Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
Add more filters










Publication year range
1.
FASEB J ; 38(10): e23691, 2024 May 31.
Article in English | MEDLINE | ID: mdl-38780525

ABSTRACT

Heme is a prosthetic group of proteins involved in vital physiological processes. It participates, for example, in redox reactions crucial for cell metabolism due to the variable oxidation state of its central iron atom. However, excessive heme can be cytotoxic due to its prooxidant properties. Therefore, the control of intracellular heme levels ensures the survival of organisms, especially those that deal with high concentrations of heme during their lives, such as hematophagous insects. The export of heme initially attributed to the feline leukemia virus C receptor (FLVCR) has recently been called into question, following the discovery of choline uptake by the same receptor in mammals. Here, we found that RpFLVCR is a heme exporter in the midgut of the hematophagous insect Rhodnius prolixus, a vector for Chagas disease. Silencing RpFLVCR decreased hemolymphatic heme levels and increased the levels of intracellular dicysteinyl-biliverdin, indicating heme retention inside midgut cells. FLVCR silencing led to increased expression of heme oxygenase (HO), ferritin, and mitoferrin mRNAs while downregulating the iron importers Malvolio 1 and 2. In contrast, HO gene silencing increased FLVCR and Malvolio expression and downregulated ferritin, revealing crosstalk between heme degradation/export and iron transport/storage pathways. Furthermore, RpFLVCR silencing strongly increased oxidant production and lipid peroxidation, reduced cytochrome c oxidase activity, and activated mitochondrial biogenesis, effects not observed in RpHO-silenced insects. These data support FLVCR function as a heme exporter, playing a pivotal role in heme/iron metabolism and maintenance of redox balance, especially in an organism adapted to face extremely high concentrations of heme.


Subject(s)
Heme , Mitochondria , Oxidation-Reduction , Rhodnius , Animals , Heme/metabolism , Rhodnius/metabolism , Mitochondria/metabolism , Receptors, Virus/metabolism , Receptors, Virus/genetics , Leukemia Virus, Feline/metabolism , Insect Proteins/metabolism , Insect Proteins/genetics
2.
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
3.
Retrovirology ; 21(1): 3, 2024 Feb 12.
Article in English | MEDLINE | ID: mdl-38347535

ABSTRACT

Endogenous retroviruses (ERV) are indicators of vertebrate evolutionary history and play important roles as homeostatic regulators. ERV long terminal repeat (LTR) elements may act as cis-activating promoters or trans-activating enhancer elements modifying gene transcription distant from LTR insertion sites. We previously documented that endogenous feline leukemia virus (FeLV)-LTR copy number variation in individual cats tracks inversely with susceptibility to virulent FeLV disease. To evaluate FeLV-LTR insertion characteristics, we assessed enFeLV-LTR integration site diversity in 20 cats from three genetically distinct populations using a baited linker-mediated PCR approach. We documented 765 individual integration sites unequally represented among individuals. Only three LTR integration sites were shared among all individuals, while 412 sites were unique to a single individual. When primary fibroblast cultures were challenged with exogenous FeLV, we found significantly increased expression of both exogenous and endogenous FeLV orthologs, supporting previous findings of potential exFeLV-enFeLV interactions; however, viral challenge did not elicit transcriptional changes in genes associated with the vast majority of integration sites. This study assesses FeLV-LTR integration sites in individual animals, providing unique transposome genotypes. Further, we document substantial individual variation in LTR integration site locations, even in a highly inbred population, and provide a framework for understanding potential endogenous retroviral element position influence on host gene transcription.


Subject(s)
Endogenous Retroviruses , Leukemia, Feline , Humans , Animals , Cats , Leukemia Virus, Feline/genetics , Leukemia Virus, Feline/metabolism , DNA Copy Number Variations , Terminal Repeat Sequences , Endogenous Retroviruses/genetics , Promoter Regions, Genetic , Leukemia, Feline/genetics
4.
N Z Vet J ; 71(1): 1-7, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36178295

ABSTRACT

Feline leukaemia virus (FeLV) is a retrovirus that infects domestic and wild cats around the world. FeLV infection is associated with the development of neoplasms, bone marrow disorders and immunosuppression. Viral subgroups arise from mutations in the FeLV genome or from recombination of FeLV with ancestral endogenous retroviruses in the cat genome. The retroviral endogenisation process has allowed generation of a diversity of endogenous viruses, both functional and defective. These elements may be part of the normal functioning of the feline genome and may also interact with FeLV to form recombinant FeLV subgroups, enhance pathogenicity of viral subgroups, or inhibit and/or regulate other retroviral infections. Recombination of the env gene occurs most frequently and appears to be the most significant in terms of both the quantity and diversification of pathogenic effects in the viral population, as well as affecting cell tropism and types of disease that occur in infected cats. This review focuses on available information regarding genetic diversity, pathogenesis and diagnosis of FeLV as a result of the interaction between endogenous and exogenous viruses.


