiviral disease. However, extensive studies are hampered because most currently available animal models for SIV infection in natural hosts are either highly endangered species (such as SMs, mandrills, and chimpanzees) or difficult to obtain (AGMs from Africa). A Caribbean AGM model for SIV infection is therefore a significant new resource, due to the availability of these monkeys and to the fact that Caribbean AGM colonies are currently present in the United States. Caribbean AGMs are an appropriate model for studying SIVagm infection. Our data clearly show that Caribbean AGMs are susceptible to SIVagm.sab infection in vitro and in vivo. SIVagm.sab infection in Caribbean AGMs faithfully reproduces the viral parameters of SIVagm infection in AGMs from Africa. This contradicts anecdotal reports that Caribbean AGMs are resistant to SIV, which were based on several lines of evidence. (i) First, there is the absence of SIV infection in Caribbean AGM colonies despite repeated large-scale serological surveys (7). Our results support the hypothesis that the founder Caribbean AGMs were probably SIVagm negative when separated from AGMs in Africa and brought to the Caribbean islands, rather than being naturally resistant to SIV (8, 32). Their young age at the time of separation may be the key to their SIV negativity, as it has been demonstrated that AGMs become SIVagm positive at sexual maturity (45), and no vertical transmission of SIVagm has been described to date (41). (ii) Second, previous attempts to infect Caribbean monkeys with SIV resulted in significantly less efficient viremia compared with AGMs from Africa, and some of the animals failed to become persistently infected (13). The reason for these failures may be the use of SIVagm strains belonging to different subtypes. Prior studies used SIVagm.ver, which naturally infects a different AGM subspecies, the vervets (Chlorocebus pygerythrus). (iii) Finally, differences in CD4 and CD8 expression reported by Holznagel et al. suggested that a modification of the immune system of Caribbean AGMs may have occurred as the result of them being separated from their African relatives for 200 to 400 years (20). However, we did not

8 VOL. 80, 2006 CARIBBEAN AGMs AS A MODEL FOR SIV INFECTION 4865 FIG. 7. CD4 T-cell and B-cell dynamics in LNs in SIVagm.sab-infected Caribbean AGMs. A significant (P 0.05) decrease in CD4 T-cell percentages was observed in the LNs at day 28 post-sivagm.sab inoculation. The levels of CD4 T cells remained decreased at day 200, but the difference was not significant compared with preinfection values (a). Concomitant relative increase and return to normal levels of CD20 cells in the same compartment (b). Symbols for animals: no. EI45 (F), no. EI53 (E), no. EI52 ( ), no. EI42 (ƒ), no. EI51 ( ), and no. EI44 ( ). find any difference in CD4 T-cell phenotype regarding CD8 coexpression between Caribbean and African sabaeus monkeys. Monkeys harboring double-positive CD4/CD8 T cells were present in both populations. A possible explanation for the difference in results may rely on the use of inbred animals in the previous study (20). Also, in our study, only animals originating in St. Kitts were included. It is possible that monkeys born in Barbados might be more homogeneous in terms of CD4/CD8 expression. Regardless, no difference was found in the pattern of viral replication after SIVagm in vitro and in vivo infection of Caribbean AGMs harboring CD4 CD8 or CD4 CD8 neg, indicating that these phenotypes do not play a role in their susceptibility to infection. In order to match the virus with the host species, the species of our Caribbean AGMs was confirmed by genotyping. Based on cytochrome b typing, we demonstrated for the first time that Caribbean AGMs originating from St. Kitts cluster with sabaeus monkeys from Senegal. All Caribbean AGMs showed clear differences from three other species of AGMs, such as FIG. 8. Kinetic expression of percent HLA-DR upon CD4 (F) and CD8 (E) T lymphocytes in peripheral blood of SIVagm.sab infected Caribbean AGMs. A significant but transitory activation of CD8 T cells can be observed during the primary infection. No modification of the activation status was observed for the CD4 T cells in SIVagm.sabinfected Caribbean AGMs. vervet, tantalus, and grivet monkeys. Surprisingly, despite their putatively more diverse origin (8), Caribbean AGMs from Barbados showed a high homogeneity in their cytochrome b structure, which was identical with sabaeus species of AGM from St. Kitts and Senegal. Our study therefore genetically traced the origins of all Caribbean AGMs to West Africa (Senegal) (Fig. 1) and confirmed that the use of an SIVagm.sab strain is the best approach for SIV pathogenesis studies in this AGM species. Furthermore, there were no significant differences detected in the dynamics of blood and LN CD4 T cells between SIVagm.sab-infected Caribbean and African AGMs. Moreover, Caribbean AGMs and AGMs from Africa respond to SIVsm infection with similar patterns of T-cell activation, as showed by the dynamics of the HLA-DR on their T cells. Finally, no difference in the availability of target cells (CD4 CCR5 T cells) was observed between Caribbean and African AGMs (I. Pandrea et al., submitted for publication). A longer follow-up of the animals in our study group will be done in order to verify potential differences in viral replication, immune cell dynamics, and clinical outcomes that can occur later. However, due to the previously mentioned similarities between Caribbean-born and African-born AGMs, our study strongly supports the usefulness of the Caribbean model to study SIVagm pathogenesis. SIVagm.sab92018 as a reference strain for pathogenesis studies on AGM. This study was based on the hypothesis that SIVagm.sab92018 would replicate at high levels in Caribbean AGMs and therefore may be used as a tool to study the SIVagm infection in the natural host. In addition to the reasons above regarding matching the species of animal and the viral strain, the selection of SIVagm.sab92018 was prompted by the following rationales. (i) We previously used this strain to infect sabaeus and vervet African monkeys and showed that SIVagm.sab92018 replicates at similar levels in both AGM species, although at slightly lower levels in vervets. These two species separated millennia ago (32). Therefore, we hypothesized that Caribbean AGMs that diverged from their African relatives only 300 years ago would support high levels of viral replication when infected with SIVagm.sab. (ii) This strain was previously used to infect a large number of AGMs from Africa,

