Potential Contributions of Viral Envelope and ... - Journal of Virology

3 downloads 98 Views 221KB Size Report
nant plasmids (25 g) were transfected into 293T cells (5. 106 cells) by standard ..... Choe, H., M. Farzan, Y. Sun, N. Sullivan, B. Rollins, P. D. Ponath, L. Wu, ..... Muller, H. Muller-Hermelink, D. Fuchs, H. Wachter, M. Daniel, R. Desro- siers, and ...
JOURNAL OF VIROLOGY, Nov. 1998, p. 8650–8658 0022-538X/98/$04.0010 Copyright © 1998, American Society for Microbiology. All Rights Reserved.

Vol. 72, No. 11

Potential Contributions of Viral Envelope and Host Genetic Factors in a Human Immunodeficiency Virus Type 1-Infected Long-Term Survivor KATHIE GROVIT-FERBAS,1,2 JOHN FERBAS,1,2 VAHEIDEH GUDEMAN,1,3 SAEED SADEGHI,2 MATTHEW BIDWELL GOETZ,1,2 JANIS V. GIORGI,1 IRVIN S. Y. CHEN,1,3* 1,2 AND WILLIAM A. O’BRIEN † Division of Hematology-Oncology, Department of Medicine1 and Department of Microbiology and Immunology,3 UCLA School of Medicine and UCLA AIDS Institute, Los Angeles, California 90095-1678, and VA Medical Center, Los Angeles, California 900732 Received 1 April 1998/Accepted 20 July 1998

The lack of clinical progression in some individuals despite prolonged human immunodeficiency virus type 1 (HIV-1) infection may result from infection with less-pathogenic viral strains. To address this question, we examined the HIV-1 envelope protein from a donor with a low viral burden, stable CD41 T-lymphocyte counts, and little evidence of CD81 T-cell expansion, activation, or immune activity. To avoid potential changes in envelope function resulting from selection in vitro, envelope clones were constructed by using viral RNA isolated from uncultured peripheral blood mononuclear cells (PBMC). The data showed that recombinant viruses containing envelope sequences derived from RNA isolated from patient PBMC replicated poorly in primary CD41 T cells but demonstrated efficient growth in macrophages. The unusual phenotype of these viruses could not be explained solely by differential utilization of coreceptors since the chimeric viruses, as well as an uncloned isolate obtained from the same visit date, can utilize CCR5. In addition, the donor’s own cells appeared resistant to infection with chimeric viruses containing autologous envelope sequences. Genotype analysis revealed that the donor was heterozygous for the previously described 32-bp deletion in CCR5 which may be linked with prolonged survival in HIV-1-infected individuals. These data suggest that the changes in envelope sequences confer properties of viral attenuation, which together with the CCR5 1/D32 genotype could account for the long-term survival of this patient. 86, 89). There are a number of potential explanations for the phenomenon of slow progression, including infection with attenuated viral strains demonstrating limited capacity for highlevel replication in vivo (21, 54, 58, 64). We report here the functional characteristics of chimeric viruses containing env sequences from an LTS infected with a virus that repeatedly replicates with slower growth kinetics in coculture relative to viral isolates from other LTS and recent seroconverters. This finding suggests that the donor may be infected with a virus which possesses a selective growth disadvantage. Our studies demonstrate both HIV-1 attenuation and an apparently diminished susceptibility to infection in the donor as potential factors for slow clinical progression in this patient.

The natural history of human immunodeficiency virus type 1 (HIV-1) infection is variable among individuals and is governed by the link between viral replication and host-specific factors. In fact, several studies have demonstrated that higher levels of HIV-1 replication are associated with more-rapid clinical progression (15, 18, 46, 65, 66, 74, 79, 82, 85). Furthermore, it is well documented that HIV-1 phenotypes associated with enhanced replication have been linked with clinical progression (14, 36, 52, 72, 87, 92, 95). In contrast, progression rates are slower when there are effective immunologic responses associated with restraint of HIV-1 replication, including production of antiviral cytokines (9, 12, 13, 59), cytotoxic T-lymphocyte responses (6, 43, 96), and antibody-mediated suppression (5, 27, 44, 70, 77). Furthermore, recent use of more-potent and intensive antiretroviral chemotherapy has resulted in reductions in HIV-1 RNA load in plasma and in decreased rates of clinical progression for some patients (24, 31, 32, 40, 42, 51). Interestingly, there are individuals who remain clinically asymptomatic and demonstrate stable immunologic function in the absence of any therapeutic intervention more than 10 years after primary HIV-1 infection. Cohort studies estimate that this group of long-term survivors (LTS) or long-term nonprogressors constitutes approximately 1 to 5% of the infected population (7, 38, 58, 71,

MATERIALS AND METHODS Description of donor 6. The clinical course of this patient, referred to as donor 6 (based on a previous publication [34]), was described. In brief, this individual tested positive for HIV-1 in December 1984 at the time of first presentation to the San Francisco Men’s Health Study Clinic, with a CD41 T-cell count of 576 cells/mm3 and with a median CD41 T-cell count of 582 since that time. Viral burden at the time of testing was 6,686 copies/ml. Viral isolation in CD41 T lymphocytes. CD41 T lymphocytes from seropositive individuals were selected onto commercially available flasks coated with anti-CD4 antibodies (Applied Immune Sciences [AIS]). For this purpose, 30 3 106 peripheral blood mononuclear cells (PBMC) were added to each flask and after a 1-h incubation, the nonadherent cells were removed. Seven million allogeneic CD41 T lymphocytes previously activated with anti-CD3 (Ortho) antibody (see below for methods) (45) were added to the adherent cell population, and the cultures were maintained in the presence of 5,000 U of interleukin-2 (Chiron) per ml for 21 to 28 days. Culture supernatants were sampled every 7 days, at which time fresh medium was added to the cultures. Preparation of CD41 lymphocytes and macrophages for viral replication studies. CD41 lymphocytes from a total of six HIV-1-seronegative Red Cross Blood Bank donors were selected onto individual AIS flasks as outlined above.

* Corresponding author. Mailing address: 11-934 Factor, Department of Medicine, UCLA School of Medicine and the UCLA AIDS Institute, Los Angeles, CA 90095-1678. Phone: (310) 825-4793; Fax: (310) 794-7682. E-mail: [email protected]. † Present address: University of Texas Medical Branch, Galveston, TX 77555. 8650

VOL. 72, 1998

FACTORS CONTRIBUTING TO LONG-TERM SURVIVAL WITH HIV-1

8651

FIG. 1. Virus isolated from CD41 T lymphocytes of donor 6 grows slowly in culture relative to that of other HIV-infected individuals. CD41 lymphocytes from HIV-infected donors were cocultured with a pool of allogeneic CD41 T cells and sampled at the times indicated. CD41 lymphocytes were selected onto AIS flasks coated with anti-CD4 antibodies. Thirty million PBMC were added to each flask, and after a 1-h incubation, the nonadherent cells were removed. Seven million allogeneic CD41 lymphocytes activated with anti-CD3 antibody (45) were added to the adherent cell population, and the cultures were maintained in the presence of 5,000 U of interleukin-2 (Chiron)/ml for 21 to 28 days. Culture supernatants were sampled every 7 days, at which time fresh medium was added to the cultures. (A) CD41 lymphocytes from eight HIV-infected individuals. Donors are as indicated on the figure. Donor 13, a rapid progressor to disease, is included for the purposes of comparison. (B) CD41 lymphocytes from donor 6 were tested on four different occasions.

