Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 275
Filter
1.
J Immunol ; 208(2): 444-453, 2022 01 15.
Article in English | MEDLINE | ID: mdl-34893529

ABSTRACT

SAMHD1 is a potent HIV-1 restriction factor that blocks reverse transcription in monocytes, dendritic cells and resting CD4+ T cells by decreasing intracellular dNTP pools. However, SAMHD1 may diminish innate immune sensing and Ag presentation, resulting in a weaker adaptive immune response. To date, the role of SAMHD1 on antiretroviral immunity remains unclear, as mouse SAMHD1 had no impact on murine retrovirus replication in prior in vivo studies. Here, we show that SAMHD1 significantly inhibits acute Friend retrovirus infection in mice. Pretreatment with LPS, a significant driver of inflammation during HIV-1 infection, further unmasked a role for SAMHD1 in influencing immune responses. LPS treatment in vivo doubled the intracellular dNTP levels in immune compartments of SAMHD1 knockout but not wild-type mice. SAMHD1 knockout mice exhibited higher plasma infectious viremia and proviral DNA loads than wild-type mice at 7 d postinfection (dpi), and proviral loads inversely correlated with a stronger CD8+ T cell response. SAMHD1 deficiency was also associated with weaker NK, CD4+ T and CD8+ T cell responses by 14 dpi and weaker neutralizing Ab responses by 28 dpi. Intriguingly, SAMHD1 influenced these cell-mediated immune (14 dpi) and neutralizing Ab (28 dpi) responses in male but not female mice. Our findings formally demonstrate SAMHD1 as an antiretroviral factor in vivo that could promote adaptive immune responses in a sex-dependent manner. The requirement for LPS to unravel the SAMHD1 immunological phenotype suggests that comorbidities associated with a "leaky" gut barrier may influence the antiviral function of SAMHD1 in vivo.


Subject(s)
Adaptive Immunity/immunology , Friend murine leukemia virus/growth & development , Lipopolysaccharides/pharmacology , Retroviridae Infections/prevention & control , SAM Domain and HD Domain-Containing Protein 1/genetics , Animals , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Antigen Presentation/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , DNA, Viral/blood , Female , Friend murine leukemia virus/immunology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Retroviridae Infections/virology , Reverse Transcription/genetics , SAM Domain and HD Domain-Containing Protein 1/immunology , Viral Load
2.
Viruses ; 13(11)2021 10 26.
Article in English | MEDLINE | ID: mdl-34834962

ABSTRACT

Porcine endogenous retroviruses (PERVs) are integrated in the genome of all pigs, and some of them are able to infect human cells. Therefore, PERVs pose a risk for xenotransplantation, the transplantation of pig cells, tissues, or organ to humans in order to alleviate the shortage of human donor organs. Up to 2021, a huge body of knowledge about PERVs has been accumulated regarding their biology, including replication, recombination, origin, host range, and immunosuppressive properties. Until now, no PERV transmission has been observed in clinical trials transplanting pig islet cells into diabetic humans, in preclinical trials transplanting pig cells and organs into nonhuman primates with remarkable long survival times of the transplant, and in infection experiments with several animal species. Nevertheless, in order to prevent virus transmission to the recipient, numerous strategies have been developed, including selection of PERV-C-free animals, RNA interference, antiviral drugs, vaccination, and genome editing. Furthermore, at present there are no more experimental approaches to evaluate the full risk until we move to the clinic.


Subject(s)
Endogenous Retroviruses , Retroviridae Infections/virology , Swine Diseases/virology , Swine/virology , Transplantation, Heterologous , Animals , Endogenous Retroviruses/genetics , Endogenous Retroviruses/isolation & purification , Endogenous Retroviruses/physiology , Gammaretrovirus/genetics , Host Specificity , Immunosuppressive Agents , Retroviridae Infections/drug therapy , Retroviridae Infections/prevention & control , Retroviridae Infections/transmission , Zoonoses/virology
3.
EMBO J ; 40(16): e106540, 2021 08 16.
Article in English | MEDLINE | ID: mdl-34121210

ABSTRACT

Dendritic cells (DC) subsets, like Langerhans cells (LC), are immune cells involved in pathogen sensing. They express specific antimicrobial cellular factors that are able to restrict infection and limit further pathogen transmission. Here, we identify the alarmin S100A9 as a novel intracellular antiretroviral factor expressed in human monocyte-derived and skin-derived LC. The intracellular expression of S100A9 is decreased upon LC maturation and inversely correlates with enhanced susceptibility to HIV-1 infection of LC. Furthermore, silencing of S100A9 in primary human LC relieves HIV-1 restriction while ectopic expression of S100A9 in various cell lines promotes intrinsic resistance to both HIV-1 and MLV infection by acting on reverse transcription. Mechanistically, the intracellular expression of S100A9 alters viral capsid uncoating and reverse transcription. S100A9 also shows potent inhibitory effect against HIV-1 and MMLV reverse transcriptase (RTase) activity in vitro in a divalent cation-dependent manner. Our findings uncover an unexpected intracellular function of the human alarmin S100A9 in regulating antiretroviral immunity in Langerhans cells.


