Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
J Virol ; 75(21): 10187-99, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11581387

ABSTRACT

Transient antiretroviral treatment with tenofovir, (R)-9-(2-phosphonylmethoxypropyl)adenine, begun shortly after inoculation of rhesus macaques with the highly pathogenic simian immunodeficiency virus (SIV) isolate SIVsmE660, facilitated the development of SIV-specific lymphoproliferative responses and sustained effective control of the infection following drug discontinuation. Animals that controlled plasma viremia following transient postinoculation treatment showed substantial resistance to subsequent intravenous rechallenge with homologous (SIVsmE660) and highly heterologous (SIVmac239) SIV isolates, up to more than 1 year later, despite the absence of measurable neutralizing antibody. In some instances, resistance to rechallenge was observed despite the absence of detectable SIV-specific binding antibody and in the face of SIV lymphoproliferative responses that were low or undetectable at the time of challenge. In vivo monoclonal antibody depletion experiments demonstrated a critical role for CD8(+) lymphocytes in the control of viral replication; plasma viremia rose by as much as five log units after depletion of CD8(+) cells and returned to predepletion levels (as low as <100 copy Eq/ml) as circulating CD8(+) cells were restored. The extent of host control of replication of highly pathogenic SIV strains and the level of resistance to heterologous rechallenge achieved following transient postinoculation treatment compared favorably to the results seen after SIVsmE660 and SIVmac239 challenge with many vaccine strategies. This impressive control of viral replication was observed despite comparatively modest measured immune responses, less than those often achieved with vaccination regimens. The results help establish the underlying feasibility of efforts to develop vaccines for the prevention of AIDS, although the exact nature of the protective host responses involved remains to be elucidated.


Subject(s)
Adenine/therapeutic use , Anti-HIV Agents/therapeutic use , CD8-Positive T-Lymphocytes/physiology , Organophosphonates , Organophosphorus Compounds/therapeutic use , Simian Acquired Immunodeficiency Syndrome/immunology , Adenine/analogs & derivatives , Animals , Female , Macaca mulatta , Simian Acquired Immunodeficiency Syndrome/drug therapy , Tenofovir , Virus Replication
2.
J Virol ; 75(8): 4019-22, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11264395

ABSTRACT

We analyzed virus sequences in two monkeys infected with SIVmac239 and two monkeys infected with SHIVnef that maintained high, persisting viral loads. Sequence changes were observed consistently at four loci in all four animals: a single nucleotide change in the Lys-tRNA primer binding site in the 5' long terminal repeat; two nucleotide changes that resulted in two amino acid changes in the pol gene product; and a single nucleotide change in the region of the simian immunodeficiency virus genome where the rev and env genes overlap, resulting in changes in the predicted amino acid sequences of both gene products. None of these mutations were seen in short-term cultures of CEMx174 cells infected with SIVmac239 or SHIVnef. At all four positions in all four animals, the new sequences represented consensus sequences for primate lentiviruses, whereas the inoculum sequences at these four loci have either never been or rarely been reported outside of SIVmac239. Thus, although cloned SIVmac239 is consistently pathogenic and consistently induces high viral load set points, it is clearly less than optimal at these four nucleotide positions.


Subject(s)
Evolution, Molecular , Macaca mulatta/virology , Mutation/genetics , Simian Immunodeficiency Virus/genetics , Simian Immunodeficiency Virus/pathogenicity , Amino Acid Sequence , Animals , Base Sequence , Cell Line , Consensus Sequence/genetics , Gene Products, env/chemistry , Gene Products, env/genetics , Gene Products, nef/physiology , Gene Products, pol/chemistry , Gene Products, pol/genetics , Gene Products, rev/chemistry , Gene Products, rev/genetics , Gene Products, tat/chemistry , Gene Products, tat/genetics , HIV-1/genetics , HIV-1/pathogenicity , HIV-1/physiology , Molecular Sequence Data , RNA, Transfer, Lys/genetics , RNA, Viral/genetics , RNA, Viral/metabolism , Sequence Alignment , Simian Immunodeficiency Virus/classification , Simian Immunodeficiency Virus/physiology , Terminal Repeat Sequences/genetics , Viral Load , nef Gene Products, Human Immunodeficiency Virus , rev Gene Products, Human Immunodeficiency Virus , tat Gene Products, Human Immunodeficiency Virus
3.
J Virol ; 74(17): 7745-54, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10933680