Each flask was treated with anti-CD3 antibody and interleukin-2 for 5 days. Pools of three donors each were prepared at the end of the incubation period and were cryopreserved for future use. Pools of three donors were used to minimize host genetic differences in infectivity potential among different donors. Cells were used for infection within 2 days after thaw. Macrophages (MA) were prepared as previously described (75). Cells from three donors were cultured for each experiment, and the cells were pooled and plated for infections on day 6. As stated above, pools of cells limited the variability seen with different donors. This was particularly important since the MA could not be cryopreserved. Infections were performed on 7-day-old MA. The MA donors were different from the CD41 T-cell donors. Amplification of viral RNA sequences. A blood sample was obtained from donor 6 in December 1994. Cell-associated RNA was isolated from PBMC using TRI-Reagent (Molecular Research Center). Purified RNA was diluted to the limit of detection for env-specific nested reverse transcription-PCR (RT-PCR). The 962-bp amplified fragment included the region encoding V1 to V3 of the HIV-1 envelope. The outer primers used for PCR (Oligos etc) were as follows: sense A2 (HIVNL4-3 [1], nucleotide positions 6320 to 6340) and antisense MFN 20 (HIVNL4-3, nucleotide positions 7352 to 7333). The inner primers were as follows: sense KPN (HIVNL4-3, nucleotide positions 6343 to 6364) and antisense MFN 17 (HIVNL4-3, nucleotide positions 7327 to 7305). Primer KPN includes the KpnI restriction site, and primer MFN 17 includes the MstII restriction site used for subsequent cloning (see below). MFN 20 was used to prime the RT reaction, using Superscript II Moloney murine leukemia virus reverse transcriptase (Gibco) in 50 mM Tris (pH 8.3), 75 mM KCl, and 3 mM MgCl2. PCR samples were gel purified and cloned into the pCRII vector (Invitrogen). Construction of full-length HIV-1 molecular clones. Patient-derived envelope sequences were excised from the pCRII vector after digestion with KpnI and MstII. The fragments were gel purified as above, and full-length HIV-1 chimeric clones were constructed. Clones were screened on the basis of restriction digestion. Sequence analysis. Plasmid DNA from full-length clones was prepared using an anion-exchange resin membrane (Qiagen, Inc.), and it was subjected to cycle sequencing with an automated DNA sequencer (Applied Biosystems, Inc.). Nucleotide sequences were aligned by Clustal analysis. Generation and testing of chimeric virus stocks. Full-length HIV-1 recombinant plasmids (25 mg) were transfected into 293T cells (5 3 106 cells) by standard methods (83). Virus supernatants were collected at 48 h and tested for p24 antigen (Coulter). Alternatively, 25 mg of DNA was electroporated into 5 3 106 phytohemagglutinin-stimulated PBMC, which were immediately cocultured with

an additional 5 3 106 PBMC from the same donor. Twenty-four-hour harvests were performed on days 4 to 7, and p24 antigen testing was performed. Viral growth kinetic studies were performed on allogeneic pools of purified monocytederived MA and anti-CD3-stimulated CD41 T cells or on autologous MA and anti-CD3-stimulated CD41 T cells. HIV-1 DNA formation was determined by quantitative PCR as previously described (75). The T-cell line-adapted HIVNL4-3 (1) and/or MA-tropic HIVSX (75), encoding Env from the brain-derived, MAtropic HIVJRFL, were used as controls. All PCR assays (Perkin-Elmer) and p24 determinations were made in duplicate. Autologous infectivity assays were performed on MA and anti-CD3-stimulated CD41 T cells isolated from donor 6. Serial half-log dilutions of viral stocks were applied to the cells for 2 h, and supernatants were changed at day 7 and harvested at day 14 to determine the 50% tissue culture infective dose/ml for each cell type. Coreceptor usage. Coreceptor usage was determined after infection of human osteosarcoma cells (HOS) (gift of D. Littman) expressing CD4 and one of the following chemokine receptors: CCR1, CCR2, CCR3, CCR4, CCR5, GPR15 (BONZO), STRL33 (BOB), or CXCR4. In addition, the cells contain the green fluorescence protein (GFP) under the control of the HIV long terminal repeat. Cultures infected by HIV-1 chimeras resulted in the production of GFP at 48 h postinfection. GFP expression was measured in formaldehyde-fixed (2%) samples by flow cytometry (Becton Dickinson; FACScan). PCR analysis of CCR5 from genomic DNA. Genomic DNA was isolated from PBMC by using DNAzol according to the manufacturer’s instructions (GIBCO BRL). Primers (CCR51 and CCR52) and amplification conditions were as described previously (94). One-tenth of the amplified product was run on a 4% NuSieve GTG agarose (FMC) gel in the presence of 10 mg of ethidium bromide/ ml. These primers yield a 172-bp fragment for the CCR5 wild-type allele and a 142-bp fragment for the CCR5 D32 allele (94).

RESULTS Slow viral growth kinetics of a virus isolated from an LTS. We have previously demonstrated that virus isolated from an LTS displays delayed growth in vitro (donor 6), suggesting infection with a growth-attenuated virus. Moreover, this donor has maintained relatively stable CD41 T-cell levels since he was first identified in 1984, with little evidence of CD81 cell activation (as measured by CD38 expression) or immune ac-

8652

GROVIT-FERBAS ET AL.

J. VIROL.

FIG. 2. Viral envelope sequences of the clones are homogeneous. Plasmid DNA from full-length clones were prepared by using an anion-exchange resin membrane (Qiagen, Inc.) and were subjected to cycle sequencing with an automated DNA sequencer (Applied Biosystems, Inc.). Potential N-linked glycosylation sites are underlined. The numbering system is based on the sequence of gp120 from HIVJRFL. Dashes indicate that the corresponding sequences of the recombinant clones containing donor 6 envelope sequences are identical to those of HIVJRFL. Amino acid substitutions are indicated, and deletions are identified by a boldface dot. (A) V1 sequences. (B) V2 sequences. (C) V3 sequences.

tivity (as measured by cytotoxic T-lymphocyte activity against gag, pol, or env targets) (34). In the current study, we examined the kinetics of virus replication in the context of eight other donors (Fig. 1A) from our first report (34). As expected, virus from donor 6 grew slowly relative to that from five of the other HIV-1-infected LTS (donors 1, 3, 4, 5, and 8) and one recent seroconverter (donor 13). Donor 6 was tested an additional two times for virus growth patterns to confirm this observation. In all instances (Fig. 1B) virus isolation still required 14 to 21 days to reach a concentration of greater than 1 ng of p24/ml, reinforcing the presence of a poorly replicating virus. Donor 2 virus also replicated with significantly diminished growth kinetics relative to that of the other donors, but on two additional occasions we were unable to recover virus and elected not to examine this individual further in our current study. Recombinant viruses containing envelope sequences from donor 6 demonstrate growth attenuation in primary CD41 T cells but not in MA. We examined whether viral envelope sequences from donor 6 contribute to the slow in vitro growth phenotype we observed with virus isolated from this LTS. For this purpose, the C1 to V3 regions of the envelope were amplified from PBMC-derived RNA. This region of the envelope was chosen for study based on the numerous phenotypic properties which have been mapped to this region, including viral tropism, phenotypic diversity, and in vitro growth characteristics (17, 23, 36, 39, 75, 76, 87). Purified RNA was diluted to the limit of detection for env-specific RT-PCR (envelope region C1 to V3). We chose to use RNA to avoid potential changes in envelope function which may result from passage and selection during the isolation of virus in vitro. The function of the am-

plified env gene product was evaluated by inserting RT-PCR fragments into the corresponding region of an infectious molecular clone (pNL4-3). The nucleotide analysis of the cloned envelope sequences for V1, V2, and V3 demonstrated that they were closely related and comprised a relatively homogeneous population (Fig. 2). In order to determine whether chimeric viruses containing env sequences from this donor recapitulated the growth attenuation phenotype observed with uncloned virus, virus stocks were generated and infection of allogeneic CD41 T cells was performed (Fig. 3). As with the uncloned virus shown in Fig. 1B, the recombinant viruses PBMC 7 (Fig. 3A), PBMC 8 (Fig. 3B), and PBMC 17 (Fig. 3D) also demonstrated significantly delayed kinetics of replication relative to that of the control strain HIVSX which contains envelope sequences from the MA-tropic strain HIVJRFL cloned into HIVNL4-3 (75). This virus strain uses CCR5 and is similar to typical MA-tropic HIV-1 strains. In contrast, the recombinant virus PBMC 14 mirrored the control virus HIVSX in its growth kinetics (Fig. 3C). Since three of the four recombinant viruses examined exhibited growth kinetics similar to that of the uncloned virus, we reasoned that circulating virus might be growth attenuated in CD41 T cells but might replicate efficiently in the other major target cell for HIV-1 infection, the monocyte-derived MA. In order to examine this hypothesis, replication kinetics were examined on pools of allogeneic MA and evaluated in the context of infection in CD41 T cells (Fig. 4). Most MA-tropic strains of HIV-1, like HIVSX (76), replicate comparably in both MA and CD41 T cells. In contrast, the chimeric viruses