Subject(s)
Alarmins/genetics , Calgranulin B/genetics , HIV-1/physiology , Langerhans Cells/virology , Moloney murine leukemia virus/physiology , Retroviridae Infections/prevention & control , Animals , CD4-Positive T-Lymphocytes/immunology , Cell Line , Cricetulus , HIV-1/genetics , Host-Pathogen Interactions , Humans , Langerhans Cells/immunology , Leukemia, Experimental/prevention & control , Mice , Moloney murine leukemia virus/genetics , Reverse Transcription , Transforming Growth Factor beta/immunology , Tumor Virus Infections/prevention & control , Virus Replication
4.
Curr Issues Mol Biol ; 43(1): 52-64, 2021 Apr 30.
Article in English | MEDLINE | ID: mdl-33946297

ABSTRACT

Koala populations are currently declining and under threat from koala retrovirus (KoRV) infection both in the wild and in captivity. KoRV is assumed to cause immunosuppression and neoplastic diseases, favoring chlamydiosis in koalas. Currently, 10 KoRV subtypes have been identified, including an endogenous subtype (KoRV-A) and nine exogenous subtypes (KoRV-B to KoRV-J). The host's immune response acts as a safeguard against pathogens. Therefore, a proper understanding of the immune response mechanisms against infection is of great importance for the host's survival, as well as for the development of therapeutic and prophylactic interventions. A vaccine is an important protective as well as being a therapeutic tool against infectious disease, and several studies have shown promise for the development of an effective vaccine against KoRV. Moreover, CRISPR/Cas9-based genome editing has opened a new window for gene therapy, and it appears to be a potential therapeutic tool in many viral infections, which could also be investigated for the treatment of KoRV infection. Here, we discuss the recent advances made in the understanding of the immune response in KoRV infection, as well as the progress towards vaccine development against KoRV infection in koalas.


Subject(s)
Cytokines/immunology , Phascolarctidae/virology , Retroviridae Infections/prevention & control , Retroviridae/immunology , Toll-Like Receptors/immunology , Vaccination/methods , Animals , Cytokines/metabolism , Phascolarctidae/immunology , Retroviridae Infections/immunology , Retroviridae Infections/virology , Toll-Like Receptors/metabolism
5.
Mar Drugs ; 19(5)2021 Apr 28.
Article in English | MEDLINE | ID: mdl-33925208

ABSTRACT

Walleye dermal sarcoma virus (WDSV) is a type of retrovirus, which affects most of the adult walleye fishes during the spawning time. The virus causes multiple epithelial tumors on the fish's skin and fins that are liable for more than 50% of the mortality rate of fish around the world. Till now, no effective antiviral drug or vaccine candidates have been developed that can block the progression of the disease caused by the pathogen. It was found that the 582-amino-acid (aa) residues long internal structural gag polyprotein of the virus plays an important role in virus budding and virion maturation outside of the cell. Inhibition of the protein can block the budding and virion maturation process and can be developed as an antiviral drug candidate against the virus. Therefore, the study aimed to identify potential natural antiviral drug candidates from the tropical mangrove marine plant Avicennia alba, which will be able to block the budding and virion maturation process by inhibiting the activity of the gag protein of the virus. Initially, a homology modeling approach was applied to identify the 3D structure, followed by refinement and validation of the protein. The refined protein structures were then utilized for molecular docking simulation. Eleven phytochemical compounds have been isolated from the marine plant and docked against the virus gag polyprotein. Three compounds, namely Friedlein (CID244297), Phytosterols (CID12303662), and 1-Triacontanol (CID68972) have been selected based on their docking score -8.5 kcal/mol, -8.0 kcal/mol and -7.9 kcal/mol, respectively, and were evaluated through ADME (Absorption, Distribution, Metabolism and Excretion), and toxicity properties. Finally, molecular dynamics (MD) simulation was applied to confirm the binding stability of the protein-ligands complex structure. The ADME and toxicity analysis reveal the efficacy and non-toxic properties of the compounds, where MD simulation confirmed the binding stability of the selected three compounds with the targeted protein. This computational study revealed the virtuous value of the selected three compounds against the targeted gag polyprotein and will be effective and promising antiviral candidates against the pathogen in a significant and worthwhile manner. Although in vitro and in vivo study is required for further evaluation of the compounds against the targeted protein.