ABSTRACT

An effective vaccine for AIDS may require development of novel vectors capable of eliciting long-lasting immune responses. Here we report the development and use of replication-competent and replication-defective strains of recombinant herpes simplex virus (HSV) that express envelope and Nef antigens of simian immunodeficiency virus (SIV). The HSV recombinants induced antienvelope antibody responses that persisted at relatively stable levels for months after the last administration. Two of seven rhesus monkeys vaccinated with recombinant HSV were solidly protected, and another showed a sustained reduction in viral load following rectal challenge with pathogenic SIVmac239 at 22 weeks following the last vaccine administration. HSV vectors thus show great promise for being able to elicit persistent immune responses and to provide durable protection against AIDS.


Subject(s)
SAIDS Vaccines/immunology , Simian Acquired Immunodeficiency Syndrome/prevention & control , Simian Immunodeficiency Virus/immunology , Simplexvirus/immunology , Vaccines, Synthetic/immunology , Animals , Antibodies, Viral/biosynthesis , Injections, Intravenous , Macaca mulatta , SAIDS Vaccines/biosynthesis , SAIDS Vaccines/genetics , Simian Acquired Immunodeficiency Syndrome/immunology , Simplexvirus/genetics , Simplexvirus/metabolism , Vaccines, Attenuated/biosynthesis , Vaccines, Attenuated/genetics , Vaccines, Attenuated/immunology , Vaccines, Synthetic/biosynthesis , Vaccines, Synthetic/genetics , Viral Envelope Proteins/immunology , Viral Envelope Proteins/metabolism , Viral Load , Virus Replication
4.
J Virol ; 74(1): 584-90, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10590154

ABSTRACT

All members of the Herpesviridae family contain sequences for a highly conserved glycoprotein B (gB) gene. We investigated the phylogenetic relationships of gB sequences from eight independent rhadinovirus isolates obtained from three species: rhesus (Macaca mulatta), cynomologus (Macaca fasicularis), and pig-tailed (Macaca nemestrina) macaques. Samples were derived from monkeys housed at four separate facilities. Analysis of these eight independent gB sequences revealed five regions of heterogeneity within the 823- to 829-amino-acid polypeptides: residues 1 to 65, 120 to 185, 255 to 300, 352 to 393, and 412 to 457. The remaining regions of gB were highly conserved among the different macaque isolates. Overall divergence among these gene sequences ranged from 0.1 to 7.2% at the amino acid level. Phylogenetic trees constructed with our macaque rhadinovirus gB sequences and those derived from additional subfamilies or genera (alpha, beta, gamma-1, and gamma-2) revealed that the macaque gB sequences branched with other gamma-2 herpesvirus gB sequences and that within the gamma-2 genera, the macaque gB sequences clustered as a distinct branch. The eight macaque rhadinovirus gB sequences were all approximately equidistant from Kaposi sarcoma-associated herpesvirus (KSHV) gB sequences and had a shorter evolutionary distance to KSHV gB sequences than to any other herpesvirus, including the gamma-2 herpesvirus saimiri (HVS) of New World squirrel monkeys. The macaque gB sequences did not cluster according to the facility of origin, but did cluster according to the species of origin, displaying less intraspecies divergence (0.1 to 2.9%) than interspecies divergence (3.3 to 7.2%). These results demonstrate a close relatedness of rhadinovirus isolates from different macaque species.


Subject(s)
Glycoproteins/genetics , Rhadinovirus/genetics , Amino Acid Sequence , Animals , Base Sequence , DNA Primers , Glycoproteins/chemistry , Macaca , Molecular Sequence Data , Open Reading Frames , Phylogeny , Polymerase Chain Reaction , Sequence Homology, Amino Acid , Species Specificity
5.
J Virol ; 73(12): 10320-8, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10559350

