Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
Vaccine ; 22(23-24): 2974-84, 2004 Aug 13.
Article in English | MEDLINE | ID: mdl-15356916

ABSTRACT

Cell-surface CCR5 is a major coreceptor with CD4 glycoprotein, mediating cellular entry of CCR5 strains of HIV-1 or SIV. We targeted the SIV CCR5 coreceptor in a combined CCR5-SIV antigen immunization strategy. Rhesus macaques were immunized i.m. with the 70 kDa heat shock protein (HSP70) covalently linked to the CCR5 peptides, SIV gpl20 and p27. Intravenous challenge with SIV mac 8980 prevented SIV infection or decreased the viral load with the CCR5-SIV combined vaccine. CC chemokines and antibodies which block and downmodulateCCR5 were induced, as well as immune responses to the subunit SIV antigens. This novel vaccination strategy complements cognate immunity to SIV with innate immunity to the CCR5 coreceptor of SIV.


Subject(s)
Receptors, CCR5/immunology , SAIDS Vaccines/therapeutic use , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Acquired Immunodeficiency Syndrome/prevention & control , Amino Acid Sequence , Animals , Antibodies, Viral/analysis , Antibodies, Viral/biosynthesis , Antigens, Surface/immunology , Antigens, Viral/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Gene Products, rex/genetics , Gene Products, rex/immunology , HSP70 Heat-Shock Proteins/metabolism , Immunization Schedule , Immunoglobulin A/analysis , Immunoglobulin A/biosynthesis , Immunoglobulin G/analysis , Immunoglobulin G/biosynthesis , Kinetics , Macaca mulatta , Membrane Glycoproteins/genetics , Membrane Glycoproteins/immunology , Molecular Sequence Data , Mycobacterium tuberculosis/immunology , T-Lymphocytes/immunology , Viral Envelope Proteins/genetics , Viral Envelope Proteins/immunology
2.
AIDS ; 15(16): 2085-92, 2001 Nov 09.
Article in English | MEDLINE | ID: mdl-11684927

ABSTRACT

OBJECTIVE: A comparative study of the replication kinetics of different HIV-1 variants (including SIV(cpz)) was undertaken to determine which viral characteristics were associated with sustained plasma viraemia in chimpanzees. DESIGN: Plasma samples from chimpanzees infected with six different HIV-1 clade B isolates were compared with plasma samples from SIV(cpz-ant)-infected chimpanzees. METHODS: A pan-clade quantitative competitive reverse transcriptase-polymerase chain reaction assay was developed based on conserved primer sequences recognizing M, N and O human lentiviruses as well as different SIV(cpz) isolates. RESULTS: Important differences between early kinetics in the human lentivirus isolates as well as compared with the chimpanzee isolate SIV(cpz-ant) were observed. R5-dependent non-syncytium-inducing (NSI) isolates (5016, Ba-L, SIV(cpz)) were found to have relatively higher viral loads than the syncytium-inducing (SI), X4-dependent primary (SF2), T cell-adapted (IIIB) or X4/R5 (Han2, DH12) SI primary isolates. CONCLUSION: Infection of chimpanzees with NSI R5-utilizing isolates correlated with persistent viraemia (approximately 10(4) RNA equivalents/ml) in contrast to transient viraemia observed after infection with SI X4-utilizing isolates.


Subject(s)
Ape Diseases/virology , HIV Infections/virology , HIV-1/physiology , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/physiology , Viral Load , Animals , Giant Cells/physiology , HIV-1/classification , Humans , Pan troglodytes , RNA, Viral/blood , Reverse Transcriptase Polymerase Chain Reaction , Simian Immunodeficiency Virus/classification
3.
J Med Primatol ; 30(4): 197-206, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11555138

ABSTRACT

The efficacy of a multicomponent vaccination with modified vaccinia Ankara constructs (rMVA) expressing structural and regulatory genes of simian immunodeficiency virus (SIV(mac251/32H/J5)) was investigated in cynomolgus monkeys, following challenge with a pathogenic SIV. Vaccination with rMVA-J5 performed at week 0, 12, and 24 induced a moderate proliferative response to whole SIV, a detectable humoral response to all but Nef SIV antigens, and failed to induce neutralizing antibodies. Two months after the last boost, the monkeys were challenged intravenously with 50 MID50 of SIV(mac251). All control monkeys, previously inoculated with non-recombinant MVA, were infected by week two and seroconverted by weeks four to eight. In contrast a sharp increase of both humoral and proliferative responses at two weeks post-challenge was observed in vaccinated monkeys compared to control monkeys. Although all vaccinated monkeys were infected, vaccination with rMVA-J5 appeared to partially control viral replication during the acute and late phase of infection as judged by cell- and plasma-associated viral load.


