Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
1.
PLoS Med ; 11(4): e1001623, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24714396

ABSTRACT

Kumanan Wilson and colleagues explain how the rapid response to XMRV as a novel pathogen has highlighted some challenges pertaining to policy-making and editorial responsibilities. The impact on policy and the propagation of the initial scientific information may not cease if the evidence is disproven and retracted from the peer-reviewed literature, which creates a challenge for regulators and scientific journals. Please see later in the article for the Editors' Summary.


Subject(s)
Editorial Policies , Health Policy/legislation & jurisprudence , Policy Making , Public Health/legislation & jurisprudence , Retroviridae Infections/transmission , Xenotropic murine leukemia virus-related virus/physiology , Blood Donors , Humans , Retroviridae Infections/prevention & control , Retroviridae Infections/virology
2.
Biochem Biophys Res Commun ; 447(1): 216-22, 2014 Apr 25.
Article in English | MEDLINE | ID: mdl-24721431

ABSTRACT

Xenotropic murine leukemia virus-related virus (XMRV) is a novel gammaretrovirus that was originally isolated from human prostate cancer. It is now believed that XMRV is not the etiologic agent of prostate cancer. An analysis of murine leukemia virus (MLV) infection in various human cell lines revealed that prostate cancer cell lines are preferentially infected by XMRV, and this suggested that XMRV infection may confer some sort of growth advantage to prostate cancer cell lines. To examine this hypothesis, androgen-dependent LNCaP cells were infected with XMRV and tested for changes in certain cell growth properties. We found that XMRV-infected LNCaP cells can proliferate in the absence of the androgen dihydrotestosterone. Moreover, androgen receptor expression is significantly reduced in XMRV-infected LNCaP cells. Such alterations were not observed in uninfected and amphotropic MLV-infected LNCaP cells. This finding explains why prostate cancer cell lines are preferentially infected with XMRV.


Subject(s)
Androgens/pharmacology , Cell Proliferation/drug effects , Prostatic Neoplasms/virology , Xenotropic murine leukemia virus-related virus/physiology , Androgen Receptor Antagonists/pharmacology , Anilides/pharmacology , Animals , Cell Line, Tumor , Dihydrotestosterone/pharmacology , Humans , Male , Mice , Nitriles/pharmacology , Rats , Receptors, Androgen/biosynthesis , Receptors, Androgen/drug effects , Tosyl Compounds/pharmacology
3.
J Virol ; 87(21): 11525-37, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23966380

ABSTRACT

We previously identified two novel endogenous murine leukemia virus proviruses, PreXMRV-1 and PreXMRV-2, and showed that they most likely recombined during xenograft passaging of a human prostate tumor in mice to generate xenotropic murine leukemia virus-related virus (XMRV). To determine the recombination potential of PreXMRV-1 and PreXMRV-2, we examined the generation of replication-competent retroviruses (RCRs) over time in a cell culture system. We observed that either virus alone was noninfectious and the RNA transcripts of the viruses were undetectable in the blood and spleen of nude mice that carry them. To determine their potential to generate RCRs through recombination, we transfected PreXMRV-1 and PreXMRV-2 into 293T cells and used the virus produced to infect fresh cells; the presence of reverse transcriptase activity at 10 days postinfection indicated the presence of RCRs. Population sequencing of proviral DNA indicated that all RCRs contained the gag and 5' half of pol from PreXMRV-2 and the long terminal repeat, 3' half of pol (including integrase), and env from PreXMRV-1. All crossovers were within sequences of at least 9 identical nucleotides, and crossovers within each of two selected recombination zones of 415 nucleotides (nt) in the 5' untranslated region and 982 nt in pol were required to generate RCRs. A recombinant with the same genotype as XMRV was not detected, and our analysis indicates that the probability of generating an identical RCR is vanishingly small. In addition, the studies indicate that the process of RCR formation is primarily driven by selection for viable cis and trans elements from the parental proviruses.