9 4866 PANDREA ET AL. J. VIROL. and we had sufficient data to compare the dynamics of SIVagm.sab92018 infection (9, 14, 26, 42, 46) in order to assess the efficacy of viral replication in Caribbean AGMs and validate our new model. (iii) All necessary experimental tools to investigate SIVagm.sab92018 VLs and CD4 T-cell dynamics had already been developed in our previous studies (9, 14, 26, 42, 46), and overlapping peptides mapping gag, env, and nef genes of SIVagm.sab92018 recently became available for cellular immunology studies through the NIH. (iv) Finally, SIVagm.sab92018 is a wild-type virus. All the other SIVagm.sab strains available through the NIH AIDS Research Reference Reagent Program are laboratory-adapted strains grown on human cell lines (1, 22). Most of these strains are either not infectious in vivo, have not yet been tested in vivo, or have a truncated gp41 (SIVagm.sab1c). Recently the use of another primary isolate of SIVagm.sab from the central nervous system was reported (SIVagm.sab4br), but the in vivo replication of this virus was lower than that reported for SIVagm.sab92018 (J. S. Allan, B. K. Wilson, R. G. White, L. M. Parodi, V. L. Hodara, and L. D. Giaverdoni, 23rd Annu. Symp. Nonhuman Primate Models AIDS, abstr. 13, 2005). Combined with the above rationales, our results on VLs in vitro and in vivo demonstrate that primary SIVagm.sab92018 is an appropriate strain for pathogenesis studies using Caribbean AGMs. In conclusion, by matching the Caribbean AGMs with their species-specific virus, we have developed a model for pathogenesis studies of SIV infection in natural African NHP hosts. This new model may be extensively employed to determine the factors associated with the low pathogenicity of these viruses and with the mechanisms of immune protection against disease progression. In the context of an expanding pandemic and of repeated failures of current vaccines to induce protective immune responses, our model, developed in a nonendangered species, is an invaluable tool for the study of host factors associated with the control of lentiviral infection. ACKNOWLEDGMENTS We have no conflicting financial interests. We thank O. Diop for the SIVagm.sab92018 (00008) virus stock preparation and M. J. Dodd, J. Leblanc, K. Rasmussen, J. Bruhn, E. Benes, and C. Lanclos for excellent technical help. This study was supported by grants R01 AI (I.P.), R01 AI49080 (R.V.), P20 RR (C.A.), and P51 RR (TNPRC) from the National Institutes of Health and by a grant from the Pasteur Institute (M.M.T. and F.B.S.). REFERENCES 1. Allan, J. S., M. Short, M. E. Taylor, S. Su, V. M. Hirsch, P. R. Johnson, G. M. Shaw, and B. H. Hahn Species-specific diversity among simian immunodeficiency viruses from African green monkeys. J. Virol. 65: Anderson, S., A. T. Bankier, B. G. Barrell, M. H. de Bruijn, A. R. Coulson, J. Drouin, I. C. Eperon, D. P. Nierlich, B. A. Roe, F. Sanger, P. H. Schreier, A. J. Smith, R. Staden, and I. G. Young Sequence and organization of the human mitochondrial genome. Nature 290: Apetrei, C., B. Gormus, I. Pandrea, M. Metzger, P. ten Haaft, L. N. Martin, R. Bohm, X. Alvarez, G. Koopman, M. Murphey-Corb, R. S. Veazey, A. A. Lackner, G. Baskin, J. Heeney, and P. A. Marx Direct inoculation of simian immunodeficiency virus from sooty mangabeys in black mangabeys (Lophocebus aterrimus): first evidence of AIDS in a heterologous African species and different pathologic outcomes of experimental infection. J. Virol. 78: Beer, B., J. Denner, C. R. Brown, S. Norley, J. zur Megede, C. Coulibaly, R. Plesker, S. Holzammer, M. Baier, V. M. Hirsch, and R. Kurth Simian immunodeficiency virus of African green monkeys is apathogenic in the newborn natural host. J. Acquir. Immune Defic. Syndr. Hum. Retrovirol. 18: Broussard, S. R., S. I. Staprans, R. White, E. M. Whitehead, M. B. Feinberg, and J. S. Allan Simian immunodeficiency virus replicates to high levels in naturally infected African green monkeys without inducing immunologic or neurologic disease. J. Virol. 75: Chakrabarti, L. A., S. R. Lewin, L. Zhang, A. Gettie, A. Luckay, L. N. Martin, E. Skulsky, D. D. Ho, C. Cheng-Mayer, and P. A. Marx Normal T-cell turnover in sooty mangabeys harboring active simian immunodeficiency virus infection. J. Virol. 74: Daniel, M. D., N. L. Letvin, P. K. Sehgal, D. K. Schmidt, D. P. Silva, K. R. Solomon, F. S. Hodi, Jr., D. J. Ringler, R. D. Hunt, N. W. King, et al Prevalence of antibodies to 3 retroviruses in a captive colony of macaque monkeys. Int. J. Cancer 41: Denham, W. W History of green monkeys in the West Indies. Part I. Migration from Africa. J. Barbados Museum Historical Soc. 36: Diop, O. M., A. Gueye, M. Dias-Tavares, C. Kornfeld, A. Faye, P. Ave, M. Huerre, S. Corbet, F. Barre-Sinoussi, and M. C. Muller-Trutwin High levels of viral replication during primary simian immunodeficiency virus SIVagm infection are rapidly and strongly controlled in African green monkeys. J. Virol. 74: Fomsgaard, A., J. Allan, M. Gravell, W. T. London, V. M. Hirsch, and P. R. Johnson Molecular characterization of simian lentiviruses from East African green monkeys. J. Med. Primatol. 19: Fukasawa, M., T. Miura, A. Hasegawa, S. Morikawa, H. Tsujimoto, K. Miki, T. Kitamura, and M. Hayami Sequence of simian immunodeficiency virus from African green monkey, a new member of the HIV/SIV group. Nature 333: Goldstein, S., I. Ourmanov, C. R. Brown, B. E. Beer, W. R. Elkins, R. Plishka, A. Buckler-White, and V. M. Hirsch Wide range of viral load in healthy African green monkeys naturally infected with simian immunodeficiency virus. J. Virol. 74: Goldstein, S., C. R. Brown, I. Ourmanov, I. Pandrea, A. Buckler-White, C. Erb, J. S. Nandi, G. J. Foster, P. Autissier, J. E. Schmitz, and V. M. Hirsch Comparison of simian immunodeficiency virus SIVagmVer replication and CD4 T-cell dynamics in vervet and sabaeus African green monkeys. J. Virol. 80: Gueye, A., O. M. Diop, M. J. Ploquin, C. Kornfeld, A. Faye, M. C. Cumont, B. Hurtrel, F. Barre-Sinoussi, and M. C. Muller-Trutwin Viral load in tissues during the early and chronic phase of non-pathogenic SIVagm infection. J. Med. Primatol. 33: Hasegawa, M., H. Kishino, and T. Yano Dating of the human-ape splitting by a molecular clock of mitochondrial DNA. J. Mol. Evol. 22: Hendry, R. M., M. A. Wells, M. A. Phelan, A. L. Schneider, J. S. Epstein, and G. V. Quinnan Antibodies to simian immunodeficiency virus in African green monkeys in Africa in Lancet ii: Hirsch, V. M., T. R. Fuerst, G. Sutter, M. W. Carroll, L. C. Yang, S. Goldstein, M. Piatak, Jr., W. R. Elkins, W. G. Alvord, D. C. Montefiori, B. Moss, and J. D. Lifson Patterns of viral replication correlate with outcome in simian immunodeficiency virus (SIV)-infected macaques: effect of prior immunization with a trivalent SIV vaccine in modified vaccinia virus Ankara. J. Virol. 70: Hirsch, V. M., C. McGann, G. Dapolito, S. Goldstein, A. Ogen-Odoi, B. Biryawaho, T. Lakwo, and P. R. Johnson Identification of a new subgroup of SIVagm in tantalus monkeys. Virology 197: Holzammer, S., E. Holznagel, A. Kaul, R. Kurth, and S. Norley High virus loads in naturally and experimentally SIVagm-infected African green monkeys. Virology 283: Holznagel, E., S. Norley, S. Holzammer, C. Coulibaly, and R. Kurth Immunological changes in simian immunodeficiency virus SIVagm-infected African green monkeys (AGM): expanded cytotoxic T lymphocyte, natural killer and B cell subsets in the natural host of SIVagm. J. Gen. Virol. 83: Irwin, D. M., T. D. Kocher, and A. C. Wilson Evolution of the cytochrome b gene of mammals. J. Mol. Evol. 32: Jin, M. J., H. Hui, D. L. Robertson, M. C. Muller, F. Barre-Sinoussi, V. M. Hirsch, J. S. Allan, G. M. Shaw, P. M. Sharp, and B. H. Hahn Mosaic genome structure of simian immunodeficiency virus from West African green monkeys. EMBO J. 13: Johnson, P. R., M. Gravell, J. Allan, S. Goldstein, R. A. Olmsted, G. Dapolito, C. McGann, W. T. London, R. H. Purcell, and V. M. Hirsch Genetic diversity among simian immunodeficiency virus isolates from African green monkeys. J. Med. Primatol. 18: Jolly, C., J. E. Phillips-Conroy, T. R. Turner, S. Broussard, and J. S. Allan SIVagm incidence over two decades in a natural population of Ethiopian grivet monkeys (Cercopithecus aethiops aethiops). J. Med. Primatol. 25: Kaur, A., R. M. Grant, R. E. Means, H. McClure, M. Feinberg, and R. P. Johnson Diverse host responses and outcomes following simian immunodeficiency virus SIVmac239 infection in sooty mangabeys and rhesus macaques. J. Virol. 72: Kornfeld, C., M. J. Ploquin, I. Pandrea, A. Faye, R. Onanga, C. Apetrei, V. Poaty-Mavoungou, P. Rouquet, J. Estaquier, L. Mortara, J. F. Desoutter, C. Butor, R. Le Grand, P. Roques, F. Simon, F. Barre-Sinoussi, O. M. Diop, and M. C. Muller-Trutwin Antiinflammatory profiles during primary

