Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.178
Filter
1.
New Microbiol ; 47(1): 38-46, 2024 May.
Article in English | MEDLINE | ID: mdl-38700882

ABSTRACT

The shortage of organs for human transplantation is a topic of extreme interest, and xenotransplantation with porcine organs has been recognized as a promising solution. However, the potential spillover linked to infectious agents present in pigs remains a concern. Among these, Pig Endogenous Retroviruses (PERVs), whose proviral DNAs are integrated in the genome of all pig breeds, represent an extremely important biological risk. This study aims to evaluate PERVs distribution in several swine cell lines and samples of domestic and feral pigs. Moreover, the capacity of PERVs to infect human and non-human primate cells and to integrate in the cellular genome was tested by Real-Time PCR and by Reverse Transcriptase assay. Results indicated a widespread diffusion of PERVs both in cell lines and samples analysed: the viral genome was found in all the established cell lines, in 40% of the primary cell lines and in 60% of the tissue samples tested. The assays indicated that the virus can be transmitted from porcine to human cells: in the specific case, infected NSK and NPTr cells allow passage to human 293 and MRC-5 cells with active production of the virus demonstrable via PCR and RT assay. In light of these aspects and also the lack of studies on PERVs, it appears clear that there are still many questions to be clarified, also by means of future studies, before xenotransplantation can be considered microbiologically safe.


Subject(s)
Endogenous Retroviruses , Animals , Endogenous Retroviruses/genetics , Endogenous Retroviruses/isolation & purification , Swine , Humans , Cell Line , Retroviridae Infections/veterinary , Retroviridae Infections/virology , Retroviridae Infections/transmission
2.
Viruses ; 16(4)2024 03 23.
Article in English | MEDLINE | ID: mdl-38675836

ABSTRACT

PYHIN proteins are only found in mammals and play key roles in the defense against bacterial and viral pathogens. The corresponding gene locus shows variable deletion and expansion ranging from 0 genes in bats, over 1 in cows, and 4 in humans to a maximum of 13 in mice. While initially thought to act as cytosolic immune sensors that recognize foreign DNA, increasing evidence suggests that PYHIN proteins also inhibit viral pathogens by more direct mechanisms. Here, we examined the ability of all 13 murine PYHIN proteins to inhibit HIV-1 and murine leukemia virus (MLV). We show that overexpression of p203, p204, p205, p208, p209, p210, p211, and p212 strongly inhibits production of infectious HIV-1; p202, p207, and p213 had no significant effects, while p206 and p214 showed intermediate phenotypes. The inhibitory effects on infectious HIV-1 production correlated significantly with the suppression of reporter gene expression by a proviral Moloney MLV-eGFP construct and HIV-1 and Friend MLV LTR luciferase reporter constructs. Altogether, our data show that the antiretroviral activity of PYHIN proteins is conserved between men and mice and further support the key role of nuclear PYHIN proteins in innate antiviral immunity.


Subject(s)
HIV-1 , Leukemia Virus, Murine , Phosphoproteins , Animals , Mice , Humans , HIV-1/immunology , HIV-1/genetics , Leukemia Virus, Murine/genetics , Leukemia Virus, Murine/immunology , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Nuclear Proteins/immunology , Virus Replication , Cell Line , Retroviridae Infections/immunology , Retroviridae Infections/virology
3.
Front Cell Infect Microbiol ; 14: 1379962, 2024.
Article in English | MEDLINE | ID: mdl-38655281

ABSTRACT

The notion that viruses played a crucial role in the evolution of life is not a new concept. However, more recent insights suggest that this perception might be even more expansive, highlighting the ongoing impact of viruses on host evolution. Endogenous retroviruses (ERVs) are considered genomic remnants of ancient viral infections acquired throughout vertebrate evolution. Their exogenous counterparts once infected the host's germline cells, eventually leading to the permanent endogenization of their respective proviruses. The success of ERV colonization is evident so that it constitutes 8% of the human genome. Emerging genomic studies indicate that endogenous retroviruses are not merely remnants of past infections but rather play a corollary role, despite not fully understood, in host genetic regulation. This review presents some evidence supporting the crucial role of endogenous retroviruses in regulating host genetics. We explore the involvement of human ERVs (HERVs) in key physiological processes, from their precise and orchestrated activities during cellular differentiation and pluripotency to their contributions to aging and cellular senescence. Additionally, we discuss the costs associated with hosting a substantial amount of preserved viral genetic material.