Subject(s)
Recombination, Genetic , Virus Replication , Xenotropic murine leukemia virus-related virus/physiology , Animal Structures/virology , Animals , Cell Line , Crosses, Genetic , DNA, Viral/chemistry , DNA, Viral/genetics , Mice , Mice, Inbred BALB C , Mice, Nude , Molecular Sequence Data , Sequence Analysis, DNA , Viral Proteins/genetics , Xenotropic murine leukemia virus-related virus/genetics
4.
J Virol ; 87(18): 10094-104, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23843647

ABSTRACT

Xenotropic mouse leukemia viruses (X-MLVs) are broadly infectious for mammals except most of the classical strains of laboratory mice. These gammaretroviruses rely on the XPR1 receptor for entry, and the unique resistance of laboratory mice is due to two mutations in different putative XPR1 extracellular loops. Cells from avian species differ in susceptibility to X-MLVs, and 2 replacement mutations in the virus-resistant chicken XPR1 (K496Q and Q579E) distinguish it from the more permissive duck and quail receptors. These substitutions align with the two mutations that disable the laboratory mouse XPR1. Mutagenesis of the chicken and duck genes confirms that residues at both sites are critical for virus entry. Among 32 avian species, the 2 disabling XPR1 mutations are found together only in the chicken, an omnivorous, ground-dwelling fowl that was domesticated in India and/or Southeast Asia, which is also where X-MLV-infected house mice evolved. The receptor-disabling mutations are also present separately in 5 additional fowl and raptor species, all of which are native to areas of Asia populated by the virus-infected subspecies Mus musculus castaneus. Phylogenetic analysis showed that the avian XPR1 gene is under positive selection at sites implicated in receptor function, suggesting a defensive role for XPR1 in the avian lineage. Contact between bird species and virus-infected mice may thus have favored selection of mouse virus-resistant receptor orthologs in the birds, and our data suggest that similar receptor-disabling mutations were fixed in mammalian and avian species exposed to similar virus challenges.


Subject(s)
Receptors, G-Protein-Coupled/genetics , Receptors, Virus/genetics , Retroviridae Infections/genetics , Selection, Genetic , Virus Internalization , Xenotropic murine leukemia virus-related virus/physiology , Animals , Asia , Chickens , DNA Mutational Analysis , Disease Resistance , Ducks , Mice , Molecular Sequence Data , Poultry Diseases/genetics , Poultry Diseases/immunology , Receptors, G-Protein-Coupled/metabolism , Receptors, Virus/metabolism , Retroviridae Infections/immunology , Sequence Analysis, DNA , Xenotropic and Polytropic Retrovirus Receptor
5.
Appl Environ Microbiol ; 80(8): 2617-22, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24532072

ABSTRACT

Xenotropic murine leukemia virus-related virus (XMRV) represents a novel γ-retrovirus that is capable of infecting human cells and has been classified as a biosafety level 2 (BSL-2) organism. Hence, XMRV represents a potential risk for personnel in laboratories worldwide. Here, we measured the stability of XMRV and its susceptibility to alcohol-based disinfectants. To this end, we exposed an infectious XMRV reporter virus encoding a secretable luciferase to different temperatures, pH values, and disinfectants and infected XMRV-permissive Raji B cells to measure residual viral infectivity. We found that 1 min treatment of XMRV particles at 60°C is sufficient to reduce infectivity by 99.9%. XMRV infectivity was maximal at a neutral pH but was reduced by 86% at pH 4 and 99.9% at pH 10. The common hand and surface disinfectants ethanol and isopropanol as well as the cell fixation reagent paraformaldehyde abrogated XMRV infectivity entirely, as indicated by a reduction of infectivity exceeding 99.99%. Our findings provide evidence of specific means to inactivate XMRV. Their application will help to prevent unintended XMRV contamination of cell cultures in laboratories and minimize the risk for laboratory personnel and health care workers to become infected with this biosafety level 2 organism.


Subject(s)
Alcohols/pharmacology , Disinfectants/pharmacology , Environmental Microbiology , Microbial Viability/drug effects , Xenotropic murine leukemia virus-related virus/drug effects , Xenotropic murine leukemia virus-related virus/physiology , B-Lymphocytes/virology , Cell Line , Humans , Hydrogen-Ion Concentration , Temperature
6.
J Virol ; 86(6): 3152-66, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22238316