Subject(s)
Cat Diseases , Endogenous Retroviruses , Leukemia, Feline , Retroviridae Infections , Cats , Animals , Leukemia Virus, Feline/genetics , Leukemia Virus, Feline/metabolism , Endogenous Retroviruses/genetics , Leukemia, Feline/genetics , Genes, env , Retroviridae Infections/veterinary , Retroviridae Infections/genetics , Cat Diseases/genetics
5.
J Virol ; 95(23): e0007021, 2021 11 09.
Article in English | MEDLINE | ID: mdl-34495702

ABSTRACT

Endogenous retroviruses (ERVs) are increasingly recognized for biological impacts on host cell function and susceptibility to infectious agents, particularly in relation to interactions with exogenous retroviral progenitors (XRVs). ERVs can simultaneously promote and restrict XRV infections using mechanisms that are virus and host specific. The majority of endogenous-exogenous retroviral interactions have been evaluated in experimental mouse or chicken systems, which are limited in their ability to extend findings to naturally infected outbred animals. Feline leukemia virus (FeLV) has a relatively well-characterized endogenous retrovirus with a coexisting virulent exogenous counterpart and is endemic worldwide in domestic cats. We have previously documented an association between endogenous FeLV (enFeLV) long terminal repeat (LTR) copy number and abrogated exogenous FeLV in naturally infected cats and experimental infections in tissue culture. Analyses described here examine limited FeLV replication in experimentally infected peripheral blood mononuclear cells, which correlates with higher enFeLV transcripts in these cells compared to fibroblasts. We further examine NCBI Sequence Read Archive RNA transcripts to evaluate enFeLV transcripts and RNA interference (RNAi) precursors. We find that lymphoid-derived tissues, which are experimentally less permissive to exogenous FeLV infection, transcribe higher levels of enFeLV under basal conditions. Transcription of enFeLV-LTR segments is significantly greater than that of other enFeLV genes. We documented transcription of a 21-nucleotide (nt) microRNA (miRNA) just 3' to the enFeLV 5'-LTR in the feline miRNAome of all data sets evaluated (n = 27). Our findings point to important biological functions of enFeLV transcription linked to solo LTRs distributed within the domestic cat genome, with potential impacts on domestic cat exogenous FeLV susceptibility and pathogenesis. IMPORTANCE Endogenous retroviruses (ERVs) are increasingly implicated in host cellular processes and susceptibility to infectious agents, specifically regarding interactions with exogenous retroviral progenitors (XRVs). Exogenous feline leukemia virus (FeLV) and its endogenous counterpart (enFeLV) represent a well-characterized, naturally occurring XRV-ERV dyad. We have previously documented an abrogated FeLV infection in both naturally infected cats and experimental fibroblast infections that harbor higher enFeLV proviral loads. Using an in silico approach, we provide evidence of miRNA transcription that is produced in tissues that are most important for FeLV infection, replication, and transmission. Our findings point to important biological functions of enFeLV transcription linked to solo-LTRs distributed within the feline genome, with potential impacts on domestic cat exogenous FeLV susceptibility and pathogenesis. This body of work provides additional evidence of RNA interference (RNAi) as a mechanism of viral interference and is a demonstration of ERV exaptation by the host to defend against related XRVs.


Subject(s)
Leukemia Virus, Feline/genetics , Leukemia Virus, Feline/metabolism , Leukemia, Feline/virology , RNA, Small Interfering/metabolism , RNA, Viral/genetics , Animals , Cats/genetics , Endogenous Retroviruses , Fibroblasts , Leukocytes, Mononuclear , Lymphoid Tissue , Mice , MicroRNAs , RNA, Small Interfering/genetics , Terminal Repeat Sequences , Transcriptome , Virus Replication
6.
J Virol ; 94(21)2020 10 14.
Article in English | MEDLINE | ID: mdl-32817213