10 VOL. 80, 2006 CARIBBEAN AGMs AS A MODEL FOR SIV INFECTION 4867 SIV infection in African green monkeys are associated with protection against AIDS. J. Clin. Investig. 115: Lairmore, M. D., D. E. Hofheinz, N. L. Letvin, C. S. Stoner, S. Pearlman, and G. P. Toedter Detection of simian immunodeficiency virus and human immunodeficiency virus type 2 capsid antigens by a monoclonal antibody-based antigen capture assay. AIDS Res. Hum. Retrovir. 9: Lifson, J. D., M. A. Nowak, S. Goldstein, J. L. Rossio, A. Kinter, G. Vasquez, T. A. Wiltrout, C. Brown, D. Schneider, L. Wahl, A. L. Lloyd, J. Williams, W. R. Elkins, A. S. Fauci, and V. M. Hirsch The extent of early viral replication is a critical determinant of the natural history of simian immunodeficiency virus infection. J. Virol. 71: Ling, B., C. Apetrei, I. Pandrea, R. S. Veazey, A. A. Lackner, B. Gormus, and P. A. Marx Classic AIDS in a sooty mangabey after an 18-year natural infection. J. Virol. 78: Ling, B., M. L. Santiago, S. Meleth, B. Gormus, H. M. McClure, C. Apetrei, B. H. Hahn, and P. A. Marx Noninvasive detection of new simian immunodeficiency virus lineages in captive sooty mangabeys: ability to amplify virion RNA from fecal samples correlates with viral load in plasma. J. Virol. 77: McClure, H. M., D. C. Anderson, T. P. Gordon, A. A. Ansari, P. N. Fultz, S. A. Klumpp, P. Emau, and M. Isahakia Natural simian immunodeficiency virus infection in nonhuman primates. Top. Primatol. 3: Muller, M. C., and F. Barre-Sinoussi SIVagm: genetic and biological features associated with replication. Front. Biosci. 8:d1170 d Muller, M. C., N. K. Saksena, E. Nerrienet, C. Chappey, V. M. Herve, J. P. Durand, P. Legal-Campodonico, M. C. Lang, J. P. Digoutte, A. J. Georges, et al Simian immunodeficiency viruses from central and western Africa: evidence for a new species-specific lentivirus in tantalus monkeys. J. Virol. 67: Murayama, Y., A. Amano, R. Mukai, H. Shibata, S. Matsunaga, H. Takahashi, Y. Yoshikawa, M. Hayami, and A. Noguchi CD4 and CD8 expressions in African green monkey helper T lymphocytes: implication for resistance to SIV infection. Int. Immunol. 9: National Research Council Guide for the care and use of laboratory animals. National Academy Press, Washington, D.C. 36. Norley, S., and R. Kurth The role of the immune response during SIVagm infection of the African green monkey natural host. Front. Biosci. 9: Norley, S. G., G. Kraus, J. Ennen, J. Bonilla, H. Konig, and R. Kurth Immunological studies of the basis for the apathogenicity of simian immunodeficiency virus from African green monkeys. Proc. Natl. Acad. Sci. USA 87: Nowak, M. A., A. L. Lloyd, G. M. Vasquez, T. A. Wiltrout, L. M. Wahl, N. Bischofberger, J. Williams, A. Kinter, A. S. Fauci, V. M. Hirsch, and J. D. Lifson Viral dynamics of primary viremia and antiretroviral therapy in simian immunodeficiency virus infection. J. Virol. 71: Onanga, R., C. Kornfeld, I. Pandrea, J. Estaquier, S. Souquiere, P. Rouquet, V. P. Mavoungou, O. Bourry, S. M Boup, F. Barre-Sinoussi, F. Simon, C. Apetrei, P. Roques, and M. C. Muller-Trutwin High levels of viral replication contrast with only transient changes in CD4 and CD8 cell numbers during the early phase of experimental infection with simian immunodeficiency virus SIVmnd-1 in Mandrillus sphinx. J. Virol. 76: Onanga, R., S. Souquiere, M. Makuwa, A. Mouinga-Ondeme, F. Simon, C. Apetrei, and P. Roques Primary simian immunodeficiency virus SIVmnd-2 infection in mandrills (Mandrillus sphinx). J. Virol. 80: Otsyula, M. G., A. Gettie, M. Suleman, R. Tarara, I. Mohamed, and P. Marx Apparent lack of vertical transmission of simian immunodeficiency virus (SIV) in naturally infected African green monkeys, Cercopithecus aethiops. Ann. Trop. Med. Parasitol. 89: Pandrea, I., C. Kornfeld, M. J.-Y. Ploquin, C. Apetrei, A. Faye, P. Rouquet, P. Roques, F. Simon, F. Barré-Sinoussi, M. C. Müller-Trutwin, and O. M. Diop Impact of viral factors on very early in vivo replication profiles in simian immunodeficiency virus SIVagm-infected African green monkeys. J. Virol. 79: Pandrea, I., R. Onanga, C. Kornfeld, P. Rouquet, O. Bourry, S. Clifford, P. T. Telfer, K. Abernethy, L. T. White, P. Ngari, M. Muller-Trutwin, P. Roques, P. A. Marx, F. Simon, and C. Apetrei High levels of SIVmnd-1 replication in chronically infected Mandrillus sphinx. Virology 317: Pandrea, I., R. Onanga, P. Rouquet, O. Bourry, P. Ngari, E. J. Wickings, P. Roques, and C. Apetrei Chronic SIV infection ultimately causes immunodeficiency in African non-human primates. AIDS 15: Phillips-Conroy, J. E., C. J. Jolly, B. Petros, J. S. Allan, and R. C. Desrosiers Sexual transmission of SIVagm in wild grivet monkeys. J. Med. Primatol. 23: Ploquin, M. J., O. M. Diop, N. Sol-Foulon, L. Mortara, A. Faye, M. A. Soares, E. Nerrienet, R. Le Grand, Y. Van Kooyk, A. Amara, O. Schwartz, F. Barre-Sinoussi, and M. C. Muller-Trutwin DC-SIGN from African green monkeys is expressed in lymph nodes and mediates infection in trans of simian immunodeficiency virus SIVagm. J. Virol. 78: Rey-Cuille, M. A., J. L. Berthier, M. C. Bomsel-Demontoy, Y. Chaduc, L. Montagnier, A. G. Hovanessian, and L. A. Chakrabarti Simian immunodeficiency virus replicates to high levels in sooty mangabeys without inducing disease. J. Virol. 72: Saitou, N., and M. Nei The neighbor-joining method: a new method for reconstructing phylogenetic trees. Mol. Biol. Evol. 4: Silvestri, G., A. Fedanov, S. Germon, N. Kozyr, W. J. Kaiser, D. A. Garber, H. McClure, M. B. Feinberg, and S. I. Staprans Divergent host responses during primary simian immunodeficiency virus SIVsm infection of natural sooty mangabey and nonnatural rhesus macaque hosts. J. Virol. 79: Silvestri, G., D. L. Sodora, R. A. Koup, M. Paiardini, S. P. O Neil, H. M. McClure, S. I. Staprans, and M. B. Feinberg Nonpathogenic SIV infection of sooty mangabeys is characterized by limited bystander immunopathology despite chronic high-level viremia. Immunity 18: Simon, F., S. Souquiere, F. Damond, A. Kfutwah, M. Makuwa, E. Leroy, P. Rouquet, J. L. Berthier, J. Rigoulet, A. Lecu, P. T. Telfer, I. Pandrea, J. C. Plantier, F. Barre-Sinoussi, P. Roques, M. C. Muller-Trutwin, and C. Apetrei Synthetic peptide strategy for the detection of and discrimination among highly divergent primate lentiviruses. AIDS Res. Hum. Retrovir. 17: Smith, S. M., B. Holland, C. Russo, P. J. Dailey, P. A. Marx, and R. I. Connor Retrospective analysis of viral load and SIV antibody responses in rhesus macaques infected with pathogenic SIV: predictive value for disease progression. AIDS Res. Hum. Retrovir. 15: Staprans, S. I., P. J. Dailey, A. Rosenthal, C. Horton, R. M. Grant, N. Lerche, and M. B. Feinberg Simian immunodeficiency virus disease course is predicted by the extent of virus replication during primary infection. J. Virol. 73: Swofford, D PAUP* v 4: phylogenetic analysis using parsimony (*and other methods). Sinauer Associates, Sunderland, Massachusetts. 55. Traina-Dorge, V., J. Blanchard, L. Martin, and M. Murphey-Corb Immunodeficiency and lymphoproliferative disease in an African green monkey dually infected with SIV and STLV-I. AIDS Res. Hum. Retrovir. 8: Veazey, R. S., R. J. Shattock, M. Pope, J. C. Kirijan, J. Jones, Q. Hu, T. Ketas, P. A. Marx, P. J. Klasse, D. R. Burton, and J. P. Moore Prevention of virus transmission to macaque monkeys by a vaginally applied monoclonal antibody to HIV-1 gp120. Nat. Med. 9: Veazey, R. S., I. C. Tham, K. G. Mansfield, M. DeMaria, A. E. Forand, D. E. Shvetz, L. V. Chalifoux, P. K. Sehgal, and A. A. Lackner Identifying the target cell in primary simian immunodeficiency virus (SIV) infection: highly activated memory CD4 T cells are rapidly eliminated in early SIV infection in vivo. J. Virol. 74: Watson, A., J. Ranchalis, B. Travis, J. McClure, W. Sutton, P. R. Johnson, S. L. Hu, and N. L. Haigwood Plasma viremia in macaques infected with simian immunodeficiency virus: plasma viral load early in infection predicts survival. J. Virol. 71:

Early Divergence in Lymphoid Tissue Apoptosis between Pathogenic and Nonpathogenic Simian Immunodeficiency Virus Infections of Nonhuman Primates

Early Divergence in Lymphoid Tissue Apoptosis between Pathogenic and Nonpathogenic Simian Immunodeficiency Virus Infections of Nonhuman Primates JOURNAL OF VIROLOGY, Feb. 2008, p. 1175 1184 Vol. 82, No. 3 0022-538X/08/$08.00 0 doi:10.1128/jvi.00450-07 Copyright 2008, American Society for Microbiology. All Rights Reserved. Early Divergence in Lymphoid

More information

Classic AIDS in a Sooty Mangabey after an 18-Year Natural Infection

Classic AIDS in a Sooty Mangabey after an 18-Year Natural Infection JOURNAL OF VIROLOGY, Aug. 2004, p. 8902 8908 Vol. 78, No. 16 0022-538X/04/$08.00 0 DOI: 10.1128/JVI.78.16.8902 8908.2004 Copyright 2004, American Society for Microbiology. All Rights Reserved. Classic

More information

ACCEPTED. Unité de Physiopathologie des Infections Lentivirales, Institut Pasteur, Paris, France Institut Pasteur, Dakar, Sénégal

ACCEPTED. Unité de Physiopathologie des Infections Lentivirales, Institut Pasteur, Paris, France Institut Pasteur, Dakar, Sénégal JVI Accepts, published online ahead of print on 21 November 2007 J. Virol. doi:10.1128/jvi.00450-07 Copyright 2007, American Society for Microbiology and/or the Listed Authors/Institutions. All Rights

More information

INTRODUCTION. Gabon. Gabon

INTRODUCTION. Gabon. Gabon Journal of General Virology (2009), 90, 488 499 DOI 10.1099/vir.0.005181-0 Simian immunodeficiency virus types 1 and 2 (SIV mnd 1 and 2) have different pathogenic potentials in rhesus macaques upon experimental

More information

Primate models of SIV infection: Differenct susceptibility of African green monkeys and macaques to develop AIDS

Primate models of SIV infection: Differenct susceptibility of African green monkeys and macaques to develop AIDS Les Pensières Animal Models, 2011 Primate models of SIV infection: Differenct susceptibility of African green monkeys and macaques to develop AIDS Michaela Müller-Trutwin Unité de Régulation des Infections

More information

Received 19 March 2002/Accepted 16 July 2002

Received 19 March 2002/Accepted 16 July 2002 JOURNAL OF VIROLOGY, Oct. 2002, p. 10256 10263 Vol. 76, No. 20 0022-538X/02/$04.00 0 DOI: 10.1128/JVI.76.20.10256 10263.2002 Copyright 2002, American Society for Microbiology. All Rights Reserved. High

More information

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

Where the Wild Things Are: Pathogenesis of SIV Infection in African Nonhuman Primate Hosts Curr HIV/AIDS Rep (2010) 7:28 36 DOI 10.1007/s11904-009-0034-8 Where the Wild Things Are: Pathogenesis of SIV Infection in African Nonhuman Primate Hosts Ivona Pandrea & Cristian Apetrei Published online:

More information

Running head: Infectious transmitted/founder clone of SIVagm

Running head: Infectious transmitted/founder clone of SIVagm JVI Accepts, published online ahead of print on September 0 J. Virol. doi:.1/jvi.0- Copyright 0, American Society for Microbiology and/or the Listed Authors/Institutions. All Rights Reserved. 1 1 1 1 1

More information

Virus Subtype-Specific Features of Natural Simian Immunodeficiency Virus SIV smm Infection in Sooty Mangabeys

Virus Subtype-Specific Features of Natural Simian Immunodeficiency Virus SIV smm Infection in Sooty Mangabeys JOURNAL OF VIROLOGY, Aug. 2007, p. 7913 7923 Vol. 81, No. 15 0022-538X/07/$08.00 0 doi:10.1128/jvi.00281-07 Copyright 2007, American Society for Microbiology. All Rights Reserved. Virus Subtype-Specific

More information

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

Mother-to-Infant Transmission of Simian Immunodeficiency Virus Is Rare in Sooty Mangabeys and Is Associated with Low Viremia JOURNAL OF VIROLOGY, June 2011, p. 5757 5763 Vol. 85, No. 12 0022-538X/11/$12.00 doi:10.1128/jvi.02690-10 Copyright 2011, American Society for Microbiology. All Rights Reserved. Mother-to-Infant Transmission

More information

Risks and Management of SIV in Mandrils (Mandrillus sphinx) and Drills (Mandrillus leucophaeus)

Risks and Management of SIV in Mandrils (Mandrillus sphinx) and Drills (Mandrillus leucophaeus) Risks and Management of SIV in Mandrils (Mandrillus sphinx) and Drills (Mandrillus leucophaeus) A. Background Simian Immunodeficiency Viruses (SIV) are primate lentiviruses, which infect a wide variety

More information

Wide Range of Viral Load in Healthy African Green Monkeys Naturally Infected with Simian Immunodeficiency Virus

Wide Range of Viral Load in Healthy African Green Monkeys Naturally Infected with Simian Immunodeficiency Virus JOURNAL OF VIROLOGY, Dec. 2000, p. 11744 11753 Vol. 74, No. 24 0022-538X/00/$04.00 0 Wide Range of Viral Load in Healthy African Green Monkeys Naturally Infected with Simian Immunodeficiency Virus SIMOY

More information

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

Treatment with IL-7 Prevents the Decline of Circulating CD4 + T Cells during the Acute Phase of SIV Infection in Rhesus Macaques SUPPORTING INFORMATION FOR: Treatment with IL-7 Prevents the Decline of Circulating CD4 + T Cells during the Acute Phase of SIV Infection in Rhesus Macaques Lia Vassena, 1,2 Huiyi Miao, 1 Raffaello Cimbro,

More information

Functional Cure of SIVagm Infection in Rhesus Macaques Results in Complete Recovery of CD4 + T Cells and Is Reverted by CD8 + Cell Depletion

Functional Cure of SIVagm Infection in Rhesus Macaques Results in Complete Recovery of CD4 + T Cells and Is Reverted by CD8 + Cell Depletion Functional Cure of SIVagm Infection in Rhesus Macaques Results in Complete Recovery of CD4 + T Cells and Is Reverted by CD8 + Cell Depletion Ivona Pandrea 1,2,3, Thaidra Gaufin 4, Rajeev Gautam 4, Jan

More information

Micropathology Ltd. University of Warwick Science Park, Venture Centre, Sir William Lyons Road, Coventry CV4 7EZ

Micropathology Ltd. University of Warwick Science Park, Venture Centre, Sir William Lyons Road, Coventry CV4 7EZ www.micropathology.com info@micropathology.com Micropathology Ltd Tel 24hrs: +44 (0) 24-76 323222 Fax / Ans: +44 (0) 24-76 - 323333 University of Warwick Science Park, Venture Centre, Sir William Lyons

More information

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

SUPPLEMENTARY INFORMATION. Divergent TLR7/9 signaling and type I interferon production distinguish SUPPLEMENTARY INFOATION Divergent TLR7/9 signaling and type I interferon production distinguish pathogenic and non-pathogenic AIDS-virus infections Judith N. Mandl, Ashley P. Barry, Thomas H. Vanderford,

More information

HIV-1 p24 ELISA Pair Set Cat#: orb54951 (ELISA Manual)

HIV-1 p24 ELISA Pair Set Cat#: orb54951 (ELISA Manual) HIV-1 p24 ELISA Pair Set Cat#: orb54951 (ELISA Manual) BACKGROUND Human Immunodeficiency Virus ( HIV ) can be divided into two major types, HIV type 1 (HIV-1) and HIV type 2 (HIV-2). HIV-1 is related to

More information

An Evolutionary Story about HIV

An Evolutionary Story about HIV An Evolutionary Story about HIV Charles Goodnight University of Vermont Based on Freeman and Herron Evolutionary Analysis The Aids Epidemic HIV has infected 60 million people. 1/3 have died so far Worst

More information

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

Fayth K. Yoshimura, Ph.D. September 7, of 7 HIV - BASIC PROPERTIES 1 of 7 I. Viral Origin. A. Retrovirus - animal lentiviruses. HIV - BASIC PROPERTIES 1. HIV is a member of the Retrovirus family and more specifically it is a member of the Lentivirus genus of this family.