Subject(s)
Macaca fascicularis/virology , Simian Immunodeficiency Virus/pathogenicity , Vaccinia virus/genetics , Viral Vaccines/immunology , Animals , Antibody Formation , Female , Gene Expression Regulation , Gene Products, env , Immunity, Cellular , Infusions, Intravenous , Retroviridae Proteins, Oncogenic , Simian Acquired Immunodeficiency Syndrome/prevention & control , Vaccination , Vaccinia virus/immunology , Viral Fusion Proteins , Viral Load , Virus Replication
4.
J Med Primatol ; 30(4): 207-14, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11555139

ABSTRACT

Various simian immunodeficiency virus (SIV)sm/mac and simian/human immunodeficiency virus (SHIV) strains are used in different macaque species to study AIDS pathogenesis, as well as to evaluate candidate vaccine and anti-retroviral drugs efficacy. In this study we investigated the effect of route of infection, species of macaques and nature of virus stock on early plasma viral RNA load. We monitored the plasma RNA concentrations of 63 rhesus (Macaca mulatta) and cynomolgus macaques (Macaca fascicularis) infected with well-characterised virus stocks administered either by oral, rectal, vaginal or intravenous (i.v.) routes. In SIV(mac)-infected macaques, no significant difference in plasma RNA loads was observed between the rectal, oral and i.v. routes of infection. Cynomolgus macaques developed lower steady state SIV plasma RNA concentrations compared with rhesus macaques and no significant difference was observed between rectal and i.v. routes of infection. In SHIV(89.6p)-infected macaques, no difference between species or between route of infection was observed with this particular chimeric virus.


Subject(s)
AIDS Vaccines , Acquired Immunodeficiency Syndrome/physiopathology , HIV Infections/virology , Macaca fascicularis/virology , Macaca mulatta/virology , RNA/analysis , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/pathogenicity , Animals , Chimera , Gene Products, env/analysis , HIV Infections/immunology , Humans , Retroviridae Proteins, Oncogenic/analysis , Simian Acquired Immunodeficiency Syndrome/immunology , Viral Fusion Proteins/analysis , Viral Load
5.
J Virol ; 75(17): 8137-46, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11483759

ABSTRACT

The nef genes of human immunodeficiency virus and simian immunodeficiency virus (SIV) overlap about 80% of the U3 region of the 3' long terminal repeat (LTR) and contain several essential cis-acting elements (here referred to as the TPI region): a T-rich region, the polypurine tract, and attachment (att) sequences required for integration. We inactivated the TPI region in the nef reading frame of the pathogenic SIVmac239 clone (239wt) by 13 silent point mutations. To restore viral infectivity, intact cis-regulatory elements were inserted just downstream of the mutated nef gene. The resulting SIV genome contains U3 regions that are 384 bp shorter than the 517-bp 239wt U3 region. Overall, elimination of the duplicated Nef coding sequences truncates the proviral genome by 350 bp. Nonetheless, it contains all known coding sequences and cis-acting elements. The TPI mutant virus expressed functional Nef and replicated like 239wt in all cell culture assays and in vivo in rhesus macaques. Notably, these SIVmac constructs allow us to study Nef function in the context of replication-competent viruses without the restrictions of overlapping LTR sequences and important cis-acting elements. The genomes of all known primate lentiviruses contain a large overlap between nef and the U3 region. We demonstrate that this conserved genomic organization is not obligatory for efficient viral replication and pathogenicity.


Subject(s)
Genes, nef , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/physiology , Simian Immunodeficiency Virus/pathogenicity , Terminal Repeat Sequences/genetics , Virus Replication , Animals , Cell Line , Gene Products, nef/metabolism , Macaca mulatta , Mutation , Polymerase Chain Reaction , Simian Immunodeficiency Virus/genetics , Transfection
6.
J Infect Dis ; 184(2): 136-43, 2001 Jul 15.
Article in English | MEDLINE | ID: mdl-11424009

ABSTRACT

Recent evidence suggests a much higher prevalence of human immunodeficiency virus type 1 (HIV-1) recombinants than previously anticipated. These recombinants arise from secondary HIV infections in individuals already infected with the virus. It remains unclear why some individuals acquire secondary HIV-1 infections and others do not. To address this question, a study was undertaken of a small cohort of chimpanzees with well-defined HIV-1 infection. After exposure to an infectious dose of heterologous primary isolate, 4 of 8 HIV-1 seropositive chimpanzees resisted secondary infection, whereas 2 naive controls became readily infected. Only animals who were immunologically boosted were protected. Protection from heterologous secondary exposure appeared to be related to the repertoire of the cytolytic CD8(+) T cell responses to HIV-1. Data suggested that immunologic boosting by HIV-1 antigens or exposure to subinfectious doses of virus may be important events in sustaining sufficient immunity to prevent secondary infections from occurring.


