Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Viruses ; 11(9)2019 09 07.
Article in English | MEDLINE | ID: mdl-31500260

ABSTRACT

Feline immunodeficiency virus (FIV) is a naturally occurring T-cell tropic lentiviral disease of felids with many similarities to HIV/AIDS in humans. Similar to primate lentiviral-host interactions, feline APOBEC3 (A3) has been shown to inhibit FIV infection in a host-specific manner and feline A3 degradation is mediated by FIV Vif. Further, infection of felids with non-native FIV strains results in restricted viral replication in both experimental and naturally occurring infections. However, the link between molecular A3-Vif interactions and A3 biological activity during FIV infection has not been well characterized. We thus examined expression of the feline A3 genes A3Z2, A3Z3 and A3Z2-Z3 during experimental infection of domestic cats with host-adapted domestic cat FIV (referred to as FIV) and non-adapted Puma concolor FIV (referred to as puma lentivirus, PLV). We determined A3 expression in different tissues and blood cells from uninfected, FIV-infected, PLV-infected and FIV/PLV co-infected cats; and in purified blood cell subpopulations from FIV-infected and uninfected cats. Additionally, we evaluated regulation of A3 expression by cytokines, mitogens, and FIV infection in cultured cells. In all feline cells and tissues studied, there was a striking difference in expression between the A3 genes which encode FIV inhibitors, with A3Z3 mRNA abundance exceeding that of A3Z2-Z3 by 300-fold or more. Interferon-alpha treatment of cat T cells resulted in upregulation of A3 expression, while treatment with interferon-gamma enhanced expression in cat cell lines. In cats, secondary lymphoid organs and peripheral blood mononuclear cells (PBMC) had the highest basal A3 expression levels and A3 genes were differentially expressed among blood T cells, B cells, and monocytes. Acute FIV and PLV infection of cats, and FIV infection of primary PBMC resulted in no detectable change in A3 expression with the exception of significantly elevated A3 expression in the thymus, the site of highest FIV replication. We conclude that cat A3 expression is regulated by cytokine treatment but, by and large, lentiviral infection did not appear to alter expression. Differences in A3 expression in different blood cell subsets did not appear to impact FIV viral replication kinetics within these cells. Furthermore, the relative abundance of A3Z3 mRNA compared to A3Z2-Z3 suggests that A3Z3 may be the major active anti-lentiviral APOBEC3 gene product in domestic cats.


Subject(s)
Cytosine Deaminase/immunology , Feline Acquired Immunodeficiency Syndrome/enzymology , Immunodeficiency Virus, Feline/physiology , Lentivirus Infections/veterinary , Animals , B-Lymphocytes/immunology , Cats , Cytosine Deaminase/genetics , Feline Acquired Immunodeficiency Syndrome/genetics , Feline Acquired Immunodeficiency Syndrome/immunology , Feline Acquired Immunodeficiency Syndrome/virology , Host-Pathogen Interactions , Immunodeficiency Virus, Feline/genetics , Lentivirus Infections/enzymology , Lentivirus Infections/genetics , Lentivirus Infections/immunology , T-Lymphocytes/immunology , Virus Replication
2.
PLoS One ; 12(9): e0185138, 2017.
Article in English | MEDLINE | ID: mdl-28934316

ABSTRACT

Feline immunodeficiency virus (FIV) is the feline analogue of human immunodeficiency virus (HIV) and features many hallmarks of HIV infection and pathogenesis, including the development of concurrent oral lesions. While HIV is typically transmitted via parenteral transmucosal contact, recent studies prove that oral transmission can occur, and that saliva from infected individuals contains significant amounts of HIV RNA and DNA. While it is accepted that FIV is primarily transmitted by biting, few studies have evaluated FIV oral infection kinetics and transmission mechanisms over the last 20 years. Modern quantitative analyses applied to natural FIV oral infection could significantly further our understanding of lentiviral oral disease and transmission. We therefore characterized FIV salivary viral kinetics and antibody secretions to more fully document oral viral pathogenesis. Our results demonstrate that: (i) saliva of FIV-infected cats contains infectious virus particles, FIV viral RNA at levels equivalent to circulation, and lower but significant amounts of FIV proviral DNA; (ii) the ratio of FIV RNA to DNA is significantly higher in saliva than in circulation; (iii) FIV viral load in oral lymphoid tissues (tonsil, lymph nodes) is significantly higher than mucosal tissues (buccal mucosa, salivary gland, tongue); (iv) salivary IgG antibodies increase significantly over time in FIV-infected cats, while salivary IgA levels remain static; and, (v) saliva from naïve Specific Pathogen Free cats inhibits FIV growth in vitro. Collectively, these results suggest that oral lymphoid tissues serve as a site for enhanced FIV replication, resulting in accumulation of FIV particles and FIV-infected cells in saliva. Failure to induce a virus-specific oral mucosal antibody response, and/or viral capability to overcome inhibitory components in saliva may perpetuate chronic oral cavity infection. Based upon these findings, we propose a model of oral FIV pathogenesis and suggest alternative diagnostic modalities and translational approaches to study oral HIV infection.