Subject(s)
Endogenous Retroviruses , Endogenous Retroviruses/genetics , Endogenous Retroviruses/physiology , Humans , Animals , Cell Differentiation , Host-Pathogen Interactions/genetics , Host Microbial Interactions/genetics , Retroviridae Infections/virology , Cellular Senescence/genetics , Proviruses/genetics , Proviruses/physiology , Evolution, Molecular
4.
J Virol ; 98(4): e0177123, 2024 Apr 16.
Article in English | MEDLINE | ID: mdl-38440982

ABSTRACT

Endogenous retroviruses (ERVs) are remnants of ancestral viral infections. Feline leukemia virus (FeLV) is an exogenous and endogenous retrovirus in domestic cats. It is classified into several subgroups (A, B, C, D, E, and T) based on viral receptor interference properties or receptor usage. ERV-derived molecules benefit animals, conferring resistance to infectious diseases. However, the soluble protein encoded by the defective envelope (env) gene of endogenous FeLV (enFeLV) functions as a co-factor in FeLV subgroup T infections. Therefore, whether the gene emerged to facilitate viral infection is unclear. Based on the properties of ERV-derived molecules, we hypothesized that the defective env genes possess antiviral activity that would be advantageous to the host because FeLV subgroup B (FeLV-B), a recombinant virus derived from enFeLV env, is restricted to viral transmission among domestic cats. When soluble truncated Env proteins from enFeLV were tested for their inhibitory effects against enFeLV and FeLV-B, they inhibited viral infection. Notably, this antiviral machinery was extended to infection with the Gibbon ape leukemia virus, Koala retrovirus A, and Hervey pteropid gammaretrovirus. Although these viruses used feline phosphate transporter 1 (fePit1) and phosphate transporter 2 as receptors, the inhibitory mechanism involved competitive receptor binding in a fePit1-dependent manner. The shift in receptor usage might have occurred to avoid the inhibitory effect. Overall, these findings highlight the possible emergence of soluble truncated Env proteins from enFeLV as a restriction factor against retroviral infection and will help in developing host immunity and antiviral defense by controlling retroviral spread.IMPORTANCERetroviruses are unique in using reverse transcriptase to convert RNA genomes into DNA, infecting germ cells, and transmitting to offspring. Numerous ancient retroviral sequences are known as endogenous retroviruses (ERVs). The soluble Env protein derived from ERVs functions as a co-factor that assists in FeLV-T infection. However, herein, we show that the soluble Env protein exhibits antiviral activity and provides resistance to mammalian retrovirus infection through competitive receptor binding. In particular, this finding may explain why FeLV-B transmission is not observed among domestic cats. ERV-derived molecules can benefit animals in an evolutionary arms race, highlighting the double-edged-sword nature of ERVs.


Subject(s)
Gene Products, env , Leukemia Virus, Feline , Leukemia, Feline , Animals , Cats , Endogenous Retroviruses/genetics , Endogenous Retroviruses/metabolism , Gene Products, env/genetics , Gene Products, env/metabolism , Leukemia Virus, Feline/classification , Leukemia Virus, Feline/genetics , Leukemia Virus, Feline/metabolism , Leukemia Virus, Gibbon Ape/genetics , Leukemia Virus, Gibbon Ape/metabolism , Leukemia, Feline/genetics , Leukemia, Feline/metabolism , Leukemia, Feline/virology , Phosphate Transport Proteins/genetics , Phosphate Transport Proteins/metabolism , Receptors, Virus/metabolism , Retroviridae Infections/metabolism , Retroviridae Infections/virology , Solubility , Female
5.
J Virol ; 97(3): e0180322, 2023 03 30.
Article in English | MEDLINE | ID: mdl-36779757