Subject(s)
Antiviral Agents/pharmacology , Avicennia/chemistry , Epsilonretrovirus/drug effects , Fish Diseases/prevention & control , Plant Extracts/pharmacology , Retroviridae Infections/veterinary , Tumor Virus Infections/veterinary , Animals , Antiviral Agents/isolation & purification , Epsilonretrovirus/metabolism , Epsilonretrovirus/pathogenicity , Fish Diseases/virology , Gene Products, gag/antagonists & inhibitors , Gene Products, gag/metabolism , Ligands , Molecular Docking Simulation , Molecular Dynamics Simulation , Plant Extracts/isolation & purification , Protein Conformation , Retroviridae Infections/prevention & control , Retroviridae Infections/virology , Structure-Activity Relationship , Tumor Virus Infections/prevention & control , Tumor Virus Infections/virology , Virus Release/drug effects
6.
Viruses ; 13(3)2021 03 10.
Article in English | MEDLINE | ID: mdl-33801908

ABSTRACT

An evolutionary arms race has been ongoing between retroviruses and their primate hosts for millions of years. Within the last century, a zoonotic transmission introduced the Human Immunodeficiency Virus (HIV-1), a retrovirus, to the human population that has claimed the lives of millions of individuals and is still infecting over a million people every year. To counteract retroviruses such as this, primates including humans have evolved an innate immune sensor for the retroviral capsid lattice known as TRIM5α. Although the molecular basis for its ability to restrict retroviruses is debated, it is currently accepted that TRIM5α forms higher-order assemblies around the incoming retroviral capsid that are not only disruptive for the virus lifecycle, but also trigger the activation of an antiviral state. More recently, it was discovered that TRIM5α restriction is broader than previously thought because it restricts not only the human retroelement LINE-1, but also the tick-borne flaviviruses, an emergent group of RNA viruses that have vastly different strategies for replication compared to retroviruses. This review focuses on the underlying mechanisms of TRIM5α-mediated restriction of retroelements and flaviviruses and how they differ from the more widely known ability of TRIM5α to restrict retroviruses.


Subject(s)
Capsid/immunology , Immunity, Innate , RNA Viruses/immunology , RNA Viruses/metabolism , Tripartite Motif Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Antiviral Restriction Factors , Capsid/metabolism , Carrier Proteins/genetics , Flavivirus/immunology , Flavivirus/metabolism , Humans , RNA Viruses/classification , RNA Viruses/genetics , Retroviridae/immunology , Retroviridae/metabolism , Retroviridae Infections/immunology , Retroviridae Infections/prevention & control , Tripartite Motif Proteins/genetics , Tripartite Motif Proteins/immunology , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/immunology
7.
Virol J ; 17(1): 168, 2020 10 31.
Article in English | MEDLINE | ID: mdl-33129323

ABSTRACT

Koala retrovirus (KoRV) is believed to be in an active state of endogenization into the koala genome. KoRV is present as both an endogenous and exogenous infection in all koalas in northern Australia. KoRV has been linked to koala pathologies including neoplasia and increased susceptibility to Chlamydia. A KoRV vaccine recently trialled in 10 northern koalas improved antibody response and reduced viral load. This communication reports the expression of key immune genes underlining the innate and adaptive immune response to vaccination in these northern koalas. The results showed that prior to vaccination, IL-8 was expressed at the highest levels, with at least 200-fold greater expression compared to other cytokines, while CD8 mRNA expression was significantly higher than CD4 mRNA expression level. Interferon-γ was up-regulated at both 4- and 8-weeks post-vaccination while IL-8 was down-regulated at 8-weeks post-vaccination.


Subject(s)
Cytokines/genetics , Interferon-gamma/genetics , Phascolarctidae/virology , Retroviridae Infections/immunology , Retroviridae Infections/veterinary , Retroviridae/immunology , Viral Vaccines/immunology , Animals , Antibody Formation , Australia , Cohort Studies , Cytokines/immunology , Endogenous Retroviruses/genetics , Endogenous Retroviruses/immunology , Interferon-gamma/biosynthesis , Interferon-gamma/immunology , Phascolarctidae/immunology , Retroviridae/genetics , Retroviridae Infections/prevention & control , Up-Regulation , Viral Vaccines/administration & dosage
8.
Vet Res ; 51(1): 133, 2020 Oct 19.
Article in English | MEDLINE | ID: mdl-33076991

ABSTRACT

Reticuloendotheliosis is an important immunosuppressive disease, associated with avian reticuloendotheliosis virus (REV) infection, and causes notable economic losses worldwide. Glycoprotein gp90 is an important structural protein of REV, and considered to be the most important immunogenic antigen, which can induce neutralizing antibodies against REV. In this study, an optimized suspension culture system was developed and applied to secretory express the immunogenic surface antigen gp90. To achieve an optimal glycosylation, the gp90 was designed to secretory expressed into the supernatant of the cell culture, which also occurs in the natural protein maturation procedure of REV. Serum-free culture medium was introduced to simplify the purification process and reduce the production costs. Based on the purified glycosylated gp90, an oil-emulsion subunit REV vaccine candidate was developed and evaluated in chickens. The subunit gp90-based vaccine induced fast immune responses, high levels of antibodies (REV-specific antibody, gp90-specific antibody, and neutralizing antibody against REV), and preferential T helper 2 (Th2) (interleukin-4 secretion) not Th1 (interferon-γ secretion) response. Furthermore, the viremia induced by REV infection was significantly reduced in chickens immunized with the glycosylated gp90. Overall, an optimized secretory expression system for glycosylated gp90 was developed, and the glycosylated gp90 obtained in this study retained good immunogenicity and could be an attractive vaccine candidate to protect chickens against REV horizonal infection.