ABSTRACT

The recognition of naturally occurring rhadinoviruses in macaque monkeys has spurred interest in their use as models for human infection with Kaposi sarcoma-associated herpesvirus (human herpesvirus 8). Rhesus macaques (Macaca mulatta) and pig-tailed macaques (Macaca nemestrina) were inoculated intravenously with rhadinovirus isolates derived from these species (rhesus rhadinovirus [RRV] and pig-tailed rhadinovirus [PRV]). Nine rhadinovirus antibody-negative and two rhadinovirus antibody-positive monkeys were used for these experimental inoculations. Antibody-negative animals clearly became infected following virus inoculation since they developed persisting antibody responses to virus and virus was isolated from peripheral blood on repeated occasions following inoculation. Viral sequences were also detected by PCR in lymph node, oral mucosa, skin, and peripheral blood mononuclear cells following inoculation. Experimentally infected animals developed peripheral lymphadenopathy which resolved by 12 weeks following inoculation, and these animals have subsequently remained free of disease. No increased pathogenicity was apparent from cross-species infection, i.e., inoculation of rhesus macaques with PRV or of pig-tailed macaques with RRV, whether the animals were antibody positive or negative at the time of virus inoculation. Coinoculation of additional rhesus monkeys with simian immunodeficiency virus (SIV) isolate SIVmac251 and macaque-derived rhadinovirus resulted in an attenuated antibody response to both agents and shorter mean survival compared to SIVmac251-inoculated controls (155.5 days versus 560.1 days; P < 0.019). Coinfected and immunodeficient macaques died of a variety of opportunistic infections characteristic of simian AIDS. PCR analysis of sorted peripheral blood mononuclear cells indicated a preferential tropism of RRV for CD20(+) B lymphocytes. Our results demonstrate persistent infection of macaque monkeys with RRV and PRV following experimental inoculation, but no specific disease was readily apparent from these infections even in the context of concurrent SIV infection.


Subject(s)
Herpesviridae Infections/immunology , Rhadinovirus/immunology , Tumor Virus Infections/immunology , Animals , Herpesviridae Infections/blood , Herpesviridae Infections/pathology , Herpesviridae Infections/virology , Immunocompetence/immunology , Lymphatic Diseases/immunology , Lymphatic Diseases/virology , Macaca mulatta , Macaca nemestrina , Rhadinovirus/genetics , Rhadinovirus/isolation & purification , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/immunology , Tumor Virus Infections/pathology , Tumor Virus Infections/virology
6.
J Virol ; 73(7): 5814-25, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10364333

ABSTRACT

A nef gene is present in all primate lentiviruses, including human immunodeficiency virus type 1 (HIV-1) and simian immunodeficiency virus of macaque monkeys (SIVmac). However, the nef genes of HIV-1 and SIVmac exhibit minimal sequence identity, and not all properties are shared by the two. Nef sequences of SIVmac239 were replaced by four independent nef alleles of HIV-1 in a context that was optimal for expression. The sources of the HIV-1 nef sequences included NL 4-3, a variant NL 4-3 gene derived from a recombinant-infected rhesus monkey, a patient nef allele, and a nef consensus sequence. Of 16 rhesus monkeys infected with these SHIVnef chimeras, 9 maintained high viral loads for prolonged periods, as observed with the parental SIVmac239, and 6 have died with AIDS 52 to 110 weeks postinfection. Persistent high loads were observed at similar frequencies with the four different SIV recombinants that expressed these independent HIV-1 nef alleles. Infection with other recombinant SHIVnef constructions resulted in sequence changes in infected monkeys that either created an open nef reading frame or optimized the HIV-1 nef translational context. The HIV-1 nef gene was uniformly retained in all SHIVnef-infected monkeys. These results demonstrate that HIV-1 nef can substitute for SIVmac nef in vivo to produce a pathogenic infection. However, the model suffers from an inability to consistently obtain persisting high viral loads in 100% of the infected animals, as is observed with the parental SIVmac239.


Subject(s)
Acquired Immunodeficiency Syndrome/virology , Gene Products, nef/physiology , HIV-1 , Simian Immunodeficiency Virus/physiology , Amino Acid Sequence , Animals , Base Sequence , Cell Line , DNA, Viral , Gene Expression , Gene Products, nef/genetics , Humans , Kinetics , Macaca mulatta , Molecular Sequence Data , Recombination, Genetic , Simian Immunodeficiency Virus/growth & development , nef Gene Products, Human Immunodeficiency Virus
7.
J Virol ; 73(6): 4952-61, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10233957