Subject(s)
AIDS Vaccines/administration & dosage , Acquired Immunodeficiency Syndrome/immunology , Acquired Immunodeficiency Syndrome/prevention & control , T-Lymphocytes, Cytotoxic/immunology , AIDS Vaccines/immunology , Animals , HIV Infections/immunology , Pan troglodytes
7.
Vaccine ; 19(25-26): 3526-36, 2001 May 14.
Article in English | MEDLINE | ID: mdl-11348720

ABSTRACT

The immunogenicity of two vector-based vaccines, either given alone or in a prime-boost regimen, was investigated. Cynomolgus macaques were immunised with modified vaccinia virus Ankara (MVA) expressing simian immunodeficiency virus (SIV)macJ5 env, gag-pol, nef, rev, and tat genes (MVA-SIVmac) or primed with a Semliki forest virus (SFV) vaccine expressing the same genes (SFV-SIVmac) and boosted with MVA-SIVmac. Generally, antibody responses, T-cell proliferative responses and cytotoxic T-cell responses remained low or undetectable in vaccinees receiving MVA-SIVmac or SFV-SIVmac alone. In contrast, monkeys who first received SFV-SIVmac twice and then were boosted with MVA-SIVmac showed increased antibody responses as well as high T-cell proliferative responses. Three of these vaccinees had cytotoxic T-lymphocytes directed against three or four of the gene products. No evidence of protection was seen against an intrarectal heterologous SIVsm challenge given 3 months after the last immunisation. The study demonstrates a prime-boost strategy that efficiently induces both humoral and cellular immune responses.


Subject(s)
SAIDS Vaccines/administration & dosage , Semliki forest virus/immunology , Simian Immunodeficiency Virus/immunology , Vaccinia virus/immunology , AIDS Vaccines/administration & dosage , AIDS Vaccines/isolation & purification , Animals , Antibodies, Viral/biosynthesis , Antibody Specificity , Genetic Vectors , Immunity, Cellular , Immunization, Secondary , Lymphocyte Activation , Lymphocyte Count , Macaca fascicularis , RNA, Viral/blood , SAIDS Vaccines/genetics , Semliki forest virus/genetics , T-Lymphocytes/immunology , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Vaccinia virus/genetics
8.
AIDS Res Hum Retroviruses ; 17(7): 631-5, 2001 May 01.
Article in English | MEDLINE | ID: mdl-11375059

ABSTRACT

Treatment of human immunodeficiency virus (HIV-1) with drugs targeted to the reverse transcriptase (RT) rapidly selects for drug-resistant virus. It is essential to develop a suitable animal model that allows the study of the emergence and reversal of drug resistance. A monkey model was previously developed on the basis of a hybrid virus (RT-SHIV) of simian immunodeficiency virus (SIV) with its RT exchanged for HIV-1 RT. In the present study cynomolgus monkeys infected with RT-SHIV were treated with varying doses of the non-nucleoside RT inhibitor nevirapine. The drug was administered for 2-3 weeks, in agreement with clinical experience of resistance development during nevirapine monotherapy. This resulted in the selection of mutants with Y181C and K103N changes in RT, which correspond to the HIV-1 mutations in nevirapine-resistant HIV-1 patients. The mutants coexisted at varying levels with wild-type virus and fluctuations in the proportion of mutants could be closely monitored. Low-dose treatment was not more efficient in induction of mutations than a virus-inhibiting dose. Structured therapy interruptions could be performed. The monkey RT-SHIV infection offers an in vivo model to determine effects of therapies on resistance development.


Subject(s)
Disease Models, Animal , HIV-1/drug effects , Nevirapine/pharmacology , Reverse Transcriptase Inhibitors/pharmacology , Animals , Chimera , Dose-Response Relationship, Drug , Drug Resistance/genetics , HIV Infections/drug therapy , HIV Reverse Transcriptase/antagonists & inhibitors , HIV Reverse Transcriptase/genetics , HIV-1/genetics , Macaca fascicularis , RNA, Viral/blood , Simian Immunodeficiency Virus/genetics
9.
Vaccine ; 19(20-22): 2862-77, 2001 Apr 06.
Article in English | MEDLINE | ID: mdl-11282197