ABSTRACT

The isolation of the Koala retrovirus-like virus from Australian megabats and the identification of endogenous retroviruses in the bat genome have raised questions on bat susceptibility to retroviruses in general. To answer this, we studied the susceptibility of 12 cell lines from 11 bat species to four well-studied retroviruses (human and simian immunodeficiency viruses [HIV and SIV] and murine leukemia viruses [B- and N-MLV]). Systematic comparison of retroviral susceptibility among bats revealed that megabat cell lines were overall less susceptible to the four retroviruses than microbat cell lines, particularly to HIV-1 infection, whereas lineage-specific differences were observed for MLV susceptibility. Quantitative PCR of reverse transcription (RT) products, infection in heterokaryon cells, and point mutation analysis of the capsid (CA) revealed that (i) HIV-1 and MLV replication were blocked at the nuclear transport of the pre-integration complexes and before and/or during RT, respectively, and (ii) the observed lineage-specific restriction can be attributed to a dominant cellular factor constrained by specific positions in CA. Investigation of bat homologs of the three previously reported post-entry restriction factors constrained by the same residues in CA, tripartite motif-protein 5α (TRIM5α), myxovirus resistance 2/B (Mx2/MxB), and carboxy terminus-truncated cleavage and polyadenylation factor 6 (CPSF6-358), demonstrated poor anti-HIV-1 activity in megabat cells, whereas megabat TRIM5α restricted MLV infection, suggesting that the major known CA-dependent restriction factors were not dominant in the observed lineage-specific susceptibility to HIV-1 in bat cells. Therefore, HIV-1 susceptibility of megabat cells may be determined in a manner distinct from that of primate cells. IMPORTANCE Recent studies have demonstrated the circulation of gammaretroviruses among megabats in Australia and the bats' resistance to HIV-1 infection; however, the origins of these viruses in megabats and the contribution of bats to retrovirus spread to other mammalian species remains unclear. To determine the intrinsic susceptibility of bat cells to HIV-1 infection, we investigated 12 cell lines isolated from 11 bat species. We report that lineage-specific retrovirus restriction in the bat cell lines can be attributed to CA-dependent factors. However, in the megabat cell lines examined, factors known to bind capsid and block infection in primate cell culture, including homologs of TRIM5α, Mx2/MxB, and CPSF6, failed to exhibit significant anti-HIV-1 activities. These results suggested that the HIV-1 susceptibility of megabat cells occurs in a manner distinct from that of primate cells, where cellular factors, other than major known CA-dependent restriction factors, with lineage-specific functions could recognize retroviral proteins in megabats.


Subject(s)
Capsid , Chiroptera , Disease Susceptibility , Retroviridae , Animals , Humans , Mice , Australia , Capsid/metabolism , Capsid Proteins/genetics , Capsid Proteins/metabolism , Chiroptera/virology , Retroviridae/classification , Retroviridae/metabolism , Retroviridae Infections/metabolism , Retroviridae Infections/virology , Disease Susceptibility/metabolism , Disease Susceptibility/virology , Cell Line , Species Specificity , Antiviral Restriction Factors/metabolism
6.
Vet Ital ; 59(4)2023 Dec 31.
Article in English | MEDLINE | ID: mdl-38685825

ABSTRACT

Fowl Pox Viruses (FPV) infect chickens and turkeys giving rise to pock lesions on various body parts like combs, wattles, legs, shanks, eyes, mouth etc. The birds, affected with FPV, also show anemia and ruffled appearance which are clinical symptoms of Reticuloendotheliosis. Interestingly, the field strains of FPV are integrated with the provirus of Reticuloendotheliosis Virus (REV). Due to this integration, the infected birds, upon replication of FPV, give rise to free REV virions, causing severe immunosuppression and anemia. Pox scabs, collected from the infected birds, not only show positive PCR results upon performing FPV-specific 4b core protein gene PCR but also show positive results for the PCR of REV-specific env gene and FPV-REV 5'LTR junction. Homogenized suspension of the pock lesions, upon inoculating to the Chorio-allantoic Membrane (CAM) of 10 days old specific pathogen-free embryonated chicken eggs, produces characteristic pock lesions in serial passages. But the lesions also harbor REV mRNA or free virion, which can be identified by performing REV-specific env gene PCR using REV RNA from FPV-infected CAMs. The study suggests successful replication and availability of REV mRNA and free virion alongside the FPV virus, although the CAM is an ill-suited medium for any retroviral (like REV) growth and replication.


Subject(s)
Chickens , Fowlpox virus , Poultry Diseases , Reticuloendotheliosis virus , Animals , Reticuloendotheliosis virus/isolation & purification , Chickens/virology , Poultry Diseases/virology , Fowlpox virus/genetics , Fowlpox virus/isolation & purification , Specific Pathogen-Free Organisms , Chick Embryo , Fowlpox/virology , Chorioallantoic Membrane/virology , Retroviridae Infections/veterinary , Retroviridae Infections/virology
7.
Proc Natl Acad Sci U S A ; 119(33): e2122680119, 2022 08 16.
Article in English | MEDLINE | ID: mdl-35943984

