Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 57
Filter
Add more filters

Country/Region as subject
Publication year range
1.
J Gen Virol ; 98(11): 2759-2770, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28984228

ABSTRACT

Identifying molecular determinants of virulence attenuation in live attenuated canine parvovirus (CPV) vaccines is important for assuring their safety. To this end, we identified mutations in the attenuated CPV 9985-46 vaccine strain that arose during serial passage in Crandell-Rees feline kidney cells by comparison with the wild-type counterpart, as well as minimal determinants of the loss of virulence. Four amino acid substitutions (N93K, G300V, T389N and V562L) in VP2 of strain 9985-46 significantly restricted infection in canine A72 cells. Using an infectious molecular clone system, we constructed isogenic CPVs of the parental virulent 9985 strain carrying single or double mutations. We observed that only a single amino acid substitution in VP2, G300V or T389N, attenuated the virulent parental virus. Combinations of these mutations further attenuated CPV to a level comparable to that of 9985-46. Strains with G300V/T389N substitutions did not induce clinical symptoms in experimentally infected pups, and their ability to infect canine cells was highly restricted. We found that another G300V/V562L double mutation decreased affinity of the virus for canine cells, although its pathogenicity to dogs was maintained. These results indicate that mutation of residue 300, which plays a critical role in host tropism, is not sufficient for viral attenuation in vivo, and that attenuation of 9985-46 strain is defined by at least two mutations in residues 300 and 389 of the VP2 capsid protein. This finding is relevant for quality control of the vaccine and provides insight into the rational design of second-generation live attenuated vaccine candidates.


Subject(s)
Amino Acid Substitution , Capsid Proteins/genetics , Capsid Proteins/metabolism , Parvovirus, Canine/genetics , Parvovirus, Canine/pathogenicity , Viral Vaccines/genetics , Animals , Animals, Newborn , Cell Line , DNA Mutational Analysis , Dog Diseases/pathology , Dog Diseases/virology , Dogs , Parvoviridae Infections/pathology , Parvoviridae Infections/veterinary , Parvoviridae Infections/virology , Serial Passage , Vaccines, Attenuated/genetics , Virulence Factors/genetics , Virulence Factors/metabolism
2.
J Gen Virol ; 98(7): 1730-1738, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28691897

ABSTRACT

The protective efficacy of intranasal (IN) administration of inactivated feline calicivirus (FCV) vaccine against homologous or heterologous FCV infection was investigated. Groups of cats immunized with the experimental inactivated, non-adjuvanted FCV vaccine via either the IN or subcutaneous (SC) route were exposed to homologous or highly heterologous FCV. Both the IN and SC immunization protocols established robust protection against homologous FCV infection. Although neither immunization regimen conferred protection against the heterologous strain, clinical scores and virus titres of oral swabs were lower in cats in the IN group compared to those in the SC group, accompanying a faster neutralizing antibody response against the heterologous virus in cats in the IN group. The IN group secreted more IgA specific to FCV proteins in oral washes (lavage fluids from the oral cavity) than the SC group. IN immunization with an inactivated whole FCV particle, which protects cats from homologous virus exposure and shortens the period of heterologous virus shedding, may serve as a better platform for anti-FCV vaccine.


Subject(s)
Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Calicivirus, Feline/immunology , Vaccination/veterinary , Vaccines, Inactivated/immunology , Viral Vaccines/immunology , Administration, Intranasal , Animals , Caliciviridae Infections/immunology , Caliciviridae Infections/prevention & control , Cat Diseases/immunology , Cat Diseases/prevention & control , Cat Diseases/virology , Cats , Immunoglobulin A/immunology
3.
J Gen Virol ; 97(5): 1249-1260, 2016 05.
Article in English | MEDLINE | ID: mdl-26850058

ABSTRACT

Previously, we reported that a new genetically diverse CCR5 (R5) tropic simian/human immunodeficiency virus (SHIV-MK38) adapted to rhesus monkeys became more neutralization resistant to SHIV-infected plasma than did the parental SHIV-KS661 clone. Here, to clarify the significance of the neutralization-resistant phenotype of SHIV in a macaque model, we initially investigated the precise neutralization phenotype of the SHIVs, including SHIV-MK38 molecular clones, using SHIV-MK38-infected plasma, a pooled plasma of human immunodeficiency virus (HIV)-infected individuals, soluble CD4 and anti-HIV-1 neutralizing mAbs, the epitopes of which were known. The results show that SHIV-KS661 had tier 1 neutralization sensitivity, but monkey-adapted R5 tropic SHIV-MK38 acquired neutralization resistance similar to that of tier 2 or 3 as a clone virus. Sequence analysis of the env gene suggested that the neutralization-resistant phenotype of SHIV-MK38 was acquired by conformational changes in Env associated with the net charge and potential N-linked glycosylation sites. To examine the relationship between neutralization phenotype and stably persistent infection in monkeys, we performed in vivo rectal inoculation experiments using a SHIV-MK38 molecular clone. The results showed that one of three rhesus monkeys exhibited durable infection with a plasma viral load of 105 copies ml- 1 despite the high antibody responses that occurred in the host. Whilst further improvements are required in the development of a challenge virus, it will be useful to generate a neutralization-resistant R5 tropic molecular clone of the SHIV-89.6 lineage commonly used for vaccine development - a result that can be used to explore the foundation of AIDS pathogenesis.