ABSTRACT

While feline leukemia virus (FeLV) has been shown to infect felid species other than the endemic domestic cat host, differences in FeLV susceptibility among species has not been evaluated. Previous reports have noted a negative correlation between endogenous FeLV (enFeLV) copy number and exogenous FeLV (exFeLV) infection outcomes in domestic cats. Since felids outside the genus Felis do not harbor enFeLV genomes, we hypothesized absence of enFeLV results in more severe disease consequences in felid species lacking these genomic elements. We infected primary fibroblasts isolated from domestic cats (Felis catus) and pumas (Puma concolor) with FeLV and quantitated proviral and viral antigen loads. Domestic cat enFeLV env and long terminal repeat (LTR) copy numbers were determined for each individual and compared to FeLV viral outcomes. FeLV proviral and antigen levels were also measured in 6 naturally infected domestic cats and 11 naturally infected Florida panthers (P. concolor coryi). We demonstrated that puma fibroblasts are more permissive to FeLV than domestic cat cells, and domestic cat FeLV restriction was highly related to enFeLV-LTR copy number. Terminal tissues from FeLV-infected Florida panthers and domestic cats had similar exFeLV proviral copy numbers, but Florida panther tissues have higher FeLV antigen loads. Our work indicates that enFeLV-LTR elements negatively correlate with exogenous FeLV replication. Further, Puma concolor samples lacking enFeLV are more permissive to FeLV infection than domestic cat samples, suggesting that endogenization can play a beneficial role in mitigating exogenous retroviral infections. Conversely, presence of endogenous retroelements may relate to new host susceptibility during viral spillover events.IMPORTANCE Feline leukemia virus (FeLV) can infect a variety of felid species. Only the primary domestic cat host and related small cat species harbor a related endogenous virus in their genomes. Previous studies noted a negative association between the endogenous virus copy number and exogenous virus infection in domestic cats. This report shows that puma cells, which lack endogenous FeLV, produce more virus more rapidly than domestic cat fibroblasts following cell culture challenge. We document a strong association between domestic cat cell susceptibility and FeLV long terminal repeat (LTR) copy number, similar to observations in natural FeLV infections. Viral replication does not, however, correlate with FeLV env copy number, suggesting that this effect is specific to FeLV-LTR elements. This discovery indicates a protective capacity of the endogenous virus against the exogenous form, either via direct interference or indirectly via gene regulation, and may suggest evolutionary outcomes of retroviral endogenization.


Subject(s)
DNA Copy Number Variations , Gene Products, env/genetics , Leukemia Virus, Feline/genetics , Leukemia Virus, Feline/pathogenicity , Leukemia, Feline/virology , Puma/virology , Animals , Bone Marrow/pathology , Bone Marrow/virology , Cats , Female , Fibroblasts/pathology , Fibroblasts/virology , Gene Products, env/metabolism , Host Specificity , Leukemia Virus, Feline/metabolism , Leukemia, Feline/pathology , Lymph Nodes/pathology , Lymph Nodes/virology , Male , Primary Cell Culture , Spleen/pathology , Spleen/virology , Terminal Repeat Sequences , Thymus Gland/pathology , Thymus Gland/virology , Viral Load , Virus Replication/genetics
7.
J Virol ; 93(13)2019 07 01.
Article in English | MEDLINE | ID: mdl-30996094

ABSTRACT

Feline leukemia virus (FeLV) is horizontally transmitted among cats and causes a variety of hematopoietic disorders. Five subgroups of FeLV, A to D and T, each with distinct receptor usages, have been described. Recently, we identified a new FeLV Env (TG35-2) gene from a pseudotyped virus that does not belong to any known subgroup. FeLV-A is the primary virus from which other subgroups have emerged via mutation or recombination of the subgroup A env gene. Retrovirus entry into cells is mediated by the interaction of envelope protein (Env) with specific cell surface receptors. Here, phenotypic screening of a human/hamster radiation hybrid panel identified SLC19A1, a feline reduced folate carrier (RFC) and potential receptor for TG35-2-phenotypic virus. RFC is a multipass transmembrane protein. Feline and human RFC cDNAs conferred susceptibility to TG35-2-pseudotyped virus when introduced into nonpermissive cells but did not render these cells permissive to other FeLV subgroups or feline endogenous retrovirus. Moreover, human cells with genomic deletion of RFC were nonpermissive for TG35-2-pseudotyped virus infection, but the introduction of feline and human cDNAs rendered them permissive. Mutation analysis of FeLV Env demonstrated that amino acid substitutions within variable region A altered the specificity of the Env-receptor interaction. We isolated and reconstructed the full-length infectious TG35-2-phenotypic provirus from a naturally FeLV-infected cat, from which the FeLV Env (TG35-2) gene was previously isolated, and compared the replication of the virus in hematopoietic cell lines with that of FeLV-A 61E by measuring the viral RNA copy numbers. These results provide a tool for further investigation of FeLV infectious disease.IMPORTANCE Feline leukemia virus (FeLV) is a member of the genus Gammaretrovirus, which causes malignant diseases in cats. The most prevalent FeLV among cats is FeLV subgroup A (FeLV-A), and specific binding of FeLV-A Env to its viral receptor, thiamine transporter feTHTR1, is the first step of infection. In infected cats, novel variants of FeLV with altered receptor specificity for viral entry have emerged by mutation or recombination of the env gene. A novel FeLV variant arose from a subtle mutation of FeLV-A Env, which altered the specific interaction of the virus with its receptor. RFC, a folate transporter, is a potential receptor for the novel FeLV variant. The perturbation of specific retrovirus-receptor interactions under selective pressure by the host results in the emergence of novel viruses.