ABSTRACT

Koala retrovirus (KoRV) subtype A (KoRV-A) is currently in transition from exogenous virus to endogenous viral element, providing an ideal system to elucidate retroviral-host coevolution. We characterized KoRV geography using fecal DNA from 192 samples across 20 populations throughout the koala's range. We reveal an abrupt change in KoRV genetics and incidence at the Victoria/New South Wales state border. In northern koalas, pol gene copies were ubiquitously present at above five per cell, consistent with endogenous KoRV. In southern koalas, pol copies were detected in only 25.8% of koalas and always at copy numbers below one, while the env gene was detected in all animals and in a majority at copy numbers above one per cell. These results suggest that southern koalas carry partial endogenous KoRV-like sequences. Deep sequencing of the env hypervariable region revealed three putatively endogenous KoRV-A sequences in northern koalas and a single, distinct sequence present in all southern koalas. Among northern populations, env sequence diversity decreased with distance from the equator, suggesting infectious KoRV-A invaded the koala genome in northern Australia and then spread south. The exogenous KoRV subtypes (B to K), two novel subtypes, and intermediate subtypes were detected in all northern koala populations but were strikingly absent from all southern animals tested. Apart from KoRV subtype D, these exogenous subtypes were generally locally prevalent but geographically restricted, producing KoRV genetic differentiation among northern populations. This suggests that sporadic evolution and local transmission of the exogenous subtypes have occurred within northern Australia, but this has not extended into animals within southern Australia.


Subject(s)
Endogenous Retroviruses , Evolution, Molecular , Gammaretrovirus , Phascolarctidae , Animals , Endogenous Retroviruses/genetics , Gammaretrovirus/genetics , Genetic Variation , New South Wales , Phascolarctidae/virology , Retroviridae Infections/transmission , Retroviridae Infections/veterinary , Retroviridae Infections/virology , Victoria
8.
Proc Natl Acad Sci U S A ; 119(26): e2114441119, 2022 06 28.
Article in English | MEDLINE | ID: mdl-35749360

ABSTRACT

Host genetic resistance to viral infection controls the pathogenicity and epidemic dynamics of infectious diseases. Refrex-1 is a restriction factor against feline leukemia virus subgroup D (FeLV-D) and an endogenous retrovirus (ERV) in domestic cats (ERV-DC). Refrex-1 is encoded by a subset of ERV-DC loci with truncated envelope genes and secreted from cells as a soluble protein. Here, we identified the copper transporter CTR1 as the entry receptor for FeLV-D and genotype I ERV-DCs. We also identified CTR1 as a receptor for primate ERVs from crab-eating macaques and rhesus macaques, which were found in a search of intact envelope genes capable of forming infectious viruses. Refrex-1 counteracted infection by FeLV-D and ERV-DCs via competition for the entry receptor CTR1; the antiviral effects extended to primate ERVs with CTR1-dependent entry. Furthermore, truncated ERV envelope genes found in chimpanzee, bonobo, gorilla, crab-eating macaque, and rhesus macaque genomes could also block infection by feline and primate retroviruses. Genetic analyses showed that these ERV envelope genes were acquired in a species- or genus-specific manner during host evolution. These results indicated that soluble envelope proteins could suppress retroviral infection across species boundaries, suggesting that they function to control retroviral spread. Our findings revealed that several mammalian species acquired antiviral machinery from various ancient retroviruses, leading to convergent evolution for host defense.


Subject(s)
Copper Transporter 1 , Genes, env , Leukemia Virus, Feline , Leukemia, Feline , Retroviridae Infections , Animals , Cats , Copper Transporter 1/genetics , Evolution, Molecular , Host-Pathogen Interactions , Leukemia Virus, Feline/physiology , Leukemia, Feline/genetics , Leukemia, Feline/virology , Macaca mulatta , Retroviridae Infections/genetics , Retroviridae Infections/virology
9.
Proc Natl Acad Sci U S A ; 119(25): e2201844119, 2022 06 21.
Article in English | MEDLINE | ID: mdl-35696585

ABSTRACT

Retroviruses have left their legacy in host genomes over millions of years as endogenous retroviruses (ERVs), and their structure, diversity, and prevalence provide insights into the historical dynamics of retrovirus-host interactions. In bioinformatic analyses of koala (Phascolarctos cinereus) whole-genome sequences, we identify a recently expanded ERV lineage (phaCin-ß) that is related to the New World squirrel monkey retrovirus. This ERV expansion shares many parallels with the ongoing koala retrovirus (KoRV) invasion of the koala genome, including highly similar and mostly intact sequences, and polymorphic ERV loci in the sampled koala population. The recent phaCin-ß ERV colonization of the koala genome appears to predate the current KoRV invasion, but polymorphic ERVs and divergence comparisons between these two lineages predict a currently uncharacterized, possibly still extant, phaCin-ß retrovirus. The genomics approach to ERV-guided discovery of novel retroviruses in host species provides a strong incentive to search for phaCin-ß retroviruses in the Australasian fauna.