Subject(s)
Viral Proteins/metabolism , Animals , Cell Line , Cloning, Molecular , HIV , Humans , Macaca mulatta , Models, Molecular , Protein Conformation , RNA, Viral/genetics , Simian Immunodeficiency Virus , Viral Proteins/genetics , Virus Replication
4.
Biochem Biophys Res Commun ; 448(1): 95-100, 2014 May 23.
Article in English | MEDLINE | ID: mdl-24769207

ABSTRACT

ISGylation, an ubiquitin-like post-translational modification by ISG15, has been reported to participate in the interferon (IFN)-mediated antiviral response. In this study, we analyzed the functional role of ISGylation in dengue virus 2 (DENV-2) replication. Overexpression of ISG15 was found to significantly suppress the amount of extracellular infectious virus released, while intracellular viral RNA was unaffected. This effect was not observed with a conjugation-defective ISG15 mutant. In addition, extracellular virus infectivity was decreased by ISG15 overexpression. To further clarify the role of ISGylation in the anti-DENV-2 response, we depleted endogenous ISG15 by RNA interference and analyzed the virus production in the absence or presence of type-I IFN. Results showed a significant reduction in extracellular DENV-2 RNA levels for cells treated with IFN, and that these DENV-2 RNA levels could be partially restored by the ISG15 knockdown. Among various DENV-2 proteins, NS3 and NS5 were subjected to the ISGylation. These results demonstrate that IFN-inducible ISGylation suppresses DENV-2 particle release, and that ISG15 is one of the mediators of IFN-induced inhibition of DENV-2 replication. ISG15 therefore functions as a host antiviral factor against DENV-2 infection.


Subject(s)
Cytokines/metabolism , Dengue Virus/physiology , Interferon Type I/pharmacology , Ubiquitins/metabolism , Virus Replication/drug effects , Animals , Cell Line , Cricetinae , Dengue Virus/drug effects , HEK293 Cells , HeLa Cells , Humans , RNA Interference , Viral Nonstructural Proteins/metabolism
5.
J Virol ; 87(8): 4789-93, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23408611

ABSTRACT

To elucidate the mode of viral persistence in primate lentivirus-infected individuals during combination antiretroviral therapy (cART), four simian immunodeficiency virus 239-infected monkeys were treated with cART for 1 year. The viral env genes prepared from total RNA extracted from the mesenteric lymph nodes collected at the completion of therapy were assessed by single genome amplification. Analyses of nucleotide substitutions and phylogeny revealed no viral evolution during cART.


Subject(s)
Anti-Retroviral Agents/administration & dosage , Evolution, Molecular , Lymph Nodes/virology , Simian Acquired Immunodeficiency Syndrome/drug therapy , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/classification , Simian Immunodeficiency Virus/genetics , Animals , Antiretroviral Therapy, Highly Active , Gene Products, env/genetics , Macaca mulatta , Phylogeny , Point Mutation , Sequence Analysis, DNA , Simian Immunodeficiency Virus/isolation & purification
6.
J Virol ; 87(1): 482-8, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23097434

ABSTRACT

We have recently isolated a rhesus macaque cytotoxic T cell line, 2N5.1, that specifically recognizes an N-myristoylated 5-mer peptide (C(14)-Gly-Gly-Ala-Ile-Ser [C14nef5]) derived from the simian immunodeficiency virus (SIV) Nef protein. Such C14nef5-specific T cells expand in the circulation of SIV-infected monkeys, underscoring the capacity of T cells to recognize viral lipopeptides; however, the molecular basis for the lipopeptide antigen presentation remains to be elucidated. Here, functional studies indicated that the putative antigen-presenting molecule for 2N5.1 was likely to have two separate antigen-binding sites, one for interaction with a C(14)-saturated acyl chain and the other for anchorage of the C-terminal serine residue. Mutants with alanine substitutions for the second glycine residue and the fourth isoleucine residue were not recognized by 2N5.1 but interfered with the presentation of C14nef5 to 2N5.1, indicating that these structural analogues retained the ability to interact with the antigen-presenting molecules. In contrast to the highly specific recognition of C14nef5 by 2N5.1, an additional cytotoxic T cell line, SN45, established independently from a C14nef5-stimulated T cell culture, showed superb reactivity to both C14nef5 and an N-myristoylated Nef 4-mer peptide, and therefore, the C-terminal serine residue was dispensable for the recognition of lipopeptides by the SN45 T cells. Furthermore, the mutants with alanine substitutions were indeed recognized by the SN45 T cells. Given that N-myristoylation of the Nef protein occurs in the conserved motifs and is critical for viral pathogenesis, these observations predict that the lipopeptide-specific T cell response is difficult for viruses to avoid by simply introducing amino acid mutations.