VOL. 72, 1998

FACTORS CONTRIBUTING TO LONG-TERM SURVIVAL WITH HIV-1

8653

higher in MA than in CD41 T cells), levels of viral cDNA from chimeric viruses containing envelope sequences from donor 6 that demonstrated growth attenuation in CD41 T cells were 6.5-fold (PBMC 17) and 15-fold (PBMC 7) higher in MA than in CD41 T cells (Table 1). In contrast, PBMC 14 cDNA levels were only 2.5-fold higher in MA than in CD41 T cells and most closely resembled those of the typical MA-tropic strain HIVSX. This finding is consistent with the growth-attenuation phenotype observed in CD41 T cells and extends the above findings to demonstrate that differences in infectivity are most likely due to differences in viral entry. LTS envelope-containing viruses use CCR5 for infection. Viral entry is influenced by a number of parameters, including the potential use of a wide spectrum of newly described second receptors (2, 10, 26, 28, 29, 33, 60). Since we had observed a preferential replication in MA over primary CD41 T cells, we hypothesized that this phenotype might be linked to coreceptor use. Therefore, we characterized the use of known coreceptors by our chimeric viruses. For these experiments, we used stable HOS cell transfectants (GHOST cells) which coexpress CD4 and the chemokine receptors CCR1 through CCR5, CXCR4, BOB (STRL33), or BONZO (GPR15). Despite differences in

FIG. 3. Growth kinetics of recombinant viruses on allogeneic CD41 T cells recapitulate growth patterns of the uncloned virus. Infections were performed in 96-well microtiter plates. CD41 lymphocytes from three donors were selected onto individual, commercially available AIS flasks coated with anti-CD4 antibodies. Each flask was treated with anti-CD3 antibody and interleukin-2 for 5 days. A pool of the three donors was prepared at the end of the incubation period, and it was cryopreserved for future use. Cells were used for infection within 2 days after thaw. For infections, 2 3 105 cells were incubated with virus made by electroporation of PBMC for 2 h as described in Materials and Methods. Clones are as indicated. Data points represent the averages of two independent experiments.

PBMC 7, 8, and 17 demonstrated better replication in MA than in CD41 T cells (compare Fig. 3 with Fig. 4). Since there was no evidence for increased cell death in these cultures relative to the control HIVSX, these differences could not be explained on the basis of decreases in cell viability in the CD4 T-cell cultures. These data are consistent with our hypothesis that donor 6 is infected with a virus that demonstrates growth attenuation in CD41 T cells. Differences in replication kinetics are linked to the efficiency of viral entry. The experiments described above revealed an increase in growth kinetics in MA versus primary CD41 T cells. Since these chimeric viruses contained env sequences from donor 6, it appeared likely that the differences we observed were due to restrictions at the level of entry. As such, viral entry was examined by infecting purified CD41 T cells and MA with the recombinant viruses PBMC 7, 14, and 17. The T-cell line-tropic HIVNL4-3 and the MA-tropic HIVSX were included as controls. Viral entry was examined by quantitative PCR for early viral cDNA synthesis, an early step following viral entry as previously described (75). Whereas levels of viral cDNA for the MA-tropic virus HIVSX were essentially equivalent in both CD41 T cells and MA (1.4-fold

FIG. 4. Recombinant viruses are capable of replicating in MA. Infections were performed in 96-well microtiter plates. For infections, 2 3 105 cells were incubated with virus made by electroporation of PBMC for 2 h as described in Materials and Methods. MA from three donors were prepared, and the cells were pooled and plated for infection on day 6. Infections were performed on 7-day-old MA. The MA donors were different from the CD41 T-cell donors in Fig. 3. Clones are as indicated. Data points represent the averages of at least two independent experiments. If error bars are drawn, data points represent the averages of three independent experiments.

8654

GROVIT-FERBAS ET AL.

J. VIROL.

TABLE 1. Defects in replication of chimeric viruses in CD41 T lymphocytes are correlated with decreased entry Virusb

NL SX PBMC 7 PBMC 14 PBMC 17

Copies of HIV-1 DNA/mg of total DNAa CD41 T cellsc

MAd

590 140 37 160 60

40 200 585 400 385

a HIV DNA was measured by quantitative PCR 18 h after infection and are expressed as copies of HIV DNA/mg of total DNA. Heat-inactivated controls were negative (data not shown). b Infections were performed in 96-well microtiter plates. Cells (2 3 105) were infected for 2 h with 20 ng of the recombinant viruses generated in 293T cells. c CD41 T cells were prepared as described in the legend to Fig. 2. d MA were prepared as described in the legend to Fig. 3.

infectivity of MA versus CD41 T cells, the recombinant viruses we tested, as well as the uncloned virus obtained from the same visit date, used only the b-chemokine receptor CCR5 (Fig. 5). These data indicate that differences in infectivity patterns with these viruses are not due to differential use of known coreceptors. Chimeric viruses replicate to lower titers in the cells of donor 6. Our data thus far implicated viral envelope sequences in the growth-attenuated phenotype we have observed with a virus from an LTS. It was possible, however, that a host determinant might influence this donor’s long-term survival. In order to address the latter possibility, we examined viral replication in cells from donor 6. We found that chimeric viruses replicated less efficiently than HIVSX in CD41 T cells from donor 6 (Fig. 6). These data suggest a decreased susceptibility to infection of both CD41 T cells and MA isolated from donor 6 by autologous as well as heterologous (HIVNL4-3 and HIVSX) envelope-containing viruses. Previous reports have indicated that 20% of the Caucasian population are heterozygous for a 32-bp deletion in the CCR5 allele (1/D32) (22, 61, 84). Whereas this mutation has been demonstrated to confer protection from infection on individuals who are D32/D32 (61, 84), the heterozygous condition (1/D32) has been reported to correlate with delayed progression to disease (22, 47, 69). Since the data described above suggested a potential for decreased susceptibility of the donor’s own cells to infection with HIV-1 containing autologous envelope sequences, the CCR5 allele was examined. As shown in Fig. 7, donor 6 was heterozygous (lane 4) for the CCR5 mutant allele. This finding is consistent with the decreased susceptibility to infection with the recombinant CCR5-tropic viruses reported above. It is interesting to note that CD41 T cells from donor 6 were also more resistant to infection with a CXCR4-tropic virus (HIVNL4-3) relative to allogeneic CD41 T cells. This finding raises the possibility that there are other blocks to infection in the CD41 T cells of donor 6 not identified by these studies. DISCUSSION Despite extensive study, much remains to be learned about the virologic determinants of HIV pathogenesis. We describe here the infection of an LTS with a CD41 T-cell growthattenuated virus. Although it has been reported that viral isolates from LTS generally replicate poorly in culture, the kinetics of replication were much slower than those seen for other LTS we studied as controls (Fig. 1A). Furthermore, it may be important that viral growth attenuation was observed in CD41