Subject(s)
Antigens, Viral/immunology , Chickens , Poultry Diseases/virology , Reticuloendotheliosis Viruses, Avian/immunology , Retroviridae Infections/veterinary , Tumor Virus Infections/veterinary , Viral Envelope Proteins/immunology , Viral Vaccines/immunology , Animals , Antigens, Surface/immunology , Glycosylation , Immunogenicity, Vaccine , Poultry Diseases/prevention & control , Retroviridae Infections/prevention & control , Retroviridae Infections/virology , Tumor Virus Infections/prevention & control , Tumor Virus Infections/virology
9.
FEMS Microbiol Rev ; 44(5): 583-605, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32556174

ABSTRACT

The iconic Australian marsupial, the koala (Phascolarctos cinereus), has suffered dramatic population declines as a result of habitat loss and fragmentation, disease, vehicle collision mortality, dog attacks, bushfires and climate change. In 2012, koalas were officially declared vulnerable by the Australian government and listed as a threatened species. In response, research into diseases affecting koalas has expanded rapidly. The two major pathogens affecting koalas are Chlamydia pecorum, leading to chlamydial disease and koala retrovirus (KoRV). In the last eight years, these pathogens and their diseases have received focused study regarding their sources, genetics, prevalence, disease presentation and transmission. This has led to vast improvements in pathogen detection and treatment, including the ongoing development of vaccines for each as a management and control strategy. This review will summarize and highlight the important advances made in understanding and combating C. pecorum and KoRV in koalas, since they were declared a threatened species. With complementary advances having also been made from the koala genome sequence and in our understanding of the koala immune system, we are primed to make a significant positive impact on koala health into the future.


Subject(s)
Chlamydia Infections/veterinary , Phascolarctidae/immunology , Retroviridae Infections/veterinary , Animals , Australia , Chlamydia , Chlamydia Infections/diagnosis , Chlamydia Infections/prevention & control , Chlamydia Infections/therapy , Endangered Species , Phascolarctidae/microbiology , Phascolarctidae/virology , Retroviridae , Retroviridae Infections/diagnosis , Retroviridae Infections/prevention & control , Retroviridae Infections/therapy , Viral Vaccines
10.
Viruses ; 11(10)2019 10 10.
Article in English | MEDLINE | ID: mdl-31658739

ABSTRACT

Simian foamy viruses (SFV) infect a wide range of Old World and Neotropical primates (NP). Unlike Old World primates, little is known about the diversity and prevalence of SFV in NP, mainly from a free-living population. Phylogenetic analyses have shown that SFV coevolved with their hosts. However, viral strains infecting Leontopithecus chrysomelas did not behave as expected for this hypothesis. The purpose of this study was to determine the eco-epidemiological profile and molecular characterization of SFV in a recently captured invasive population of L. chrysomelas located in Niteroi/RJ using buccal swab as an alternative collection method. A prevalence of 34.8% (32/92) and a mean viral load of 4.7 log copies of SFV/106 cells were observed. With respect to time since capture, SFV prevalence was significantly higher in the group of animals sampled over 6 months after capture (55.2%) than in those more recently captured (25.4%) (p = 0.005). Infected solitary animals can contribute to SFV transmission between different groups in the population. SFV strains formed two distinct clades within the SFV infecting the Cebidae family. This is the first study to use buccal swabs as a tool to study SFV diversity and prevalence in a recently free-living NP population upon recent capture.


Subject(s)
Leontopithecus/virology , Retroviridae Infections , Simian foamy virus , Animals , Animals, Wild/virology , Brazil/epidemiology , Genes, Viral , Monkey Diseases/virology , Phylogeny , Prevalence , Retroviridae Infections/epidemiology , Retroviridae Infections/prevention & control , Retroviridae Infections/transmission , Simian foamy virus/classification , Simian foamy virus/genetics , Simian foamy virus/isolation & purification
11.
PLoS Pathog ; 15(9): e1008043, 2019 09.
Article in English | MEDLINE | ID: mdl-31568492

