INTRODUCTION. Gabon. Gabon

Size: px
Start display at page:

Download "INTRODUCTION. Gabon. Gabon"

Transcription

1 Journal of General Virology (2009), 90, DOI /vir Simian immunodeficiency virus types 1 and 2 (SIV mnd 1 and 2) have different pathogenic potentials in rhesus macaques upon experimental cross-species transmission Sandrine Souquière, 1 Richard Onanga, 1 Maria Makuwa, 1 Ivona Pandrea, 1,2 Paul Ngari, 1 Pierre Rouquet, 3 Olivier Bourry, 3 Mirdad Kazanji, 1 Cristian Apetrei, 1,2 François Simon 1,4 and Pierre Roques 1,5 Correspondence Sandrine Souquière souquiere@cirmf.org sandsouquiere@yahoo.fr 1 Laboratoire de Rétrovirologie, Centre International de Recherches Médicales (CIRMF), Franceville, Gabon 2 Tulane National Primate Research Center, Covington, LA 70433, USA 3 Centre de Primatologie, Centre International de Recherches Médicales (CIRMF), Franceville, Gabon 4 Laboratoire de Virologie, Hôpital St Louis, Paris, France 5 Service de Neurovirologie, CEA imeti, Fontenay aux Roses, France Received 30 June 2008 Accepted 12 October 2008 The mandrill (Mandrillus sphinx) is naturally infected by two types of simian immunodeficiency virus (SIV): SIVmnd types 1 and 2. Both of these viruses cause long-term, non-progressive infections in their natural host despite high plasma viral loads. This study assessed the susceptibility of rhesus macaques to infection by these two types of SIVmnd and compared the virological and basic immunological characteristics of the resulting infections with those observed in natural infection in mandrills. Whilst both SIVmnd types induced similar levels of virus replication during acute infection in both mandrills and macaques, they produced a more pronounced CD4 + T-cell depletion in rhesus macaques that persisted longer during the initial stage of infection. Pro-inflammatory cytokine responses were also induced at higher levels in rhesus macaques early in the infection. During the chronic phase of infection in mandrills, which in this case was followed for up to 2 years after infection, high levels of chronic virus replication did not induce significant changes in CD4 + or CD8 + T-cell counts. In rhesus macaques, the overall chronic virus replication level was lower than in mandrills. At the end of the follow-up period, although the viral loads of SIVmnd-1 and SIVmnd-2 were relatively similar in rhesus macaques, only SIVmnd-1-infected rhesus macaques showed significant CD4 + T-cell depletion, in the context of higher levels of CD4 + and CD8 + T-cell activation, compared with SIVmnd-infected mandrills. The demonstration of the ability of both SIVmnd types to induce persistent infections in rhesus macaques calls for a careful assessment of the potential of these two viruses to emerge as new human pathogens. INTRODUCTION African non-human primates (NHPs) are the natural hosts of simian immunodeficiency viruses (SIVs) (Apetrei et al., 2004; Hahn et al., 2000; VandeWoude & Apetrei, 2006). To date, more than 40 different SIVs have been described and they infect different African species of monkeys and apes at high levels of prevalence (Apetrei et al., 2004; Hahn et al., 2000; VandeWoude & Apetrei, 2006). Unlike pathogenic human immunodeficiency virus (HIV) in humans and SIV in macaques, natural SIV infections generally do not progress to acquired immune deficiency syndrome (AIDS) (Chakrabarti, 2004; Hirsch, 2004; Muller & Barré-Sinoussi, 2003; Norley et al., 1999; Onanga et al., 2002, 2006; Pandrea et al., 2008b; Silvestri, 2005). In fact, only a handful of cases of immunodeficiency have been described to date in African NHP species, all of which have occurred in captive animals (Ling et al., 2004; Pandrea et al., 2001; Traina-Dorge et al., 1992). This lack of SIV-related disease progression in natural NHP hosts does not appear to be due to better infection control, as natural SIV infections are characterized by high levels of virus replication and G 2009 SGM Printed in Great Britain

2 SIVmnd types 1 and 2 infection of rhesus macaques immune responses that are not necessarily stronger than those observed in pathogenic infections (Dunham et al., 2006; Wang et al., 2006). It is currently believed that this observed lack of pathogenicity is due to the host s ability to control the deleterious side effects of the SIV infection. Supporting this view is the observation that natural infections are characterized by only a transient depletion of peripheral CD4 + T cells during primary infection and CD4 + T-cell restoration during the chronic infection to near-baseline levels (Kaur et al., 1998; Kornfeld et al., 2005; Pandrea et al., 2005, 2006; Silvestri et al., 2005). Also, natural SIV infections are characterized by only transient and moderate increases in immune activation and proliferation during acute infection, with a return to baseline levels during the chronic infection (Kaur et al., 1998; Kornfeld et al., 2005; Onanga et al., 2006; Pandrea et al., 2006, 2007; Silvestri et al., 2005). Finally, normal levels of apoptosis are observed throughout natural SIV infections (Estaquier et al., 1994; Pandrea et al., 2007; Silvestri et al., 2003). However, at mucosal sites, CD4 + T-cell depletion is more pronounced and prolonged, and restoration is only partial throughout the chronic infection (Gordon et al., 2007; Pandrea et al., 2007). In striking contrast to the general lack of disease progression of SIV infection in natural hosts, pathogenic HIV and SIVmac infections in humans and Asian macaques, respectively, lead to CD4 + T-cell depletion and invariable progression to AIDS (Hirsch & Johnson, 1994). HIV-1 and HIV-2 originated from independent cross-species transmissions of SIVcpz, a virus that naturally infects chimpanzees in central Africa (Keele et al., 2006; Peeters et al., 2002), and SIVsmm, a virus that naturally infects sooty mangabeys in western Africa (Hirsch et al., 1989; Marx et al., 1991), respectively. The emergence of SIVmac occurred by accidental transmission of SIVsmm from sooty mangabeys to various macaque species housed in US-based primate centres (Apetrei et al., 2005, 2006). Although it is not yet clear what mechanisms led to this dramatic increase in the pathogenicity of SIVs in these new hosts, serial passage may have been involved in this transformation (Apetrei et al., 2006; Marx et al., 2001). Experimental infections involving the cross-species transmission of various SIVs have resulted in widely variable outcomes. In addition to the pathogenic infections described previously, rhesus macaques infected with SIVagm from African green monkeys (genus Chlorocebus) (Pandrea et al., 2007), SIVrcm from red-capped mangabeys (Cercocebus torquatus) (Smith et al., 1998), SIVlhoest from l Hoesti monkeys (Cercopithecus lhoesti lhoesti) (Hirsch et al., 1999), SIVsyk from Sykes monkeys (Cercopithecus albogularis) (Hirsch et al., 1993) and SIVtal from talapoin monkeys (Miopithecus talapoin) (Osterhaus et al., 1999) developed transient infections characterized by an active SIV replication during the acute infection and complete control of virus replication during the chronic infection. Conversely, cross-species SIV transmission to pig-tailed macaques resulted in progressive disease in most circumstances, with cases of AIDS in pig-tailed macaques being described following experimental infections with SIVsun from solatus monkeys (Cercopithecus lhoesti solatus) (Beer et al., 2005), SIVlhoest (Beer et al., 2005), SIVagm from vervet (Hirsch et al., 1995) and sabaeus monkeys (I. Pandrea, unpublished observation) and SIVrcm (C. Apetrei, unpublished observation). Mandrills (Mandrillus sphinx) are endemic in Gabon, central Africa, and are to date the only NHP species shown to be infected with two types of SIV: SIVmnd type 1 (SIVmnd-1) (Souquiere et al., 2001; Tsujimoto et al., 1988, 1989) and SIVmnd type 2 (SIVmnd-2) (Hu et al., 2003; Souquiere et al., 2001; Takehisa et al., 2001). These two SIVmnd types have different origins, with SIVmnd-1 belonging to the SIVlhoest lineage and clustering together with SIVlhoest from l Hoesti monkeys and SIVsun from solatus monkeys (Beer et al., 1999; Hirsch et al., 1999; VandeWoude & Apetrei, 2006), and SIVmnd-2 being closer to other Papionini viruses, such as SIVdrl from drills (Mandrillus leuchophaeus) and SIVrcm (Hu et al., 2003; Souquiere et al., 2001; Takehisa et al., 2001). SIVmnd-2 is a recombinant virus, harbouring gag, pol, vif, vpx and tat sequences that are closely related to SIVrcm sequences, whereas its env and nef sequences are closely related to SIVmnd-1 (Hu et al., 2003; Souquière et al., 2001). Although SIVmnd-1 and SIVmnd-2 have different origins, these two viruses have identical virological characteristics in their natural hosts (Onanga et al., 2002, 2006). Thus, during experimental infection, high peak viral loads (VLs) and high levels of chronic virus replication are associated with only transient decreases in peripheral CD4 + T-cell counts. The aim of this study was to investigate the dynamics of SIVmnd infections in rhesus macaques. We found that both SIVmnd-1 and SIVmnd-2 induced persistent infections in experimentally infected rhesus macaques. Although the levels of chronic SIVmnd-1 and SIVmnd-2 replication were comparable, a continuous CD4 + T-cell depletion was observed only in SIVmnd-1-infected rhesus macaques. METHODS Animals and infections. Four captive mandrills and six rhesus macaques of Chinese origins were included in this study. All of these animals, which tested negative for SIV and simian T-lymphotropic virus, were housed at the CIRMF Primate Center and handled under Biosafety level 3 conditions in accordance with the NIH Guide for the Care and Use of Laboratory Animals. The mandrills comprised two females: 2I (9 years old) and 12D4 (7 years old), and two males: 5A4 (10 years old) and 2H (13 years old). All rhesus macaques were male: (8 years old), 93183A (3 years old), P9110A (6 years old), (8 years old), 93183B (3 years old) and 93046A (6 years old). All animal protocols and procedures were approved by the Gabonese Ethics Committee for Animal Experimentation. The viral inocula used consisted of plasma obtained directly from acutely infected mandrills at peak viraemia following experimental 489