T cells, a major target for HIV infection in vivo (20, 63). This phenotype was reproduced with chimeric viruses containing RNA-derived env sequences between C1 and V3. Close examination revealed a diminished susceptibility to replicate in CD41 T cells in three of four recombinant viruses, although infection did occur in MA. Entry was also less efficient in CD41 T cells than in MA. Finally, we observed a decrease in the ability to infect cells isolated from donor 6 by viruses containing envelope sequences derived from his own PBMC RNA as well as those from other unrelated HIV-1 strains. This observation may be related to the heterozygous 1/D32 genotype. LTS almost uniformly demonstrate a low viral burden (8, 34), but factors in addition to production rate may also influence the speed of clinical progression. These include infection

FIG. 5. Recombinant viruses use CCR5 for entry in a single round of infection. Coreceptor usage was determined after infection of HOS cells (gift of D. Littman) expressing CD4 and one of the following chemokine receptors: CCR1, CCR2, CCR3, CCR4, CCR5, GPR15 (BONZO), STRL33 (BOB), or CXCR4. In addition, the cells contain the GFP under the control of the HIV long terminal repeat. Cultures infected by HIV-1 chimeras resulted in the production of GFP 48 h postinfection. GFP expression was measured in paraformaldehyde-fixed (2%) samples by flow cytometry (Becton Dickinson; FACScan). GHOST cells were infected with 50 to 70 ng of recombinant virus and cultured for 2 days. (A) PBMC 7. (B) PBMC 14. (C) PBMC 17. Rows: 1, GHOST CD4 cells; 2, GHOST CD4 1 CCR1 cells; 3, GHOST CD4 1 CCR2b; 4, GHOST CD4 1 CCR3; 5, GHOST CD4 1 CCR4; 6, GHOST CD4 1 CCR5; 7, GHOST CD4 1 BOB; 8, GHOST CD4 1 BONZO; 9, GHOST CD4 1 CXCR4. In this assay, approximately 10 to 20% of the cells will be positive in a single round of infection. Infections performed with vesicular stomatitis virus protein-G pseudotyped envelope yielded an average of 10% positive cells in all nine cell lines (data not shown).

VOL. 72, 1998

FACTORS CONTRIBUTING TO LONG-TERM SURVIVAL WITH HIV-1

8655

FIG. 6. Cells from donor 6 are less susceptible to infection by virus containing autologous envelope sequences as well as heterologous envelope sequences. Infections were performed in 96-well microtiter plates. For infections, 2 3 105 cells were incubated with 50 pg of virus for 2 h. Allogeneic MA (B) and CD41 T cells (A) were used as pools as described in the legends to Fig. 3 and 4. Infections were performed on 7-day-old MA. The MA donors were different from the CD41 T-cell donors. MA and CD41 T cells from donor 6 were prepared according to the same methods. Clones are as indicated. Samples were tested for p24 production at day 14.

with attenuated strains (8, 21, 54, 64, 67, 68, 78), the extent of viral diversity (25, 41, 62, 73, 81, 92), and the relative cytopathicity (14, 19, 52, 72, 88, 95) of the infecting and/or evolving virus. Among all these factors, the potential for infection with an attenuated virus has received the most attention. Experimental evidence for HIV attenuation has been described in vitro, following alteration of the auxiliary genes of HIV-1, including nef (11, 50), vif (35, 37, 91), vpr (4, 80), or vpu (30, 55). Furthermore, viruses containing deletions in the nef (53) or vpr (57) gene of simian immunodeficiency virus SIVmaq have been shown to protect adult rhesus macaques against challenge with wild-type SIV, although neonatal macaques were not similarly protected (3). In addition, a lack of intact nef sequences in HIV-1 infection in vivo has been described in some (21, 54, 64, 67, 68), but not all (48), LTS. There is also a report of an attenuated Rev variant in an LTS, but this mutation was also detected in two of four progressors (49). Finally, Connor et al. observed defects in either synthesis or processing of envelope precursor protein (gp160) in clones derived from four of six LTS (16). We report here on viral env sequences spanning C1 through V3 which confer growth attenuation onto primary CD41 T cells but not onto primary MA. Furthermore, we demonstrate that these sequences are responsible for differences in viral entry. We tested the hypothesis that the block to highly productive infection in CD41 T cells occurs at the level of interaction between viral gp120 and the coreceptor molecule at the cell surface. All chimeric viruses tested, as well as an uncloned culture isolate from this individual, used CCR5 as a coreceptor for infection. It is of interest that CCR5 was used by the viral isolates examined in this study, since this molecule is expressed on both activated CD41 T cells and MA (94). Consequently, our studies cannot exclude the possibility that viruses encoding envelope proteins from donor 6 may use a novel receptor that is present on MA but absent on activated CD41 T cells. In this model, circulating virus may be able to use CCR5 but might preferentially utilize an additional, undefined receptor on MA. As such, these viruses would have a distinct growth advantage in MA, since they could enter these cells via two different pathways.

Interestingly, we found that donor 6 was heterozygous (1/ D32) for the 32-bp deletion in CCR5. This genetic marker has been implicated as an HIV-1 survival factor in some studies (22, 47). Individuals with the homozygous D32/D32 genotype are resistant to infection with CCR5-tropic viruses due to defects in entry (16, 47). Therefore, it follows that heterozygotes might demonstrate diminished susceptibility to infection with these same viruses. Other viral strains use different chemokine receptors for entry, which may also have an impact on clinical progression. For instance, a point mutation in CCR2 was linked to long-term survival (90); however, recent data indicate that this phenomenon may be a surrogate marker for a mutation in the CCR5 regulatory region (56). Finally, it has been reported that a polymorphism in the 39 untranslated region of the gene for the ligand (SDF-1) for the coreceptor molecule CXCR4 correlates with survival (93). Although a consensus regarding these analyses is still evolving, they clearly demonstrate that host genetics are likely to play a major role in the rate of disease progression by impacting the interaction of HIV-1 and its cognate receptors at the level of viral entry.

FIG. 7. Donor 6 is CCR5 1/D32. Genomic DNA was extracted as described in Materials and Methods. One-tenth of the amplified product was run on a 4% NuSieve agarose gel in the presence of ethidium bromide. Lanes 1 to 3 contain control samples and are labeled accordingly. Lane 4 contains the sample from donor 6. A 172-bp band is indicative of the 1/1 condition (lane 3), a 140-bp band is indicative of the D32/D32 condition (lane 2), and both bands indicate the heterozygous condition 1/D32 (lanes 1 and 4).

8656

GROVIT-FERBAS ET AL.

The data presented here support the hypothesis that viral entry may be a critical determinant of long-term survival. Our results indicate that genetic differences in viral envelope sequences which result in inefficient entry into cells may be important determinants of long-term survival. While the current study focused on envelope sequences, it is also possible that other viral genes may be involved in the growth attenuation phenotype of uncloned virus from this individual. Given the impact of host genetic factors, it appears that both viral and genetic influences are at play in this particular individual’s delayed clinical progression. Taken together, studies of delayed progressors to AIDS will continue to offer insights into the mechanisms of viral persistence in the absence of clinical progression.

J. VIROL.

15.

16.

17. 18. 19.

ACKNOWLEDGMENTS We express our sincere thanks to the volunteer who donated specimens for this study. We thank Stephanie Lu, Christine Yeramian, and Mary Ann Hausner for expert technical assistance; the staff of the UCLA Multicenter AIDS Cohort Study, especially Roger Detels, Denis Miles, and Martin Majchrowicz; and the Medical Research Service of the Department of Veterans Affairs. We thank Betty Poon and Sheila Stewart for valuable reagents, helpful discussions, and critical reading of the manuscript. This work was supported by grants UO1-AI-35040, UO1-AI-37613, UO1-AI-28697, VA103, and T32-AI-07988 (K.G.-F.).