Subject(s)
Genes, env/genetics , Leukemia Virus, Feline/genetics , Receptors, Virus/genetics , Reduced Folate Carrier Protein/genetics , Viral Envelope Proteins/genetics , Virus Internalization , Amino Acid Sequence , Animals , Cats , Cell Line , Cricetinae , Endogenous Retroviruses/metabolism , Gene Products, env/genetics , HeLa Cells , Humans , Leukemia Virus, Feline/metabolism , Leukemia, Feline/virology , Phylogeny , Proviruses , RNA, Viral/genetics , Receptors, Virus/metabolism , Reduced Folate Carrier Protein/classification , Reduced Folate Carrier Protein/metabolism , Sequence Alignment , Virus Replication
8.
Biologicals ; 49: 76-80, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28734742

ABSTRACT

The purpose of this study was to compare the efficacy of three FeLV vaccines, under identical conditions in a laboratory challenge model that closely mimics natural infection. Four groups of cats (n = 20 per group) were administered two doses of vaccine, 21 days apart, starting at 9-10 weeks of age (Purevax® FeLV, Versifel® FeLV, Nobivac® feline 2-FeLV, and a placebo). Cats were challenged 3 weeks later with a virulent, heterologous FeLV isolate. FeLV antigenemia was determined at weekly intervals from 3 to 15 weeks postchallenge. Circulating proviral DNA was determined on terminal PBMC samples. Following challenge, persistent antigenemia developed in 15 (75%) placebo-vaccinated cats, 3 (15%) cats in the Versifel FeLV vaccinated group, and 1 cat (5%) each in the Purevax FeLV and the Nobivac FeLV vaccinated groups. The prevented fractions for three vaccine groups were 93%, 93%, and 80% respectively. The adjusted p-values for all vaccine group comparisons fail to approach statistical significance. There was excellent agreement between proviral FeLV DNA in circulating PBMCs and persistent antigenemia. It is shown that when cats are managed under the same conditions during a virulent challenge, via the normal route of infection, the tested vaccines all show a comparable degree of protection.


Subject(s)
Feline Acquired Immunodeficiency Syndrome , Leukemia Virus, Feline/immunology , Leukocytes, Mononuclear , Viral Vaccines/pharmacology , Animals , Cats , DNA, Viral/blood , DNA, Viral/immunology , Feline Acquired Immunodeficiency Syndrome/blood , Feline Acquired Immunodeficiency Syndrome/immunology , Feline Acquired Immunodeficiency Syndrome/prevention & control , Female , Leukemia Virus, Feline/metabolism , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Male , Vaccines, Inactivated/immunology , Vaccines, Inactivated/pharmacology , Viral Vaccines/immunology
9.
Arch Virol ; 162(4): 1031-1036, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28005210

ABSTRACT

Oncogene-containing retroviruses are generated by recombination events between viral and cellular sequences, a phenomenon called "oncogene capture". The captured cellular genes, referred to as "v-onc" genes, then acquire new oncogenic properties. We report a novel feline leukemia virus (FeLV), designated "FeLV-AKT", that has captured feline c-AKT1 in feline lymphoma. FeLV-AKT contains a gag-AKT fusion gene that encodes the myristoylated Gag matrix protein and the kinase domain of feline c-AKT1, but not its pleckstrin homology domain. Therefore, it differs structurally from the v-Akt gene of murine retrovirus AKT8. AKT may be involved in the mechanisms underlying malignant diseases in cats.


Subject(s)
Cat Diseases/genetics , Leukemia Virus, Feline/genetics , Proto-Oncogene Proteins c-akt/genetics , Recombination, Genetic , Retroviridae Infections/veterinary , Tumor Virus Infections/veterinary , Animals , Cat Diseases/enzymology , Cat Diseases/virology , Cats , Leukemia Virus, Feline/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Retroviridae Infections/enzymology , Retroviridae Infections/genetics , Retroviridae Infections/virology , Tumor Virus Infections/enzymology , Tumor Virus Infections/genetics , Tumor Virus Infections/virology
10.
BMC Vet Res ; 11: 90, 2015 Apr 10.
Article in English | MEDLINE | ID: mdl-25879730