3 S. Souquière and others infection, to avoid potential problems due to virus selection through in vitro culture, as described previously (Onanga et al., 2002, 2006). Mandrills 2I and 5A4 and rhesus macaques 93057, 93183A and P9110A were inoculated with 500 ml plasma containing RNA copies of SIVmnd-1 (strains 16C and 12A3) (Pandrea et al., 2008a). Mandrills 12D4 and 2H and rhesus macaques 93036, 93183B and 93046A were inoculated with 400 ml plasma containing RNA copies of SIVmnd-2 (strain 10I) (Onanga et al., 2006). Animals were anaesthetized with ketamine/hcl for handling and were inoculated via the saphenous vein. Specimen collection. Blood samples were collected on days 0, 4, 7, 11, 14, 28, 32, 60, 120, 180, 270, 360 and 750 days post-infection (p.i.) in EDTA K2 tubes for mandrills, and on days 0, 4, 7, 11, 28, 32, 60, 90, 120, 180, 330, 360 and 750 p.i. for rhesus macaques. Blood was used for flow cytometry, and peripheral blood mononuclear cells (PBMCs) were isolated by Ficoll-Hypaque gradient centrifugation (Sigma- Aldrich), aliquotted in 10 % DMSO (Sigma-Aldrich) in fetal bovine serum (Gibco-BRL) and frozen at 280 uc. Plasma was centrifuged at 3000 g for 10 min, dispensed into 1 ml aliquots and frozen at 280 uc. Excisional inguinal lymph nodes (LNs) were collected from mandrills and rhesus macaques on days 0, 7, 28, 60 and 360 p.i. Cells from LNs were separated by passage through a mesh screen. The cells obtained were washed in RPMI (Gibco-BRL), centrifuged for 10 min at 1500 g, aliquotted in 10 % DMSO in bovine fetal serum and frozen at 80 uc. Kinetics of antibody production. Anti-SIVmnd antibody dynamics were monitored using a peptide-based ELISA detecting antibodies against SIVmnd-specific peptides mapping the V3 region of the env glycoprotein (Simon et al., 2001). The same SIVmnd GB1 peptide was used to evaluate the presence of anti-sivmnd antibodies in both types of infection, as the two types of SIVmnd share the same env sequences as a consequence of recombination events (Souquiere et al., 2001) and our preliminary studies showed a similar sensitivity of peptide-based ELISA for both SIVmnd types (Simon et al., 2001). The test was performed as described previously (Simon et al., 2001). SIVmnd RNA quantification. RNA was extracted from 150 ml plasma using a QiaAmp Viral RNA Mini kit (Qiagen) and eluted in 50 ml TE buffer, as recommended by the manufacturer. For LNs, we used a QiaAmp RNA Isolation kit (Qiagen) to extract RNA from frozen aliquots of cells. Real-time PCR assays specific for each virus type were developed for SIVmnd quantification. Briefly, quantification by real-time RT-PCR was performed with 5 ml extracted RNA, using a QuantiTect SYBR Green RT-PCR kit (Qiagen) in capillary tubes, with the LightCycler System (Roche Diagnostics). For both viral strains, quantification was based on amplification of a 230 bp fragment located in the integrase region. Specific sets of primers were designed for each viral strain. The primers used for SIVmnd-1 were M17IF: 59-AAACAGATTGTGC- AAAGTTGCA-39 and M17IR: 59-CCCATTATCTGTGTGTAGTTT- ACTAATA-39. The primers used for SIVmnd-2 quantification were M26IF: 59-GCAAAGGAGATAGTAGCTCAGTGTC-39 and M26IR: 59-GCCATTATCTGTATGTAAATGTTTCACT-39. Primers were used at a final concentration of 1 mm and the final MgCl 2 concentration was 2.5 mm. The amplification protocol for SIVmnd-1 quantification consisted of reverse transcription (20 min at 50 uc), followed by denaturation and activation of HotStart Taq DNA polymerase (15 min at 95 uc) and cdna amplification (50 cycles of denaturation for 15 s at 95 uc, annealing for 15 s at 58 uc and elongation for 22 s at 72 uc). For SIVmnd-2 quantification, the conditions were the same except that the annealing temperature was 60 uc. Data were analysed using LightCycler Software version 3.5. The RNA copy number was determined by comparison with an external standard curve and was expressed as RNA copies ml 21 for plasma and RNA copies per 10 6 cells for LNs. Standards, consisting of non-virus-specific tailored competitor RNA complementary to sequences specific for SIVmnd-1 or SIVmnd-2, were added at both the 59 and 39 ends. All molecular constructs were produced by Mobidab Molekularbiologie GmbH & Co. The MO4-01 RNA standard for SIVmnd-1 was 230 bp and the MO4-02 RNA standard for SIVmnd-2 was 233 bp. Each standard was subjected to 10-fold dilutions to generate solutions with RNA copies. The detection limit of the SIVmnd quantification assays was 250 RNA copies (ml plasma) 21. Lymphocyte studies and flow cytometry. Lymphocyte subsets were analysed by three-colour fluorescence-activated cell sorter analysis (FACSCalibur; Becton Dickinson). For staining, 50 ml cell suspension ( PBMCs) was mixed with 5 ml of different combinations of the following monoclonal antibodies: fluorescein isothiocyanate-conjugated CD3 (clone SP-34), phycoerythrin (PE)- conjugated CD4 (clone M-T477), PE-conjugated CD8 (clone Leu-2a) and peridinin chlorophyll protein-conjugated HLA-DR (clone L- 243) (all from BD Biosciences). The cells were incubated for 20 min at 4 uc in the dark and then washed twice with PBS and resuspended in 300 ml CellFix (Becton Dickinson). An irrelevant anti-mouse IgG1 mab (MOPC-31C; BD Biosciences) was used as a negative control. Analysis was performed using FlowJo software version (TreeStar) Gamma interferon (IFN-c) assay. Quantification of IFN-c in plasma at days 0, 3, 7, 10, 14 and 28 p.i. was evaluated using a commercial ELISA kit (Monkey IFN-c ELISA kit; Cell Sciences) according to the manufacturer s instructions. The sensitivity of the kit was 2 pg (ml plasma) 21. Statistical analysis. Comparisons between macaques and mandrills were performed using a non-parametric Mann Whitney U-test with Statistica software (StatSoft France, version 7.1; RESULTS Antibody response profile in infected mandrills and rhesus macaques V3 ELISA testing showed that all SIVmnd-1- and SIVmnd- 2-infected rhesus macaques and the four mandrills had seroconverted by day 28 p.i. (Fig. 1a, b). However, anti-v3 antibody titres were significantly higher for rhesus macaques than for mandrills at day 360 (P,0.02) (Fig. 1c). Plasma SIVmnd-1 and SIVmnd-2 dynamics in rhesus macaques and mandrills Acute infection showed common replication patterns for both viruses in mandrills and rhesus macaques. In mandrills, plasma VLs peaked between days 7 and 11 and ranged from to RNA copies ml 21 (Fig. 2a). In rhesus macaques, plasma VLs ranged from to RNA copies ml 21 between days 7 and 14 (Fig. 2b). No significant VL difference was found during acute infection between mandrills and rhesus macaques. Interestingly, in three rhesus macaques (93057, 93183A and 93183B), plasma VLs decreased just before reaching the 490 Journal of General Virology 90