ABSTRACT

Three different deletion mutants of simian immunodeficiency virus (SIV) that vary in their levels of attenuation were tested for the ability to protect against mucosal challenge with pathogenic SIV. Four female rhesus monkeys were vaccinated by intravenous inoculation with SIVmac239Delta3, four with SIVmac239Delta3X, and four with SIVmac239Delta4. These three vaccine strains exhibit increasing levels of attenuation: Delta3 < Delta3X

Subject(s)
SAIDS Vaccines/immunology , Vagina/virology , Animals , Antibodies, Viral/blood , CD4 Lymphocyte Count , Female , Macaca mulatta , T-Lymphocytes, Cytotoxic/immunology , Vaccines, Attenuated/immunology
8.
AIDS Res Hum Retroviruses ; 15(1): 11-21, 1999 Jan 01.
Article in English | MEDLINE | ID: mdl-10024048

ABSTRACT

We engineered recombinant derivatives of simian immunodeficiency virus (SIV) to express enhanced green fluorescent protein (EGFP). Replacement of vpr sequences with EGFP resulted in a genome that did not produce detectable levels of replication-competent virus. Replication-competent virus and bright fluorescence of infected cells were obtained with two other constructs, one in which SIV nef sequences were replaced by EGFP and another in which EGFP was inserted into the SIV nef locus and HIV-1 nef sequences were expressed by downstream placement of an internal ribosomal entry site. These strains were infectious in rhesus monkeys and green fluorescing cells were detected in the tissues of infected monkeys by FACS analysis and by direct microscopic visualization. EGFP sequences were absent from recovered virus by 8 weeks following infection. We conclude that recombinant SIV that is engineered to express EGFP can be used to directly detect productively infected cells and aid in the immunophenotypic characterization of these cells within the first 2 weeks of infection of rhesus monkeys.


Subject(s)
Luminescent Proteins/genetics , Recombinant Fusion Proteins/metabolism , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/genetics , Animals , Base Sequence , Cell Line , Flow Cytometry , Green Fluorescent Proteins , Immunohistochemistry , In Situ Hybridization , Indicators and Reagents/metabolism , Luminescent Proteins/metabolism , Macaca mulatta , Microscopy, Fluorescence , Molecular Sequence Data , Polymerase Chain Reaction/methods , Recombinant Fusion Proteins/genetics , Simian Immunodeficiency Virus/metabolism , Simian Immunodeficiency Virus/physiology , Transfection , Viral Load , Virus Replication
9.
J Virol ; 73(2): 1341-9, 1999 Feb.
Article in English | MEDLINE | ID: mdl-9882339

ABSTRACT

Kaposi's sarcoma-associated herpesvirus (KSHV) has been consistently identified in Kaposi's sarcomas (KS), body cavity-based lymphomas (BCBL), and some forms of Castleman's disease. Previous serological tests with KS patient sera have detected lytic-cycle polypeptides from KSHV-infected BCBL cells. We have found that these polypeptides are predominantly encoded by the K8.1 open reading frame, which is present in the same genomic position as virion envelope glycoproteins of other gammaherpesviruses. The cDNA of K8.1 from BCBL-1 cells was found to encode a glycosylated protein with an apparent molecular mass of 37 kDa. K8.1 was found to be expressed during lytic KSHV replication in BCBL-1 cells and was localized on the surface of cells and virions. The results of immunofluorescence and immunoelectron microscopy suggest that KSHV acquires K8.1 protein on its virion surface during the process of budding at the plasma cell membrane. When KSHV K8.1 derived from mammalian cells was used as an antigen in immunoblot tests, antibodies to K8.1 were detected in 18 of 20 KS patients and in 0 of 10 KS-negative control subjects. These results demonstrate that the K8.1 gene encodes a KSHV virion-associated glycoprotein and suggest that antibodies to K8.1 may prove useful as contributory serological markers for infection by KSHV.