Subject(s)
Gene Products, nef/immunology , Lipopeptides/immunology , Oligopeptides/immunology , Simian Immunodeficiency Virus/immunology , T-Lymphocytes, Cytotoxic/immunology , Amino Acid Substitution , Animals , Gene Products, nef/metabolism , Lipopeptides/metabolism , Macaca mulatta , Molecular Sequence Data , Mutant Proteins/immunology , Mutant Proteins/metabolism , Oligopeptides/metabolism , Protein Binding , Sequence Analysis, DNA
7.
J Virol ; 87(21): 11447-61, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23966385

ABSTRACT

Human immunodeficiency virus type 1 (HIV-1) replication in macaque cells is restricted mainly by antiviral cellular APOBEC3, TRIM5α/TRIM5CypA, and tetherin proteins. For basic and clinical HIV-1/AIDS studies, efforts to construct macaque-tropic HIV-1 (HIV-1mt) have been made by us and others. Although rhesus macaques are commonly and successfully used as infection models, no HIV-1 derivatives suitable for in vivo rhesus research are available to date. In this study, to obtain novel HIV-1mt clones that are resistant to major restriction factors, we altered Gag and Vpu of our best HIV-1mt clone described previously. First, by sequence- and structure-guided mutagenesis, three amino acid residues in Gag-capsid (CA) (M94L/R98S/G114Q) were found to be responsible for viral growth enhancement in a macaque cell line. Results of in vitro TRIM5α susceptibility testing of HIV-1mt carrying these substitutions correlated well with the increased viral replication potential in macaque peripheral blood mononuclear cells (PBMCs) with different TRIM5 alleles, suggesting that the three amino acids in HIV-1mt CA are involved in the interaction with TRIM5α. Second, we replaced the transmembrane domain of Vpu of this clone with the corresponding region of simian immunodeficiency virus SIVgsn166 Vpu. The resultant clone, MN4/LSDQgtu, was able to antagonize macaque but not human tetherin, and its Vpu effectively functioned during viral replication in a macaque cell line. Notably, MN4/LSDQgtu grew comparably to SIVmac239 and much better than any of our other HIV-1mt clones in rhesus macaque PBMCs. In sum, MN4/LSDQgtu is the first HIV-1 derivative that exhibits resistance to the major restriction factors in rhesus macaque cells.


Subject(s)
HIV-1/physiology , Macaca mulatta/virology , Viral Tropism , Virus Replication , Animals , Cells, Cultured , Gene Products, gag/genetics , Gene Products, gag/metabolism , HIV-1/genetics , HIV-1/immunology , Human Immunodeficiency Virus Proteins/genetics , Human Immunodeficiency Virus Proteins/metabolism , Humans , Leukocytes, Mononuclear/virology , Mutant Proteins/genetics , Mutant Proteins/metabolism , Viral Regulatory and Accessory Proteins/genetics , Viral Regulatory and Accessory Proteins/metabolism
8.
Infect Immun ; 81(1): 311-6, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23132493

ABSTRACT

Human CD1b molecules contain a maze of hydrophobic pockets and a tunnel capable of accommodating the unusually long, branched acyl chain of mycolic acids, an essential fatty acid component of the cell wall of mycobacteria. It has been accepted that CD1b-bound mycolic acids constitute a scaffold for mycolate-containing (glyco)lipids stimulating CD1b-restricted T cells. Remarkable homology in amino acid sequence is observed between human and monkey CD1b molecules, and indeed, monkey CD1b molecules are able to bind glucose monomycolate (GMM), a glucosylated species of mycolic acids, and present it to specific human T cells in vitro. Nevertheless, we found, unexpectedly, that Mycobacterium bovis bacillus Calmette-Guerin (BCG)-vaccinated monkeys exhibited GMM-specific T cell responses that were restricted by CD1c rather than CD1b molecules. GMM-specific, CD1c-restricted T cells were detected in the circulation of all 4 rhesus macaque monkeys tested after but not before vaccination with BCG. The circulating GMM-specific T cells were detected broadly in both CD4(+) and CD8(+) cell populations, and upon antigenic stimulation, a majority of the GMM-specific T cells produced both gamma interferon (IFN-γ) and tumor necrosis factor alpha (TNF-α), two major host protective cytokines functioning against infection with mycobacteria. Furthermore, the GMM-specific T cells were able to extravasate and approach the site of infection where CD1c(+) cells accumulated. These observations indicate a previously inconceivable role for primate CD1c molecules in eliciting T cell responses to mycolate-containing antigens.