20. 21.

22.

REFERENCES 1. Adachi, A., E. Gendelman, S. Koenig, T. Folks, R. Willey, A. Rabson, and M. A. Martin. 1986. Production of acquired immunodeficiency syndromeassociated retrovirus in human and nonhuman cells transfected with an infectious molecular clone. J. Virol. 59:284–291. 2. Alkhatib, G., C. Combadiere, C. C. Broder, Y. Feng, P. E. Kennedy, P. M. Murphy, and E. A. Berger. 1996. CC CKR5: a RANTES, MIP-1a, MIP-1b receptor as a fusion cofactor for macrophage-tropic HIV-1. Science 272: 1955–1958. 3. Baba, T. W., Y. S. Jeong, D. Pennick, R. Bronson, M. F. Greene, and R. M. Ruprecht. 1995. Pathogenicity of live, attenuated SIV after mucosal infection of neonatal macaques. Science 267:1820–1825. 4. Balliet, J. W., D. L. Kolson, G. Eiger, F. M. Kim, K. A. McGann, A. Srinivasan, and R. Collman. 1994. Distinct effects in primary macrophages and lymphocytes of the human immunodeficiency virus type 1 accessory genes vpr, vpu, and nef: mutational analysis of a primary HIV-1 isolate. Virology 200:623–631. 5. Binley, J. M., P. J. Klasse, Y. Cao, I. Jones, M. Markowitz, D. D. Ho, and J. P. Moore. 1997. Differential regulation of the antibody responses to Gag and Env proteins of human immunodeficiency virus type 1. J. Virol. 71:2799– 2809. 6. Borrow, P., H. Lewicki, X. Wei, M. S. Horwitz, N. Peffer, H. Meyers, J. A. Nelson, J. E. Gairin, B. H. Hahn, M. B. A. Oldstone, and G. M. Shaw. 1997. Antiviral pressure exerted by HIV-1-specific cytotoxic T lymphocytes (CTLs) during primary infection demonstrated by rapid selection of CTL escape virus. Nat. Med. 3:205–211. 7. Buchbinder, S. P., M. H. Katz, N. A. Hessol, P. M. O’Malley, and S. D. Holmberg. 1994. Long-term HIV-1 infection without immunologic progression. AIDS 8:1123–1128. 8. Cao, Y., L. Qin, Z. Linqi, J. Safrit, and D. D. Ho. 1995. Virologic and immunologic characterization of long-term survivors of human immunodeficiency virus type 1 infection. N. Engl. J. Med. 332:201–208. 9. Chen, Y., and P. Gupta. 1997. CD81 T-cell mediated suppression of HIV-1 infection may not be due to chemokines RANTES, MIP-1a and MIP-1b. AIDS 10:1434–1435. 10. Choe, H., M. Farzan, Y. Sun, N. Sullivan, B. Rollins, P. D. Ponath, L. Wu, C. R. Mackay, G. LaRosa, W. Newman, N. Gerard, C. Gerard, and J. Sodroski. 1996. The b-chemokine receptors CCR3 and CCR5 facilitate infection by primary HIV-1 isolates. Cell 85:1135–1148. 11. Chowers, M. Y., C. A. Spina, T. J. Kwoh, N. J. Fitch, D. D. Richman, and J. C. Guatelli. 1994. Optimal infectivity in vitro of human immunodeficiency virus type 1 requires an intact nef gene. J. Virol. 68:2906–2914. 12. Clerici, M., C. Balotta, D. Trabattoni, L. Papango, S. Ruzzante, S. Rusconi, M. L. Fusi, M. C. Colombo, and M. Galli. 1997. Chemokine production in HIV-seropositive long-term asymptomatic individuals. AIDS 10:1432–1433. 13. Cocchi, F., A. L. DeVico, A. Garzino-Demo, S. K. Arya, R. C. Gallo, and P. Lusso. 1995. Identification of RANTES, MIP-1a, and MIP-1b as the major HIV-suppressive factors produced by CD81 T cells. Science 270:1811–1815. 14. Connor, R. I., and D. D. Ho. 1994. Human immunodeficiency virus type 1

23.

24.

25. 26.

27.

28. 29.

30. 31. 32.

33. 34.

35. 36.

variants with increased replicative capacity develop during the asymptomatic stage before disease progression. J. Virol. 68:4400–4408. Connor, R. I., H. Mohri, Y. Cao, and D. D. Ho. 1993. Increased viral burden and cytopathicity correlate temporally with CD41 T-lymphocyte decline and clinical progression in human immunodeficiency virus type 1-infected individuals. J. Virol. 67:1772–1777. Connor, R. I., W. A. Paxton, K. E. Sheridan, and R. A. Koup. 1996. Macrophages and CD41 T lymphocytes from two multiply exposed, uninfected individuals resist infection with primary non-syncytium-inducing isolates of human immunodeficiency virus type 1. J. Virol. 70:8758–8764. Connor, R. I., K. E. Sheridan, D. Ceradini, S. Choe, and N. R. Landau. 1997. Change in coreceptor usage correlates with disease progression in HIV-1 infected individuals. J. Exp. Med. 185:621–628. Coombs, R. W., A. C. Collier, J. P. Allain, B. Nikora, M. Leuther, G. F. Gjerset, and L. Corey. 1989. Plasma viremia in human immunodeficiency virus infection. N. Engl. J. Med. 321:1626–1631. Cornelissen, M., G. Mulder-Kampinga, J. Veenstra, F. Zorgdrager, C. Kuiken, S. Hartman, J. Dekker, L. van der Hoek, C. Sol, and R. Coutinho. 1995. Syncytium-inducing (SI) phenotype suppression at seroconversion after intramuscular inoculation of a non-syncytium-inducing/SI phenotypically mixed human immunodeficiency virus population. J. Virol. 69:1810–1818. Dalgleish, A. G., P. C. L. Beverley, P. R. Clapham, D. H. Crawford, M. F. Greaves, and R. A. Weiss. 1984. The CD4 (T4) antigen is an essential component of the receptor for the AIDS retrovirus. Nature 312:763–767. Deacon, N. J., A. Tsykin, A. Solomon, K. Smith, M. Ludford-Menting, D. J. Hooker, D. A. McPhee, A. L. Greenway, A. Ellett, and C. Chatfield. 1995. Genomic structure of an attenuated quasi species of HIV-1 from a blood transfusion donor and recipients. Science 270:988–991. Dean, M., M. Carrington, C. Winkler, G. A. Huttley, M. W. Smith, R. Allikmets, J. J. Goedert, S. P. Buchbinder, E. Vittinghoff, E. Gomperts, S. Donfield, D. Vlahov, R. Kaslow, A. Saah, C. Rinaldo, R. Detels, and S. J. O’Brien. 1996. Genetic restriction of HIV-1 infection and progression to AIDS by a deletion allele of the CKR5 structural gene. Science 273:1856– 1862. DeJong, J.-J., J. Goudsmit, W. Keulen, B. Klaver, W. Krone, M. Tersmette, and A. deRonde. 1992. Human immunodeficiency virus type 1 clones chimeric for the envelope V3 domain differ in syncytium formation and replication capacity. J. Virol. 66:757–765. Delta Coordinating Committee. 1996. Delta: a randomised double-blind controlled trial comparing combinations of zidovudine plus didanosine or zalcitabine with zidovudine alone in HIV-infected individuals. Lancet 348: 283–291. Delwart, E. L., H. W. Sheppard, B. D. Walker, J. Goudsmit, and J. I. Mullins. 1994. Human immunodeficiency virus type 1 evolution in vivo tracked by DNA heteroduplex mobility assays. J. Virol. 68:6672–6683. Deng, H., R. Liu, W. Ellmeier, S. Choe, D. Unutmaz, M. Burkhart, P. Di Marzio, S. Marmon, R. E. Sutton, C. M. Hill, C. B. Davis, S. C. Peiper, T. J. Schall, D. R. Littman, and N. R. Landau. 1996. Identification of a major co-receptor for primary isolates of HIV-1. Nature 381:661–666. Devash, Y., T. A. Calvelli, D. G. Wood, K. J. Reagan, and A. Rubinstein. 1990. Vertical transmission of human immunodeficiency virus is correlated with the absence of high-affinity/avidity maternal antibodies to the gp120 principal neutralizing domain. Proc. Natl. Acad. Sci. USA 87:3445–3449. Doranz, B. J., J. F. Berson, J. Rucker, and R. W. Doms. 1997. Chemokine receptors as fusion cofactors for human immunodeficiency virus type 1 (HIV-1). Immunol. Res. 16:15–28. Dragic, T., V. Litwin, G. P. Allaway, S. R. Martin, Y. Huang, K. A. Nagashima, C. Cayanan, P. J. Maddon, R. A. Koup, J. P. Moore, and W. A. Paxton. 1996. HIV-1 entry into CD41 cells is mediated by the chemokine receptor CC-CKR-5. Nature 381:667–673. Du, B., A. Wolf, S. Lee, and E. Terwilliger. 1993. Changes in the host range and growth potential of an HIV-1 clone are conferred by the vpu gene. Virology 195:260–264. Enger, C., N. Graham, Y. Peng, J. S. Chmiel, L. A. Kingsley, R. Detels, and A. Munoz. 1996. Survival from early, intermediate, and late stages of HIV infection. JAMA 275:1329–1334. Eron, J. J., S. L. Benoit, J. Jemsek, R. D. MacArthur, J. Santana, J. B. Quinn, D. R. Kuritzkes, M. A. Fallon, and M. Rubin. 1995. Treatment with lamivudine, zidovudine, or both in HIV-positive patients with 200 to 500 CD41 cells per cubic millimeter. N. Engl. J. Med. 333:1662–1669. Feng, Y., C. C. Broder, P. E. Kennedy, and E. A. Berger. 1996. HIV-1 entry cofactor: functional cDNA cloning of a seven-transmembrane, G proteincoupled receptor. Science 272:872–877. Ferbas, J., A. H. Kaplan, M. A. Hausner, L. E. Hultin, J. L. Matud, Z. Liu, D. L. Panicali, H. Nerng-Ho, R. Detels, and J. V. Giorgi. 1995. Virus burden in long-term survivors of human immunodeficiency virus (HIV) infection is a determinant of anti-HIV CD81 lymphocyte activity. J. Infect. Dis. 172: 329–339. Fisher, A. G., B. Ensoli, L. Ivanoff, M. Chamberlain, S. Petteway, L. Ratner, R. C. Ballo, and S. F. Wong. 1987. The sor gene of HIV-1 is required for efficient virus transmission in vitro. Science 237:888–893. Fouchier, R. A. M., M. Groenink, N. A. Kootstra, M. Tersmette, H. G.