Subject(s)
Glycoproteins/genetics , Herpesvirus 8, Human/genetics , Sarcoma, Kaposi/virology , Viral Envelope Proteins/genetics , Viral Proteins , Amino Acid Sequence , Animals , Antibodies, Viral/immunology , Base Sequence , COS Cells , Cloning, Molecular , DNA, Viral , Glycoproteins/metabolism , Herpesvirus 8, Human/immunology , Herpesvirus 8, Human/metabolism , Humans , Molecular Sequence Data , Prevalence , Rabbits , Sarcoma, Kaposi/epidemiology , Sarcoma, Kaposi/immunology , Sarcoma, Kaposi/metabolism , Sequence Analysis, DNA , Tumor Cells, Cultured , Viral Envelope Proteins/metabolism , Virion
10.
J Virol ; 72(8): 6770-6, 1998 Aug.
Article in English | MEDLINE | ID: mdl-9658125

ABSTRACT

The product of open reading frame 14 (orf14) of herpesvirus saimiri (HVS) exhibits significant homology with mouse mammary tumor virus superantigen. orf14 encodes a 50-kDa secreted glycoprotein, as shown previously (Z. Yao, E. Maraskovsky, M. K. Spriggs, J. I. Cohen, R. J. Armitage, and M. R. Alderson, J. Immunol. 156:3260-3266, 1996). orf14 expressed from recombinant baculovirus powerfully induces proliferation of CD4-positive cells originating from several different species. To study the role of orf14 in transformation, a mutant form of HVS (HVS Deltaorf14) was constructed with a deletion in the orf14 gene. The transforming potential of HVS Deltaorf14 was tested in cell culture and in common marmosets. Parental HVS subgroup C strain 488 immortalized common marmoset T lymphocytes in vitro to interleukin-2-independent growth, while the HVS Deltaorf14 mutant did not produce such a growth transformation. In addition, HVS Deltaorf14 was nononcogenic in common marmosets. In contrast to other nononcogenic HVS mutant viruses which were repeatedly isolated from peripheral blood mononuclear cells of infected marmosets for more than 5 months, HVS Deltaorf14 did not persist at a high level in vivo. These results demonstrate that orf14 of HVS is not required for replication but is required for transformation and for high-level persistence in vivo.


Subject(s)
Cell Transformation, Viral , Herpesviridae Infections/virology , Herpesvirus 2, Saimiriine/physiology , Lymphoma/virology , Oncogene Proteins, Viral/physiology , Tumor Virus Infections/virology , Virus Latency , Animals , Aotidae , Callithrix , Cell Division , Cell Line , Disease Models, Animal , Herpesviridae Infections/immunology , Herpesvirus 2, Saimiriine/genetics , Herpesvirus 2, Saimiriine/immunology , Humans , Lymphoma/immunology , Macaca mulatta , Mutagenesis , Oncogene Proteins, Viral/genetics , Open Reading Frames , Rabbits , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/physiology , Saimiri , Spodoptera , T-Lymphocytes/cytology , Tumor Virus Infections/immunology
11.
J Virol ; 72(4): 2607-14, 1998 Apr.
Article in English | MEDLINE | ID: mdl-9525577

ABSTRACT

The proline-rich SH3-binding (SH3B) motif of the tyrosine kinase-interacting protein (Tip) of herpesvirus saimiri (HVS) is required for binding to the cellular Src family kinase Lck. We constructed a mutant form of HVS in which prolines in the SH3B motif of Tip were altered to alanines. This mutant form of Tip was incapable of binding to Lck. The mutant virus, HVS/Tip mSH3B, retained its ability to immortalize common marmoset lymphocytes in culture. In fact, common marmoset lymphocytes immortalized by the HVS/Tip mSH3B mutant displayed increased expression of HLA-DR lymphocyte activation marker, an altered pattern of tyrosine phosphorylation, increased expression of the tyrosine kinase Lyn, and a shift in electrophoretic mobility of Lck compared to cells immortalized by wild-type HVS. Experimental infection of common marmosets resulted in fulminant lymphoma with both HVS/Tip mSH3B and wild-type HVS. However, HVS/Tip mSH3B produced greater infiltration of affected organs by proliferating lymphoid cells compared to wild-type HVS. These results demonstrate that Tip binding to Lck is not necessary for transformation and that abrogation of Tip binding to Lck alters the characteristics of transformed cells and the severity of the pathologic lesions.