Subject(s)
Betaretrovirus , Endogenous Retroviruses , Host Microbial Interactions , Phascolarctidae , Retroviridae Infections , Animals , Betaretrovirus/genetics , Endogenous Retroviruses/genetics , Evolution, Molecular , Genome , Genomics , Phascolarctidae/genetics , Phascolarctidae/virology , Retroviridae Infections/veterinary , Retroviridae Infections/virology
10.
Viruses ; 14(2)2022 02 05.
Article in English | MEDLINE | ID: mdl-35215917

ABSTRACT

Retroviruses must selectively recognize their unspliced RNA genome (gRNA) among abundant cellular and spliced viral RNAs to assemble into newly formed viral particles. Retroviral gRNA packaging is governed by Gag precursors that also orchestrate all the aspects of viral assembly. Retroviral life cycles, and especially the HIV-1 one, have been previously extensively analyzed by several methods, most of them based on molecular biology and biochemistry approaches. Despite these efforts, the spatio-temporal mechanisms leading to gRNA packaging and viral assembly are only partially understood. Nevertheless, in these last decades, progress in novel bioimaging microscopic approaches (as FFS, FRAP, TIRF, and wide-field microscopy) have allowed for the tracking of retroviral Gag and gRNA in living cells, thus providing important insights at high spatial and temporal resolution of the events regulating the late phases of the retroviral life cycle. Here, the implementation of these recent bioimaging tools based on highly performing strategies to label fluorescent macromolecules is described. This report also summarizes recent gains in the current understanding of the mechanisms employed by retroviral Gag polyproteins to regulate molecular mechanisms enabling gRNA packaging and the formation of retroviral particles, highlighting variations and similarities among the different retroviruses.


Subject(s)
Capsid/metabolism , Gene Products, gag/metabolism , Retroviridae Infections/virology , Retroviridae/physiology , Virus Assembly , Animals , Gene Products, gag/genetics , Genome, Viral , Humans , Retroviridae/genetics , Virion/genetics , Virion/physiology
11.
Nat Commun ; 13(1): 463, 2022 01 24.
Article in English | MEDLINE | ID: mdl-35075135

ABSTRACT

Germ cells are essential to pass DNA from one generation to the next. In human reproduction, germ cell development begins with the specification of primordial germ cells (PGCs) and a failure to specify PGCs leads to human infertility. Recent studies have revealed that the transcription factor network required for PGC specification has diverged in mammals, and this has a significant impact on our understanding of human reproduction. Here, we reveal that the Hominidae-specific Transposable Elements (TEs) LTR5Hs, may serve as TEENhancers (TE Embedded eNhancers) to facilitate PGC specification. LTR5Hs TEENhancers become transcriptionally active during PGC specification both in vivo and in vitro with epigenetic reprogramming leading to increased chromatin accessibility, localized DNA demethylation, enrichment of H3K27ac, and occupation of key hPGC transcription factors. Inactivation of LTR5Hs TEENhancers with KRAB mediated CRISPRi has a significant impact on germ cell specification. In summary, our data reveals the essential role of Hominidae-specific LTR5Hs TEENhancers in human germ cell development.


Subject(s)
Endogenous Retroviruses/physiology , Hominidae/virology , Reproduction , Retroelements , Retroviridae Infections/virology , Animals , Endogenous Retroviruses/genetics , Enhancer Elements, Genetic , Gene Expression Regulation, Developmental , Germ Cells/physiology , Germ Cells/virology , Hominidae/genetics , Hominidae/physiology , Humans , Retroviridae Infections/physiopathology , Transcription Factors/genetics , Transcription Factors/metabolism
12.
Cell Rep ; 38(3): 110279, 2022 01 18.
Article in English | MEDLINE | ID: mdl-35045303

ABSTRACT

Murine leukemia virus (MLV)-presenting cells form stable intercellular contacts with target cells during infection of lymphoid tissue, indicating a role of cell-cell contacts in retrovirus dissemination. Whether host cell adhesion proteins are required for retrovirus spread in vivo remains unknown. Here, we demonstrate that the lymphocyte-function-associated-antigen-1 (LFA1) and its ligand intercellular-adhesion-molecule-1 (ICAM1) are important for cell-contact-dependent transmission of MLV between leukocytes. Infection experiments in LFA1- and ICAM1-deficient mice demonstrate a defect in MLV spread within lymph nodes. Co-culture of primary leukocytes reveals a specific requirement for ICAM1 on donor cells and LFA1 on target cells for cell-contact-dependent spread through trans- and cis-infection. Importantly, adoptive transfer experiments combined with a newly established MLV-fusion assay confirm that the directed LFA1-ICAM1 interaction is important for retrovirus fusion and transmission in vivo. Taken together, our data provide insights on how retroviruses exploit host proteins and the biology of cell-cell interactions for dissemination.