ABSTRACT

Recent evidence suggests that a CD8-mediated cytotoxic T cell response against the Tat protein of human immunodeficiency virus (HIV)/simian immunodeficiency virus (SIV) controls primary infection after pathogenic virus challenge, and correlates with the status of long-term nonprogressor in humans. Due to the presence of unmethylated CpG sequences, DNA vaccination can boost the innate immunity driving more potent T cell-mediated immune responses. Therefore, cynomolgus monkeys were vaccinated with a tat-expressing vector containing defined unmethylated CpG sequences (pCV-tat). Here it is shown that the intramuscular inoculation of the pCV-tat contained primary infection with the highly pathogenic SHIV89.6P virus preventing the CD4(+) T cell decline in all the vaccinated monkeys. Undetectable virus replication and negative virus isolation correlated in all cases with the presence of anti-Tat CTLs. However, a CD8-mediated non cytolytic antiviral activity was also present in all protected animals. Of note, this activity was absent in the controls but was present in the monkey inoculated with the CpG-rich vector alone that was partially protected against viral challenge (i.e. no virus replication but positive virus isolation). These results suggest that a CTL response against Tat protects against primary infection by blocking virus replication at its early stage, in the absence of sterilizing immunity. Nevertheless, the boost of the innate immunity by CpG sequences can contribute to this protection both by driving more potent CTL responses and by inducing other CD8-mediated antiviral activities. Thus, the CpG-rich tat DNA vaccine may represent a promising candidate for preventive and therapeutic vaccination against AIDS.


Subject(s)
AIDS Vaccines/immunology , Acquired Immunodeficiency Syndrome/prevention & control , Dinucleoside Phosphates/administration & dosage , Gene Products, tat/immunology , Vaccines, DNA/immunology , Animals , DNA Methylation , Gene Products, tat/genetics , HIV Antibodies/blood , Macaca fascicularis , Vaccination , tat Gene Products, Human Immunodeficiency Virus
10.
Virology ; 279(2): 527-38, 2001 Jan 20.
Article in English | MEDLINE | ID: mdl-11162808

ABSTRACT

It has been suggested that the presence of immunoglobulin and complement receptors on rectal epithelium may facilitate the entry of HIV complexed to nonneutralizing antibody. We tested this hypothesis using simian immunodeficiency virus (SIV) infection of rhesus macaques. First, in a pilot study, a nonneutralizing IgG fraction of macaque anti-SIV gp120 was shown to enhance the immunogenicity of SIV envelope following rectal immunization. The same antibody was then mixed with a subinfectious dose of SIV and the occurrence of rectal infection was compared with virus alone. Animals were not infected overtly and were rechallenged with a 10-fold higher dose of virus with and without addition of antibody. There was no evidence of antibody-mediated infection, since equal numbers of macaques became infected, regardless of the presence of antibody. In addition, the application of immune complexes did not alter significantly the subsequent virus load or the immune responses generated. In seronegative animals, in which virus and proviral DNA were undetectable in PBMC and tissues, SIV-specific T-cell responses and antibody-secreting cells were found in systemic and gut-associated sites. Our results show that nonneutralizing antibody neither facilitated nor enhanced rectal infection with SIV, in the small number of animals used, despite the consistent trend for this antibody to enhance antibody responses to gp120 following rectal immunization with immune-complexed antigen. However, mucosal exposure to subinfectious doses of virus primed both systemic and local immunity, regardless of addition of nonneutralizing antibody.


Subject(s)
Rectum/immunology , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Immunodeficiency Virus , Animals , Antibodies, Viral/administration & dosage , Antibodies, Viral/blood , Antigens, Viral/immunology , Cytotoxicity, Immunologic , HIV Envelope Protein gp120/immunology , Immunity, Active , Immunity, Cellular , Immunoglobulin G/analysis , Intestinal Mucosa/immunology , Intestinal Mucosa/virology , Leukocytes, Mononuclear/virology , Macaca mulatta , Rectum/virology , Simian Acquired Immunodeficiency Syndrome/virology , Viral Load
11.
J Med Primatol ; 30(5): 243-53, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11990239

ABSTRACT

In a longitudinal study we address the hypothesis that resis tance to disease progression in lentivirus-infected chimpanzees is related to potent non-cytotoxic suppression of virus replication. In a long-term follow-up, the viral suppressive capacity in two simian immunodeficiency virus (SIV)cpz-infected chimpanzees was correlated with two polymerase chain reaction (PCR)- and two culture-based virus load measurements. In both animals, quantitative virus isolation (QVI) tended to decline slowly, whereas in vitro virus suppression was sustained or increased over time. In general, plasma virus loads in SIVcpz-infected animals were maintained for extended periods of time. Based on current assays that measure virus suppressive capacity in peripheral blood, it was not possible to conclude that virus suppression played a major role in the maintenance of the disease-free state in lentivirus-infected chimpanzees.