4 SIVmnd types 1 and 2 infection of rhesus macaques Fig. 1. Anti-V3 antibody titres in mandrills (a) and rhesus macaques (b) infected with SIVmnd-1 (red) and SIVmnd- 2 (blue). (c) Comparison of Anti-V3 antibody titres between mandrills and rhesus macaques at day 360 using a Mann Whitney U-test. peak (Fig. 2b). This biphasic profile was confirmed in several independent plasma VL determinations. The post-acute and chronic replication patterns were very different between mandrills and rhesus macaques. In mandrills, plasma VLs reached a set point by day 28 p.i., which was maintained during the follow-up period. The set-point VLs were remarkably stable with the exception of some points around day 180 p.i., when VLs decreased to reach 10 4 RNA copies ml 21. At the 2-year follow-up, plasma VLs were to RNA copies ml 21 (mean: ) for both SIVmnd-1- and SIVmnd-2- infected mandrills (Fig. 2a). In rhesus macaques, plasma VLs decreased below the threshold of detection (250 SIVmnd RNA copies ml 21 ) between days 90 and 330. However, this control of SIVmnd-1 and SIVmnd-2 replication in rhesus macaques was only transient. Blips of detectable VL were observed for both viruses in rhesus macaques. At the end of the follow-up period, 2 years after infection, all rhesus macaques but one (93036) showed detectable VLs that ranged between and RNA copies ml 21 (mean: ) (Fig. 2b). Plasma VL dynamics in rhesus macaques showed no significant differences for SIVmnd-1 and SIVmnd-2. LN RNA VL dynamics SIVmnd RNA loads in LNs generally paralleled that of plasma VL dynamics (Fig. 2c, d). At day 7 p.i., RNA copies ranged from to per 10 6 cells for mandrills and to per 10 6 cells for rhesus macaques. At day 60 p.i., in all cases, LN RNA VL decreased moderately by around 1 log. After 1 year of infection, there was no significant difference between rhesus macaques and mandrills except for two rhesus macaques (93057 and 93183A) that had a very low LN VL ( RNA copies per 10 6 cells)

5 S. Souquière and others Fig. 2. Dynamics of plasma VL (viral RNA copies ml 1 ) in mandrills (a) and rhesus macaques (b) and of lymph node RNA loads in mandrills (c) and rhesus macaques (d). Animals infected with SIVmnd-1 are indicated in red and animals infected with SIVmnd-2 are indicated in blue. Real-time PCR detection limits: 250 copies ml 1 (a, b) and 250 copies per 10 6 cells (c, d). SIVmnd-1 and SIVmnd-2 have a different impact on CD4 + and CD8 + T cells in mandrills and rhesus macaques In spite of the fact that acute virus replication patterns were similar between mandrills and rhesus macaques, their impact on CD4 + T cells was different for the two species. A moderate, transient increase in CD4 + T cells was observed in two mandrills just before the peak VL, followed by moderate decreases in CD4 + T cells occurring after the peak of virus replication in two of the four mandrills (Fig. 3a). During the chronic infection, CD4 + T-cell counts returned to close to the baseline levels in all mandrills (Fig. 3a). As previously reported, we did not observe any difference in CD4 + T-cell counts between mandrills infected with SIVmnd-1 and SIVmnd-2 (Fig. 3a). Conversely, in both SIVmnd-1- and SIVmnd-2-infected rhesus macaques, a significant decrease in CD4 + T-cell counts occurred during week 1 p.i., which was followed by a partial CD4 + T-cell restoration. Moreover, differences in CD4 + T-cell counts were recorded between SIVmnd-1- and SIVmnd-2-infected rhesus macaques. Thus, during the acute infection, CD4 + T-cell depletion appeared to be more important in SIVmnd-2-infected macaques (93036, 93183B and 93046A), in which % of the CD4 + T cells were depleted (Table 1). In SIVmnd-1-infected rhesus macaques, only % of CD4 + T cells were depleted during the acute infection (Table 1). However, these differences were not significant (P50.12). Conversely, during the chronic infection, after a 2-year follow-up period, the SIVmnd-2- infected rhesus macaques maintained their CD4 + T-cell counts, whereas the SIVmnd-1-infected rhesus macaques showed significant CD4 + T-cell depletion (Fig. 4). Thus, the mean decrease in SIVmnd-1-infected rhesus macaques was significantly higher (67 %) than in SIVmnd-2-infected rhesus macaques (14 %) (P,0.05). Only moderate variations in CD8 + T-cell levels were observed in SIVmnd-infected mandrills during acute or chronic infection (Fig. 3c). Conversely, in four out of six macaques (both SIVmnd-1- and SIVmnd-2-infected), CD8 + T-cell counts increased considerably after the peak of virus replication, and frequent transient increases were noted during the follow-up period (Fig. 3d). At 2 years p.i., CD8 + T cells returned to baseline levels in all SIVmnd-1- and SIVmnd-2-infected rhesus macaques, except for P9110A. Interestingly, this decrease in CD8 + T-cell levels corresponded to an increase in virus replication, with plasma VLs being consistently detectable at 2 years p.i. (Fig. 2b). 492 Journal of General Virology 90

6 SIVmnd types 1 and 2 infection of rhesus macaques Fig. 3. Variations in peripheral CD4 + and CD8 + T-cell counts in mandrills (a, c) and macaques (b, d) compared with baseline. Animals infected with SIVmnd-1 are indicated in red, whilst animals infected with SIVmnd-2 are indicated in blue. The means are shown as solid lines. Table 1. Number of CD4 + T cells in macaques infected by SIVmnd-1 and SIVmnd-2 at day 0 and on the day corresponding to the peak VL, and percentage decrease from baseline SIVmnd type Macaque no. No. CD4 + T cells ml 1 (day 0) No. CD4 + T cells ml 1 (day p.i.) Decrease from baseline (%) SIVmnd (day 11) A (day 7) 49 P9110A (day 7) 64 SIVmnd (day 14) B (day 4) A (day 7)