Subject(s)
Intercellular Adhesion Molecule-1/metabolism , Leukemia Virus, Murine/pathogenicity , Leukemia, Experimental/virology , Lymphocyte Function-Associated Antigen-1/metabolism , Retroviridae Infections/virology , Animals , Host-Pathogen Interactions/physiology , Lymphocytes/virology , Macrophages/virology , Mice , Mice, Inbred C57BL , Retroviridae Infections/transmission , Tumor Virus Infections/transmission , Tumor Virus Infections/virology
13.
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
14.
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
15.
Pol J Vet Sci ; 24(3): 439-443, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34730307

ABSTRACT

Reticuloendotheliosis virus (REV) is an avian oncogenic retrovirus that causes atrophy of immune organs, such as the spleen, thymus, and bursa of Fabricius, leading to severe immunosuppression. However, there is limited information describing the genes or microRNAs (miRNAs) that play a role in replicating REV-spleen necrosis virus (SNV). Our previous miRNA and RNA sequencing data showed that the expression of gga-miR-222b-5p was significantly upregulated in REV-SNV-infected chicken spleens of 7, 14, and 21 dpi compared to non-infected chicken spleens, but mitogen-activated protein kinase 10 (MAPK10), which is related to innate immunity, had the opposite expression pattern. To understand chicken cellular miRNA function in the virus-host interactions during REV infection, we used quantitative reverse transcription PCR (qRT-PCR) to determine whether the expression of gga-miR-222b-5p and MAPK10 in the spleen of specific-pathogen-free chickens at 28, 35, and 42 dpi was consistent with the first 3 time points, and dual-luciferase reporter assay was used to determine the targeting relationship between gga-miR-222b-5p and MAPK10. Results show that MAPK10 was downregulated at all 3 time points; however, significant difference (p⟨0.01) was noted only at 35 dpi. Moreover, the expression of gga-miR-222b-5p was upregulated; however, significant difference (p⟨0.01) was observed only at 28 and 35 dpi. A dual-luciferase reporter assay showed that MAPK10 is a direct target of gga-miR-222b-5p. This study suggests that gga-miR-222b-5p may target MAPK10 to promote the REV-SNV-induced tumorigenesis via the RLRs signaling pathway.


Subject(s)
MicroRNAs/metabolism , Mitogen-Activated Protein Kinase 10/metabolism , Retroviridae Infections/veterinary , Spleen/metabolism , Trager duck spleen necrosis virus/immunology , Transcriptome , Animals , Chickens , Down-Regulation , Gene Expression Regulation , Humans , MicroRNAs/genetics , Mitogen-Activated Protein Kinase 10/genetics , Retroviridae Infections/virology , Specific Pathogen-Free Organisms , Tumor Virus Infections/veterinary , Tumor Virus Infections/virology
16.
Front Immunol ; 12: 729017, 2021.
Article in English | MEDLINE | ID: mdl-34603301

ABSTRACT

Piscine orthoreovirus (PRV-1) infection causes heart and skeletal muscle inflammation (HSMI) in farmed Atlantic salmon (Salmo salar). The virus is also associated with focal melanized changes in white skeletal muscle where PRV-1 infection of macrophages appears to be important. In this study, we studied the macrophage polarization into M1 (pro-inflammatory) and M2 (anti-inflammatory) phenotypes during experimentally induced HSMI. The immune response in heart with HSMI lesions was characterized by CD8+ and MHC-I expressing cells and not by polarized macrophages. Fluorescent in situ hybridization (FISH) assays revealed localization of PRV-1 in a few M1 macrophages in both heart and skeletal muscle. M2 type macrophages were widely scattered in the heart and were more abundant in heart compared to the skeletal muscle. However, the M2 macrophages did not co-stain for PRV-1. There was a strong cellular immune response to the infection in the heart compared to that of the skeletal muscle, seen as increased MHC-I expression, partly in cells also containing PRV-1 RNA, and a high number of cytotoxic CD8+ granzyme producing cells that targeted PRV-1. In skeletal muscle, MHC-I expressing cells and CD8+ cells were dispersed between myocytes, but these cells did not stain for PRV-1. Gene expression analysis by RT-qPCR complied with the FISH results and confirmed a drop in level of PRV-1 following the cell mediated immune response. Overall, the results indicated that M1 macrophages do not contribute to the initial development of HSMI. However, large numbers of M2 macrophages reside in the heart and may contribute to the subsequent fast recovery following clearance of PRV-1 infection.