Subject(s)
Ape Diseases/immunology , CD8-Positive T-Lymphocytes/immunology , Pan troglodytes , Simian Immunodeficiency Virus/immunology , Animals , Ape Diseases/virology , Chemokine CCL4 , Chemokine CCL5/biosynthesis , Chemokine CCL5/blood , Disease Progression , Genes, MHC Class I/immunology , Genes, MHC Class II/immunology , Longitudinal Studies , Macrophage Inflammatory Proteins/biosynthesis , Macrophage Inflammatory Proteins/blood , Viral Load/veterinary
12.
J Med Primatol ; 29(3-4): 193-208, 2000 Aug.
Article in English | MEDLINE | ID: mdl-11085582

ABSTRACT

The Tat protein of human immunodeficiency virus (HIV) is produced very early after infection, plays a key role in the virus life cycle and in acquired immunodeficiency syndrome (AIDS) pathogenesis, is immunogenic and well conserved among all virus clades. Notably, a Tat-specific immune response correlates with non-progression to AIDS. Here, we show that a vaccine based on the Tat protein of HIV blocks primary infection with the simian/human immunodeficiency virus (SHIV)89.6P and prevents the CD4 T cell decline and disease onset in cynomolgus monkeys. No signs of virus replication were found in five out of seven vaccinated macaques for almost 1 year of follow-up. Since the inoculated virus (derived from rhesus or from cynomolgus macaques) is shown to be highly pathogenic in cynomolgus macaques, the results indicate efficacy of Tat vaccination in protection against highly pathogenic virus challenge. Finally, the studies of the Tat-specific immunological responses indicate a correlation of protection with a cytotoxic T cell response. Thus, a Tat-based vaccine is a promising candidate for preventive and therapeutic vaccination in humans.


Subject(s)
AIDS Vaccines/pharmacology , Gene Products, tat/immunology , HIV Infections/immunology , HIV/pathogenicity , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Immunodeficiency Virus/pathogenicity , Virus Replication/drug effects , Animals , CD4 Lymphocyte Count , Chimera , Cytotoxicity, Immunologic , Disease Progression , HIV/genetics , HIV/physiology , HIV Infections/prevention & control , HIV Infections/virology , HIV-1/immunology , Humans , Macaca fascicularis , Macaca mulatta , Simian Acquired Immunodeficiency Syndrome/prevention & control , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/genetics , Simian Immunodeficiency Virus/physiology , T-Lymphocytes, Cytotoxic/immunology , Time Factors , tat Gene Products, Human Immunodeficiency Virus
13.
J Med Primatol ; 29(3-4): 268-73, 2000 Aug.
Article in English | MEDLINE | ID: mdl-11085589

ABSTRACT

A large number of recombinant of viral and bacterial systems have been engineered as vectors to express foreign genes for vaccination and/or gene therapy. A common problem is the immune response to the vector itself. The presence of anti-vector immune responses may preclude sufficient 'priming' or immunogenicity if pre-existing immune responses are present, or they may impair optimal 'boosting' upon repeated immunization or delivery with the same vector. To circumvent this problem we developed a strategy using different chimeric vectors which share only the expression of common specific antigens desired for immunization. This approach not only has the advantage of avoiding increased anti-vector responses, but allows the use of combinations of vectors which could subsequently present the same or related antigen differently to the immune system as well as at alternative sites to induce the optimal type of immunity against the pathogen of interest.


Subject(s)
Antigens, Viral/immunology , SAIDS Vaccines/immunology , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Immunodeficiency Virus/immunology , Vaccines, Synthetic/immunology , Animals , Antigens, Viral/genetics , Chimera , Genes, env , Genes, gag , Genes, nef , Genes, pol , Genes, rev , Genes, tat , Macaca mulatta , Simian Acquired Immunodeficiency Syndrome/prevention & control
14.
J Gen Virol ; 81(Pt 7): 1719-26, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10859377

ABSTRACT

Of the viral factors that are proposed to influence the rate of progression to AIDS, the role of infectious dose remains unresolved. Intravenous infection of outbred Macaca mulatta with various doses of simian immunodeficiency virus isolate 8980 (SIV(8980)) revealed an endpoint from which an infectious dose 50 (ID(50)) was defined. In the six infected animals, the time to develop AIDS was variable with a spectrum of rapid, intermediate and slow progressors. High and sustained plasma viraemia with marked loss of CD4(+) T-cells was a distinguishing feature between rapid versus intermediate and slow progressors. Animals that received the highest doses did not develop the highest sustained viral loads, nor did they progress more rapidly to disease. Similarly, animals infected with lower doses did not uniformly develop lower viral loads or progress more slowly to AIDS. Furthermore, compiled data from more than 21 animals infected with different doses of the same virus administered by the same route failed to reveal any correlation of infectious dose with survival. Indeed, host factors of these outbred animals, rather than dose of the initial inoculum, were probably an important factor influencing the rate of disease progression in each individual animal. Comparison of animals infected with SIV(B670), from which SIV(8980) was derived, revealed marked differences in disease progression. Clearly, although dose did not influence viral loads nor disease progression, the virulence of the initial inoculum was a major determinant of the rate of progression to AIDS.