Subject(s)
Antigens, CD1/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Glycolipids/immunology , Macaca mulatta/immunology , Mycobacterium Infections/immunology , Mycobacterium/immunology , Animals , Antigen Presentation/immunology , Antigens, Bacterial/immunology , Antigens, Bacterial/metabolism , Antigens, CD1/metabolism , BCG Vaccine/immunology , BCG Vaccine/metabolism , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Glycolipids/metabolism , Interferon-gamma/immunology , Interferon-gamma/metabolism , Macaca mulatta/metabolism , Mycobacterium/metabolism , Mycobacterium Infections/metabolism , Mycobacterium Infections/microbiology , Mycobacterium bovis/immunology , Mycobacterium bovis/metabolism , Mycolic Acids/immunology , Mycolic Acids/metabolism , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/metabolism
9.
J Gen Virol ; 94(Pt 10): 2202-2207, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23851439

ABSTRACT

To investigate the potential role of non-human primates (NHPs) in a dengue virus (DENV) epidemic, we conducted serological and genomic studies using plasma samples collected from 100 cynomolgus monkeys (Macaca fascicularis) in an animal breeding facility in the Philippines. An ELISA revealed 21 monkeys with a positive IgM reaction and 19 positive for IgG. Five of the monkeys were positive for both IgM and IgG. Of the 21 IgM-positive samples, a neutralization assay identified seven containing DENV-specific antibodies. We amplified the viral non-structural 1 (NS1) gene in two and the envelope (E) gene in one of these seven samples by RT-PCR. Phylogenetic analyses revealed that these DENV genes belonged to the epidemic DENV-2 family, not the sylvatic DENV family. These results suggest that NHPs may serve as a reservoir of epidemic DENV; therefore, the ecology of the urban DENV infection cycle should be investigated in these animals in detail.


Subject(s)
Dengue Virus/immunology , Dengue/veterinary , Epidemics/veterinary , Macaca fascicularis , Monkey Diseases/epidemiology , Animals , Antibodies, Viral/blood , Dengue/blood , Dengue/epidemiology , Dengue/virology , Dengue Virus/classification , Dengue Virus/genetics , Immunoglobulin G/blood , Immunoglobulin M/blood , Molecular Sequence Data , Monkey Diseases/blood , Monkey Diseases/virology , Philippines/epidemiology , Phylogeny
10.
J Gen Virol ; 94(Pt 12): 2710-2716, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24026672

ABSTRACT

Simian-human immunodeficiency virus (SHIV) carrying the envelope from the clade B clinical human immunodeficiency virus type 1 (HIV-1) isolate MNA, designated SHIV MNA, was generated through intracellular homologous recombination. SHIV MNA inherited biological properties from the parental HIV-1, including CCR5 co-receptor preference, resistance to neutralization by the anti-V3 loop mAb KD-247 and loss of resistance in the presence of the CD4-mimic small-molecule YYA-021. SHIV MNA showed productive replication in rhesus macaque PBMCs. Experimental infection of a rhesus macaque with SHIV MNA caused a transient but high titre of plasma viral RNA and a moderate antibody response. Immunoglobulin in the plasma at 24 weeks post-infection was capable of neutralizing SHIV MNA in the presence but not in the absence of YYA-021. SHIV MNA could serve a model for development of novel therapeutic interventions based on CD4-mimic-mediated conversion of envelope protein susceptible to antibody neutralization.