Subject(s)
CD8-Positive T-Lymphocytes/virology , Fish Diseases/virology , Heart/virology , Macrophages/virology , Orthoreovirus/pathogenicity , Retroviridae Infections/virology , Salmo salar/virology , Animals , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Fish Diseases/immunology , Fish Diseases/metabolism , Host-Pathogen Interactions , Immunity, Cellular , Macrophages/immunology , Macrophages/metabolism , Muscle, Skeletal/immunology , Muscle, Skeletal/metabolism , Muscle, Skeletal/virology , Myocardium/immunology , Myocardium/metabolism , Orthoreovirus/immunology , Phenotype , Retroviridae Infections/immunology , Retroviridae Infections/metabolism , Salmo salar/immunology , Salmo salar/metabolism , Time Factors , Viral Load
17.
Front Immunol ; 12: 728677, 2021.
Article in English | MEDLINE | ID: mdl-34691035

ABSTRACT

Multiple Sclerosis (MS) is an inflammatory disease of the central nervous system. Sardinia, an Italian island, is one of the areas with the highest global prevalence of MS. Genetic factors have been widely explored to explain this greater prevalence among some populations; the genetic makeup of the Sardinians appears to make them more likely to develop autoimmune diseases. A strong association between MS and some infections have been reported globally. The most robust evidence indicating the role of infections is MS development concerns the Epstein-Barr virus (EBV). Anti-EBV antibodies in patients once infected by EBV are associated with the development of MS years later. These features have also been noted in Sardinian patients with MS. Many groups have found an increased expression of the Human endogenous retroviruses (HERV) family in patients with MS. A role in pathogenesis, prognosis, and prediction of treatment response has been proposed for HERV. A European multi-centre study has shown that their presence was variable among populations, ranging from 59% to 100% of patients, with higher HERV expression noted in Sardinian patients with MS. The mycobacterium avium subspecies paratuberculosis (MAP) DNA and antibodies against MAP2694 protein were found to be associated with MS in Sardinian patients. More recently, this association has also been reported in Japanese patients with MS. In this study, we analysed the role of infectious factors in Sardinian patients with MS and compared it with the findings reported in other populations.


Subject(s)
Autoimmunity , Epstein-Barr Virus Infections/epidemiology , Global Health , Multiple Sclerosis/epidemiology , Paratuberculosis/epidemiology , Retroviridae Infections/epidemiology , Endogenous Retroviruses/immunology , Endogenous Retroviruses/pathogenicity , Epstein-Barr Virus Infections/immunology , Epstein-Barr Virus Infections/virology , Herpesvirus 4, Human/immunology , Herpesvirus 4, Human/pathogenicity , Host-Pathogen Interactions , Humans , Italy/epidemiology , Multiple Sclerosis/immunology , Multiple Sclerosis/microbiology , Multiple Sclerosis/virology , Mycobacterium avium subsp. paratuberculosis/immunology , Mycobacterium avium subsp. paratuberculosis/pathogenicity , Paratuberculosis/immunology , Paratuberculosis/microbiology , Prognosis , Retroviridae Infections/immunology , Retroviridae Infections/virology , Risk Assessment , Risk Factors
18.
Proc Natl Acad Sci U S A ; 118(38)2021 09 21.
Article in English | MEDLINE | ID: mdl-34493581

ABSTRACT

Koala populations are currently in rapid decline across Australia, with infectious diseases being a contributing cause. The koala retrovirus (KoRV) is a gammaretrovirus present in both captive and wild koala colonies that presents an additional challenge for koala conservation in addition to habitat loss, climate change, and other factors. Currently, nine different subtypes (A to I) have been identified; however, KoRV genetic diversity analyses have been limited. KoRV is thought to be exogenously transmitted between individuals, with KoRV-A also being endogenous and transmitted through the germline. The mechanisms of exogenous KoRV transmission are yet to be extensively investigated. Here, deep sequencing was employed on 109 captive koalas of known pedigree, housed in two institutions from Southeast Queensland, to provide a detailed analysis of KoRV transmission dynamics and genetic diversity. The final dataset included 421 unique KoRV sequences, along with the finding of an additional subtype (KoRV-K). Our analysis suggests that exogenous transmission of KoRV occurs primarily between dam and joey, with evidence provided for multiple subtypes, including nonendogenized KoRV-A. No evidence of sexual transmission was observed, with mating partners found to share a similar number of sequences as unrelated koala pairs. Importantly, both distinct captive colonies showed similar trends. These findings indicate that breeding strategies or antiretroviral treatment of females could be employed as effective management approaches in combating KoRV transmission.