Subject(s)
Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/pathogenicity , Animals , Macaca mulatta , RNA, Viral/blood , Virulence
15.
J Virol ; 74(4): 1704-11, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10644340

ABSTRACT

A limited period of chemotherapy during primary immunodeficiency virus infection might provide a long-term clinical benefit even if treatment is initiated at a time point when virus is already detectable in plasma. To evaluate this strategy, we infected rhesus macaques with the pathogenic simian/human immunodeficiency virus RT-SHIV and treated them with the antiretroviral drug (R)-9-(2-phosphonylmethoxypropyl)adenine (PMPA) for 8 weeks starting 7 or 14 days postinfection. PMPA treatment suppressed viral replication efficiently in all of the monkeys. After chemotherapy ended, virus replication rebounded and viral RNA in plasma reached levels comparable to that of the controls in four of the six monkeys. However, in the other two animals, virus loads peaked only moderately after withdrawal of the drug and then declined to low or even undetectable levels. These low levels of viremia remained stable for at least 31 weeks after cessation of therapy. At this time point, these two monkeys were challenged with SIV(8980) to evaluate whether the host responses which were able to keep RT-SHIV replication under control were also sufficient to protect against infection with a highly pathogenic heterologous virus. Both monkeys proved to be protected against the heterologous virus. In one of the two animals, low levels of SIV(8980) replication were detected. Thus, by chemotherapy during the acute phase of pathogenic virus replication, we could achieve not only persistent virus load suppression in two out of six monkeys but also protection from subsequent heterologous challenge. By this chemotherapeutic attenuation, the replication kinetics of attenuated viruses could be mimicked and a vaccination effect similar to that induced by live attenuated simian immunodeficiency virus vaccines was achieved.


Subject(s)
Adenine/analogs & derivatives , Anti-HIV Agents/therapeutic use , HIV-1 , Membrane Glycoproteins , Organophosphonates , Organophosphorus Compounds/therapeutic use , Reverse Transcriptase Inhibitors/therapeutic use , Simian Acquired Immunodeficiency Syndrome/drug therapy , Simian Immunodeficiency Virus , Viral Envelope Proteins , Viral Load , Adenine/adverse effects , Adenine/therapeutic use , Animals , Anti-HIV Agents/adverse effects , Cells, Cultured , HIV Envelope Protein gp120/immunology , HIV Reverse Transcriptase/antagonists & inhibitors , HIV Reverse Transcriptase/genetics , HIV-1/enzymology , HIV-1/genetics , Humans , Lymphocyte Subsets , Macaca mulatta , Organophosphorus Compounds/adverse effects , RNA, Viral/blood , Recombination, Genetic , Reverse Transcriptase Inhibitors/adverse effects , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Acquired Immunodeficiency Syndrome/physiopathology , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/enzymology , Simian Immunodeficiency Virus/genetics , Simian Immunodeficiency Virus/immunology , Tenofovir
16.
J Virol ; 74(9): 4017-27, 2000 May.
Article in English | MEDLINE | ID: mdl-10756013

ABSTRACT

Current strategies in human immunodeficiency virus type 1 (HIV-1) vaccine development are often based on the production of different vaccine antigens according to particular genetic clades of HIV-1 variants. To determine if virus virulence or genetic distance had a greater impact on HIV-1 vaccine efficacy, we designed a series of heterologous chimeric simian/human immunodeficiency virus (SHIV) challenge experiments in HIV-1 subunit-vaccinated rhesus macaques. Of a total of 22 animals, 10 nonimmunized animals served as controls; the remainder were vaccinated with the CCR5 binding envelope of HIV-1(W6.1D). In the first study, heterologous challenge included two nonpathogenic SHIV chimeras encoding the envelopes of the divergent clade B HIV-1(han2) and HIV-1(sf13) strains. In the second study, all immunized animals were rechallenged with SHIV(89. 6p), a virus closely related to the vaccine strain but highly virulent. Protection from either of the divergent SHIV(sf13) or SHIV(han2) challenges was demonstrated in the majority of the vaccinated animals. In contrast, upon challenge with the more related but virulent SHIV(89.6p), protection was achieved in only one of the previously protected vaccinees. A secondary but beneficial effect of immunization on virus load and CD4(+) T-cell counts was observed despite failure to protect from infection. In addition to revealing different levels of protective immunity, these results suggest the importance of developing vaccine strategies capable of protecting from particularly virulent variants of HIV-1.