7 S. Souquière and others was observed in mandrills (Fig. 6a), whereas significant increases (up to 10-fold) in the levels of IFN-c were observed in rhesus macaques (Fig. 6b) (P50.01). In both species, these increases corresponded to the peak of virus replication. Fig. 4. Comparison of CD4 + T-cells counts on days 0 and 750 in rhesus macaques. Macaques infected with SIVmnd-1 are indicated in red, whilst macaques infected with SIVmnd-2 are indicated in blue. Increases in T-cell immune activation are associated with CD4 + T-cell depletion in SIVmnd- 1-infected rhesus macaques In both SIVmnd-1- and SIVmnd-2-infected mandrills, we showed that, with the exception of a transient increase in HLA-DR expression (a marker of immune activation) by both CD4 + and CD8 + T cells during the acute infection, corresponding to the peak of virus replication, there was no significant variation in HLA-DR expression in CD4 + or CD8 + T cells (Fig. 5a, b). Conversely, in rhesus macaques infected with SIVmnd-1 and SIVmnd-2, there was a significant increase in immune activation of both CD4 + and CD8 + T cells, as illustrated by the significant increase in HLA-DR expression during acute and chronic infection. At the end of the follow-up period, a significant increase in immune activation of both CD4 + and CD8 + T cells was observed in both SIVmnd-1- and SIVmnd-2-infected rhesus macaques (Fig. 5c, d). This increase was highly significant for both CD4 + and CD8 + T cells (P50.01) (Fig. 5). Differences in the dynamics of pro-inflammatory cytokines between mandrills and rhesus macaques infected with SIVmnd In order to investigate whether differences in inflammation are responsible for the differences in immune activation levels between SIVmnd-infected mandrills and rhesus macaques, we measured the dynamics of a pro-inflammatory cytokine (IFN-c) in the plasma of SIVmnd-infected monkeys during acute infection. As shown in Fig. 6, there were significant differences in the dynamics of plasma IFNc between mandrills and rhesus macaques. Thus, only a modest increase (less than twofold) in the levels of IFN-c DISCUSSION Systematic studies of cross-species-transmitted SIV infections are critical in order to understand the mechanisms responsible for the spectacular increase in pathogenicity that has been observed for some of these viruses. Here, we showed that SIVmnd-1 and SIVmnd-2, two viruses that naturally infect mandrills in the wild, induced persistent infections in rhesus macaques with relatively similar replication patterns. However, the pathogenic outcomes of these two infections were different: SIVmnd-2-infected rhesus macaques maintained their CD4 + T-cell levels during a 2-year follow-up period, whilst significant CD4 + T-cell depletion occurred in SIVmnd-1-infected rhesus macaques, in the context of a higher T-cell immune activation, suggesting a more pathogenic outcome. Data published prior to this study on the outcome of crossspecies-transmitted infections in Asian primates identified some interesting patterns. The first was a general tendency towards controlled infection in rhesus macaques infected with SIVs from African primates, which includes Sykes monkeys, African green monkeys, talapoin monkeys and mangabeys. The second was a general tendency towards persistent, progressive infection in pig-tailed macaques after infection with various SIVs. For example, earlier studies reported that exposure of rhesus macaques to SIVsmm, the virus naturally infecting sooty mangabeys, resulted in persistent infection and rapid progression to AIDS (Murphey-Corb et al., 1986). In fact, accidental transmission of SIVsmm to rhesus macaques led to the development of the current animal model for AIDS research (Apetrei et al., 2005, 2006; Gormus et al., 2004). However, as shown in subsequent studies, the current reference strains for use in macaques resulted from serial passage of SIVsmm, a factor that contributed to this increased pathogenicity (Apetrei et al., 2006; Mansfield et al., 1995). In fact, the intrinsic pathogenic potential of primary SIVsmm isolates in rhesus macaques is significantly lower than initially believed (C. Apetrei, unpublished data), similar to other studies that have reported that SIVsyk (Hirsch et al., 1993), SIVagm (Pandrea et al., 2007), SIVtal (Osterhaus et al., 1999) and SIVrcm (Smith et al., 1998) replicate only transiently in rhesus macaques. In all of these cases, replication only occurred during acute infection, and both VLs and viral cultures were generally negative starting from relatively early time points of chronic infection. As mentioned above, pig-tailed macaques seem to be more susceptible to cross-species-transmitted viruses than rhesus macaques. Progression to AIDS has been reported to occur 494 Journal of General Virology 90

8 SIVmnd types 1 and 2 infection of rhesus macaques Fig. 5. Dynamics of immune activation, as illustrated by changes in HLA-DR expression in peripheral CD4 + and CD8 + T cells from mandrills (a, b) and macaques (c, d). Animals infected with SIVmnd-1 are indicated in red, whilst animals infected with SIVmnd-2 are indicated in blue. Comparison of HLA-DR expression on CD4 + and CD8 + T cells between mandrills and rhesus macaques at day 750 was carried out using a Mann Whitney U-test. *, P50.01; **, P Fig. 6. Quantification of IFN-c in the plasma of SIVmnd-1- and SIVmnd-2-infected mandrills (a) and rhesus macaques (b). Monkeys infected with SIVmnd-1 are indicated in red, whilst monkeys infected with SIVmnd-2 are indicated in blue. The detection limit was 2 pg ml

9 S. Souquière and others in pig-tailed macaques after exposure to SIVsmm (Fultz, 1991), SIVagm (Hirsch et al., 1995), SIVlhoest and SIVsun (Beer et al., 2005). However, it is important to note that disease progression in pig-tailed macaques upon SIV crossspecies transmission is not a constant feature: in the same experiment, some animals may progress to AIDS, whereas others may control the infection, for example in SIVagm infection (Hirsch et al., 1995). Interestingly, rhesus macaques infected with viruses that are known to naturally infect monkey species phylogenetically close to rhesus macaques (within the Papionini tribe) have generated discordant data. For example, SIVsmm produced a persistent infection (McClure et al., 1989; Murphey-Corb et al., 1986), whilst SIVrcm from redcapped mangabeys resulted in a controlled infection (Smith et al., 1998) Mandrills are also phylogenetically close to rhesus macaques, as both belong to the Papionini tribe (Harris & Disotell, 1998). Mandrills are naturally infected by two SIV types, SIVmnd-1 and SIVmnd-2, which have different origins. SIVmnd-2 is closely related phylogenetically to other Papionini SIVs, such as SIVrcm and SIVdrl from drills (Clewley et al., 1998; Hu et al., 2003; Souquiere et al., 2001). In its current form, SIVmnd-2 emerged following recombination of the ancestral SIVrcm-related virus with the SIVmnd-1 ancestor. This recombination was probably ancient, as the recombinant virus was also transmitted to the related drill monkey (Clewley et al., 1998; Hu et al., 2003). In contrast, it is considered that SIVmnd-1 emerged following cross-species transmission from the sympatric l Hoesti supergroup. The cross-species transmission event resulting in the emergence of SIVmnd-1 was probably ancient, before the subspeciation of mandrills, which occurred about 1 million years ago (Telfer et al., 2003). Previously, we demonstrated that the pathogenic potential of SIVmnd-1 in mandrills is not different from that of other species-specific SIVs (Muller & Barré-Sinoussi, 2003; Pandrea et al., 2006; Silvestri et al., 2003) and we reported that there was no pathogenic difference between SIVmnd-1 and SIVmnd-2 in mandrills (Onanga et al., 2002, 2006). In the present study, we first confirmed these findings and showed again, although in a limited number of mandrills, that there was no discernible difference in biological or clinical outcome between SIVmnd-1 and SIVmnd-2. The main objective of this study, however, was to compare the outcome of the cross-species transmission of these viruses in rhesus macaques. We have shown here that both SIVmnd-1 and SIVmnd-2 induce persistent infections in rhesus macaques. We found different patterns of T-cell activation and antibody production between SIVmnd-infected rhesus macaques and mandrills. In addition, we observed differences in the secretion of pro-inflammatory cytokines, as illustrated by the production of IFN-c, which was significantly higher in rhesus macaques than in mandrills. These findings confirm previously reported divergent host responses between pathogenic and non-pathogenic SIV infections (Silvestri et al., 2005). Moreover, during the follow-up period, a significant and persistent CD4 + T-cell depletion was observed in SIVmnd- 1-infected rhesus macaques. This result was somewhat unexpected, as SIVmnd-1 is a virus acquired by mandrills through cross-species transmission (Beer et al., 1999; Souquiere et al., 2001) and is more closely related to Cercopithecus-specific SIVs. Based on the results of previous studies, we would have expected Cercopithecusspecific SIVs to be associated with lower pathogenicity in rhesus macaques. However, SIVmnd-1-infected rhesus macaques showed immunological signs of a more pathogenic infection, as supported by continuous CD4 + T-cell decline. Together with the similar pathogenicity of SIVmnd-1 and SIVmnd-2 in mandrills, these results suggest that SIVmnd-1 is currently well adapted for replication and persistence in Papionini hosts. SIVmnd-1 adaptation to its new mandrill host is probably the best explanation for the relatively high pathogenicity of this virus in rhesus macaques, as illustrated by persistent infection and CD4 + T-cell depletion. Immune activation of T cells and plasma IFN-c secretion were identical for both types of SIVmnd in rhesus macaques, which is apparently in disagreement with the different pathogenic outcomes of the two infections. Nevertheless, our results did not exclude the possibility that SIVmnd-2-infected rhesus macaques may progress to AIDS after a longer incubation period beyond the followup period in this study. Alternatively, it is possible that the pathogenicity of these two viruses differs because of different mechanisms of CD4 + T-cell depletion in SIVmnd-1 and SIVmnd-2 infections. A previous study reported CXCR4 co-receptor usage for the SIVmnd-1 strain GB1, from which the virus strains used in the present study are derived (Schols & De Clercq, 1998), and CXCR4- tropic SIVs are more prone to induce CD4 + T-cell depletion at extra-intestinal mucosal sites. In contrast, CCR5-tropic SIVs, such as SIVmnd-2, are more prone to induce intestinal depletion of effector memory CD4 + T cells, as described previously for SIVsmm strains (Brown et al., 2007; Nishimura et al., 2007). In order to address this question, investigation of the mucosal pathogenicity of these two viruses is needed. The difference in pathogenic potential for rhesus macaques between SIVmnd-2, which induced persistent infection, and SIVrcm, which is controlled in rhesus macaques, may also appear somewhat surprising in the context of the close phylogenetic relationship between these two viruses. However, this difference could also be associated with the fact that these viruses utilize different receptors, specifically CCR2b in the case of SIVrcm (Chen et al., 1998) and CCR5 in the case of SIVmnd-2 (Hu et al., 2003). In addition, this difference may be related to the recombinant nature of SIVmnd-2, which has thus gained an evolutionary advantage compared with SIVrcm. Similar recombination 496 Journal of General Virology 90