More information

Received 4 December 2001/Accepted 29 April 2002

Received 4 December 2001/Accepted 29 April 2002 JOURNAL OF VIROLOGY, Aug. 2002, p. 8433 8445 Vol. 76, No. 16 0022-538X/02/$04.00 0 DOI: 10.1128/JVI.76.16.8433 8445.2002 Copyright 2002, American Society for Microbiology. All Rights Reserved. The Relationship

More information

Simian immunodeficiency viruses (SIVs) from eastern and southern Africa: detection of a SIVagm variant from a chacma baboon

Simian immunodeficiency viruses (SIVs) from eastern and southern Africa: detection of a SIVagm variant from a chacma baboon Journal of General Virology (1998), 79, 1809 1814. Printed in Great Britain.... SHORT COMMUNICATION Simian immunodeficiency viruses (SIVs) from eastern and southern Africa: detection of a SIVagm variant

More information

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

Severe Depletion of Mucosal CD4 + T Cells in AIDS-Free Simian Immunodeficiency Virus-Infected Sooty Mangabeys This information is current as of September 1, 2018. References Subscription Permissions Email Alerts Severe Depletion of Mucosal CD4 + T Cells in AIDS-Free Simian Immunodeficiency Virus-Infected Sooty

More information

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

Micro 301 HIV/AIDS. Since its discovery 31 years ago 12/3/ Acquired Immunodeficiency Syndrome (AIDS) has killed >32 million people Micro 301 HIV/AIDS Shiu-Lok Hu hus@uw.edu December 3, 2012 Since its discovery 31 years ago Acquired Immunodeficiency Syndrome (AIDS) has killed >32 million people In 2011 34.0 million [31.4 35.9 million]

More information

Risks and Management of SIV in De Brazza Guenon (Cercopithecus neglectus)

Risks and Management of SIV in De Brazza Guenon (Cercopithecus neglectus) Risks and Management of SIV in De Brazza Guenon (Cercopithecus neglectus) NB: This paper is produced specifically for SIV in De Brazza Monkeys. There are significant species differences and therefore the

More information

Received 1 August 2002/Accepted 22 October 2002

Received 1 August 2002/Accepted 22 October 2002 JOURNAL OF VIROLOGY, Feb. 2003, p. 2214 2226 Vol. 77, No. 3 0022-538X/03/$08.00 0 DOI: 10.1128/JVI.77.3.2214 2226.2003 Copyright 2003, American Society for Microbiology. All Rights Reserved. Noninvasive

More information

Over thousands of years, species-specific

Over thousands of years, species-specific T1 Natural SIV Hosts: Showing AIDS the Door Ann Chahroudi, 1,2 Steven E. Bosinger, 2 Thomas H. Vanderford, 2 Mirko Paiardini, 2,3 * Guido Silvestri 2,3 * Many species of African nonhuman primates are naturally

More information

reproduced or photocopying

reproduced or photocopying AIDS Reviews 2004;6:40-53 What Can Natural Infection of African Monkeys with Simian Immunodeficiency Virus Tell us About the Pathogenesis of AIDS? Vanessa M. Hirsch Laboratory of Molecular Microbiology,

More information

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

Application of μmacs Streptavidin MicroBeads for the analysis of HIV-1 directly from patient plasma Excerpt from MACS&more Vol 8 1/2004 Application of μmacs Streptavidin MicroBeads for the analysis of HIV-1 directly from patient plasma L. Davis Lupo and Salvatore T. Butera HIV and Retrovirology Branch,

More information

Chronic inflammation and immune activation in human immunodeficiency

Chronic inflammation and immune activation in human immunodeficiency Plasmacytoid Dendritic Cell Infection and Sensing Capacity during Pathogenic and Nonpathogenic Simian Immunodeficiency Virus Infection Simon P. Jochems, a,b Beatrice Jacquelin, a Lise Chauveau, c Nicolas

More information

CD4-Like Immunological Function by CD4 T Cells in Multiple Natural Hosts of Simian Immunodeficiency Virus

CD4-Like Immunological Function by CD4 T Cells in Multiple Natural Hosts of Simian Immunodeficiency Virus JOURNAL OF VIROLOGY, Sept. 2011, p. 8702 8708 Vol. 85, No. 17 0022-538X/11/$12.00 doi:10.1128/jvi.00332-11 Copyright 2011, American Society for Microbiology. All Rights Reserved. CD4-Like Immunological

More information

Human Immunodeficiency Virus type 1 (HIV-1) p24 / Capsid Protein p24 ELISA Pair Set

Human Immunodeficiency Virus type 1 (HIV-1) p24 / Capsid Protein p24 ELISA Pair Set Human Immunodeficiency Virus type 1 (HIV-1) p24 / Capsid Protein p24 ELISA Pair Set Catalog Number : SEK11695 To achieve the best assay results, this manual must be read carefully before using this product

More information

GOVX-B11: A Clade B HIV Vaccine for the Developed World

GOVX-B11: A Clade B HIV Vaccine for the Developed World GeoVax Labs, Inc. 19 Lake Park Drive Suite 3 Atlanta, GA 3 (678) 384-72 GOVX-B11: A Clade B HIV Vaccine for the Developed World Executive summary: GOVX-B11 is a Clade B HIV vaccine targeted for use in

More information

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

X/01/$ DOI: /JVI Copyright 2001, American Society for Microbiology. All Rights Reserved. JOURNAL OF VIROLOGY, Apr. 2001, p. 3753 3765 Vol. 75, No. 8 0022-538X/01/$04.00 0 DOI: 10.1128/JVI.75.8.3753 3765.2001 Copyright 2001, American Society for Microbiology. All Rights Reserved. Route of Simian

More information

Molecular Epidemiology of Simian Immunodeficiency Virus SIVsm in U.S. Primate Centers Unravels the Origin of SIVmac and SIVstm

Molecular Epidemiology of Simian Immunodeficiency Virus SIVsm in U.S. Primate Centers Unravels the Origin of SIVmac and SIVstm JOURNAL OF VIROLOGY, July 2005, p. 8991 9005 Vol. 79, No. 14 0022-538X/05/$08.00 0 doi:10.1128/jvi.79.14.8991 9005.2005 Copyright 2005, American Society for Microbiology. All Rights Reserved. Molecular

More information

DC-SIGN from African Green Monkeys Is Expressed in Lymph Nodes and Mediates Infection in trans of Simian Immunodeficiency Virus SIVagm

DC-SIGN from African Green Monkeys Is Expressed in Lymph Nodes and Mediates Infection in trans of Simian Immunodeficiency Virus SIVagm JOURNAL OF VIROLOGY, Jan. 2004, p. 798 810 Vol. 78, No. 2 0022-538X/04/$08.00 0 DOI: 10.1128/JVI.78.2.798 810.2004 Copyright 2004, American Society for Microbiology. All Rights Reserved. DC-SIGN from African

More information

Received 7 November 2007/Accepted 15 January 2008

Received 7 November 2007/Accepted 15 January 2008 JOURNAL OF VIROLOGY, Apr. 2008, p. 3725 3735 Vol. 82, No. 7 0022-538X/08/$08.00 0 doi:10.1128/jvi.02408-07 Copyright 2008, American Society for Microbiology. All Rights Reserved. Short-Lived Infected Cells

More information

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

Low immune activation despite high levels of pathogenic HIV-1 results in long-term asymptomatic disease Low immune activation despite high levels of pathogenic HIV-1 results in long-term asymptomatic disease Shailesh K. Choudhary 1 *, Nienke Vrisekoop 2 *, Christine A. Jansen 2, Sigrid A. Otto 2, Hanneke

More information

The use of nonhuman primates in biomedical research has led to the isolation of many

The use of nonhuman primates in biomedical research has led to the isolation of many JVI Accepts, published online ahead of print on 29 September 2010 J. Virol. doi:10.1128/jvi.01928-10 Copyright 2010, American Society for Microbiology and/or the Listed Authors/Institutions. All Rights

More information

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

VIRAL TITER COUNTS. The best methods of measuring infectious lentiviral titer VIRAL TITER COUNTS The best methods of measuring infectious lentiviral titer FLUORESCENCE CYCLES qpcr of Viral RNA SUMMARY Viral vectors are now routinely used for gene transduction in a wide variety of

More information

Supporting Information

Supporting Information Supporting Information Sui et al..7/pnas.997 Pre-CLP CM9 LA9 SL Tat# Pol Vif % Tetramer + CD + CD + Vac+IL- +IL- Vac Fig. S. Frequencies of six different CD + CD + Mamu-A*-tetramer + cells were measured

More information

JOURNAL OF VIROLOGY, Feb. 1999, p Vol. 73, No. 2. Copyright 1999, American Society for Microbiology. All Rights Reserved.