ABSTRACT

Immunization vectors based on cytomegalovirus (CMV) have attracted a lot of interest in recent years because of their high efficacy in the simian immunodeficiency virus (SIV) macaque model, which has been attributed to their ability to induce strong, unusually broad, and unconventionally restricted CD8+ T cell responses. To evaluate the ability of CMV-based vectors to mediate protection by other immune mechanisms, we evaluated a mouse CMV (MCMV)-based vector encoding Friend virus (FV) envelope (Env), which lacks any known CD8+ T cell epitopes, for its protective efficacy in the FV mouse model. When we immunized highly FV-susceptible mice with the Env-encoding MCMV vector (MCMV.env), we could detect high frequencies of Env-specific CD4+ T cells after a single immunization. While the control of an early FV challenge infection was highly variable, an FV infection applied later after immunization was tightly controlled by almost all immunized mice. Protection of mice correlated with their ability to mount a robust anamnestic neutralizing antibody response upon FV infection, but Env-specific CD4+ T cells also produced appreciable levels of interferon γ. Depletion and transfer experiments underlined the important role of antibodies for control of FV infection but also showed that while no Env-specific CD8+ T cells were induced by the MCMV.env vaccine, the presence of CD8+ T cells at the time of FV challenge was required. The immunity induced by MCMV.env immunization was long-lasting, but was restricted to MCMV naïve animals. Taken together, our results demonstrate a novel mode of action of a CMV-based vaccine for anti-retrovirus immunization that confers strong protection from retrovirus challenge, which is conferred by CD4+ T cells and antibodies.


Subject(s)
Friend murine leukemia virus/immunology , Muromegalovirus/immunology , Viral Vaccines/immunology , Adoptive Transfer , Animals , Antibodies, Viral/biosynthesis , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/virology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/virology , Female , Friend murine leukemia virus/genetics , Friend murine leukemia virus/pathogenicity , Gene Products, env/genetics , Gene Products, env/immunology , Genetic Vectors , Immunization , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Muromegalovirus/genetics , Retroviridae Infections/immunology , Retroviridae Infections/prevention & control , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology , Viral Vaccines/genetics
12.
J Virol ; 93(21)2019 11 01.
Article in English | MEDLINE | ID: mdl-31375593

ABSTRACT

Adenovirus (AdV)-based vectors are popular experimental vaccine vectors, but despite their ability to induce strong immune responses, their application is impeded by widespread preexisting immunity against many AdV types that can impair or even abrogate the induction of transgene-specific immune responses. Therefore, the development of vectors based on AdV types with a low seroprevalence is important for effective AdV-based immunization in humans. We investigated the immunization efficacy of vectors based on AdV type 48 (Ad48) and Ad50 in the ovalbumin (ova) model as well as the Friend retrovirus (FV) model, which allows testing of the protective effect of vaccine-induced immunity. Using ova-encoding vectors, we found a significantly lower induction of ova-specific CD8+ T cells and antibody responses by Ad48- and Ad50-based vectors than by Ad5-based vectors. Similarly, we found a reduced induction of FV-specific CD8+ T cell responses in Ad48- and Ad50.Leader-Gag-immunized mice compared with that in Ad5-immunized mice; however, some of those mice were able to control the FV infection, and protection correlated with the level of neutralizing antibodies 10 days after FV challenge. Analyses of the AdV-specific antibodies and CD8+ T cells induced by the individual AdV types revealed a high level of cross-reactivity, and the efficacy of Ad48-based immunization was impaired in Ad5-preimmune mice. Our results show that the immunity induced by Ad48- and Ad50-based vectors is reduced compared to that induced by Ad5 and is sufficient to control FV infection in only some of the immunized mice. A high level of cross-reactivity suggests that AdV preimmunity must be considered even when applying rare AdV-based vectors.IMPORTANCE AdV-based vectors are important tools for the development of vaccines against a wide range of pathogens. While AdV vectors are generally considered safe and highly effective, their application can be severely impaired by preexisting immunity due to the widespread seroprevalence of some AdV types. The characterization of different AdV types with regard to immunogenicity and efficacy in challenge models is of great importance for the development of improved AdV-based vectors that allow for efficient immunization despite anti-AdV immunity. We show that the immunity induced by an Ad48-based vector is inferior to that induced by an Ad5-based vector but can still mediate the control of an FV infection in highly FV-susceptible mice. However, the efficacy of Ad48-based immunization was impaired in Ad5-preimmune mice. Importantly, we found cross-reactivity of both the humoral and cellular immune responses raised by the individual AdV types, suggesting that switching to a different AdV type may not be sufficient to circumvent preexisting anti-AdV immunity.