ABSTRACT

BACKGROUND: Cats infected with exogenous feline leukemia virus (exFeLV) have a higher chance of lymphoma development than uninfected cats. Furthermore, an increased exFeLV transcription has been detected in lymphomas compared to non-malignant tissues. The possible mechanisms of lymphoma development by exFeLV are insertional mutagenesis or persistent stimulation of host immune cells by viral antigens, bringing them at risk for malignant transformation. Vaccination of cats against exFeLV has in recent years decreased the overall infection rate in most countries. Nevertheless, an increasing number of lymphomas have been diagnosed among exFeLV-negative cats. Endogenous feline leukemia virus (enFeLV) is another retrovirus for which transcription has been observed in cat lymphomas. EnFeLV provirus elements are present in the germline of various cat species and share a high sequence similarity with exFeLV but, due to mutations, are incapable of producing infectious viral particles. However, recombination between exFeLV and enFeLV could produce infectious particles. RESULTS: We examined the FeLV expression in cats that have developed malignant lymphomas and discussed the possible mechanisms that could have induced malignant transformation. For expression analysis we used next-generation RNA-sequencing (RNA-Seq) and for validation reverse transcription quantitative PCR (RT-qPCR). First, we showed that there was no expression of exFeLV in all samples, which eliminates the possibility of recombination between exFeLV and enFeLV. Next, we analyzed the difference in expression of three enFeLV genes between control and lymphoma samples. Our analysis showed an average of 3.40-fold decreased viral expression for the three genes in lymphoma compared to control samples. The results were confirmed by RT-qPCR. CONCLUSIONS: There is a decreased expression of enFeLV genes in lymphomas versus control samples, which contradicts previous observations for the exFeLV. Our results suggest that a persistent stimulation of host immune cells is not an appropriate mechanism responsible for malignant transformation caused by feline endogenous retroviruses.


Subject(s)
Cat Diseases/virology , Gene Expression Regulation, Viral/physiology , Leukemia Virus, Feline/metabolism , Lymphoma/veterinary , Retroviridae Infections/veterinary , Tumor Virus Infections/veterinary , Animals , Base Sequence , Case-Control Studies , Cat Diseases/pathology , Cats , Female , Leukemia Virus, Feline/genetics , Lymphoma/virology , Male , RNA, Viral/genetics , RNA, Viral/metabolism , Retroviridae Infections/virology , Reverse Transcriptase Polymerase Chain Reaction , Tumor Virus Infections/virology
11.
Virology ; 475: 110-9, 2015 Jan 15.
Article in English | MEDLINE | ID: mdl-25462351

ABSTRACT

Previously, we found that mutation of glutamine to proline in the endoproteolytic cleavage signal of the PERV-C envelope (RQKK to RPKK) resulted in non-infectious vectors. Here, we show that RPKK results in a non-infectious vector when placed in not only a PERV envelope, but also the envelope of a related gammaretrovirus, FeLV-B. The amino acid substitutions do not prevent envelope precursor cleavage, viral core and genome assembly, or receptor binding. Rather, the mutations result in the formation of hyperglycosylated glycoprotein and a reduction in the reverse transcribed minus strand synthesis and undetectable 2-LTR circular DNA in cells exposed to vectors with these mutated envelopes. Our findings suggest novel functions associated with the cleavage signal sequence that may affect trafficking through the glycosylation machinery of the cell. Further, the glycosylation status of the envelope appears to impact post-binding events of the viral life cycle, either membrane fusion, internalization, or reverse transcription.


Subject(s)
Endogenous Retroviruses/metabolism , Gene Expression Regulation, Viral/physiology , Glycoproteins/metabolism , Leukemia Virus, Feline/metabolism , Viral Envelope Proteins/metabolism , Virus Replication/physiology , Animals , Cell Line , DNA, Viral/biosynthesis , Endogenous Retroviruses/genetics , Glycoproteins/genetics , Leukemia Virus, Feline/genetics , Mutation , Protein Binding , Swine , Viral Envelope Proteins/genetics , Virus Replication/genetics
12.
Mol Ther ; 18(7): 1310-7, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20372106

ABSTRACT

Lentiviral vectors are useful for transducing primitive hematopoietic cells. We examined four envelope proteins for their ability to mediate lentiviral transduction of mobilized human CD34(+) peripheral blood cells. Lentiviral particles encoding green fluorescent protein (GFP) were pseudotyped with the vesicular stomatitis virus envelope glycoprotein (VSV-G), the amphotropic (AMPHO) murine leukemia virus envelope protein, the endogenous feline leukemia viral envelope protein or the feline leukemia virus type C envelope protein. Because the relative amount of genome RNA per ml was similar for each pseudotype, we transduced CD34(+) cells with a fixed volume of each vector preparation. Following an overnight transduction, CD34(+) cells were transplanted into immunodeficient mice which were sacrificed 12 weeks later. The average percentages of engrafted human CD45(+) cells in total bone marrow were comparable to that of the control, mock-transduced group (37-45%). Lenti-particles pseudotyped with the VSV-G envelope protein transduced engrafting cells two- to tenfold better than particles pseudotyped with any of the gamma-retroviral envelope proteins. There was no correlation between receptor mRNA levels for the gamma-retroviral vectors and transduction efficiency of primitive hematopoietic cells. These results support the use of the VSV-G envelope protein for the development of lentiviral producer cell lines for manufacture of clinical-grade vector.