JOURNAL OF VIROLOGY, Feb. 1999, p Vol. 73, No. 2. Copyright 1999, American Society for Microbiology. All Rights Reserved. JOURNAL OF VIROLOGY, Feb. 1999, p. 1036 1045 Vol. 73, No. 2 0022-538X/99/$04.00 0 Copyright 1999, American Society for Microbiology. All Rights Reserved. Characterization of a Novel Simian Immunodeficiency

More information

Edinburgh Research Explorer

Edinburgh Research Explorer Edinburgh Research Explorer Characterization of a novel simian immunodeficiency virus (SIV) from L'Hoest monkeys (Cercopithecus l'hoesti) Citation for published version: Hirsch, VM, Campbell, BJ, Bailes,

More information

Human Immunodeficiency Virus. Acquired Immune Deficiency Syndrome AIDS

Human Immunodeficiency Virus. Acquired Immune Deficiency Syndrome AIDS Human Immunodeficiency Virus Acquired Immune Deficiency Syndrome AIDS Sudden outbreak in USA of opportunistic infections and cancers in young men in 1981 Pneumocystis carinii pneumonia (PCP), Kaposi s

More information

Received 8 October 1997/Accepted 5 January 1998

Received 8 October 1997/Accepted 5 January 1998 JOURNAL OF VIROLOGY, Apr. 1998, p. 3248 3258 Vol. 72, No. 4 0022-538X/98/$04.00 0 Copyright 1998, American Society for Microbiology In Vivo Replication Capacity Rather Than In Vitro Macrophage Tropism

More information

Understanding HIV. Transmitted/Founder Viruses. Brandon Keele SAIC-Frederick National Cancer Institute

Understanding HIV. Transmitted/Founder Viruses. Brandon Keele SAIC-Frederick National Cancer Institute Understanding HIV Transmission Utilizing Transmitted/Founder Viruses Brandon Keele SAIC-Frederick National Cancer Institute AIDS Vaccine 2011 15 September 2011 Overview Several years ago, the CHAVI sought

More information

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

Massive infection and loss of memory CD4 + T cells in multiple tissues during acute SIV infection Massive infection and loss of memory CD4 + T cells in multiple tissues during acute SIV infection Joseph J. Mattapallil 1, Daniel C. Douek 2, Brenna Hill 2, Yoshiaki Nishimura 3, Malcolm Martin 3 & Mario

More information

Natural Infection of a Household Pet Red-Capped Mangabey (Cercocebus torquatus torquatus) with a New Simian Immunodeficiency Virus

Natural Infection of a Household Pet Red-Capped Mangabey (Cercocebus torquatus torquatus) with a New Simian Immunodeficiency Virus JOURNAL OF VIROLOGY, Jan. 1998, p. 600 608 Vol. 72, No. 1 0022-538X/98/$04.00 0 Copyright 1998, American Society for Microbiology Natural Infection of a Household Pet Red-Capped Mangabey (Cercocebus torquatus

More information

Experimental SIV infection of rhesus macaques (RMs),

Experimental SIV infection of rhesus macaques (RMs), Depletion of CD8 Cells in Sooty Mangabey Monkeys Naturally Infected with Simian Immunodeficiency Virus Reveals Limited Role for Immune Control of Virus Replication in a Natural Host Species 1 Ashley P.

More information

Lentiviruses: HIV-1 Pathogenesis

Lentiviruses: HIV-1 Pathogenesis Lentiviruses: HIV-1 Pathogenesis Human Immunodeficiency Virus, HIV, computer graphic by Russell Kightley Tsafi Pe ery, Ph.D. Departments of Medicine and Biochemistry & Molecular Biology NJMS, UMDNJ. e-mail:

More information

Divergent Host Responses during Primary Simian Immunodeficiency Virus SIVsm Infection of Natural Sooty Mangabey and Nonnatural Rhesus Macaque Hosts

Divergent Host Responses during Primary Simian Immunodeficiency Virus SIVsm Infection of Natural Sooty Mangabey and Nonnatural Rhesus Macaque Hosts JOURNAL OF VIROLOGY, Apr. 2005, p. 4043 4054 Vol. 79, No. 7 0022-538X/05/$08.00 0 doi:10.1128/jvi.79.7.4043 4054.2005 Copyright 2005, American Society for Microbiology. All Rights Reserved. Divergent Host

More information

Human Immunodeficiency Virus

Human Immunodeficiency Virus Human Immunodeficiency Virus Virion Genome Genes and proteins Viruses and hosts Diseases Distinctive characteristics Viruses and hosts Lentivirus from Latin lentis (slow), for slow progression of disease

More information

Dynamics of T- and B-Lymphocyte Turnover in a Natural Host of Simian Immunodeficiency Virus

Dynamics of T- and B-Lymphocyte Turnover in a Natural Host of Simian Immunodeficiency Virus JOURNAL OF VIROLOGY, Feb. 2008, p. 1084 1093 Vol. 82, No. 3 0022-538X/08/$08.00 0 doi:10.1128/jvi.02197-07 Copyright 2008, American Society for Microbiology. All Rights Reserved. Dynamics of T- and B-Lymphocyte

More information

Supporting Information

Supporting Information Supporting Information Hatziioannou et al. 10.1073/pnas.0812587106 SI Text Viral Constructs and Viral Stock Generation. To generate the HIV-1 constructs used throughout these studies, the env gene in an

More information

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

David Verhoeven, Sumathi Sankaran, Melanie Silvey, and Satya Dandekar* JOURNAL OF VIROLOGY, Apr. 2008, p. 4016 4027 Vol. 82, No. 8 0022-538X/08/$08.00 0 doi:10.1128/jvi.02164-07 Copyright 2008, American Society for Microbiology. All Rights Reserved. Antiviral Therapy during

More information

HIV Update in Laboratory Testing. Patricia Slev, PhD, D(ABCC)

HIV Update in Laboratory Testing. Patricia Slev, PhD, D(ABCC) HIV Update in Laboratory Testing Patricia Slev, PhD, D(ABCC) Objectives Explain the advances in HIV diagnostics, including fourth generation Ag/Ab combination HIV screening assays Describe the new CDC

More information

Genome and other Sequence Information from Primate Models of HIV Infection

Genome and other Sequence Information from Primate Models of HIV Infection V12242009 Genome and other Sequence Information from Primate Models of HIV Infection Jeffrey Rogers 1, Carlos Bustamente 2, Richard, Gibbs 1, R. Paul Johnson 3, Welkin Johnson 3, Devin Locke 4, Keith Mansfield

More information

HIV: A DISCOVERY OPENING THE ROAD TO NOVEL SCIENTIFIC KNOWLEDGE AND GLOBAL HEALTH IMPROVEMENT

HIV: A DISCOVERY OPENING THE ROAD TO NOVEL SCIENTIFIC KNOWLEDGE AND GLOBAL HEALTH IMPROVEMENT HIV: A DISCOVERY OPENING THE ROAD TO NOVEL SCIENTIFIC KNOWLEDGE AND GLOBAL HEALTH IMPROVEMENT Nobel Lecture, December 7, 2008 by Françoise Barré-Sinoussi Institut Pasteur, Unit of Regulation of Retroviral

More information

Received 29 August 2002/Accepted 3 December 2002

Received 29 August 2002/Accepted 3 December 2002 JOURNAL OF VIROLOGY, Mar. 2003, p. 3099 3118 Vol. 77, No. 5 0022-538X/03/$08.00 0 DOI: 10.1128/JVI.77.5.3099 3118.2003 Copyright 2003, American Society for Microbiology. All Rights Reserved. Simian-Human