Subject(s)
AIDS Vaccines/immunology , HIV Infections/prevention & control , HIV-1/immunology , HIV-1/pathogenicity , Animals , Disease Models, Animal , HIV Envelope Protein gp120/genetics , HIV Envelope Protein gp120/immunology , HIV Envelope Protein gp41/genetics , HIV Infections/immunology , HIV Infections/virology , Humans , Macaca mulatta , Simian Immunodeficiency Virus/genetics , Vaccines, Attenuated/immunology , Virulence
17.
J Infect Dis ; 180(5): 1494-502, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10515808

ABSTRACT

Human immunodeficiency virus type 2 (HIV-2) and the closely related simian immunodeficiency viruses (SIVs) frequently use the orphan receptor BOB/GPR15 in addition to the chemokine receptor CCR5 for efficient entry and replication. However, the role of BOB/GPR15 in replication and pathogenesis of HIV-2 and SIV in vivo is unclear. This study shows that a single amino acid substitution in the V3 loop of the pathogenic SIVmac239 clone, 321P-->S, impaired the ability to use BOB/GPR15 for entry and replication but had little effect on the ability to use CCR5. This envelope variant replicated with an efficiency comparable with the parental SIVmac239 isolate in rhesus macaques. Furthermore, the mutant genotype and phenotype remained stable even after the onset of immunodeficiency. These results suggest that this cofactor plays only a minor role for the pathogenicity of the HIV-2/SIVmac/SIVsm group of primate lentiviruses.


Subject(s)
Receptors, CCR5/metabolism , Receptors, G-Protein-Coupled , Receptors, Peptide/metabolism , Receptors, Virus/metabolism , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/physiology , Virus Replication , Animals , Cell Line , Genes, env/genetics , Leukocytes, Mononuclear/virology , Macaca mulatta , Simian Acquired Immunodeficiency Syndrome/pathology , Simian Immunodeficiency Virus/genetics , Simian Immunodeficiency Virus/metabolism , Simian Immunodeficiency Virus/pathogenicity
18.
Virology ; 257(1): 138-55, 1999 Apr 25.
Article in English | MEDLINE | ID: mdl-10208928

ABSTRACT

Variants of the pathogenic SIVmac239 clone with changes in Nef were analyzed to assess the functional relevance of two highly conserved regions in Nef in vitro and in vivo. Changes in a region with an acidic charge (Aci-Nef), or a potential protein kinase C phosphorylation site (PKC-Nef), impaired the ability of Nef to down-regulate CD4 and MHC class I surface expression and to alter CD3-initiated signal transduction in Jurkat T cells. The Aci-Nef, but not the PKC-Nef, associated with the previously described p65 phosphoprotein. SIV containing Aci-Nef, but not SIV containing PKC-Nef, showed reduced infectivity and replication in cell culture systems. One of two rhesus macaques infected with the PKC-Nef mutant virus showed rapid reversion and progressed to disease. In the second animal no reversions and nonprogressive infection was observed. In one of two macaques infected with the Aci-Nef variant, the mutations were stable during the first 40 weeks after infection. Thereafter, variants evolved in which up to six of the eight mutated positions in Nef were reverted and functional activity in vitro was partially restored. These changes occurred concomitantly with increasing viral load and disease progression. The second animal infected with the Aci-Nef variant showed no reversions and remained asymptomatic. Our study suggests that the acidic region and conserved PKC phosphorylation site in Nef are important for SIV replication in rhesus macaques and for several in vitro Nef functions. An almost wild-type activity in in vitro infectivity and replication assays seems insufficient to confer a full nef-positive phenotype in vivo.