Subject(s)
Antibodies, Neutralizing/immunology , CD4 Antigens/immunology , HIV-1/immunology , Molecular Mimicry/immunology , Recombination, Genetic , Simian Immunodeficiency Virus/immunology , Virus Replication , Animals , HIV Antibodies/immunology , HIV-1/genetics , HIV-1/pathogenicity , HIV-1/physiology , Humans , Leukocytes, Mononuclear/virology , Macaca mulatta , Neutralization Tests , RNA, Viral/blood , Receptors, CCR5/immunology , Receptors, CCR5/physiology , Simian Immunodeficiency Virus/genetics , Simian Immunodeficiency Virus/pathogenicity , Simian Immunodeficiency Virus/physiology
11.
Biochem Biophys Res Commun ; 441(1): 108-13, 2013 Nov 08.
Article in English | MEDLINE | ID: mdl-24134838

ABSTRACT

Trehalose 6,6'-dimycolate (TDM) is a major glycolipid of the cell wall of mycobacteria with remarkable adjuvant functions. To avoid detection by the host innate immune system, invading mycobacteria down-regulate the expression of TDM by utilizing host-derived glucose as a competitive substrate for their mycolyltransferases; however, this enzymatic reaction results in the concomitant biosynthesis of glucose monomycolate (GMM) which is recognized by the acquired immune system. GMM-specific, CD1-restricted T cell responses have been detected in the peripheral blood of infected human subjects and monkeys as well as in secondary lymphoid organs of small animals, such as guinea pigs and human CD1-transgenic mice. Nevertheless, it remains to be determined how tissues respond at the site where GMM is produced. Here we found that rhesus macaques vaccinated with Mycobacterium bovis bacillus Calmette-Guerin mounted a chemokine response in GMM-challenged skin that was favorable for recruiting T helper (Th)1 T cells. Indeed, the expression of interferon-γ, but not Th2 or Th17 cytokines, was prominent in the GMM-injected tissue. The GMM-elicited tissue response was also associated with the expression of monocyte/macrophage-attracting CC chemokines, such as CCL2, CCL4 and CCL8. Furthermore, the skin response to GMM involved the up-regulated expression of granulysin and perforin. Given that GMM is produced primarily by pathogenic mycobacteria proliferating within the host, the Th1-skewed tissue response to GMM may function efficiently at the site of infection.


Subject(s)
Glycolipids/immunology , Macaca mulatta/immunology , Macaca mulatta/microbiology , Mycobacterium Infections/microbiology , Mycobacterium/immunology , Organ Specificity/immunology , Th1 Cells/immunology , Animals , Chemokines/blood , Humans , Interferon-gamma/metabolism , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/microbiology , Macaca mulatta/blood , Macrophages/immunology , Mice , Models, Biological , Mycobacterium Infections/blood , Mycobacterium Infections/immunology , Skin/immunology , Skin/microbiology , Skin/pathology
12.
Bioorg Med Chem ; 21(24): 7884-9, 2013 Dec 15.
Article in English | MEDLINE | ID: mdl-24189188

ABSTRACT

To date, several small molecules of CD4 mimics, which can suppress competitively the interaction between an HIV-1 envelope glycoprotein gp120 and a cellular surface protein CD4, have been reported as viral entry inhibitors. A lead compound 2 (YYA-021) with relatively high potency and low cytotoxicity has been identified previously by SAR studies. In the present study, the pharmacokinetics of the intravenous administration of compound 2 in rats and rhesus macaques is reported. The half-lives of compound 2 in blood in rats and rhesus macaques suggest that compound 2 shows wide tissue distribution and relatively high distribution volumes. A few hours after the injection, both plasma concentrations of compound 2 maintained micromolar levels, indicating it might have promise for intravenous administration when used combinatorially with anti-gp120 monoclonal antibodies.


Subject(s)
CD4 Antigens/chemistry , HIV Fusion Inhibitors/pharmacokinetics , Molecular Mimicry , Oxamic Acid/analogs & derivatives , Piperidines/pharmacokinetics , Administration, Intravenous , Adsorption , Animals , CD4 Antigens/metabolism , HIV Fusion Inhibitors/administration & dosage , HIV Fusion Inhibitors/chemistry , Half-Life , Macaca mulatta , Molecular Structure , Oxamic Acid/administration & dosage , Oxamic Acid/chemistry , Oxamic Acid/pharmacokinetics , Piperidines/administration & dosage , Piperidines/chemistry , Rats , Rats, Sprague-Dawley , Surface Properties , Tissue Distribution
13.
J Immunol ; 187(2): 608-12, 2011 Jul 15.
Article in English | MEDLINE | ID: mdl-21653836

ABSTRACT

The use of the host cellular machinery is essential for pathogenic viruses to replicate in host cells. HIV and SIV borrow the host-derived N-myristoyl-transferase and its substrate, myristoyl-CoA, for coupling a saturated C(14) fatty acid (myristic acid) to the N-terminal glycine residue of the Nef protein. This biochemical reaction, referred to as N-myristoylation, assists its targeting to the plasma membrane, thereby supporting the immunosuppressive activity proposed for the Nef protein. In this study, we show that the host immunity is equipped with CTLs capable of sensing N-myristoylation of the Nef protein. A rhesus macaque CD8(+) T cell line was established that specifically recognized N-myristoylated, but not unmodified, peptides of the Nef protein. Furthermore, the population size of N-myristoylated Nef peptide-specific T cells was found to increase significantly in the circulation of SIV-infected monkeys. Thus, these results identify N-myristoylated viral peptides as a novel class of CTL target Ag.