More information

HIV/AIDS. Biology of HIV. Research Feature. Related Links. See Also

HIV/AIDS. Biology of HIV. Research Feature. Related Links. See Also 6/1/2011 Biology of HIV Biology of HIV HIV belongs to a class of viruses known as retroviruses. Retroviruses are viruses that contain RNA (ribonucleic acid) as their genetic material. After infecting a

More information

Trends in molecular diagnostics

Trends in molecular diagnostics Trends in molecular diagnostics Detection of target genes of interest Quantification Infectious diseases HIV Hepatitis C & B TB / MAC Cytomegalovirus Herpes simplex Varicella zoster CT/GC HPV Profiling

More information

The Struggle with Infectious Disease. Lecture 6

The Struggle with Infectious Disease. Lecture 6 The Struggle with Infectious Disease Lecture 6 HIV/AIDS It is generally believed that: Human Immunodeficiency Virus --------- causes ------------- Acquired Immunodeficiency Syndrome History of HIV HIV

More information

HIV-1 Viral Load Real Time (RG)

HIV-1 Viral Load Real Time (RG) -1 Viral Load Real Time (RG) Real Time RT-PCR type 1 RNA quantification assay MSP Reg. pending Valdense 3616. 11700. Montevideo. Uruguay. phone (598) 2 336 83 01. Fax (598) 2 336 71 60. Info@atgen.com.uy

More information

HIV Anti-HIV Neutralizing Antibodies

HIV Anti-HIV Neutralizing Antibodies ,**/ The Japanese Society for AIDS Research The Journal of AIDS Research : HIV HIV Anti-HIV Neutralizing Antibodies * Junji SHIBATA and Shuzo MATSUSHITA * Division of Clinical Retrovirology and Infectious

More information

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

HLA-DR CD38 CD4 T Lymphocytes Have Elevated CCR5 Expression and Produce the Majority of R5-Tropic HIV-1 RNA In Vivo JOURNAL OF VIROLOGY, Oct. 2011, p. 10189 10200 Vol. 85, No. 19 0022-538X/11/$12.00 doi:10.1128/jvi.02529-10 Copyright 2011, American Society for Microbiology. All Rights Reserved. HLA-DR CD38 CD4 T Lymphocytes

More information

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

Supplemental Materials and Methods Plasmids and viruses Quantitative Reverse Transcription PCR Generation of molecular standard for quantitative PCR Supplemental Materials and Methods Plasmids and viruses To generate pseudotyped viruses, the previously described recombinant plasmids pnl4-3-δnef-gfp or pnl4-3-δ6-drgfp and a vector expressing HIV-1 X4

More information

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

On an individual level. Time since infection. NEJM, April HIV-1 evolution in response to immune selection pressures HIV-1 evolution in response to immune selection pressures BISC 441 guest lecture Zabrina Brumme, Ph.D. Assistant Professor, Faculty of Health Sciences Simon Fraser University http://www3.niaid.nih.gov/topics/hivaids/understanding/biology/structure.htm

More information

Chronic HIV-1 Infection Frequently Fails to Protect against Superinfection

Chronic HIV-1 Infection Frequently Fails to Protect against Superinfection Chronic HIV-1 Infection Frequently Fails to Protect against Superinfection Anne Piantadosi 1,2[, Bhavna Chohan 1,2[, Vrasha Chohan 3, R. Scott McClelland 3,4,5, Julie Overbaugh 1,2* 1 Division of Human

More information

A VACCINE FOR HIV BIOE 301 LECTURE 10 MITALI BANERJEE HAART

A VACCINE FOR HIV BIOE 301 LECTURE 10 MITALI BANERJEE HAART BIOE 301 LECTURE 10 MITALI BANERJEE A VACCINE FOR HIV HIV HAART Visit wikipedia.org and learn the mechanism of action of the five classes of antiretroviral drugs. (1) Reverse transcriptase inhibitors (RTIs)

More information

Received 1 August 2005/Accepted 25 October 2005

Received 1 August 2005/Accepted 25 October 2005 JOURNAL OF VIROLOGY, Jan. 2006, p. 634 642 Vol. 80, No. 2 0022-538X/06/$08.00 0 doi:10.1128/jvi.80.2.634 642.2006 Copyright 2006, American Society for Microbiology. All Rights Reserved. Perturbations of

More information

Received 19 July 2002/Accepted 24 September 2002

Received 19 July 2002/Accepted 24 September 2002 JOURNAL OF VIROLOGY, Jan. 2003, p. 744 748 Vol. 77, No. 1 0022-538X/03/$08.00 0 DOI: 10.1128/JVI.77.1.744 748.2003 Copyright 2003, American Society for Microbiology. All Rights Reserved. Partial Molecular

More information

Immunodeficiency. (2 of 2)

Immunodeficiency. (2 of 2) Immunodeficiency (2 of 2) Acquired (secondary) immunodeficiencies More common Many causes such as therapy, cancer, sarcoidosis, malnutrition, infection & renal disease The most common of which is therapy-related

More information

HIV 101: Fundamentals of HIV Infection

HIV 101: Fundamentals of HIV Infection HIV 101: Fundamentals of HIV Infection David H. Spach, MD Professor of Medicine University of Washington Seattle, Washington Learning Objectives After attending this presentation, learners will be able

More information

Copyright information:

Copyright information: Reduced Simian Immunodeficiency Virus Replication in Macrophages of Sooty Mangabeys Is Associated with Increased Expression of Host Restriction Factors Kiran Mir-Hudgeons, Emory University Maud Mavigner,

More information

The Discovery of SIV and Development of Monkey Models for the Study of HIV/AIDS. Ronald C Desrosiers University of Miami Miller School of Medicine

The Discovery of SIV and Development of Monkey Models for the Study of HIV/AIDS. Ronald C Desrosiers University of Miami Miller School of Medicine The Discovery of SIV and Development of Monkey Models for the Study of HIV/AIDS Ronald C Desrosiers University of Miami Miller School of Medicine 10 FEBRUARY 1984 Infectious Diseases Branch, National

More information

Mosaic Genomes of the Six Major Primate Lentivirus Lineages Revealed by Phylogenetic Analyses

Mosaic Genomes of the Six Major Primate Lentivirus Lineages Revealed by Phylogenetic Analyses REFERENCES CONTENT ALERTS Mosaic Genomes of the Six Major Primate Lentivirus Lineages Revealed by Phylogenetic Analyses Marco Salemi, Tulio De Oliveira, Valerie Courgnaud, Vincent Moulton, Barbara Holland,

More information

Received 19 September 2007/Accepted 21 November 2007

Received 19 September 2007/Accepted 21 November 2007 JOURNAL OF VIROLOGY, Feb. 2008, p. 1723 1738 Vol. 82, No. 4 0022-538X/08/$08.00 0 doi:10.1128/jvi.02084-07 Copyright 2008, American Society for Microbiology. All Rights Reserved. Patterns of CD8 Immunodominance

More information

ALTHOUGH disease develops within 10 years in

ALTHOUGH disease develops within 10 years in 228 THE NEW ENGLAND JOURNAL OF MEDICINE Jan. 26, 1995 BRIEF REPORT: ABSENCE OF INTACT nef SEQUENCES IN A LONG-TERM SURVIVOR WITH NONPROGRESSIVE HIV-1 INFECTION FRANK KIRCHHOFF, PH.D., THOMAS C. GREENOUGH,

More information

NOTES. Michael D. George,* David Verhoeven, Sumathi Sankaran, Tiffany Glavan, Elizabeth Reay, and Satya Dandekar

NOTES. Michael D. George,* David Verhoeven, Sumathi Sankaran, Tiffany Glavan, Elizabeth Reay, and Satya Dandekar CLINICAL AND VACCINE IMMUNOLOGY, Feb. 2009, p. 277 281 Vol. 16, No. 2 1556-6811/09/$08.00 0 doi:10.1128/cvi.00265-08 Copyright 2009, American Society for Microbiology. All Rights Reserved. NOTES Heightened

More information

Rajesh Kannangai Phone: ; Fax: ; *Corresponding author

Rajesh Kannangai   Phone: ; Fax: ; *Corresponding author Amino acid sequence divergence of Tat protein (exon1) of subtype B and C HIV-1 strains: Does it have implications for vaccine development? Abraham Joseph Kandathil 1, Rajesh Kannangai 1, *, Oriapadickal