Subject(s)
Gene Products, nef/metabolism , Protein Kinase C/metabolism , Simian Immunodeficiency Virus/physiology , Virus Replication , Amino Acid Sequence , Animals , CD4 Antigens/biosynthesis , COS Cells , Gene Products, nef/genetics , HIV-1 , Histocompatibility Antigens Class I/biosynthesis , Hydrogen-Ion Concentration , Macaca mulatta , Molecular Sequence Data , Mutagenesis, Site-Directed , Phosphorylation , Simian Immunodeficiency Virus/genetics , Surface Properties , nef Gene Products, Human Immunodeficiency Virus
19.
J Virol ; 73(4): 3292-300, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10074183

ABSTRACT

The kinetics of T-helper immune responses generated in 16 mature outbred rhesus monkeys (Macaca mulatta) within a 10-month period by three different human immunodeficiency virus type 1 (HIV-1) vaccine strategies were compared. Immune responses to monomeric recombinant gp120SF2 (rgp120) when the protein was expressed in vivo by DNA immunization or when it was delivered as a subunit protein vaccine formulated either with the MF59 adjuvant or by incorporation into immune-stimulating complexes (ISCOMs) were compared. Virus-neutralizing antibodies (NA) against HIV-1SF2 reached similar titers in the two rgp120SF2 protein-immunized groups, but the responses showed different kinetics, while NA were delayed and their levels were low in the DNA-immunized animals. Antigen-specific gamma interferon (IFN-gamma) T-helper (type 1-like) responses were detected in the DNA-immunized group, but only after the fourth immunization, and the rgp120/MF59 group generated both IFN-gamma and interleukin-4 (IL-4) (type 2-like) responses that appeared after the third immunization. In contrast, rgp120/ISCOM-immunized animals rapidly developed marked IL-2, IFN-gamma (type 1-like), and IL-4 responses that peaked after the second immunization. To determine which type of immune responses correlated with protection from infection, all animals were challenged intravenously with 50 50% infective doses of a rhesus cell-propagated, in vivo-titrated stock of a chimeric simian immunodeficiency virus-HIVSF13 construct. Protection was observed in the two groups receiving the rgp120 subunit vaccines. Half of the animals in the ISCOM group were completely protected from infection. In other subunit vaccinees there was evidence by multiple assays that virus detected at 2 weeks postchallenge was effectively cleared. Early induction of potent type 1- as well as type 2-like T-helper responses induced the most-effective immunity.


Subject(s)
AIDS Vaccines/immunology , Acquired Immunodeficiency Syndrome/immunology , HIV Envelope Protein gp120/immunology , HIV-1/immunology , ISCOMs/immunology , Immunity, Cellular , Polysorbates/pharmacology , Squalene/immunology , Squalene/pharmacology , AIDS Vaccines/chemistry , AIDS Vaccines/pharmacology , Acquired Immunodeficiency Syndrome/prevention & control , Adjuvants, Immunologic/pharmacology , Animals , Chemistry, Pharmaceutical , HIV Envelope Protein gp120/pharmacology , Humans , ISCOMs/pharmacology , Macaca mulatta , Recombinant Proteins/immunology , Recombinant Proteins/pharmacology
20.
J Virol ; 72(12): 10281-5, 1998 Dec.
Article in English | MEDLINE | ID: mdl-9811776

ABSTRACT

To determine if a specific pathogenic threshold of plasma viral RNA could be defined irrespective of virus strain, RNA levels in the plasma of more than 50 infected rhesus macaques (Macaca mulatta) were measured. Animals were inoculated intravenously with either simian immunodeficiency virus (SIV) or simian-human immunodeficiency virus (SHIV) strains of known pathogenic potential (SIV8980, SIVsmm-3, SIVmac32H/J5, SIVmac32H/1XC, reverse transcriptase-SHIV, SHIV89.6p) or with attenuated strains (SHIVW6.1D, SHIVsf13, SHIVhan-2, SIVmacDeltanef, SHIVsf33). In animals inoculated with nonpathogenic strains, shortly after the primary peak of viremia viral RNA levels declined and remained below 10(4) RNA equivalents/ml of plasma between 6 and 12 weeks postinoculation. Animals infected with documented pathogenic strains maintained viral RNA levels higher than 10(5) RNA equivalents/ml of plasma. In animals infected with strains with low virulence, a decline in plasma RNA levels was observed, but with notable individual variation. Our results demonstrate that the disease-causing potential was predicted and determined by a threshold plasma virus load which remained greater than 10(5) RNA equivalents/ml of plasma 6 to 12 weeks after inoculation. A threshold virus load value which remained below 10(4) RNA equivalents/ml of plasma was indicative of a nonpathogenic course of infection.


Subject(s)
HIV/pathogenicity , Simian Immunodeficiency Virus/pathogenicity , Animals , Base Sequence , Chimera/genetics , Chimera/immunology , DNA Primers/genetics , Disease Models, Animal , HIV/genetics , HIV/isolation & purification , HIV Infections/virology , Humans , Macaca mulatta , RNA, Viral/blood , RNA, Viral/genetics , Reverse Transcriptase Polymerase Chain Reaction , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/genetics , Simian Immunodeficiency Virus/isolation & purification , Species Specificity , Viremia/virology , Virulence
SELECTION OF CITATIONS
SEARCH DETAIL
...