Subject(s)
Feline Acquired Immunodeficiency Syndrome/etiology , Immunodeficiency Virus, Feline/physiology , Mouth/virology , Saliva/virology , Animals , Antibody Specificity , Cats , DNA, Viral/blood , DNA, Viral/metabolism , Feline Acquired Immunodeficiency Syndrome/blood , Feline Acquired Immunodeficiency Syndrome/transmission , Immunodeficiency Virus, Feline/immunology , Immunoglobulin A/immunology , RNA, Viral/blood , RNA, Viral/metabolism , Viral Load
3.
Virology ; 436(2): 284-94, 2013 Feb 20.
Article in English | MEDLINE | ID: mdl-23290868

ABSTRACT

Four cats (24%) experimentally infected with FIV unexpectedly developed neoplastic changes within four months of inoculation. While FIV has previously been associated with neoplasia, the rapidity and high attack rate seen here is highly unusual. PCR for antigen receptor rearrangements (PARR) detected clonally rearranged T cells in two animals diagnosed with B cell follicular lymphoma by classical means. All cats were negative for feline leukemia virus; gamma-herpesvirus DNA was not amplified using degenerate primers. FIV proviral load in neoplastic tissue was two orders of magnitude lower than in the periphery, lower in neoplastic vs non-neoplastic lymph node, and clonal integration was not detected. We hypothesize that neoplasia was secondary to FIV immune dysregulation, and show that PARR can augment our capacity to phenotype these tumors and distinguish follicular hyperplasia from lymphoma. Age of exposure and relative virulence of the inoculum likely contributed to this unusual presentation of FIV infection.


Subject(s)
Immunodeficiency Virus, Feline/pathogenicity , Lentivirus Infections/complications , Lentivirus Infections/immunology , Lymphoma, B-Cell/epidemiology , Lymphoma, B-Cell/etiology , Animal Experimentation , Animals , Cats , Lentivirus Infections/virology
4.
PLoS One ; 6(8): e24020, 2011.
Article in English | MEDLINE | ID: mdl-21887365

ABSTRACT

Chimeric viruses constructed between a highly pathogenic Feline Immunodeficiency Virus isolate (FIV-C36) and a less pathogenic but neurotropic strain (FIV-PPR) have been used to map viral genetic determinants of in vivo pathogenicity. Chimeric virus FIV-PCenv, which contains FIV-C36 genome from the 3' region of pol to upstream of the 3'LTR on an FIV-PPR backbone, was previously shown to be replication-competent in vivo, inducing altered CD4(+) T-cell and neutrophil profiles intermediate between parental strains following a delay in viral replication during initial infection. Examination of FIV-PCenv proviral sequences recovered at week 11 post-infection revealed two changes compared to initial viral inoculum; the most significant being arginine to histidine in the integrase region of Pol at residue 813 (R813H). Pooled plasma from the initial in vivo study was used to inoculate a second cohort of cats to determine whether similar virulence and kinetics could be established following primary infection. Viral replication kinetics and immunocyte profiles were monitored in blood, bone marrow, and saliva over a one-year period. Passaged FIV-PCenv again displayed intermediate phenotype between parental strains, but unlike primary experiments, the onset of acute viremia was not delayed. CD4/8 alterations were noted in all groups of animals, though significant changes from controls were delayed in FIV-PPR infected animals compared to FIV-C36 and FIV-PCenv. In vivo passage of FIV-PCenv increased replication-competence relative to the initial molecularly-cloned chimera in association with one adaptive nucleotide change in the 5' end of the genome relative to primary tissue culture inoculum, while mutations in the 3' end of the genome were not detected. The results are consistent with the interpretation that 3' elements contribute to heightened virulence of FIV-C36, and that integrase residue 813 plays an important role in facilitating successful in vivo replication.