Subject(s)
Herpesvirus 2, Saimiriine/immunology , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/metabolism , Phosphoproteins/genetics , Phosphoproteins/immunology , Point Mutation , T-Lymphocytes/immunology , Viral Proteins/genetics , Viral Proteins/immunology , Animals , Aotidae , Binding Sites , Callithrix , Cell Line , Cell Line, Transformed , Cells, Cultured , Flow Cytometry , Herpesvirus 2, Saimiriine/genetics , Herpesvirus 2, Saimiriine/metabolism , Lymphocyte Activation , NF-kappa B/metabolism , Phosphoproteins/metabolism , Promoter Regions, Genetic , Recombination, Genetic , Signal Transduction , T-Lymphocytes/metabolism , T-Lymphocytes/virology , Viral Proteins/metabolism , src-Family Kinases/genetics
12.
J Virol ; 72(2): 1308-13, 1998 Feb.
Article in English | MEDLINE | ID: mdl-9445031

ABSTRACT

Mutant forms of herpesvirus saimiri (HVS) subgroup C strain 488 with deletions in either STP-C488 or Tip were constructed. The transforming potentials of the HVS mutants were tested in cell culture and in common marmosets. Parental HVS subgroup C strain 488 immortalized common marmoset T lymphocytes in vitro to interleukin-2-independent growth, but neither of the deletion mutants produced such growth transformation. Wild-type HVS produced fatal lymphoma within 19 to 20 days of experimental infection of common marmosets, while HVS deltaSTP-C488 and HVS deltaTip were nononcogenic. Virus was repeatedly isolated from the peripheral blood of marmosets infected with mutant virus for more than 5 months. These results demonstrate that STP-C488 and Tip are not required for replication or persistence, but each is essential for transformation in cell culture and for lymphoma induction in common marmosets.


Subject(s)
Gene Deletion , Genes, Viral , Herpesviridae Infections/virology , Herpesvirus 2, Saimiriine/genetics , Oncogene Proteins, Viral/genetics , Phosphoproteins/genetics , T-Lymphocytes/virology , Tumor Virus Infections/virology , Viral Proteins/genetics , Animals , Herpesvirus 2, Saimiriine/pathogenicity , Virulence/genetics
13.
J Virol ; 72(2): 1431-7, 1998 Feb.
Article in English | MEDLINE | ID: mdl-9445045

ABSTRACT

Deletion mutants of the pathogenic clone of simian immunodeficiency virus isolate 239 (SIVmac239) were derived that are missing nef, vpr, and upstream sequences (US) in the U3 region of the LTR (SIVmac239 delta3), nef, vpx, and US (SIVmac239 delta3x), and nef, vpr, vpx, and US (SIVmac239 delta4). These multiply deleted derivatives replicated well in the continuously growing CEMx174 cell line and were infectious for rhesus monkeys. However, on the basis of virus load measurements, strength of antibody responses, and lack of disease progression, these mutants were highly attenuated. Measurements of cell-associated viral load agreed well with assays of plasma viral RNA load and with the strengths of the antibody responses; thus, these measurements likely reflected the extent of viral replication in vivo. A derivative of SIVmac239 lacking vif sequences (SIVmac239 delta vif) could be consistently grown only in a vif-complementing cell line. This delta vif virus appeared to be very weakly infectious for rhesus monkeys on the basis of sensitive antibody tests only. The weak antibody responses elicited by SIVmac239 delta vif were apparently in response to low levels of replicating virus since they were not elicited by heat-inactivated virus and the anti-SIV antibody responses persisted for greater than 1 year. These results, and the results of previous studies, allow a rank ordering of the relative virulence of nine mutant strains of SIVmac according to the following order: delta vpr > delta vpx > delta vpr delta vpx approximately delta nef > delta3 > delta3x > or = delta4 > delta vif > delta5. The results also demonstrate that almost any desired level of attenuation can be achieved, ranging from still pathogenic in a significant proportion of animals (delta vpr and delta vpx) to not detectably infectious (delta5), simply by varying the number and location of deletions in these five loci.