Subject(s)
Genetic Vectors/genetics , Hematopoietic Stem Cells/metabolism , Lentivirus/genetics , Transduction, Genetic/methods , Animals , Antigens, CD34/metabolism , Cell Line , HeLa Cells , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/cytology , Humans , Leukemia Virus, Feline/metabolism , Leukemia Virus, Murine/metabolism , Membrane Glycoproteins/genetics , Mice , Viral Envelope Proteins/genetics
13.
Gene Ther ; 17(6): 799-804, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20237508

ABSTRACT

Adverse events linked to perturbations of cellular genes by vector insertion reported in gene therapy trials and animal models have prompted attempts to better understand the mechanisms directing viral vector integration. The integration profiles of vectors based on MLV, ASLV, SIV and HIV have all been shown to be non-random, and novel vectors with a safer integration pattern have been sought. Recently, we developed a producer cell line called CatPac that packages standard MoMLV vectors with feline leukemia virus (FeLV) gag, pol and env gene products. We now report the integration profile of this vector, asking if the FeLV integrase and capsid proteins could modify the MoMLV integration profile, potentially resulting in a less genotoxic pattern. We transduced rhesus macaque CD34+ hematopoietic progenitor cells with CatPac or standard MoMLV vectors, and determined their integration profile by LAM-PCR. We obtained 184 and 175 unique integration sites (ISs) respectively for CatPac and standard MoMLV vectors, and these were compared with 10 000 in silico-generated random IS. The integration profile for CatPac vector was similar to MoMLV and equally non-random, with a propensity for integration near transcription start sites and in highly dense gene regions. We found an IS for CatPac vector localized 715 nucleotides upstream of LMO-2, the gene involved in the acute lymphoblastic leukemia developed by X-SCID patients treated by gene therapy using MoMLV vectors. In conclusion, we found that replacement of MoMLV env, gag and pol gene products with FeLV did not alter the basic integration profile. Thus, there appears to be no safety advantage for this packaging system. However, considering the stability and efficacy of CatPac vectors, further development is warranted, using potentially safer vector backbones, for instance those with a SIN configuration.


Subject(s)
Gene Transfer Techniques/adverse effects , Genetic Vectors/adverse effects , Hematopoietic Stem Cells/virology , Integrases/genetics , Leukemia Virus, Feline/genetics , Moloney murine leukemia virus/genetics , Virus Integration , Animals , Capsid , Capsid Proteins/genetics , Leukemia Virus, Feline/metabolism , Macaca mulatta , Transduction, Genetic
14.
J Virol ; 81(13): 7274-9, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17428873

ABSTRACT

Infection with a recombinant murine-feline gammaretrovirus, MoFe2, or with the parent virus, Moloney murine leukemia virus, caused significant reduction in B-lymphoid differentiation of bone marrow at 2 to 8 weeks postinfection. The suppression was selective, in that myeloid potential was significantly increased by infection. Analysis of cell surface markers and immunoglobulin H gene rearrangements in an in vitro model demonstrated normal B-lymphoid differentiation after infection but significantly reduced viability of differentiating cells. This reduction in viability may confer a selective advantage on undifferentiated lymphoid progenitors in the bone marrow of gammaretrovirus-infected animals and thereby contribute to the establishment of a premalignant state.


Subject(s)
B-Lymphocytes/metabolism , Cell Differentiation , Leukemia Virus, Feline/metabolism , Moloney murine leukemia virus/metabolism , Myeloid Progenitor Cells/metabolism , Retroviridae Infections/metabolism , Tumor Virus Infections/mortality , Animals , B-Lymphocytes/pathology , B-Lymphocytes/virology , Bone Marrow/metabolism , Bone Marrow/pathology , Bone Marrow/virology , Cats , Hematopoiesis, Extramedullary , Mice , Myeloid Progenitor Cells/pathology , Myeloid Progenitor Cells/virology , Retroviridae Infections/pathology , Tumor Virus Infections/pathology
15.
Virology ; 351(2): 340-8, 2006 Aug 01.
Article in English | MEDLINE | ID: mdl-16678875

ABSTRACT

Libraries of feline leukemia virus subgroup A (FeLV-A)-derived envelope (Env) proteins with random peptides incorporated into the cell-targeting region were screened for productive gene delivery to the PC-3 human prostate cell line. In order to increase the efficiency of recovering and testing functional clones, the screen was performed in the presence of a replication-competent 4070A Env-expressing virus under conditions of viral interference. The Env proteins resulting from this library screen were able to mediate gene delivery to 4070A-infected human PC-3, DU145 prostate and TE671 rhabdomyosarcoma cells in the presence, but not absence, of 4070A helper virus. FeLV-A, FeLV-B and Moloney murine leukemia virus (Mo-MuLV) Env proteins were unable to substitute for 4070A Env. Flow cytometry and Western blot analyses indicated increased cell-surface expression and virion incorporation of library-derived Env proteins in the presence of 4070A Env. Interference assays on cells infected with both 4070A and FeLV-B are consistent with the combination of library-derived and 4070A Env proteins utilizing the Pit1 receptor.