VOL. 72, 1998

37. 38.

39.

40.

41.

42.

43.

44.

45. 46.

47.

48. 49.

50.

51.

52. 53. 54. 55. 56.

FACTORS CONTRIBUTING TO LONG-TERM SURVIVAL WITH HIV-1

Huisman, F. Miedema, and H. Schuitemaker. 1992. Phenotype-associated sequence variation in the third variable domain of the human immunodeficiency virus type 1 gp120 molecule. J. Virol. 66:3183–3187. Gabuzda, D. H., K. Lawrence, E. Langhoff, E. Terwilliger, T. Dorfman, W. Haseltine, and J. Sodroski. 1992. Role of Vif in replication of human immunodeficiency virus type 1 in CD41 T lymphocytes. J. Virol. 66:6489–6495. Gange, S. J., A. Mun ¨ oz, L. K. Schrager, J. B. Margolick, J. V. Giorgi, A. J. Saah, C. R. Rinaldo, R. Detels, and J. P. Phair. 1997. Design of nested studies to identify factors related to late progression of HIV infection. J. Acquired Immune Defic. Syndr. Hum. Retrovirol. 15:S5–S9. Groenick, M., R. A. Fouchier, S. Broersen, C. H. Baker, M. Koot, A. B. van’t Wout, H. G. Huisman, F. Miedema, M. Tersmette, and H. Schuitemaker. 1993. Relation of phenotype evolution of HIV-1 to envelope configuration. Science 260:1513–1516. Gulick, R. M., J. W. Mellors, D. Havlir, J. J. Eron, C. Gonzalez, D. McMahon, D. D. Richman, F. T. Valentine, J. Rooney, L. Jonas, A. Meibohm, E. A. Emini, J. Chodakewitz, et al. 1997. Treatment with indinavir, zidovudine and lamivudine in adults with HIV infection and prior antiretroviral therapy. N. Engl. J. Med. 337:734–739. Hahn, B. H., G. M. Shaw, M. E. Taylor, R. R. Redfield, P. D. Markham, S. Z. Salahudin, F. Wong-Staal, R. C. Gallo, E. S. Parks, and W. P. Parks. 1986. Genetic variation in HTLV-III/LAV over time in patients with AIDS or at risk for AIDS. Science 232:1548–1553. Hammer, S. M., D. A. Katzenstein, M. D. Hughes, H. Gundacker, R. T. Schooley, R. H. Haubrich, W. K. Henry, M. M. Lederman, J. P. Phair, M. Niu, M. S. Hirsch, and T. C. Merigan. 1996. A trial comparing nucleoside monotherapy with combination therapy in HIV-infected adults with CD4 counts from 200–500 per cubic millimeter. N. Engl. J. Med. 335:1081–1090. Harrer, E., T. Harrer, S. Buchbinder, D. L. Mann, M. Feinberg, T. Yilma, R. P. Johnson, and B. D. Walker. 1994. HIV-1-specific cytotoxic T lymphocyte response in healthy, long-term. AIDS Res. Hum. Retroviruses 10(Suppl 2):S77–S78. Harrer, T., E. Harrer, S. A. Kalams, T. Elbeik, S. I. Staprans, M. B. Feinberg, Y. Cao, D. D. Ho, T. Yilma, A. M. Caliendo, R. P. Johnson, S. P. Buchbinder, and B. D. Walker. 1996. Strong cytotoxic T cell and weak neutralizing antibody responses in a subset of persons with stable nonprogressing HIV type 1 infection. AIDS Res. Hum. Retroviruses 12:585–592. Hausner, M. A., J. V. Giorgi, and S. Plaeger-Marshall. 1993. A reproducible method to detect CD8 T cell mediated inhibition of HIV production from naturally infected CD4 cells. J. Immunol. Methods 157:181–187. Hogervorst, E., S. Jurriaans, F. de Wolf, A. van Wijk, A. Wiersma, M. Valk, M. Roos, B. van Gemen, R. Coutinho, and F. Miedema. 1995. Predictors for non- and slow progression in human immunodeficiency virus (HIV) type 1 infection: low viral RNA copy numbers in serum and maintenance of high HIV-1 p24-specific but not V3-specific antibody levels. J. Infect. Dis. 171: 811–821. Huang, Y., W. A. Paxton, S. M. Wolinsky, A. U. Neumann, L. Zhang, T. He, S. Kang, D. Ceradini, Z. Jin, K. Yazdanbakhsh, K. Kunstman, D. Erickson, E. Dragon, N. R. Landau, J. Phair, D. D. Ho, and R. A. Koup. 1996. The role of a mutant CCR5 allele in HIV-1 transmission and disease progression. Nat. Med. 2:1240–1243. Huang, Y., L. Zhang, and D. D. Ho. 1995. Characterization of nef sequences in long-term survivors of human immunodeficiency virus type 1 infection. J. Virol. 69:93–100. Iversen, A. K., E. G. Shpaer, A. G. Rodrigo, M. S. Hirsch, B. D. Walker, H. W. Sheppard, T. C. Merigan, and J. I. Mullins. 1995. Persistence of attenuated rev genes in a human immunodeficiency virus type 1-infected asymptomatic individual. J. Virol. 69:5743–5753. Jamieson, B. D., G. M. Aldrovandi, V. Planelles, J. B. M. Jowett, L. Gao, L. M. Bloch, I. S. Y. Chen, and J. A. Zack. 1994. Requirement of human immunodeficiency virus type 1 nef for in vivo replication and pathogenicity. J. Virol. 68:3478–3485. Katzenstein, D. A., S. M. Hammer, M. D. Hughes, H. Gundacker, J. B. Jackson, S. Fiscus, S. Rasheed, T. Elbeik, R. Reichman, A. Japour, T. C. Merigan, and M. S. Hirsch. 1996. The relation of virologic and immunologic markers to clinical outcomes after nucleoside therapy in HIV-infected adults with 200 to 500 CD4 cells per cubic millimeter. New Engl. J. Med. 335:1091– 1098. Keet, I. P. M., P. Krijnen, M. Koot, J. M. A. Lange, F. Miedema, J. Goudsmit, and R. A. Coutinho. 1993. Predictors of rapid progression to AIDS in HIV-1 seroconverters. AIDS 7:51–57. Kestler, H. W., D. J. Ringler, K. Mori, D. L. Panicali, P. K. Sehgal, M. D. Daniel, and R. C. Desrosiers. 1991. Importance of the nef gene for maintenance of high virus loads and for development of AIDS. Cell 65:651–662. Kirchhoff, F., T. C. Greenough, D. B. Brettler, J. L. Sullivan, and R. C. Desrosiers. 1995. Brief report: absence of intact nef sequences in a long-term survivor with nonprogressive HIV-1 infection. N. Engl. J. Med. 332:228–232. Klimkait, T., K. Strebel, M. D. Hoggan, M. A. Martin, and J. M. Orenstein. 1990. The human immunodeficiency virus type-specific protein Vpu is required for efficient virus maturation and release. J. Virol. 64:621–629. Kostrikis, L. G., Y. Huang, J. Moore, S. M. Wolinsky, L. Zhang, Y. Gou, Deutsch L, J. P. Phair, A. U. Neumann, and D. D. Ho. 1998. A chemokine