Subject(s)
Gene Deletion , Genome, Viral , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/genetics , Animals , Genes, Viral , Macaca mulatta , Simian Immunodeficiency Virus/pathogenicity , Virulence/genetics
14.
J Virol ; 71(12): 9650-3, 1997 Dec.
Article in English | MEDLINE | ID: mdl-9371629

ABSTRACT

Recombination may be an important mechanism for increasing variation in retroviral populations. Retroviral recombination has been demonstrated in tissue culture systems by artificially creating doubly infected cells. Evidence for retroviral recombination in vivo is indirect and is based principally on the identification of apparently mosaic human immunodeficiency virus type 1 genomes from phylogenetic analyses of viral sequences. We infected a rhesus monkey with two different molecularly cloned strains of simian immunodeficiency virus. One strain of virus had a deletion in vpx and vpr, and the other strain had a deletion in nef. Each strain on its own induced low virus loads and was nonpathogenic in rhesus monkeys. When injected simultaneously into separate legs of the same monkey, persistent high virus loads and declines in CD4+ lymphocyte concentrations were observed. Analysis of proviral DNA isolated directly from peripheral blood mononuclear cells showed that full-length, nondeleted SIVmac239 predominated by 2 weeks after infection. These results provide direct experimental evidence for genetic recombination between two different retroviral strains in an infected host. The results illustrate the ease and rapidity with which recombination can occur in an infected animal and the selection that can occur for variants generated by genetic recombination.


Subject(s)
Genes, nef , Genes, vpr , Lentivirus Infections/veterinary , Recombination, Genetic , Retroviridae Proteins/genetics , Simian Immunodeficiency Virus/genetics , Viral Regulatory and Accessory Proteins/genetics , Animals , DNA, Viral , Deoxyribonucleases, Type II Site-Specific/metabolism , Lentivirus Infections/virology , Macaca mulatta , Polymerase Chain Reaction , Proviruses/genetics , Retroviridae/genetics
15.
J Virol ; 71(12): 9764-9, 1997 Dec.
Article in English | MEDLINE | ID: mdl-9371642

ABSTRACT

A herpesvirus that is related to but distinct from the Kaposi's sarcoma-associated herpesvirus (KSHV, or human herpesvirus 8) was isolated from rhesus monkeys. The sequence of 10.6 kbp from virion DNA revealed the presence of an interleukin-6 homolog similar to what is present in KSHV and a closer relatedness of the DNA polymerase and glycoprotein B reading frames to those of KSHV than to those of any other herpesvirus. This rhesus monkey herpesvirus replicated lytically and to high titers in cultured rhesus monkey fibroblasts. Antibody testing revealed a high prevalence for at least 10 years in our rhesus monkey colony and a high prevalence in two other colonies that were tested. Thus, rhesus monkeys naturally harbor a virus related to KSHV, which we have called RRV, for rhesus monkey rhadinovirus.


Subject(s)
Herpesviridae Infections/veterinary , Herpesvirus 8, Human , Macaca mulatta/virology , Rhadinovirus/isolation & purification , Sarcoma, Kaposi/virology , Tumor Virus Infections/veterinary , Animals , Antibodies, Viral/blood , Base Sequence , Cells, Cultured , Cytopathogenic Effect, Viral , DNA, Viral , DNA-Directed DNA Polymerase/genetics , Glycoproteins/genetics , Herpesviridae , Herpesviridae Infections/immunology , Herpesviridae Infections/virology , Humans , Interleukin-6/genetics , Molecular Sequence Data , Rhadinovirus/genetics , Rhadinovirus/immunology , Tumor Virus Infections/immunology , Tumor Virus Infections/virology , Viral Proteins/genetics
16.
J Virol ; 71(11): 8141-8, 1997 Nov.
Article in English | MEDLINE | ID: mdl-9343164

ABSTRACT

Three rhesus macaques, previously immunized with SIVdelta3 or SIVdelta2, each an attenuated derivative of SIVmac239, and two naive monkeys were challenged with 30,000 50% tissue culture infective doses of SHIV, an SIV/human immunodeficiency virus type 1 (HIV-1) chimeric virus bearing the dual-tropic envelope of HIV-1DH12. By several criteria, including virus isolation, serological assays, and PCR (both DNA and reverse transcriptase), SHIV levels were reduced to barely detectable levels in the circulating blood of vaccinated animals. The resistant SIV-vaccinated macaques had no preexisting neutralizing antibodies directed against SHIV, nor did they produce neutralizing antibodies at any time over a 14-month observation period following SHIV challenge. Interestingly, SIV sequences, derived from the vaccine, could be amplified from numerous tissue samples collected at the conclusion of the experiment, 60 weeks postchallenge, but SHIV-specific sequences (viz., HIV-1 env) could not. These results demonstrate that live attenuated SIV vaccines provide strong long-term protection even against challenge strains with highly divergent envelope sequences.