10 SIVmnd types 1 and 2 infection of rhesus macaques events have been described for SIVcpz, which evolved as a chimpanzee virus following recombination of viruses related to SIVrcm and SIVgsn/mon/mus (Bailes et al., 2003). This relatively high propensity for successful crossspecies transmission of the SIVmnd viruses, one already cross-species transmitted and the second a recombinant virus, suggests that host species barriers can be overcome relatively easily by cross-species-transmitted SIVs through recombination and adaptation. As SIVcpz is the direct ancestor of HIV-1, this observation calls for effective measures for prevention of cross-species transmission of SIVs to humans in central Africa, the area of endemicity of most SIVs. ACKNOWLEDGEMENTS This work was funded by the Centre International de Recherches Médicales de Franceville (CIRMF), Gabon. CIRMF is supported by the Government of Gabon, Total-Elf Gabon and the Ministère de la Coopération Française. We would also like to thank the staff of the CIRMF Primate Center. We thank Patricia Reed for critical reading of the manuscript. I. P. and C. A. are supported by grants RO1 AI and R21AI (I. P.) and RO1 AI (C. A.) from the National Institute of Health. REFERENCES Apetrei, C., Robertson, D. L. & Marx, P. A. (2004). The history of SIVS and AIDS: epidemiology, phylogeny and biology of isolates from naturally SIV infected non-human primates (NHP) in Africa. Front Biosci 9, Apetrei, C., Kaur, A., Lerche, N. W., Metzger, M., Pandrea, I., Hardcastle, J., Falkenstein, S., Bohm, R., Koehler, J. & other authors (2005). Molecular epidemiology of simian immunodeficiency virus SIVsm in U.S. primate centers unravels the origin of SIVmac and SIVstm. J Virol 79, Apetrei, C., Lerche, N. W., Pandrea, I., Gormus, B., Silvestri, G., Kaur, A., Robertson, D. L., Hardcastle, J., Lackner, A. A. & Marx, P. A. (2006). Kuru experiments triggered the emergence of pathogenic SIVmac. AIDS 20, Bailes, E., Gao, F., Bibollet-Ruche, F., Courgnaud, V., Peeters, M., Marx, P. A., Hahn, B. H. & Sharp, P. M. (2003). Hybrid origin of SIV in chimpanzees. Science 300, Beer, B. E., Bailes, E., Goeken, R., Dapolito, G., Coulibaly, C., Norley, S. G., Kurth, R., Gautier, J. P., Gautier-Hion, A. & other authors (1999). Simian immunodeficiency virus (SIV) from sun-tailed monkeys (Cercopithecus solatus): evidence for host-dependent evolution of SIV within the C. lhoesti superspecies. J Virol 73, Beer, B. E., Brown, C. R., Whitted, S., Goldstein, S., Goeken, R., Plishka, R.,Buckler-White,A.&Hirsch,V.M.(2005).Immunodeficiency in the absence of high viral load in pig-tailed macaques infected with simian immunodeficiency virus SIVsun or SIVlhoest. JVirol79, Brown, C. R., Czapiga, M., Kabat, J., Dang, Q., Ourmanov, I., Nishimura, Y., Martin, M. A. & Hirsch, V. M. (2007). Unique pathology in simian immunodeficiency virus-infected rapid progressor macaques is consistent with a pathogenesis distinct from that of classical AIDS. J Virol 81, Chakrabarti, L. A. (2004). The paradox of simian immunodeficiency virus infection in sooty mangabeys: active viral replication without disease progression. Front Biosci 9, Chen, Z., Gettie, A., Ho, D. D. & Marx, P. A. (1998). Primary SIVsm isolates use the CCR5 coreceptor from sooty mangabeys naturally infected in West Africa: a comparison of coreceptor usage of primary SIVsm, HIV-2, and SIVmac. Virology 246, Clewley, J. P., Lewis, J. C., Brown, D. W. & Gadsby, E. L. (1998). A novel simian immunodeficiency virus (SIVdrl) pol sequence from the drill monkey, Mandrillus leucophaeus. J Virol 72, Dunham, R., Pagliardini, P., Gordon, S., Sumpter, B., Engram, J., Moanna, A., Paiardini, M., Mandl, J. N., Lawson, B. & other authors (2006). The AIDS resistance of naturally SIV-infected sooty mangabeys is independent of cellular immunity to the virus. Blood 108, Estaquier, J., Idziorek, T., de Bels, F., Barré-Sinoussi, F., Hurtrel, B., Aubertin, A. M., Venet, A., Mehtali, M., Muchmore, E. & other authors (1994). Programmed cell death and AIDS: significance of T-cell apoptosis in pathogenic and nonpathogenic primate lentiviral infections. Proc Natl Acad Sci U S A 91, Fultz, P. N. (1991). Replication of an acutely lethal simian immunodeficiency virus activates and induces proliferation of lymphocytes. J Virol 65, Gordon, S. N., Klatt, N. R., Bosinger, S. E., Brenchley, J. M.,, Milush, J. M., Engram, J. C., Dunham, R. M., Paiardini, M., Klucking, S. & other authors (2007). Severe depletion of mucosal CD4 + T cells in AIDSfree simian immunodeficiency virus-infected sooty mangabeys. J Immunol 179, Gormus, B. J., Martin, L. N. & Baskin, G. B. (2004). A brief history of the discovery of natural simian immunodeficiency virus (SIV) infections in captive sooty mangabey monkeys. Front Biosci 9, Hahn, B. H., Shaw, G. M., De Cock, K. M. & Sharp, P. M. (2000). AIDS as a zoonosis: scientific and public health implications. Science 287, Harris, E. E. & Disotell, T. R. (1998). Nuclear gene trees and the phylogenetic relationships of the mangabeys (Primates: Papionini). Mol Biol Evol 15, Hirsch, V. M. (2004). What can natural infection of African monkeys with simian immunodeficiency virus tell us about the pathogenesis of AIDS? AIDS Rev 6, Hirsch, V. M. & Johnson, P. R. (1994). Pathogenic diversity of simian immunodeficiency viruses. Virus Res 32, Hirsch, V. M., Olmsted, R. A., Murphey-Corb, M., Purcell, R. H. & Johnson, P. R. (1989). An African primate lentivirus (SIVsm) closely related to HIV-2. Nature 339, Hirsch, V. M., Dapolito, G. A., Goldstein, S., McClure, H., Emau, P., Fultz, P. N., Isahakia, M., Lenroot, R., Myers, G. & Johnson, P. R. (1993). A distinct African lentivirus from Sykes monkeys. J Virol 67, Hirsch, V. M., Dapolito, G., Johnson, P. R., Elkins, W. R., London, W. T., Montali, R. J., Goldstein, S. & Brown, C. (1995). Induction of AIDS by simian immunodeficiency virus from an African green monkey: species-specific variation in pathogenicity correlates with the extent of in vivo replication. J Virol 69, Hirsch, V. M., Campbell, B. J., Bailes, E., Goeken, R., Brown, C., Elkins, W. R., Axthelm, M., Murphey-Corb, M. & Sharp, P. M. (1999). Characterization of a novel simian immunodeficiency virus (SIV) from l hoest monkeys (Cercopithecus l hoesti): implications for the origins of SIVmnd and other primate lentiviruses. J Virol 73, Hu, J., Switzer, W. M., Foley, B. T., Robertson, D. L., Goeken, R. M., Korber, B. T., Hirsch, V. M. & Beer, B. E. (2003). Characterization and comparison of recombinant simian immunodeficiency virus from drill (Mandrillus leucophaeus) and mandrill (Mandrillus sphinx) isolates. J Virol 77,