Subject(s)
Adenoviridae Infections/immunology , Adenoviridae/classification , Adenoviridae/immunology , Adenovirus Vaccines/administration & dosage , Antibodies, Viral/immunology , Immunity, Cellular/immunology , Retroviridae Infections/immunology , Adenoviridae Infections/prevention & control , Adenoviridae Infections/virology , Adenovirus Vaccines/immunology , Animals , Antigens, Viral/immunology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/virology , Female , Genetic Vectors/administration & dosage , Humans , Immunization , Mice , Mice, Inbred BALB C , Ovalbumin/immunology , Retroviridae/immunology , Retroviridae Infections/prevention & control , Retroviridae Infections/virology
13.
Viruses ; 11(6)2019 05 31.
Article in English | MEDLINE | ID: mdl-31159230

ABSTRACT

A field study was undertaken to (i) measure the prevalence of feline leukaemia virus (FeLV) exposure and FeLV infection in a cross-section of healthy Australian pet cats; and (ii) investigate the outcomes following natural FeLV exposure in two Australian rescue facilities. Group 1 (n = 440) consisted of healthy client-owned cats with outdoor access, predominantly from eastern Australia. Groups 2 (n = 38) and 3 (n = 51) consisted of a mixture of healthy and sick cats, group-housed in two separate rescue facilities in Sydney, Australia, tested following identification of index cases of FeLV infection in cats sourced from these facilities. Diagnostic testing for FeLV exposure/infection included p27 antigen testing using three different point-of-care FeLV kits and a laboratory-based ELISA, real-time polymerase chain reaction (qPCR) testing to detect FeLV proviral DNA in leukocytes, real-time reverse-transcription PCR (qRT-PCR) testing to detect FeLV RNA in plasma, and neutralising antibody (NAb) testing. Cats were classified as FeLV-uninfected (FeLV-unexposed and presumptively FeLV-abortive infections) or FeLV-infected (presumptively regressive and presumptively progressive infections). In Group 1, 370 FeLV-unexposed cats (370/440, 84%), 47 abortive infections (47/440, 11%), nine regressive infections (9/440, 2%), and two progressive infections (2/440, 0.5%) were identified, and 12 FeLV-uninfected cats (12/440, 3%) were unclassifiable as FeLV-unexposed or abortive infections due to insufficient samples available for NAb testing. In Groups 2 and 3, 31 FeLV-unexposed cats (31/89, 35%), eight abortive infections (8/89, 9%), 22 regressive infections (22/89; 25%), and 19 progressive infections (19/89; 21%) were discovered, and nine FeLV-uninfected cats (9/89; 10%) were unclassifiable due to insufficient samples available for NAb testing. One of the presumptively progressively-infected cats in Group 3 was likely a focal FeLV infection. Two other presumptively progressively-infected cats in Group 3 may have been classified as regressive infections with repeated testing, highlighting the difficulties associated with FeLV diagnosis when sampling cats at a single time point, even with results from a panel of FeLV tests. These results serve as a reminder to Australian veterinarians that the threat of FeLV to the general pet cat population remains high, thus vigilant FeLV testing, separate housing for FeLV-infected cats, and FeLV vaccination of at-risk cats is important, particularly in group-housed cats in shelters and rescue facilities, where outbreaks of FeLV infection can occur.


Subject(s)
Cat Diseases/virology , Leukemia Virus, Feline , Leukemia, Feline/virology , Retroviridae Infections/veterinary , Animals , Antibodies, Viral/blood , Australia/epidemiology , Cats , Cross-Sectional Studies , DNA, Viral/blood , Leukemia Virus, Feline/immunology , Leukemia Virus, Feline/isolation & purification , Leukemia, Feline/diagnosis , Leukemia, Feline/epidemiology , Leukemia, Feline/prevention & control , Retroviridae Infections/diagnosis , Retroviridae Infections/epidemiology , Retroviridae Infections/prevention & control , Viral Load/veterinary
16.
J Virol ; 92(11)2018 06 01.
Article in English | MEDLINE | ID: mdl-29593034

ABSTRACT

The apolipoprotein B editing complex 3 (APOBEC3) proteins are potent retroviral restriction factors that are under strong positive selection, both in terms of gene copy number and sequence diversity. A common feature of all the members of the APOBEC3 family is the presence of one or two cytidine deamination domains, essential for cytidine deamination of retroviral reverse transcripts as well as packaging into virions. Several studies have indicated that human and mouse APOBEC3 proteins restrict retrovirus infection via cytidine deaminase (CD)-dependent and -independent means. To understand the relative contribution of CD-independent restriction in vivo, we created strains of transgenic mice on an APOBEC3 knockout background that express a deaminase-dead mouse APOBEC3 due to point mutations in both CD domains (E73Q/E253Q). Here, we show that the CD-dead APOBEC3 can restrict murine retroviruses in vivo Moreover, unlike the wild-type protein, the mutant APOBEC3 is not packaged into virions but acts only as a cell-intrinsic restriction factor that blocks reverse transcription by incoming viruses. Finally, we show that wild-type and CD-dead mouse APOBEC3 can bind to murine leukemia virus (MLV) reverse transcriptase. Our findings suggest that the mouse APOBEC3 cytidine deaminase activity is not required for retrovirus restriction.IMPORTANCE APOBEC3 proteins are important host cellular restriction factors essential for restricting retrovirus infection by causing mutations in the virus genome and by blocking reverse transcription. While both methods of restriction function in vitro, little is known about their role during in vivo infection. By developing transgenic mice with mutations in the cytidine deamination domains needed for enzymatic activity and interaction with viral RNA, we show that APOBEC3 proteins can still restrict in vivo infection by interacting with reverse transcriptase and blocking its activity. These studies demonstrate that APOBEC3 proteins have evolved multiple means for blocking retrovirus infection and that all of these means function in vivo.