Subject(s)
HIV-1/immunology , Reassortant Viruses/immunology , SAIDS Vaccines/immunology , Simian Immunodeficiency Virus/immunology , Vaccines, Attenuated/immunology , Animals , Antibodies, Viral/biosynthesis , HIV Antigens/immunology , Macaca mulatta , Neutralization Tests , Retroviridae Infections/immunology , Retroviridae Infections/prevention & control , Retroviridae Infections/virology , Tissue Distribution , Viral Proteins/immunology , Virus Replication
17.
J Virol ; 71(3): 1880-7, 1997 Mar.
Article in English | MEDLINE | ID: mdl-9032318

ABSTRACT

Rhesus monkeys (Macaca mulatta) were infected with five strains of simian immunodeficiency virus (SIV) derived from SIVmac239 containing deletions (delta) or substitutions (subst) in NF-kappaB and Sp1 binding sites. We have shown previously that mutations in these regions still allow efficient SIVmac replication in primary lymphoid cell cultures (P. O. Ilyinskii and R. C. Desrosiers, J. Virol. 70:3118-3126, 1996). Two animals were inoculated intravenously with each mutant strain of SIVmac239: delta NFkappaB, delta Sp1234, delta NFkappaB delta Sp1234, substSp12, and substSp1234. All but one of the infected animals showed an early spike in plasma antigenemia, maintained high virus burdens, and had significant changes in lymphoid tissues, and six died with AIDS within the first 60 weeks of infection. One of the animals infected with the SIV strain delta NFkappaB delta Sp1234 showed lower levels of plasma antigenemia and lower virus burdens; the other animal infected with this same mutant strain died with AIDS 17 weeks after inoculation. No consistent novel mutations or reversions were detected in proviral sequences derived from the animals infected with the deletion mutants and the substSp12 mutant by 20 weeks postinfection. Point-mutated sequences were partially deleted in both animals infected with the substSp1234 strain. These results indicate that the NF-kappaB and Sp1 binding sites are not essential for the induction of AIDS by SIVmac239. They also provide indirect evidence for the importance of a novel enhancer element in the U3 region of the SIVmac long terminal repeat that is located immediately upstream of the NF-kappaB binding site within the C-terminal region of the nef coding sequence.


Subject(s)
NF-kappa B/metabolism , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/genetics , Sp1 Transcription Factor/metabolism , Animals , Antibodies, Viral/blood , Antigens, Viral/blood , Binding Sites , Cell Line , Disease Progression , Gene Products, gag/blood , Macaca mulatta , Mutagenesis , Simian Acquired Immunodeficiency Syndrome/blood , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Immunodeficiency Virus/immunology
18.
Science ; 258(5090): 1938-41, 1992 Dec 18.
Article in English | MEDLINE | ID: mdl-1470917

ABSTRACT

Vaccine protection against the human immunodeficiency virus (HIV) and the related simian immunodeficiency virus (SIV) in animal models is proving to be a difficult task. The difficulty is due in large part to the persistent, unrelenting nature of HIV and SIV infection once infection is initiated. SIV with a constructed deletion in the auxiliary gene nef replicates poorly in rhesus monkeys and appears to be nonpathogenic in this normally susceptible host. Rhesus monkeys vaccinated with live SIV deleted in nef were completely protected against challenge by intravenous inoculation of live, pathogenic SIV. Deletion of nef or of multiple genetic elements from HIV may provide the means for creating a safe, effective, live attenuated vaccine to protect against acquired immunodeficiency syndrome (AIDS).


Subject(s)
Genes, nef , Sequence Deletion , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Immunodeficiency Virus/genetics , Simian Immunodeficiency Virus/immunology , Vaccines, Attenuated/immunology , Viral Vaccines/immunology , Animals , Base Sequence , DNA, Viral/analysis , DNA, Viral/genetics , DNA, Viral/isolation & purification , Macaca mulatta , Molecular Sequence Data , Polymerase Chain Reaction , Simian Acquired Immunodeficiency Syndrome/prevention & control , Simian Immunodeficiency Virus/isolation & purification
SELECTION OF CITATIONS
SEARCH DETAIL
...