57.

58. 59. 60.

61.

62.

63. 64.

65. 66.

67.

68.

69. 70. 71.

72. 73. 74.

75.

76.

77.

78.

79.

8657

receptor CCR2 allele delays HIV-1 disease progression and is associated with a CCR5 promoter mutation. Nat. Med. 4:350–353. Lang, S. M., M. Weeger, C. Stahl-Hennig, C. Coulibaly, G. Hunsmann, J. Muller, H. Muller-Hermelink, D. Fuchs, H. Wachter, M. Daniel, R. Desrosiers, and B. Fleckenstein. 1993. Importance of vpr for infection of rhesus monkeys with simian immunodeficiency virus. J. Virol. 67:902–912. Learmont, J., B. Tindall, L. Evans, A. Cunningham, P. Cunningham, J. Wells, R. Penny, J. Kaldor, and D. A. Cooper. 1992. Long-term symptomless HIV-1 infection in recipients of blood products. Lancet 340:863–867. Levy, J. A., C. E. Mackewicz, and E. Barker. 1996. Controlling HIV pathogenesis: the role of the noncytotoxic anti-HIV response of CD81 T cells. Immunol. Today 17:217–224. Liao, F., G. Alkhatib, K. W. Peden, G. Sharma, E. A. Berger, and J. M. Farber. 1997. STRL33, a novel chemokine receptor-like protein, functions as a fusion cofactor for both macrophage-tropic and T cell line-tropic HIV-1. J. Exp. Med. 185:2015–2023. Liu, R., W. A. Paxton, S. Choe, D. Ceradini, S. R. Martin, R. Horuk, M. E. MacDonald, H. Stuhlmann, R. A. Koup, and N. R. Landau. 1996. Homozygous defect in HIV-1 coreceptor accounts for resistance of some multiplyexposed individuals to HIV-1 infection. Cell 86:367–377. Liu, S. L., T. Schacker, L. Musey, D. Shriner, M. J. McElrath, L. Corey, and J. I. Mullins. 1997. Divergent patterns of progression to AIDS after infection from the same source: human immunodeficiency virus type 1 evolution and antiviral responses. J. Virol. 71:4284–4295. Maddon, P. J., A. G. Dalgleish, J. S. McDougal, P. R. Clapham, R. A. Weiss, and R. Axel. 1986. The T4 gene encodes the AIDS virus receptor and is expressed in the immune system and the brain. Cell 47:333–348. Mariani, R., F. Kirchhoff, T. C. Greenough, J. L. Sullivan, R. C. Desrosiers, and J. Skowronski. 1996. High frequency of defective nef alleles in a longterm survivor with nonprogressive human immunodeficiency virus type 1 infection. J. Virol. 70:7752–7764. Mellors, J. W., L. A. Kingsley, C. R. J. Rinaldo, J. A. Todd, B. S. Hoo, R. P. Kokka, and P. Gupta. 1995. Quantitation of HIV-1 RNA in plasma predicts outcome after seroconversion. Ann. Intern. Med. 122:573–579. Mellors, J. W., A. Munoz, J. V. Giorgi, J. B. Margolick, C. J. Tassoni, P. Gupta, L. A. Kingsley, J. A. Todd, A. J. Saah, R. Detels, J. P. Phair, and C. R. Rinaldo. 1997. Plasma viral load and CD41 lymphocytes as prognostic markers of HIV-1 infection. Ann. Intern. Med. 126:946–954. Michael, N. L., G. Chang, L. A. d’Arcy, P. K. Ehrenberg, R. Mariani, M. P. Busch, D. L. Birx, and D. H. Schwartz. 1995. Defective accessory genes in a human immunodeficiency virus type 1-infected long-term survivor lacking recoverable virus. J. Virol. 69:4228–4236. Michael, N. L., G. Chang, L. A. d’Arcy, C. J. Tseng, D. L. Birx, and H. W. Sheppard. 1995. Functional characterization of human immunodeficiency virus type 1 nef genes in patients with divergent rates of disease progression. J. Virol. 69:6758–6769. Michael, N. L., G. Chang, L. G. Louie, J. R. Mascola, D. Dondero, D. L. Birx, and H. W. Sheppard. 1997. The role of viral phenotype and CCR-5 gene defects in HIV-1 transmission and disease progression. Nat. Med. 3:338–340. Moore, J. P., Y. Cao, D. D. Ho, and R. A. Koup. 1994. Development of the anti-gp120 antibody response during seroconversion to human immunodeficiency virus type 1. J. Virol. 68:5142–5155. Munoz, A., A. J. Kirby, Y. D. He, J. B. Margolick, B. R. Visscher, C. R. Rinaldo, R. A. Kaslow, and J. P. Phair. 1995. Long-term survivors with HIV-1 infection: incubation period and longitudinal patterns of CD41 lymphocytes. J. Acquired Immune Defic. Syndr. Hum. Retrovirol. 8:496–505. Nielsen, C., C. Pedersen, J. D. Lundgren, and J. Gerstoft. 1993. Biological properties of HIV isolates in primary HIV infection. AIDS 7:1035–1040. Nowak, M. A., and C. R. Bangham. 1996. Population dynamics of immune responses to persistent viruses. Science 272:74–79. O’Brien, W. A., P. M. Hartigan, D. Martin, J. Esinhart, A. Hill, S. Benoit, M. Rubin, M. S. Simberkoff, and J. D. Hamilton. 1996. Changes in plasma HIV-1 RNA and CD41 lymphocyte counts and the risk of progression to AIDS. N. Engl. J. Med. 334:426–431. O’Brien, W. A., Y. Koyanagi, A. Namazie, J.-Q. Zhao, A. Diagne, K. Idler, J. A. Zack, and I. S. Y. Chen. 1990. HIV-1 tropism for mononuclear phagocytes can be determined by regions of gp120 outside the CD4-binding domain. Nature 348:69–73. O’Brien, W. A., S.-H. Mao, Y. Cao, and J. P. Moore. 1994. Macrophagetropic and T-cell line-adapted chimeric strains of human immunodeficiency virus type 1 differ in their susceptibilities to neutralization by soluble CD4 at different temperatures. J. Virol. 68:5264–5269. Ojo-Amaize, E. A., P. Nishanian, D. E. Keith, Jr., R. L. Houghton, D. F. Heitjan, J. L. Fahey, and J. V. Giorgi. 1987. Antibodies to human immunodeficiency virus in human sera induce cell-mediated lysis of human immunodeficiency virus-infected cells. J. Immunol. 139:2458–2463. Pantaleo, G., S. Menzo, M. Vaccarezza, C. Graziosi, O. J. Cohen, J. F. Demarest, D. Montefiori, J. M. Orenstein, C. Fox, L. K. Schrager, J. B. Margolick, S. Buchbinder, J. V. Giorgi, and A. S. Fauci. 1995. Studies in subjects with long-term nonprogressive human immunodeficiency virus infection. New Engl. J. Med. 332:209–216. Piatak, M., M. S. Saag, L. C. Yang, S. J. Clark, J. C. Kappes, K.-C. Luk,