Subject(s)
Gene Products, nef/metabolism , Myristic Acid/metabolism , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Acquired Immunodeficiency Syndrome/metabolism , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/virology , Amino Acid Motifs , Animals , Cell Differentiation/immunology , Cell Line , Conserved Sequence , Epitopes, T-Lymphocyte/immunology , Epitopes, T-Lymphocyte/metabolism , Lymphocyte Activation/immunology , Macaca mulatta , Simian Acquired Immunodeficiency Syndrome/pathology , T-Lymphocytes, Cytotoxic/pathology
14.
Retrovirology ; 9: 18, 2012 Feb 25.
Article in English | MEDLINE | ID: mdl-22364292

ABSTRACT

BACKGROUND: Developing a quantitative understanding of viral kinetics is useful for determining the pathogenesis and transmissibility of the virus, predicting the course of disease, and evaluating the effects of antiviral therapy. The availability of data in clinical, animal, and cell culture studies, however, has been quite limited. Many studies of virus infection kinetics have been based solely on measures of total or infectious virus count. Here, we introduce a new mathematical model which tracks both infectious and total viral load, as well as the fraction of infected and uninfected cells within a cell culture, and apply it to analyze time-course data of an SHIV infection in vitro. RESULTS: We infected HSC-F cells with SHIV-KS661 and measured the concentration of Nef-negative (target) and Nef-positive (infected) HSC-F cells, the total viral load, and the infectious viral load daily for nine days. The experiments were repeated at four different MOIs, and the model was fitted to the full dataset simultaneously. Our analysis allowed us to extract an infected cell half-life of 14.1 h, a half-life of SHIV-KS661 infectiousness of 17.9 h, a virus burst size of 22.1 thousand RNA copies or 0.19 TCID50, and a basic reproductive number of 62.8. Furthermore, we calculated that SHIV-KS661 virus-infected cells produce at least 1 infectious virion for every 350 virions produced. CONCLUSIONS: Our method, combining in vitro experiments and a mathematical model, provides detailed quantitative insights into the kinetics of the SHIV infection which could be used to significantly improve the understanding of SHIV and HIV-1 pathogenesis. The method could also be applied to other viral infections and used to improve the in vitro determination of the effect and efficacy of antiviral compounds.


Subject(s)
HIV-1/growth & development , Simian Immunodeficiency Virus/growth & development , T-Lymphocytes/virology , Viral Load , Animals , Cell Line , HIV-1/genetics , HIV-1/pathogenicity , Macaca fascicularis , Models, Theoretical , Simian Immunodeficiency Virus/genetics , Simian Immunodeficiency Virus/pathogenicity
15.
J Virol ; 85(20): 10617-26, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21813599

ABSTRACT

Evolution of the env gene in transmitted R5-tropic human immunodeficiency virus type 1 (HIV-1) strains is the most widely accepted mechanism driving coreceptor switching. In some infected individuals, however, a shift in coreceptor utilization can occur as a result of the reemergence of a cotransmitted, but rapidly controlled, X4 virus. The latter possibility was studied by dually infecting rhesus macaques with X4 and R5 chimeric simian simian/human immunodeficiency viruses (SHIVs) and monitoring the replication status of each virus using specific primer pairs. In one of the infected monkeys, both SHIVs were potently suppressed by week 12 postinoculation, but a burst of viremia at week 51 was accompanied by an unrelenting loss of total CD4+ T cells and the development of clinical disease. PCR analyses of plasma viral RNA indicated an env gene segment containing the V3 region from the inoculated X4 SHIV had been transferred into the genetic background of the input R5 SHIV by intergenomic recombination, creating an X4 virus with novel replicative, serological, and pathogenic properties. These results indicate that the effects of retrovirus recombination in vivo can be functionally profound and may even occur when one of the recombination participants is undetectable in the circulation as cell-free virus.