11 S. Souquière and others Kaur, A., Grant, R. M., Means, R. E., McClure, H., Feinberg, M. & Johnson, R. P. (1998). Diverse host responses and outcomes following simian immunodeficiency virus SIVmac239 infection in sooty mangabeys and rhesus macaques. J Virol 72, Keele, B. F., Van Heuverswyn, F., Li, Y., Bailes, E., Takehisa, J., Santiago, M. L., Bibollet-Ruche, F., Chen, Y., Wain, L. V. & other authors (2006). Chimpanzee reservoirs of pandemic and nonpandemic HIV-1. Science 313, Kornfeld,C.,Ploquin,M.J.,Pandrea,I.,Faye,A.,Onanga,R.,Apetrei,,C., Poaty-Mavoungou, V., Rouquet, P., Estaquier, J. & other authors (2005). Antiinflammatory profiles during primary SIV infection in African green monkeys are associated with protection against AIDS. J Clin Invest 115, Ling, B., Apetrei, C., Pandrea, I., Veazey, R. S., Lackner, A. A., Gormus, B. & Marx, P. A. (2004). Classic AIDS in a sooty mangabey after an 18-year natural infection. J Virol 78, Mansfield, K. G., Lerch, N. W., Gardner, M. B. & Lackner, A. A. (1995). Origins of simian immunodeficiency virus infection in macaques at the New England Regional Primate Research Center. J Med Primatol 24, Marx, P. A., Li, Y., Lerche, N. W., Sutjipto, S., Gettie, A., Yee, J. A., Brotman, B. H., Prince, A. M., Hanson, A. & other authors (1991). Isolation of a simian immunodeficiency virus related to human immunodeficiency virus type 2 from a West African pet sooty mangabey. J Virol 65, Marx, P. A., Alcabes, P. G. & Drucker, E. (2001). Serial human passage of simian immunodeficiency virus by unsterile injections and the emergence of epidemic human immunodeficiency virus in Africa. Philos Trans R Soc Lond B Biol Sci 356, McClure, H. M., Anderson, D. C., Fultz, P. N., Ansari, A. A., Lockwood, E. & Brodie, A. (1989). Spectrum of disease in macaque monkeys chronically infected with SIV/SMM. Vet Immunol Immunopathol 21, Muller, M. C. & Barré-Sinoussi, F. (2003). SIVagm: genetic and biological features associated with replication. Front Biosci 8, d1170 d1185. Murphey-Corb,M.,Martin,L.N.,Rangan,S.R.,Baskin,G.B.,Gormus, B.J.,Wolf,R.H.,Andes,W.A.,West,M.&Montelaro,R.C.(1986).Isolation of an HTLV-III-related retrovirus from macaques with simian AIDS and its possible origin in asymptomatic mangabeys. Nature 321, Nishimura, Y., Igarashi, T., Buckler-White, A., Buckler, C., Imamichi, H., Goeken,R.M.,Lee,W.R.,Lafont,B.A.,Byrum,R.&otherauthors (2007). Loss of naive cells accompanies memory CD4 + T-cell depletion during long-term progression to AIDS in simian immunodeficiency virus-infected macaques. JVirol81, Norley, S., Beer, B., Holzammer, S., zur Megede, J. & Kurth, R. (1999). Why are the natural hosts of SIV resistant to AIDS? Immunol Lett 66, Onanga, R., Kornfeld, C., Pandrea, I., Estaquier, J., Souquière, S., Rouquet, P., Mavoungou, V. P., Bourry, O., M Boup, S. & other authors (2002). 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., Souquière, S., Makuwa, M., Mouinga-Ondeme, A., Simon, F., Apetrei, C. & Roques, P. (2006). Primary simian immunodeficiency virus SIVmnd-2 infection in mandrills (Mandrillus sphinx). J Virol 80, Osterhaus, A. D., Pedersen, N., van Amerongen, G., Frankenhuis, M. T., Marthas, M., Reay, E., Rose, T. M., Pamungkas, J. & Bosch, M. L. (1999). Isolation and partial characterization of a lentivirus from talapoin monkeys (Myopithecus talapoin). Virology 260, Pandrea, I., Onanga, R., Rouquet, P., Bourry, O., Ngari, P., Wickings, E. J., Roques, P. & Apetrei, C. (2001). Chronic SIV infection ultimately causes immunodeficiency in African non-human primates. AIDS 15, Pandrea, I., Kornfeld, C., Ploquin, M. J., Apetrei, C., Faye, A., Rouquet, P., Roques, P., Simon, F., Barré-Sinoussi, F. & other authors (2005). Impact of viral factors on very early in vivo replication profiles in simian immunodeficiency virus SIVagminfected African green monkeys. J Virol 79, Pandrea, I., Silvestri, G., Onanga, R., Veazey, R. S., Marx, P. A., Hirsch, V. & Apetrei, C. (2006). Simian immunodeficiency viruses replication dynamics in African non-human primate hosts: common patterns and species-specific differences. J Med Primatol 35, Pandrea, I. V., Gautam, R., Ribeiro, R. M., Brenchley, J. M., Butler, I. F., Pattison, M., Rasmussen, T., Marx, P. A., Silvestri, G. & other authors (2007). Acute loss of intestinal CD4 + T cells is not predictive of simian immunodeficiency virus virulence. J Immunol 179, Pandrea, I., Onanga, R., Souquière, S., Mouinga-Ondéme, A., Bourry, O., Makuwa, M., Rouquet, P., Silvestri, G., Simon, F. & other authors (2008a). Paucity of CD4 + CCR5 + T cells may prevent transmission of simian immunodeficiency virus in natural nonhuman primate hosts by breast-feeding. J Virol 82, Pandrea, I., Ribeiro, R. M., Gautam, R., Gaufin, T., Pattison, M., Barnes, M., Monjure, C., Stoulig, C., Dufour, J. & other authors (2008b). SIVagm dynamics in African green monkeys. J Virol 82, Peeters, M., Courgnaud, V., Abela, B., Auzel, P., Pourrut, X., Bibollet- Ruche, F., Loul, S., Liegeois, F., Butel, C. & other authors (2002). Risk to human health from a plethora of simian immunodeficiency viruses in primate bushmeat. Emerg Infect Dis 8, Schols, D. & De Clercq, E. (1998). The simian immunodeficiency virus mnd(gb-1) strain uses CXCR4, not CCR5, as coreceptor for entry in human cells. J Gen Virol 79, Silvestri, G. (2005). Naturally SIV-infected sooty mangabeys: are we closer to understanding why they do not develop AIDS? J Med Primatol 34, Silvestri, G., Sodora, D. L., Koup, R. A., Paiardini, M., O Neil, S. P., McClure, H. M., Staprans, S. I. & Feinberg, M. B. (2003). Nonpathogenic SIV infection of sooty mangabeys is characterized by limited bystander immunopathology despite chronic high-level viremia. Immunity 18, Silvestri, G., Fedanov, A., Germon, S., Kozyr, N., Kaiser, W. J., Garber, D. A., McClure, H., Feinberg, M. B. & Staprans, S. I. (2005). Divergent host responses during primary simian immunodeficiency virus SIVsm infection of natural sooty mangabey and nonnatural rhesus macaque hosts. J Virol 79, Simon, F., Souquière, S., Damond, F., Kfutwah, A., Makuwa, M., Leroy, E., Rouquet, P., Berthier, J. L., Rigoulet, J. & other authors (2001). Synthetic peptide strategy for the detection of and discrimination among highly divergent primate lentiviruses. AIDS Res Hum Retroviruses 17, Smith, S. M., Makuwa, M., Lee, F., Gettie, A., Russo, C. & Marx, P. A. (1998). SIVrcm infection of macaques. J Med Primatol 27, Souquière, S., Bibollet-Ruche, F., Robertson, D. L., Makuwa, M., Apetrei, C., Onanga, R., Kornfeld, C., Plantier, J. C., Gao, F. & other authors (2001). Wild Mandrillus sphinx are carriers of two types of lentivirus. J Virol 75, Takehisa, J., Harada, Y., Ndembi, N., Mboudjeka, I., Taniguchi, Y., Ngansop, C., Kuate, S., Zekeng, L., Ibuki, K. & other authors (2001). Natural infection of wild-born mandrills (Mandrillus sphinx) with two different types of simian immunodeficiency virus. AIDS Res Hum Retroviruses 17, Telfer, P. T., Souquière, S., Clifford, S. L., Abernethy, K. A., Bruford, M. W., Disotell, T. R., Sterner, K. N., Roques, P., Marx, P. A. & 498 Journal of General Virology 90