8658

80. 81. 82.

83.

84.

85.

86. 87.

88.

GROVIT-FERBAS ET AL.

B. H. Hahn, G. M. Shaw, and J. D. Lifson. 1993. High levels of HIV-1 in plasma during all stages of infection determined by competitive PCR. Science 259:1749–1754. Planelles, V., A. Haislip, E. S. Withers-Ward, S. A. Stewart, Y. Xie, N. P. Shah, and I. S. Y. Chen. 1995. A new reporter system for detection of retroviral infection. Gene Therapy 2:1–6. Richman, D. D. and S. A. Bozzett. 1993. The impact of syncytium-inducing phenotype of human immunodeficiency virus on disease progression. J. Infect. Dis. 169:968–974. Saag, M. S., M. J. Crain, W. D. Decker, S. Campbell-Hill, S. Robinson, W. E. Brown, M. Leuther, R. J. Whitley, B. H. Hahn, and G. M. Shaw. 1991. High-level viremia in adults and children infected with human immunodeficiency virus: relation to disease stage and CD41 lymphocyte levels. J. Infect. Dis. 164:72–80. Sambrook, J., E. F. Fritsch, and T. Maniatis. 1989. Calcium phosphate transfection of mammalian cells, p. 16.30–16.40. In Molecular cloning: a laboratory manual, 2nd ed. Cold Spring Harbor Laboratory, Cold Spring Harbor, N.Y. Samson, M., F. Libert, B. J. Doranz, J. Rucker, C. Liesnard, C. M. Farber, S. Saragosti, C. Lapoumeroulie, J. Cognaux, C. Forceille, G. Muyldermans, C. Verhofstede, G. Burtonboy, M. Georges, T. Imai, S. Rana, Y. Yi, R. J. Smyth, R. G. Collman, R. W. Doms, G. Vassart, and M. Parmentier. 1996. Resistance to HIV-1 infection in caucasian individuals bearing mutant alleles of the CCR-5 chemokine receptor gene. Nature 382:722–725. Schnittman, S. M., J. J. Greenhouse, M. C. Psallidopoulos, M. Baseler, N. P. Salzman, A. S. Fauci, and H. C. Lane. 1990. Increasing viral burden in CD41 T cells from patients with human immunodeficiency virus (HIV) infection reflects rapidly progressive immunosuppression and clinical disease. Ann. Intern. Med. 113:438–443. Schrager, L. K., J. Young, M. G. Fowler, B. Mathieson, and S. H. Vermund. 1994. Long-term survivors of HIV infection: definitions and research challenges. AIDS 8:S95–S108. Schuitemaker, H., M. Koot, N. A. Kootstra, M. W. Dercksen, R. E. Y. DeGoede, R. P. Van Steenwijk, J. M. A. Lange, S. J. K. M. Eeftink, F. Miedema, and M. Tersmette. 1992. Biological phenotype of human immunodeficiency virus type 1 clones at different stages of infection: progression of disease is associated with a shift from monocytotropic to T-cell-tropic virus populations. J. Virol. 66:1354–1360. Schuitemaker, H., N. A. Kootstra, R. de Goede, F. de Wolf, F. Miedema, and

J. VIROL.

89. 90.

91. 92.

93.

94.

95. 96.

M. Tersmette. 1991. Monocytotropic human immunodeficiency virus type 1 (HIV-1) variants detectable in all stages of HIV-1 infection lack T-cell line tropism and syncytium-inducing ability in primary T-cell culture. J. Virol. 65: 356–363. Sheppard, H. W., W. Lang, M. S. Ascher, E. Vittinghoff, and W. Winkelstein. 1993. The characterization of non-progressors: long-term HIV-1 infection with stable CD41 T-cell levels. AIDS 7:1159–1166. Smith, M. W., M. Dean, M. Carrington, C. Winkler, G. A. Huttley, D. A. Lomb, J. J. Goedert, T. R. O’Brien, L. P. Jacobson, R. Kaslow, S. Buchbinder, E. Vittinghoff, D. Vlahov, K. Hoots, M. W. Hilgartner, and S. J. O’Brien. 1997. Contrasting genetic influence of CCR2 and CCR5 variants on HIV-1 infection and disease progression. Hemophilia Growth and Development Study (HGDS), Multicenter AIDS Cohort Study (MACS), Multicenter Hemophilia Cohort Study (MHCS), San Francisco City Cohort (SFCC), ALIVE Study. Science 277:959–965. Strebel, K., D. Daugherty, K. Clouse, D. Cohen, T. Folks, and M. A. Martin. 1987. The HIV ’A’ (sor) gene product is essential for virus infectivity. Nature 328:728–731. Tersmette, M., R. E. Y. de Goede, B. J. M. Al, I. N. Winkel, R. A. Gruters, H. T. Cuypers, H. G. Huisman, and F. Miedema. 1988. Differential syncytium-inducing capacity of human immunodeficiency virus isolates: frequent detection of syncytium-inducing isolates in patients with acquired immunodeficiency syndrome (AIDS) and AIDS-related complex. J. Virol. 62:2026– 2032. Winkler, C., W. Modi, M. W. Smith, G. W. Nelson, X. Wu, M. Carrington, M. Dean, T. Honjo, K. Tashiro, D. Yabe, S. Buchbinder, E. Vittinghoff, J. J. Goedert, T. R. O’Brien, L. P. Jacobson, R. Detels, S. Donfield, A. Willoughby, E. Gomperts, D. Vlahov, J. Phair, and S. J. O’Brien. 1998. Genetic restriction of AIDS pathogenesis by an SDF-1 chemokine gene variant. Science 279:389–393. Wu, L., W. A. Paxton, N. Kassam, N. Ruffing, J. B. Rottman, N. Sullivan, H. Choe, J. Sodroski, W. Newman, R. A. Koup, et al. 1997. CCR5 levels and expression pattern correlate with infectability by macrophage-tropic HIV-1, in vitro. J. Exp. Med. 185:1681–1691. Zhu, T., H. Mo, N. Wang, D. Nam, Y. Cao, R. Koup, and D. Ho. 1993. Genotypic and phenotypic characterization of HIV-1 in patients with primary infection. Science 261:1179–1181. Zinkernagel, R. M. 1995. Are HIV-specific CTL responses salutary or pathogenic? Curr. Opin. Immunol. 7:462–470.