Subject(s)
3' Flanking Region/genetics , Immunodeficiency Virus, Feline/growth & development , Immunodeficiency Virus, Feline/pathogenicity , Animals , Blood/virology , Bone Marrow/virology , CD4-Positive T-Lymphocytes , CD8-Positive T-Lymphocytes , Cats , Genes, Viral , Integrases , Kinetics , Saliva/virology , Virus Replication
5.
Vet Immunol Immunopathol ; 143(3-4): 282-91, 2011 Oct 15.
Article in English | MEDLINE | ID: mdl-21715019

ABSTRACT

Feline immunodeficiency virus (FIV) is a naturally occurring lentivirus of domestic cats, and is the causative agent of feline AIDS. Similar to human immunodeficiency virus (HIV), the pathogenesis of FIV involves infection of lymphocytes and macrophages, and results in chronic progressive immune system collapse and death. Neuropathologic correlates of FIV infection have not yet been elucidated, and may be relevant to understanding HIV-associated neurologic disease (neuroAIDS). As in HIV, FIV strains have been shown to express differential tendencies towards development of clinical neuroAIDS. To interrogate viral genetic determinants that might contribute to neuropathogenicity, cats were exposed to two well-characterized FIV strains with divergent clinical phenotypes and a chimeric strain as follows: FIV(PPR) (PPR, relatively apathogenic but associated with neurologic manifestations), FIV(C36) (C36, immunopathogenic but without associated neurologic disease), and Pcenv (a chimeric virus consisting of a PPR backbone with substituted C36 env region). A sham inoculum control group was also included. Peripheral nerve conduction velocity, CNS imaging studies, viral loads and hematologic analysis were performed over a 12 month period. At termination of the study (350 days post-inoculation), brain sections were obtained from four anatomic locations known to be involved in human and primate lentiviral neuroAIDS. Histological and immunohistochemical evaluation with seven markers of inflammation revealed that Pcenv infection resulted in mild inflammation of the CNS, microglial activation, neuronal degeneration and apoptosis, while C36 and PPR strains induced minimal neuropathologic changes. Conduction velocity aberrations were noted peripherally in all three groups at 63 weeks post-infection. Pcenv viral load in this study was intermediate to the parental strains (C36 demonstrating the highest viral load and PPR the lowest). These results collectively suggest that (i) 3' C36 genomic elements contribute to viral replication characteristics, and (ii) 5' PPR genomic elements contribute to CNS manifestations. This study illustrates the potential for FIV to provide valuable information about neuroAIDS pathogenesis related to genotype and viral kinetics, as well as to identify strains useful to evaluation of therapeutic intervention.


Subject(s)
Central Nervous System/virology , Feline Acquired Immunodeficiency Syndrome/virology , Immunodeficiency Virus, Feline/physiology , Animals , Brain/pathology , Brain/virology , Brain Stem/physiopathology , Brain Stem/virology , Cats/virology , Central Nervous System/pathology , Central Nervous System/physiopathology , Evoked Potentials, Auditory, Brain Stem/physiology , Feline Acquired Immunodeficiency Syndrome/pathology , Feline Acquired Immunodeficiency Syndrome/physiopathology , Gene Products, gag/metabolism , Magnetic Resonance Imaging/veterinary , RNA, Viral/metabolism , Viral Load/veterinary
6.
Emerg Infect Dis ; 15(12): 2021-4, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19961691

ABSTRACT

Plague seroprevalence was estimated in populations of pumas and bobcats in the western United States. High levels of exposure in plague-endemic regions indicate the need to consider the ecology and pathobiology of plague in nondomestic felid hosts to better understand the role of these species in disease persistence and transmission.


Subject(s)
Lynx/microbiology , Plague/transmission , Puma/microbiology , Yersinia pestis/isolation & purification , Animals , Antibodies, Bacterial/blood , Colorado , Disease Reservoirs , Humans , Seroepidemiologic Studies , Yersinia pestis/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...