Subject(s)
Leukemia Virus, Feline/metabolism , Leukemia Virus, Murine/metabolism , Peptide Library , Viral Envelope Proteins/metabolism , Animals , Cats , Cell Line , Humans , Leukemia Virus, Feline/genetics , Leukemia Virus, Murine/genetics , Viral Envelope Proteins/genetics
16.
Vet Microbiol ; 109(3-4): 191-9, 2005 Aug 30.
Article in English | MEDLINE | ID: mdl-16023797

ABSTRACT

In a previous study, it was found that even though more male cats were infected by feline leukaemia virus (FeLV), females seemed to progress easier to overt disease. To study the effect of female hormones, 17beta-estradiol and progesterone were added in different concentrations (10(-3) M to 10(-12) M) to a culture of persistently FeLV-infected cells. The effect of both hormones was very similar. After 24 h the cell viability was very low at 10(-3) M and 10(-4) M but similar to controls at the remaining concentrations. Liberation of viral particles was estimated by the reverse transcriptase activity (RT), which was the lowest also at 10(-3) M and 10(-4) M. However, low viability could not account for this low RT, as when cells were lysed with lysis buffer RT was high. Thus, cells were dying without freeing viral particles, suggestive of apoptosis. This possibility was confirmed by staining hormone-treated cells with annexin V and propidium iodide. The FeLV antigen p27 measured in the cultures had a maximum at 10(-3) M and 10(-4) M, higher than controls and lysed cells, so the presence of p27 in the supernatant was not only due to cell lysis but a consequence of hormone effect. In conclusion, 17beta-estradiol and progesterone induce death of FeLV-infected cells at high concentrations, probably through a process of apoptosis, which might limit the spread of the infection, as infective viral particles would be hampered from budding.


Subject(s)
Cat Diseases/virology , Estradiol/pharmacology , Leukemia Virus, Feline/drug effects , Progesterone/pharmacology , Retroviridae Infections/veterinary , Tumor Virus Infections/veterinary , Animals , Apoptosis/drug effects , Cats , Cell Line , Female , Flow Cytometry/veterinary , Gene Products, gag/biosynthesis , Leukemia Virus, Feline/metabolism , Male , Retroviridae Infections/drug therapy , Retroviridae Infections/virology , Retroviridae Proteins/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction/veterinary , Tumor Virus Infections/drug therapy , Tumor Virus Infections/virology
17.
Virology ; 332(1): 272-83, 2005 Feb 05.
Article in English | MEDLINE | ID: mdl-15661159

ABSTRACT

The mechanisms responsible for effective vs. ineffective viral containment are central to immunoprevention and therapies of retroviral infections. Feline leukemia virus (FeLV) infection is unique as a naturally occurring, diametric example of effective vs. ineffective retroviral containment by the host. We developed a sensitive quantitative real-time DNA PCR assay specific for exogenous FeLV to further explore the FeLV-host relationship. By assaying p27 capsid antigen in blood and FeLV DNA in blood and tissues of successfully vaccinated, unsuccessfully vaccinated, and unvaccinated pathogen-free cats, we defined four statistically separable classes of FeLV infection, provisionally designated as abortive, regressive, latent, and progressive. These host-virus relationships were established by 8 weeks post-challenge and could be maintained for years. Real-time PCR methods offer promise in gaining deeper insight into the mechanisms of FeLV infection and immunity.


Subject(s)
Cat Diseases/virology , Leukemia Virus, Feline/metabolism , Polymerase Chain Reaction/methods , Vaccination/veterinary , Viral Vaccines/immunology , Animals , Cats , Leukemia Virus, Feline/genetics , Proliferating Cell Nuclear Antigen/immunology , RNA, Viral/analysis , RNA, Viral/isolation & purification , Retrospective Studies , Sensitivity and Specificity
18.
Virus Res ; 98(1): 1-15, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14609625

ABSTRACT

The host range of swinepox virus (SPV) is restricted to swine, although SPV has been shown to infect mammalian, non-swine cells, without recovery of infectious virus. SPV is a reasonable candidate for development as a non-productively replicating viral vector for use in non-swine, mammalian species, such as the cat. A novel SPV gene deletion (SPV 043) was created and found to be non-attenuating. This deletion was utilized to generate a stable recombinant virus expressing the Gag-Pro and Env proteins of feline leukemia virus (FeLV). Expression and replication of this vector was studied in embryonic swine kidney cells (ESK-4), and two feline cell lines, Crandell feline kidney cells (CRFK) and feline skin fibroblasts (FSF). Our results showed that feline cells were susceptible to infection by SPV and supported expression of foreign genes driven by synthetic poxvirus promoters, however, SPV viral DNA was not replicated in feline cells and infectious virus was not recovered. In addition, FeLV Gag virus-like particles were produced from both ESK-4 and CRFK cells and foreign antigens were incorporated into infectious SPV intracellular mature virions (IMV). These results suggest that SPV may have potential as a safe vaccine delivery vector for cats.