More information

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

Supplementary Figure 1. Gating strategy and quantification of integrated HIV DNA in sorted CD4 + T-cell subsets. Supplementary information HIV reservoir size and persistence are driven by T-cell survival and homeostatic proliferation. Chomont, N., M. El Far, P. Ancuta, L. Trautmann, F. A. Procopio, B. Yassine-Diab,

More information

Nef-Mediated Enhancement of Virion Infectivity and Stimulation of Viral Replication Are Fundamental Properties of Primate Lentiviruses

Nef-Mediated Enhancement of Virion Infectivity and Stimulation of Viral Replication Are Fundamental Properties of Primate Lentiviruses JOURNAL OF VIROLOGY, Dec. 2007, p. 13852 13864 Vol. 81, No. 24 0022-538X/07/$08.00 0 doi:10.1128/jvi.00904-07 Copyright 2007, American Society for Microbiology. All Rights Reserved. Nef-Mediated Enhancement

More information

Acquired Immune Deficiency Syndrome (AIDS)

Acquired Immune Deficiency Syndrome (AIDS) Acquired Immune Deficiency Syndrome (AIDS) By Jennifer Osita Disease The disease I am studying is AIDS (Acquired Immune Deficiency Syndrome) which is when the immune system is too weak to fight off many

More information

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

Antibody Dependent Cellular Cytotxic activity: Past and Future. Guido Ferrari, M.D. Duke University Medical Center Antibody Dependent Cellular Cytotxic activity: Past and Future Guido Ferrari, M.D. Duke University Medical Center Mechanism of Antibody Dependent Cellular Cytotoxicity (ADCC) ADCC Effector Cells (NK, monocytes/macrophages,

More information

HIV-1 Dual Infection and Neurocognitive Impairment

HIV-1 Dual Infection and Neurocognitive Impairment HIV-1 Dual Infection and Neurocognitive Impairment Gabriel Wagner, MD Assistant Professor of Medicine Infectious Diseases & Global Public Health UC San Diego HIV-Associated End Organ Damage Antiretroviral

More information

Isolation from African Sykes' Monkeys (Cercopithecus mitis) of a Lentivirus Related to Human and Simian Immunodeficiency Viruses

Isolation from African Sykes' Monkeys (Cercopithecus mitis) of a Lentivirus Related to Human and Simian Immunodeficiency Viruses JOURNAL OF VIROLOGY, Apr. 1991, p. 2135-2140 0022-538X/91/042135-06$02.00/0 Copyright C) 1991, American Society for Microbiology Vol. 65, No. 4 Isolation from African Sykes' Monkeys (Cercopithecus mitis)

More information

Should There be Further Efficacy Testing of T-T cell Based Vaccines that do not Induce Broadly Neutralizing Antibodies?

Should There be Further Efficacy Testing of T-T cell Based Vaccines that do not Induce Broadly Neutralizing Antibodies? Dale and Betty Bumpers Vaccine Research Center National Institute of Allergy and Infectious Diseases National Institutes of Health Department of Health and Human Services Should There be Further Efficacy

More information

The Evolutionary Rate of Nonpathogenic Simian Immunodeficiency Virus (SIVagm) Is in Agreement with a Rapid and Continuous Replication in Vivo

The Evolutionary Rate of Nonpathogenic Simian Immunodeficiency Virus (SIVagm) Is in Agreement with a Rapid and Continuous Replication in Vivo VIROLOGY 223, 89 102 (1996) ARTICLE NO. 0458 The Evolutionary Rate of Nonpathogenic Simian Immunodeficiency Virus (SIVagm) Is in Agreement with a Rapid and Continuous Replication in Vivo MICHAELA C. MÜLLER-TRUTWIN,*

More information

Are we targeting the right HIV determinants?

Are we targeting the right HIV determinants? QuickTime et un décompresseur TIFF (non compressé) sont requis pour visionner cette image. AIDS Vaccine 2009 October 22 nd 2009 - Paris Are we targeting the right HIV determinants? Françoise BARRÉ-SINOUSSI

More information

HIV Basics: Pathogenesis

HIV Basics: Pathogenesis HIV Basics: Pathogenesis Michael Saag, MD, FIDSA University of Alabama, Birmingham Director, Center for AIDS Research ACTHIV 2011: A State-of-the-Science Conference for Frontline Health Professionals Learning

More information

Received 13 August 1992/Accepted 3 December 1992

Received 13 August 1992/Accepted 3 December 1992 JOURNAL OF VIROLOGY, Mar. 1993, P. 1227-1235 0022-538X/93/031227-09$02.00/0 Copyright ) 1993, American Society for Microbiology Vol. 67, No. 3 Simian Immunodeficiency Viruses from Central and Western Africa:

More information

FENG LI, 1 CAROLINE LEROUX, 1 JODI K. CRAIGO, 1 SHEILA J. COOK, 2 CHARLES J. ISSEL, 2

FENG LI, 1 CAROLINE LEROUX, 1 JODI K. CRAIGO, 1 SHEILA J. COOK, 2 CHARLES J. ISSEL, 2 JOURNAL OF VIROLOGY, Jan. 2000, p. 573 579 Vol. 74, No. 1 0022-538X/00/$04.00 0 Copyright 2000, American Society for Microbiology. All Rights Reserved. The S2 Gene of Equine Infectious Anemia Virus Is

More information

cure research HIV & AIDS

cure research HIV & AIDS Glossary of terms HIV & AIDS cure research Antiretroviral Therapy (ART) ART involves the use of several (usually a cocktail of three or more) antiretroviral drugs to halt HIV replication. ART drugs may

More information

Current Strategies in HIV-1 Vaccine Development Using Replication-Defective Adenovirus as a Case Study

Current Strategies in HIV-1 Vaccine Development Using Replication-Defective Adenovirus as a Case Study Note: I have added some clarifying comments to the slides -- please click on Comments under View to see them. Current Strategies in HIV-1 Vaccine Development Using Replication-Defective Adenovirus as a

More information

Molecular Epidemiology of Simian T-Cell Lymphotropic Virus Type 1 in Wild and Captive Sooty Mangabeys

Molecular Epidemiology of Simian T-Cell Lymphotropic Virus Type 1 in Wild and Captive Sooty Mangabeys JOURNAL OF VIROLOGY, Feb. 2005, p. 2541 2548 Vol. 79, No. 4 0022-538X/05/$08.00 0 doi:10.1128/jvi.79.4.2541 2548.2005 Copyright 2005, American Society for Microbiology. All Rights Reserved. Molecular Epidemiology

More information

UNRAVELING THE MYSTERY BEHIND HIV/AIDS

UNRAVELING THE MYSTERY BEHIND HIV/AIDS PRESS RELEASE 3 MAR 2010 UNRAVELING THE MYSTERY BEHIND HIV/AIDS New findings by Nobel Laureate shed light on the elusive AIDS virus and may lead to effective HIV vaccine development 1. Professor Francoise

More information

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

Increased Loss of CCR5 CD45RA CD4 T Cells in CD8 Lymphocyte-Depleted Simian Immunodeficiency Virus-Infected Rhesus Monkeys JOURNAL OF VIROLOGY, June 2008, p. 5618 5630 Vol. 82, No. 11 0022-538X/08/$08.00 0 doi:10.1128/jvi.02748-07 Copyright 2008, American Society for Microbiology. All Rights Reserved. Increased Loss of CCR5

More information

Far from the limelight of the human and mouse genome projects, the Feline

Far from the limelight of the human and mouse genome projects, the Feline Carolyn Sangokoya Professor Doug Brutlag Genomics and Bioinformatics 9 March 2000 Lessons from the Felidae: A Quest to Understand HIV and other Pathogenic Immunodeficiency Viruses Far from the limelight

More information

Dynamics of lentiviral infection in vivo in the absence of adaptive immune responses

Dynamics of lentiviral infection in vivo in the absence of adaptive immune responses Dynamics of lentiviral infection in vivo in the absence of adaptive immune responses Elissa J. Schwartz Associate Professor School of Biological Sciences Department of Mathematics & Statistics Washington

More information

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

Gladstone Institutes, University of California (UCSF), San Francisco, USA Fluorescence-linked Antigen Quantification (FLAQ) Assay for Fast Quantification of HIV-1 p24 Gag Marianne Gesner, Mekhala Maiti, Robert Grant and Marielle Cavrois * Gladstone Institutes, University of

More information