Subject(s)
Cytidine Deaminase/genetics , Leukemia Virus, Murine/genetics , Retroviridae Infections/prevention & control , Reverse Transcriptase Inhibitors/metabolism , Reverse Transcription/genetics , Animals , Cell Line , Cytidine Deaminase/metabolism , Deamination/genetics , HEK293 Cells , Humans , Leukemia Virus, Murine/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , RNA, Viral/genetics , RNA-Directed DNA Polymerase/genetics
17.
Antiviral Res ; 147: 1-10, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28465147

ABSTRACT

The present study is focused on investigating the immunoprotective effects of CpG-ODN/Poly(I:C) combined with the viral glycoprotein gp90 protein against reticuloendotheliosis virus (REV) infection in chickens. REV's gp90 gene was amplified from the REV-infected cells and expressed in Escherichia coli (E.coli). The expressed products, upon purification, were inoculated into 7-day-old chickens with PBS, CpG-ODN or Poly(I:C) adjuvant; Two booster inoculations were then conducted, and then each chicken was challenged. The presence of REV-antibodies in serum was determined weekly after the first vaccination. The viremia and immunosuppressive effects of REV infection were also monitored after the challenge. The neutralizing effects of the antisera were tested in vitro. The results showed that the recombinant gene containing REV gp90 gene was expressed into the recombinant protein with a size of 51 Kilo Dalton (KD), which could be recognized by a monoclonal antibody (MAb) against the gp90 protein. The viremia and immunosuppressive effects of avian influenza virus (AIV) vaccine caused by REV challenge in CpG-ODN group and in Poly(I:C) group were dramatically decreased. REV antibody with low titers was induced in gp90 group and the inoculated chickens were partly protected. Compared with those in gp90 group, the titers and the positive ratios of REV antibody in CpG+gp90 group were significantly increased, whereas the viremia and immunosuppressive effects of AIV vaccine caused by REV infection were significantly decreased. In the Poly(I:C) +gp90 group, the viremia and immunosuppressive effects caused by REV infection were also dramatically decreased, although REV antibody responses were softly increased. The diluted antisera from the vaccinated chickens in both groups could completely inhibit the replication of REV in chick fibroblast cells (CEF). Hence, it can be concluded that CpG-ODN or the Poly(I:C) adjuvant can enhance the antiviral effects of the REV subunit vaccine against REV infection, which may result from different mechanisms.


Subject(s)
Antibodies, Viral/blood , Chickens , Oligodeoxyribonucleotides/immunology , Poly I-C/immunology , Poultry Diseases/prevention & control , Retroviridae Infections/veterinary , Viral Vaccines/immunology , Adjuvants, Immunologic/administration & dosage , Animals , Neutralization Tests , Poultry Diseases/immunology , Poultry Diseases/virology , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Reticuloendotheliosis virus , Retroviridae Infections/immunology , Retroviridae Infections/prevention & control , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/genetics , Vaccines, Subunit/immunology , Vaccines, Subunit/isolation & purification , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology , Vaccines, Synthetic/isolation & purification , Viral Envelope Proteins/genetics , Viral Envelope Proteins/immunology , Viral Vaccines/administration & dosage , Viral Vaccines/genetics , Viral Vaccines/isolation & purification , Viremia/prevention & control
18.
Retrovirology ; 14(1): 8, 2017 02 06.
Article in English | MEDLINE | ID: mdl-28166802

ABSTRACT

BACKGROUND: In the Friend retrovirus mouse model we developed potent adenovirus-based vaccines that were designed to induce either strong Friend virus GagL85-93-specific CD8+ T cell or antibody responses, respectively. To optimize the immunization outcome we evaluated vaccination strategies using combinations of these vaccines. RESULTS: While the vaccines on their own confer strong protection from a subsequent Friend virus challenge, the simple combination of the vaccines for the establishment of an optimized immunization protocol did not result in a further improvement of vaccine effectivity. We demonstrate that the co-immunization with GagL85-93/leader-gag encoding vectors together with envelope-encoding vectors abrogates the induction of GagL85-93-specific CD8+ T cells, and in successive immunization protocols the immunization with the GagL85-93/leader-gag encoding vector had to precede the immunization with an envelope encoding vector for the efficient induction of GagL85-93-specific CD8+ T cells. Importantly, the antibody response to envelope was in fact enhanced when the mice were adenovirus-experienced from a prior immunization, highlighting the expedience of this approach. CONCLUSIONS: To circumvent the immunosuppressive effect of envelope on immune responses to simultaneously or subsequently administered immunogens, we developed a two immunizations-based vaccination protocol that induces strong immune responses and confers robust protection of highly Friend virus-susceptible mice from a lethal Friend virus challenge.