12 SIVmnd types 1 and 2 infection of rhesus macaques Wickings, E. J. (2003). Molecular evidence for deep phylogenetic divergence in Mandrillus sphinx. Mol Ecol 12, Traina-Dorge, V., Blanchard, J., Martin, L. & Murphey-Corb, M. (1992). Immunodeficiency and lymphoproliferative disease in an African green monkey dually infected with SIV and STLV-I. AIDS Res Hum Retroviruses 8, Tsujimoto, H., Cooper, R. W., Kodama, T., Fukasawa, M., Miura, T., Ohta, Y., Ishikawa, K., Nakai, M., Frost, E. & other authors (1988). Isolation and characterization of simian immunodeficiency virus from mandrills in Africa and its relationship to other human and simian immunodeficiency viruses. J Virol 62, Tsujimoto, H., Hasegawa, A., Maki, N., Fukasawa, M., Miura, T., Speidel, S., Cooper, R. W., Moriyama, E. N., Gojobori, T. & Hayami, M. (1989). Sequence of a novel simian immunodeficiency virus from a wild-caught African mandrill. Nature 341, VandeWoude, S. & Apetrei, C. (2006). Going wild: lessons from naturally occurring T-lymphotropic lentiviruses. Clin Microbiol Rev 19, Wang, Z., Metcalf, B., Ribeiro, R. M., McClure, H. & Kaur, A. (2006). Th-1-type cytotoxic CD8 + T-lymphocyte responses to simian immunodeficiency virus (SIV) are a consistent feature of natural SIV infection in sooty mangabeys. J Virol 80,

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

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

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

A novel simian immunodeficiency virus from black mangabey (Lophocebus aterrimus) in the Democratic Republic of Congo

A novel simian immunodeficiency virus from black mangabey (Lophocebus aterrimus) in the Democratic Republic of Congo Journal of General Virology (2005), 86, 1967 1971 DOI 10.1099/vir.0.80697-0 Short Communication A novel simian immunodeficiency virus from black mangabey (Lophocebus aterrimus) in the Democratic Republic

More information

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

Simian Immunodeficiency Virus SIVagm.sab Infection of Caribbean African Green Monkeys: a New Model for the Study of SIV Pathogenesis in Natural Hosts JOURNAL OF VIROLOGY, May 2006, p. 4858 4867 Vol. 80, No. 10 0022-538X/06/$08.00 0 doi:10.1128/jvi.80.10.4858 4867.2006 Copyright 2006, American Society for Microbiology. All Rights Reserved. Simian Immunodeficiency

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

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

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

Wild Mandrillus sphinx Are Carriers of Two Types of Lentivirus

Wild Mandrillus sphinx Are Carriers of Two Types of Lentivirus JOURNAL OF VIROLOGY, Aug. 2001, p. 7086 7096 Vol. 75, No. 15 0022-538X/01/$04.00 0 DOI: 10.1128/JVI.75.15.7086 7096.2001 Copyright 2001, American Society for Microbiology. All Rights Reserved. Wild Mandrillus

More information

OPEN ACCESS JZAR Evidence-based practice. Evidence-based practice. Introduction. Methods. M.P. Hartley 1,* and F. Schmidt 2

OPEN ACCESS JZAR Evidence-based practice. Evidence-based practice. Introduction. Methods. M.P. Hartley 1,* and F. Schmidt 2 OPEN ACCESS JZAR Evidence-based practice Evidence-based practice The use of risk analysis methodology to generate evidence-based decision making in zoo animal disease management: using simian immunodeficiency

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

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

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

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

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

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

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

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

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

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

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

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

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

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

CONCISE COMMUNICATION

CONCISE COMMUNICATION 1791 CONCISE COMMUNICATION Detection of Diverse Variants of Human Immunodeficiency Virus 1 Groups M, N, and O and Simian Immunodeficiency Viruses from Chimpanzees by Using Generic pol and env Primer Pairs

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

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

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

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

Received 30 January 2002/Accepted 10 May 2002

Received 30 January 2002/Accepted 10 May 2002 JOURNAL OF VIROLOGY, Aug. 2002, p. 8298 8309 Vol. 76, No. 16 0022-538X/02/$04.00 0 DOI: 10.1128/JVI.76.16.8298 8309.2002 Copyright 2002, American Society for Microbiology. All Rights Reserved. Characterization

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

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

University of Pittsburgh 9044 Biomedical Science Tower Fifth Avenue Pittsburgh, PA 15261

University of Pittsburgh 9044 Biomedical Science Tower Fifth Avenue Pittsburgh, PA 15261 2/22/10 CURRICULUM VITAE NAME: Cristian Apetrei BUSINESS ADDRESS: Center for Vaccine Research University of Pittsburgh 9044 Biomedical Science Tower 3 3501 Fifth Avenue Pittsburgh, PA 15261 CITZENSHIP:

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

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

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

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

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

Immunodeficiency Virus Type 2 from a West African

Immunodeficiency Virus Type 2 from a West African JOURNAL OF VIROLOGY, Aug. 1991, p. 448-4485 22-58X/91/8448-6$2./ Copyright 1991, American Society for Microbiology Vol. 65, No. 8 Isolation of a Simian Immunodeficiency Virus Related to Human Immunodeficiency

More information

Supporting Online Material for

Supporting Online Material for www.sciencemag.org/cgi/content/full/329/5998/1487/dc1 Supporting Online Material for Island Biogeography Reveals the Deep History of SIV Michael Worobey, Paul Telfer, Sandrine Souquière, Meredith Hunter,

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

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

30 Years with AIDS: Where it Came From and Why It s Still With Us. Kevin McCarthy Ben Morris Jamie Schafer

30 Years with AIDS: Where it Came From and Why It s Still With Us. Kevin McCarthy Ben Morris Jamie Schafer 30 Years with AIDS: Where it Came From and Why It s Still With Us Kevin McCarthy Ben Morris Jamie Schafer 30 Years with AIDS: Where it Came From and Why It s Still With Us Kevin McCarthy Part I: The Origins

More information

MINIREVIEW. Simian Immunodeficiency Virus Infection of Chimpanzees

MINIREVIEW. Simian Immunodeficiency Virus Infection of Chimpanzees JOURNAL OF VIROLOGY, Apr. 2005, p. 3891 3902 Vol. 79, No. 7 0022-538X/05/$08.00 0 doi:10.1128/jvi.79.7.3891 3902.2005 Copyright 2005, American Society for Microbiology. All Rights Reserved. MINIREVIEW

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

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

Going Nowhere Fast: Lentivirus genetic sequence evolution does not correlate with phenotypic evolution.

Going Nowhere Fast: Lentivirus genetic sequence evolution does not correlate with phenotypic evolution. Going Nowhere Fast: Lentivirus genetic sequence evolution does not correlate with phenotypic evolution. Brian T. Foley, PhD btf@lanl.gov HIV Genetic Sequences, Immunology, Drug Resistance and Vaccine Trials

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

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

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

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

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

Availability of activated CD4+ T cells dictates the level of viremia in naturally SIV-infected sooty mangabeys Availability of activated CD4+ T cells dictates the level of viremia in naturally SIV-infected sooty mangabeys Nichole R. Klatt, Emory University Francois Villinger, Emory University Pavel Bostik, Emory

More information

Going Wild: Lessons from Naturally Occurring T-Lymphotropic Lentiviruses

Going Wild: Lessons from Naturally Occurring T-Lymphotropic Lentiviruses CLINICAL MICROBIOLOGY REVIEWS, Oct. 2006, p. 728 762 Vol. 19, No. 4 0893-8512/06/$08.00 0 doi:10.1128/cmr.00009-06 Copyright 2006, American Society for Microbiology. All Rights Reserved. Going Wild: Lessons

More information

Human immunodeficiency viruses HIV-1

Human immunodeficiency viruses HIV-1 Origins of HIV and the Evolution of Resistance to AIDS Jonathan L. Heeney, 1 * Angus G. Dalgleish, 2 Robin A. Weiss 3 The cross-species transmission of lentiviruses from African primates to humans has

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

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

The Presence of a vpu Gene and the Lack of Nef-Mediated Downmodulation of T Cell Receptor-CD3 Are Not Always Linked in Primate Lentiviruses

The Presence of a vpu Gene and the Lack of Nef-Mediated Downmodulation of T Cell Receptor-CD3 Are Not Always Linked in Primate Lentiviruses JOURNAL OF VIROLOGY, Jan. 2011, p. 742 752 Vol. 85, No. 2 0022-538X/11/$12.00 doi:10.1128/jvi.02087-10 Copyright 2011, American Society for Microbiology. All Rights Reserved. The Presence of a vpu Gene