Subject(s)
HIV-1/pathogenicity , Receptors, HIV/metabolism , Recombination, Genetic , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/pathogenicity , Virus Internalization , Animals , CD4 Lymphocyte Count , HIV-1/genetics , Macaca mulatta , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Immunodeficiency Virus/genetics , Viremia
16.
J Virol ; 85(24): 13195-203, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21937653

ABSTRACT

The first influenza pandemic of the 21st century was caused by novel H1N1 viruses that emerged in early 2009. An Asp-to-Gly change at position 222 of the receptor-binding protein hemagglutinin (HA) correlates with more-severe infections in humans. The amino acid at position 222 of HA contributes to receptor-binding specificity with Asp (typically found in human influenza viruses) and Gly (typically found in avian and classic H1N1 swine influenza viruses), conferring binding to human- and avian-type receptors, respectively. Here, we asked whether binding to avian-type receptors enhances influenza virus pathogenicity. We tested two 2009 pandemic H1N1 viruses possessing HA-222G (isolated from severe cases) and two viruses that possessed HA-222D. In glycan arrays, viruses possessing HA-222D preferentially bound to human-type receptors, while those encoding HA-222G bound to both avian- and human-type receptors. This difference in receptor binding correlated with efficient infection of viruses possessing HA-222G, compared to those possessing HA-222D, in human lung tissue, including alveolar type II pneumocytes, which express avian-type receptors. In a nonhuman primate model, infection with one of the viruses possessing HA-222G caused lung damage more severe than did infection with a virus encoding HA-222D, although these pathological differences were not observed for the other virus pair with either HA-222G or HA-222D. These data demonstrate that the acquisition of avian-type receptor-binding specificity may result in more-efficient infection of human alveolar type II pneumocytes and thus more-severe lung damage. Collectively, these findings suggest a new mechanism by which influenza viruses may become more pathogenic in mammals, including humans.


Subject(s)
Hemagglutinin Glycoproteins, Influenza Virus/metabolism , Influenza A Virus, H1N1 Subtype/pathogenicity , Orthomyxoviridae Infections/pathology , Orthomyxoviridae Infections/virology , Receptors, Virus/metabolism , Virus Internalization , Animals , Cell Line , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Humans , Lung/pathology , Lung/virology , Macaca , Receptors, Virus/genetics
17.
Proc Natl Acad Sci U S A ; 106(19): 8015-20, 2009 May 12.
Article in English | MEDLINE | ID: mdl-19416840

ABSTRACT

We and others have reported that the vast majority of virus-producing CD4(+) T cells during the acute infection of rhesus macaques with simian immunodeficiency virus (SIV) or CXCR4 (X4)-using simian/human immunodeficiency viruses (SHIVs) exhibited a nonactivated phenotype. These findings have been extended to show that resting CD4(+) T lymphocytes collected from SIV- or X4-SHIV-infected animals during the first 10 days of infection continue to release virus ex vivo. Furthermore, we observed high frequencies of integrated viral DNA (up to 5.1 x 10(4) DNA copies per 10(5) cells) in circulating resting CD4(+) T cells during the first 10 days of the infection. Integration of SIV DNA was detected only in memory CD4(+) T cells and SHIVs preferentially integrated into resting naïve CD4(+) T cells. Taken together, these results show that during the acute infection large numbers of resting CD4(+) T cells carry integrated nonhuman primate lentiviral DNA and are the major source of progeny virions irrespective of coreceptor usage. Prompt and sustained interventions are therefore required to block the rapid systemic dissemination of virus and prevent an otherwise fatal clinical outcome.


Subject(s)
CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/virology , DNA, Viral/metabolism , Lentivirus Infections/blood , Lentivirus Infections/virology , Animals , Calibration , Immunologic Memory , Immunophenotyping , Lentivirus Infections/metabolism , Macaca mulatta , Models, Biological , Phenotype , Simian Acquired Immunodeficiency Syndrome/blood , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/genetics , Treatment Outcome , Virus Replication
18.
Biochem Biophys Res Commun ; 408(4): 615-9, 2011 May 20.
Article in English | MEDLINE | ID: mdl-21531211

ABSTRACT

Cytotoxic T lymphocyte (CTL) responses are crucial for the control of human and simian immunodeficiency virus (HIV and SIV) replication. A promising AIDS vaccine strategy is to induce CTL memory resulting in more effective CTL responses post-viral exposure compared to those in natural HIV infections. We previously developed a CTL-inducing vaccine and showed SIV control in some vaccinated rhesus macaques. These vaccine-based SIV controllers elicited vaccine antigen-specific CTL responses dominantly in the acute phase post-challenge. Here, we examined CTL responses post-challenge in those vaccinated animals that failed to control SIV replication. Unvaccinated rhesus macaques possessing the major histocompatibility complex class I haplotype 90-088-Ij dominantly elicited SIV non-Gag antigen-specific CTL responses after SIV challenge, while those induced with Gag-specific CTL memory by prophylactic vaccination failed to control SIV replication with dominant Gag-specific CTL responses in the acute phase, indicating dominant induction of vaccine antigen-specific CTL responses post-challenge even in non-controllers. Further analysis suggested that prophylactic vaccination results in dominant induction of vaccine antigen-specific CTL responses post-viral exposure but delays SIV non-vaccine antigen-specific CTL responses. These results imply a significant influence of prophylactic vaccination on CTL immunodominance post-viral exposure, providing insights into antigen design in development of a CTL-inducing AIDS vaccine.