Subject(s)
Gene Products, env/metabolism , Gene Products, gag/metabolism , Genetic Vectors , Leukemia Virus, Feline/genetics , Virus Replication , Animals , Base Sequence , Cats , Cell Line , Gene Products, env/genetics , Gene Products, gag/genetics , Leukemia Virus, Feline/metabolism , Molecular Sequence Data , Suipoxvirus/genetics , Suipoxvirus/physiology , Swine , Virion/metabolism
19.
J Virol ; 77(4): 2717-29, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12552012

ABSTRACT

Infection of T lymphocytes by the cytopathic retrovirus feline leukemia virus subgroup T (FeLV-T) requires FeLIX, a cellular coreceptor that is encoded by an endogenous provirus and closely resembles the receptor-binding domain (RBD) of feline leukemia virus subgroup B (FeLV-B). We determined the structure of FeLV-B RBD, which has FeLIX activity, to a 2.5-A resolution by X-ray crystallography. The structure of the receptor-specific subdomain of this glycoprotein differs dramatically from that of Friend murine leukemia virus (Fr-MLV), which binds a different cell surface receptor. Remarkably, we find that Fr-MLV RBD also activates FeLV-T infection of cells expressing the Fr-MLV receptor and that FeLV-B RBD is a competitive inhibitor of infection under these conditions. These studies suggest that FeLV-T infection relies on the following property of mammalian leukemia virus RBDs: the ability to couple interaction with one of a variety of receptors to the activation of a conserved membrane fusion mechanism. A comparison of the FeLV-B and Fr-MLV RBD structures illustrates how receptor-specific regions are linked to conserved elements critical for postbinding events in virus entry.


Subject(s)
Leukemia Virus, Feline/pathogenicity , Receptors, Virus/chemistry , Receptors, Virus/metabolism , Amino Acid Sequence , Animals , Cats , Cell Line , Cricetinae , Crystallization , Crystallography, X-Ray , Glycoproteins/chemistry , Humans , Leukemia Virus, Feline/metabolism , Mice , Models, Molecular , Molecular Sequence Data , Sequence Alignment , Viral Envelope Proteins/chemistry
20.
J Virol ; 76(15): 7683-93, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12097582

ABSTRACT

The mammalian gammaretroviruses gibbon ape leukemia virus (GALV) and feline leukemia virus subgroup B (FeLV-B) can use the same receptor, Pit1, to infect human cells. A highly polymorphic nine-residue sequence within Pit1, designated region A, has been proposed as the virus binding site, because mutations in this region abolish Pit1-mediated cellular infection by GALV and FeLV-B. However, a direct correlation between region A mutations deleterious for infection and loss of virus binding has not been established. We report that cells expressing a Pit1 protein harboring mutations in region A that abolish receptor function retain the ability to bind virus, indicating that Pit1 region A is not the virus binding site. Furthermore, we have now identified a second region in Pit1, comprising residues 232 to 260 (region B), that is required for both viral entry and virus binding. Epitope-tagged Pit1 proteins were used to demonstrate that mutations in region B result in improper orientation of Pit1 in the cell membrane. Compensatory mutations in region A can restore proper orientation and full receptor function to these region B mutants. Based on these results, we propose that region A of Pit1 confers competence for viral entry by influencing the topology of the authentic binding site in the membrane and hence its accessibility to a viral envelope protein. Based on glycosylation studies and results obtained by using N- and C-terminal epitope-tagged Pit1, region A and region B mutants, and the transmembrane helices predicted with the PHD PredictProtein algorithm, we propose a new Pit1 topology model.


Subject(s)
Leukemia Virus, Feline/pathogenicity , Leukemia Virus, Gibbon Ape/pathogenicity , Receptors, Virus/chemistry , Receptors, Virus/metabolism , Algorithms , Animals , Binding Sites , Cell Line , Cell Membrane/metabolism , Flow Cytometry , Gene Expression Regulation , Humans , Leukemia Virus, Feline/metabolism , Leukemia Virus, Gibbon Ape/metabolism , Mice , Models, Molecular , Mutation
SELECTION OF CITATIONS
SEARCH DETAIL
...