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

Nef-Mediated Suppression of T Cell Activation Was Lost in a Lentiviral LineagethatGaveRisetoHIV-1

Nef-Mediated Suppression of T Cell Activation Was Lost in a Lentiviral LineagethatGaveRisetoHIV-1 Nef-Mediated Suppression of T Cell Activation Was Lost in a Lentiviral LineagethatGaveRisetoHIV-1 Michael Schindler, 1 Jan Münch, 1 Olaf Kutsch, 2 Hui Li, 2 Mario L. Santiago, 2 Frederic Bibollet-Ruche,

More information

Case Study. The origins and evolution of HIV STUDENT S GUIDE. Anne Fischer. Dean Madden [Ed.] Version 1.2

Case Study. The origins and evolution of HIV STUDENT S GUIDE. Anne Fischer. Dean Madden [Ed.] Version 1.2 STUDENT S GUIDE Case Study The origins and evolution of HIV Version 1.2 Anne Fischer Formerly of the Max Planck Institute for Evolutionary Anthropology, Leipzig Dean Madden [Ed.] NCBE, University of Reading

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

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

SUPPLEMENTARY INFORMATION

SUPPLEMENTARY INFORMATION ` SUPPLEMENTAL FIGURES doi:10.1038/nature10003 Supplemental Figure 1: RhCMV/SIV vectors establish and indefinitely maintain high frequency SIV-specific T cell responses in diverse tissues: The figure shows

More information

Complete-genome analysis of hepatitis B virus from wild-born chimpanzees in central Africa demonstrates a strain-specific geographical cluster

Complete-genome analysis of hepatitis B virus from wild-born chimpanzees in central Africa demonstrates a strain-specific geographical cluster Journal of General Virology (2007), 88, 2679 2685 DOI 10.1099/vir.0.82992-0 Short Communication Correspondence Maria Makuwa mmakuwa@yahoo.com Complete-genome analysis of hepatitis B virus from wild-born

More information

Under the Radar Screen: How Bugs Trick Our Immune Defenses

Under the Radar Screen: How Bugs Trick Our Immune Defenses Under the Radar Screen: How Bugs Trick Our Immune Defenses Session 7: Cytokines Marie-Eve Paquet and Gijsbert Grotenbreg Whitehead Institute for Biomedical Research HHV-8 Discovered in the 1980 s at the

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

Analysis of the Evolutionary Relationships of HIV-1 and SIVcpz Sequences Using Bayesian Inference: Implications for the Origin of HIV-1

Analysis of the Evolutionary Relationships of HIV-1 and SIVcpz Sequences Using Bayesian Inference: Implications for the Origin of HIV-1 Analysis of the Evolutionary Relationships of HIV-1 and SIVcpz Sequences Using Bayesian Inference: Implications for the Origin of HIV-1 D. Paraskevis,* P. Lemey,* M. Salemi,* M. Suchard, Y. Van de Peer,à

More information

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

Combined IL-21 and IFNα treatment limits inflammation and delay viral rebound in ART-treated, SIV-infected macaques Combined and IFNα treatment limits inflammation and delay viral rebound in ART-treated, SIV-infected macaques Mirko Paiardini Associate Professor, Emory University School of Medicine Yerkes National Primate

More information

Nef-mediated tetherin antagonism

Nef-mediated tetherin antagonism Nef-mediated tetherin antagonism Frank Kirchhoff Institute of Molecular Virology Ulm Medical Center, Germany 9/11 African primates represent a large reservoir for immunodeficiency viruses and chimpanzees,

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 / AIDS Pathogenesis 1 (untreated infection)

HIV-1 / AIDS Pathogenesis 1 (untreated infection) HIV-1 / AIDS Pathogenesis 1 (untreated infection) Henning Drechsler, MD VA North Texas Health Care System UT Southwestern Medical Center Objectives Characterize HIV-1 Describe the clinical course and immunopathology

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

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

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

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

Mayo Clinic HIV ecurriculum Series Essentials of HIV Medicine Module 2 HIV Virology

Mayo Clinic HIV ecurriculum Series Essentials of HIV Medicine Module 2 HIV Virology Mayo Clinic HIV ecurriculum Series Essentials of HIV Medicine Module 2 HIV Virology Eric M. Poeschla, MD Professor of Medicine College of Medicine Consultant, Division of Infectious Diseases Mayo Clinic

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

Blocking TLR7- and TLR9-mediated IFN-a Production by Plasmacytoid Dendritic Cells Does Not Diminish Immune Activation in Early SIV Infection

Blocking TLR7- and TLR9-mediated IFN-a Production by Plasmacytoid Dendritic Cells Does Not Diminish Immune Activation in Early SIV Infection Blocking TLR7- and TLR9-mediated IFN-a Production by Plasmacytoid Dendritic Cells Does Not Diminish Immune Activation in Early SIV Infection Muhamuda Kader 1,2, Amanda P. Smith 1,2, Cristiana Guiducci

More information

HIV / AIDS Pathogenesis 1

HIV / AIDS Pathogenesis 1 HIV / AIDS Pathogenesis 1 Henning Drechsler, MD VA North Texas Health Care System UT Southwestern Medical Center Objectives Characterize HIV-1 (classification, structure and life cycle) Describe the clinical

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

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

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

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

The Harvard community has made this article openly available. Please share how this access benefits you. Your story matters

The Harvard community has made this article openly available. Please share how this access benefits you. Your story matters Emergence of simian immunodeficiency virus in rhesus macaques is characterized by changes in structural and accessory genes that enhance fitness in the new host species The Harvard community has made this

More information

Fayth K. Yoshimura, Ph.D. September 7, of 7 RETROVIRUSES. 2. HTLV-II causes hairy T-cell leukemia

Fayth K. Yoshimura, Ph.D. September 7, of 7 RETROVIRUSES. 2. HTLV-II causes hairy T-cell leukemia 1 of 7 I. Diseases Caused by Retroviruses RETROVIRUSES A. Human retroviruses that cause cancers 1. HTLV-I causes adult T-cell leukemia and tropical spastic paraparesis 2. HTLV-II causes hairy T-cell leukemia

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

AIDS 111. The origins and diversification of HIV INTRODUCTION THE DEEP ROOTS OF HIV

AIDS 111. The origins and diversification of HIV INTRODUCTION THE DEEP ROOTS OF HIV AIDS 111 The origins and diversification of HIV INTRODUCTION Human immunodeficiency virus (HIV) is not one but several related viruses that have crossed into the human population on multiple occasions

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

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

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

Lecture 12. Immunology and disease: parasite antigenic diversity. and. Phylogenetic trees

Lecture 12. Immunology and disease: parasite antigenic diversity. and. Phylogenetic trees Lecture 12 Immunology and disease: parasite antigenic diversity and Phylogenetic trees Benefits of antigenic variation 2. Infect hosts with prior exposure Hosts often maintain memory against prior infections,

More information

Simian retroviruses in African apes

Simian retroviruses in African apes REVIEW 10.1111/j.1469-0691.2012.03843.x Simian retroviruses in African apes M. Peeters and E. Delaporte UMI 233, TransVIHMI, Institut de Recherche pour le Développement and University of Montpellier 1,

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

HIV Pathogenesis and Natural History. Peter W. Hunt, MD Associate Professor of Medicine University of California San Francisco

HIV Pathogenesis and Natural History. Peter W. Hunt, MD Associate Professor of Medicine University of California San Francisco HIV Pathogenesis and Natural History Peter W. Hunt, MD Associate Professor of Medicine University of California San Francisco Learning Objectives Describe key features of HIV pathogenesis and natural history

More information

The quality of chimpanzee T cell activation and SIV/HIV susceptibility. achieved via antibody-mediated TCR/CD3 stimulation ACCEPTED

The quality of chimpanzee T cell activation and SIV/HIV susceptibility. achieved via antibody-mediated TCR/CD3 stimulation ACCEPTED JVI Accepts, published online ahead of print on 30 July 08 J. Virol. doi:.1128/jvi.01319-08 Copyright 08, American Society for Microbiology and/or the Listed Authors/Institutions. All Rights Reserved.

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

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

Inves)gación básica y curación del VIH- 1

Inves)gación básica y curación del VIH- 1 Inves)gación básica y curación del VIH- 1 Javier Mar)nez- Picado (jmpicado@irsicaixa.es) 23 rd Conference on Retroviruses and Opportunis5c Infec5ons February 22-25, 2016 Boston, Massachuse8s UNIVERSITAT

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