Subject(s)
Genetic Variation/genetics , Retroviridae Infections/transmission , Retroviridae Infections/virology , Retroviridae/genetics , Animals , Evolution, Molecular , Female , Male , Phascolarctidae , Queensland
19.
Nat Commun ; 12(1): 5376, 2021 09 10.
Article in English | MEDLINE | ID: mdl-34508086

ABSTRACT

Natural killer (NK) cells are important early responders against viral infections. Changes in metabolism are crucial to fuel NK cell responses, and altered metabolism is linked to NK cell dysfunction in obesity and cancer. However, very little is known about the metabolic requirements of NK cells during acute retroviral infection and their importance for antiviral immunity. Here, using the Friend retrovirus mouse model, we show that following infection NK cells increase nutrient uptake, including amino acids and iron, and reprogram their metabolic machinery by increasing glycolysis and mitochondrial metabolism. Specific deletion of the amino acid transporter Slc7a5 has only discrete effects on NK cells, but iron deficiency profoundly impaires NK cell antiviral functions, leading to increased viral loads. Our study thus shows the requirement of nutrients and metabolism for the antiviral activity of NK cells, and has important implications for viral infections associated with altered iron levels such as HIV and SARS-CoV-2.


Subject(s)
Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Retroviridae Infections/immunology , Animals , Bone Marrow , COVID-19 , Cytokines , HIV , HIV Infections , Large Neutral Amino Acid-Transporter 1/genetics , Large Neutral Amino Acid-Transporter 1/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria , Retroviridae , Retroviridae Infections/virology , SARS-CoV-2 , Viral Load
20.
J Virol ; 95(22): e0124421, 2021 10 27.
Article in English | MEDLINE | ID: mdl-34468176

ABSTRACT

Apolipoprotein B mRNA editing enzyme catalytic subunit 3 (APOBEC3) proteins are critical for the control of infection by retroviruses. These proteins deaminate cytidines in negative-strand DNA during reverse transcription, leading to G-to-A changes in coding strands. Uracil DNA glycosylase (UNG) is a host enzyme that excises uracils in genomic DNA, which the base excision repair machinery then repairs. Whether UNG removes uracils found in retroviral DNA after APOBEC3-mediated mutation is not clear, and whether this occurs in vivo has not been demonstrated. To determine if UNG plays a role in the repair of retroviral DNA, we used APOBEC3G (A3G) transgenic mice which we showed previously had extensive deamination of murine leukemia virus (MLV) proviruses. The A3G transgene was crossed onto an Ung and mouse Apobec3 knockout background (UNG-/-APO-/-), and the mice were infected with MLV. We found that virus infection levels were decreased in A3G UNG-/-APO-/- compared with A3G APO-/- mice. Deep sequencing of the proviruses showed that there were significantly higher levels of G-to-A mutations in proviral DNA from A3G transgenic UNG-/-APO-/- than A3G transgenic APO-/- mice, suggesting that UNG plays a role in the repair of uracil-containing proviruses. In in vitro studies, we found that cytoplasmic viral DNA deaminated by APOBEC3G was uracilated. In the absence of UNG, the uracil-containing proviruses integrated at higher levels into the genome than those made in the presence of UNG. Thus, UNG also functions in the nucleus prior to integration by nicking uracil-containing viral DNA, thereby blocking integration. These data show that UNG plays a critical role in the repair of the damage inflicted by APOBEC3 deamination of reverse-transcribed DNA. IMPORTANCE While APOBEC3-mediated mutation of retroviruses is well-established, what role the host base excision repair enzymes play in correcting these mutations is not clear. This question is especially difficult to address in vivo. Here, we use a transgenic mouse developed by our lab that expresses human APOBEC3G and also lacks the endogenous uracil DNA glycosylase (Ung) gene and show that UNG removes uracils introduced by this cytidine deaminase in MLV reverse transcripts, thereby reducing G-to-A mutations in proviruses. Furthermore, our data suggest that UNG removes uracils at two stages in infection-first, in unintegrated nuclear viral reverse-transcribed DNA, resulting in its degradation; and second, in integrated proviruses, resulting in their repair. These data suggest that retroviruses damaged by host cytidine deaminases take advantage of the host DNA repair system to overcome this damage.


Subject(s)
APOBEC-3G Deaminase/immunology , DNA, Viral/immunology , Retroviridae Infections , Retroviridae , Uracil-DNA Glycosidase/immunology , Animals , DNA Repair , HEK293 Cells , Humans , Mice , Mice, Knockout , NIH 3T3 Cells , Retroviridae/genetics , Retroviridae/immunology , Retroviridae Infections/immunology , Retroviridae Infections/virology
SELECTION OF CITATIONS
SEARCH DETAIL
...