Subject(s)
Adenoviridae/genetics , Retroviridae Infections/immunology , Retroviridae Infections/prevention & control , Vaccination/methods , Viral Vaccines/administration & dosage , Viral Vaccines/immunology , Adenoviridae/immunology , Animals , Antibodies, Viral/immunology , CD8-Positive T-Lymphocytes/immunology , Disease Models, Animal , Friend murine leukemia virus/genetics , Friend murine leukemia virus/immunology , Genetic Vectors , HEK293 Cells , Humans , Mice , Retroviridae Infections/virology , Vaccines, Combined/administration & dosage , Vaccines, Combined/genetics , Vaccines, Combined/immunology , Vaccines, Synthetic/immunology , Viral Vaccines/genetics
19.
Retrovirology ; 13(1): 77, 2016 Nov 08.
Article in English | MEDLINE | ID: mdl-27821119

ABSTRACT

Natural killer (NK) cells belong to the innate immune system and protect against cancers and a variety of viruses including retroviruses by killing transformed or infected cells. They express activating and inhibitory receptors on their cell surface and often become activated after recognizing virus-infected cells. They have diverse antiviral effector functions like the release of cytotoxic granules, cytokine production and antibody dependent cellular cytotoxicity. The importance of NK cell activity in retroviral infections became evident due to the discovery of several viral strategies to escape recognition and elimination by NK cells. Mutational sequence polymorphisms as well as modulation of surface receptors and their ligands are mechanisms of the human immunodeficiency virus-1 to evade NK cell-mediated immune pressure. In Friend retrovirus infected mice the virus can manipulate molecular or cellular immune factors that in turn suppress the NK cell response. In this model NK cells lack cytokines for optimal activation and can be functionally suppressed by regulatory T cells. However, these inhibitory pathways can be overcome therapeutically to achieve full activation of NK cell responses and ultimately control dissemination of retroviral infection. One effective approach is to modulate the crosstalk between NK cells and dendritic cells, which produce NK cell-stimulating cytokines like type I interferons (IFN), IL-12, IL-15, and IL-18 upon retrovirus sensing or infection. Therapeutic administration of IFNα directly increases NK cell killing of retrovirus-infected cells. In addition, IL-2/anti-IL-2 complexes that direct IL-2 to NK cells have been shown to significantly improve control of retroviral infection by NK cells in vivo. In this review, we describe novel approaches to improve NK cell effector functions in retroviral infections. Immunotherapies that target NK cells of patients suffering from viral infections might be a promising treatment option for the future.


Subject(s)
Killer Cells, Natural/immunology , Retroviridae Infections/immunology , Retroviridae Infections/therapy , Retroviridae/immunology , Animals , Dendritic Cells/immunology , Humans , Immunotherapy/methods , Interferon-alpha/therapeutic use , Interleukin-2/therapeutic use , Killer Cells, Natural/physiology , Mice , Retroviridae/physiology , Retroviridae Infections/prevention & control , Retroviridae Infections/virology
20.
J Immunol ; 197(9): 3628-3638, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27647833

ABSTRACT

Elucidation of the immune requirements for control or elimination of retroviral infection remains an important aim. We studied the induction of adaptive immunity to neonatal infection with a murine retrovirus, under conditions leading to immunological tolerance. We found that the absence of either maternal or offspring adaptive immunity permitted efficient vertical transmission of the retrovirus. Maternal immunodeficiency allowed the retrovirus to induce central Th cell tolerance in the infected offspring. In turn, this compromised the offspring's ability to mount a protective Th cell-dependent B cell response. However, in contrast to T cells, offspring B cells were not centrally tolerized and retained their ability to respond to the infection when provided with T cell help. Thus, escape of retrovirus-specific B cells from deletional tolerance offers the opportunity to induce protective retroviral immunity by restoration of retrovirus-specific T cell help, suggesting similar T cell immunotherapies for persistent viral infections.


Subject(s)
Adoptive Transfer , B-Lymphocytes/immunology , Infectious Disease Transmission, Vertical/prevention & control , Leukemia Virus, Murine/immunology , Leukemia, Experimental/prevention & control , Retroviridae Infections/prevention & control , T-Lymphocytes/immunology , Tumor Virus Infections/prevention & control , Animals , Animals, Newborn , B-Lymphocytes/transplantation , B-Lymphocytes/virology , Cells, Cultured , Central Tolerance , Female , Leukemia, Experimental/immunology , Male , Maternal Exposure/adverse effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Pregnancy , Retroviridae Infections/immunology , Retroviridae Infections/transmission , T-Lymphocytes/transplantation , T-Lymphocytes/virology , Tumor Virus Infections/immunology , Tumor Virus Infections/transmission
SELECTION OF CITATIONS
SEARCH DETAIL
...