Subject(s)
AIDS Vaccines/immunology , Antigens, Viral/immunology , SAIDS Vaccines/immunology , Simian Acquired Immunodeficiency Syndrome/prevention & control , Simian Immunodeficiency Virus/immunology , T-Lymphocytes, Cytotoxic/immunology , AIDS Vaccines/therapeutic use , Acquired Immunodeficiency Syndrome/prevention & control , Animals , Humans , Macaca mulatta , SAIDS Vaccines/therapeutic use , Simian Acquired Immunodeficiency Syndrome/immunology
19.
J Virol ; 84(9): 4769-81, 2010 May.
Article in English | MEDLINE | ID: mdl-20147396

ABSTRACT

A new pathogenic R5-tropic simian/human immunodeficiency virus (SHIV) was generated following serial passaging in rhesus macaques. All 13 animals inoculated with SHIV(AD8) passaged lineages experienced marked depletions of CD4(+) T cells. Ten of these infected monkeys became normal progressors (NPs) and had gradual losses of both memory and naïve CD4(+) T lymphocytes, generated antiviral CD4(+) and CD8(+) T cell responses, and sustained chronic immune activation while maintaining variable levels of plasma viremia (10(2) to 10(5) RNA copies/ml for up to 3 years postinfection [p.i.]). To date, five NPs developed AIDS associated with opportunistic infections caused by Pneumocystis carinii, Mycobacterium avium, and Campylobacter coli that required euthanasia between weeks 100 and 199 p.i. Three other NPs have experienced marked depletions of circulating CD4(+) T lymphocytes (92 to 154 cells/microl) following 1 to 2 years of infection. When tested for coreceptor usage, the viruses isolated from four NPs at the time of their euthanasia remained R5 tropic. Three of the 13 SHIV(AD8)-inoculated macaques experienced a rapid-progressor syndrome characterized by sustained plasma viremia of >1 x 10(7) RNA copies/ml and rapid irreversible loss of memory CD4(+) T cells that required euthanasia between weeks 19 and 23 postinfection. The sustained viremia, associated depletion of CD4(+) T lymphocytes, and induction of AIDS make the SHIV(AD8) lineage of viruses a potentially valuable reagent for vaccine studies.


Subject(s)
Receptors, Virus , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/physiology , Viral Tropism , Virus Attachment , AIDS-Related Opportunistic Infections , Animals , CD4 Lymphocyte Count , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Campylobacter coli/isolation & purification , Macaca mulatta , Mycobacterium avium/isolation & purification , Pneumocystis carinii/isolation & purification , Serial Passage , Simian Acquired Immunodeficiency Syndrome/complications , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Immunodeficiency Virus/genetics , Viral Load , Viremia
20.
Microbiol Immunol ; 55(11): 768-73, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21895748

ABSTRACT

Major histocompatibility complex class I (MHC-I)-restricted CD8(+) cytotoxic T lymphocyte (CTL) responses are crucial for the control of human immunodeficiency virus (HIV) and simian immunodeficiency virus (SIV) replication. In particular, Gag-specific CTL responses have been shown to exert strong suppressive pressure on HIV/SIV replication. Additionally, association of Vif-specific CTL frequencies with in vitro anti-SIV efficacy has been suggested recently. Host MHC-I genotypes could affect the immunodominance patterns of these potent CTL responses. Here, Gag- and Vif-specific CTL responses during primary SIVmac239 infection were examined in three groups of Burmese rhesus macaques, each group having a different MHC-I haplotype. The first group of four macaques, which possessed the MHC-I haplotype 90-010-Ie, did not show Gag- or Vif-specific CTL responses. However, Nef-specific CTL responses were elicited, suggesting that primary SIV infection does not induce predominant CTL responses specific for Gag/Vif epitopes restricted by 90-010-Ie-derived MHC-I molecules. In contrast, Gag- and Vif-specific CTL responses were induced in the second group of two 89-075-Iw-positive animals and the third group of two 91-010-Is-positive animals. Considering the potential of prophylactic vaccination to affect CTL immunodominance post-viral exposure, these groups of macaques would be useful for evaluation of vaccine antigen-specific CTL efficacy against SIV infection.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class I/metabolism , Simian Immunodeficiency Virus/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , Gene Products, gag/immunology , Gene Products, vif/immunology , Haplotypes , Histocompatibility Antigens Class I/genetics , Macaca mulatta
SELECTION OF CITATIONS
SEARCH DETAIL