ABSTRACT

Although xenotropic murine leukemia virus-related virus (XMRV) has been previously linked to prostate cancer and myalgic encephalomyelitis/chronic fatigue syndrome, recent data indicate that results interpreted as evidence of human XMRV infection reflect laboratory contamination rather than authentic in vivo infection. Nevertheless, XMRV is a retrovirus of undefined pathogenic potential that is able to replicate in human cells. Here we describe a comprehensive analysis of two male pigtailed macaques (Macaca nemestrina) experimentally infected with XMRV. Following intravenous inoculation with >10(10) RNA copy equivalents of XMRV, viral replication was limited and transient, peaking at ≤2,200 viral RNA (vRNA) copies/ml plasma and becoming undetectable by 4 weeks postinfection, though viral DNA (vDNA) in peripheral blood mononuclear cells remained detectable through 119 days of follow-up. Similarly, vRNA was not detectable in lymph nodes by in situ hybridization despite detectable vDNA. Sequencing of cell-associated vDNA revealed extensive G-to-A hypermutation, suggestive of APOBEC-mediated viral restriction. Consistent with limited viral replication, we found transient upregulation of type I interferon responses that returned to baseline by 2 weeks postinfection, no detectable cellular immune responses, and limited or no spread to prostate tissue. Antibody responses, including neutralizing antibodies, however, were detectable by 2 weeks postinfection and maintained throughout the study. Both animals were healthy for the duration of follow-up. These findings indicate that XMRV replication and spread were limited in pigtailed macaques, predominantly by APOBEC-mediated hypermutation. Given that human APOBEC proteins restrict XMRV infection in vitro, human XMRV infection, if it occurred, would be expected to be characterized by similarly limited viral replication and spread.


Subject(s)
Disease Models, Animal , Macaca nemestrina , Retroviridae Infections/virology , Virus Replication , Xenotropic murine leukemia virus-related virus/physiology , Animals , Antibodies, Viral/immunology , Humans , Male , Phylogeny , Retroviridae Infections/immunology , Xenotropic murine leukemia virus-related virus/classification , Xenotropic murine leukemia virus-related virus/genetics
7.
J Virol ; 86(1): 328-38, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22031947

ABSTRACT

Xenotropic murine leukemia virus-related virus (XMRV) was previously reported to be associated with human prostate cancer and chronic fatigue syndrome. Our groups recently showed that XMRV was created through recombination between two endogenous murine retroviruses, PreXMRV-1 and PreXMRV-2, during the passaging of a prostate tumor xenograft in nude mice. Here, multiple approaches that led to the identification of PreXMRV-2, as well as the distribution of both parental proviruses among different mouse species, are described. The chromosomal loci of both proviruses were determined in the mouse genome, and integration site information was used to analyze the distribution of both proviruses in 48 laboratory mouse strains and 46 wild-derived strains. The strain distributions of PreXMRV-1 and PreXMRV-2 are quite different, the former being found predominantly in Asian mice and the latter in European mice, making it unlikely that the two XMRV ancestors could have recombined independently in the wild to generate an infectious virus. XMRV was not present in any of the mouse strains tested, and among the wild-derived mouse strains analyzed, not a single mouse carried both parental proviruses. Interestingly, PreXMRV-1 and PreXMRV-2 were found together in three laboratory strains, Hsd nude, NU/NU, and C57BR/cd, consistent with previous data that the recombination event that led to the generation of XMRV could have occurred only in the laboratory. The three laboratory strains carried the Xpr1(n) receptor variant nonpermissive to XMRV and xenotropic murine leukemia virus (X-MLV) infection, suggesting that the xenografted human tumor cells were required for the resulting XMRV recombinant to infect and propagate.


Subject(s)
Mice/virology , Proviruses/genetics , Retroviridae Infections/veterinary , Xenotropic murine leukemia virus-related virus/genetics , Amino Acid Sequence , Animals , Base Sequence , Chromosome Mapping , Female , Humans , Male , Mice/genetics , Mice, Inbred Strains , Molecular Sequence Data , Proviruses/isolation & purification , Proviruses/physiology , Rats , Rats, Sprague-Dawley , Receptors, Virus/chemistry , Receptors, Virus/genetics , Recombination, Genetic , Retroviridae Infections/virology , Sequence Alignment , Virus Integration , Xenotropic and Polytropic Retrovirus Receptor , Xenotropic murine leukemia virus-related virus/isolation & purification , Xenotropic murine leukemia virus-related virus/physiology
8.
Prostate ; 72(8): 886-97, 2012 Jun 01.
Article in English | MEDLINE | ID: mdl-21932423

ABSTRACT

BACKGROUND: Xenotropic murine leukemia virus-related retrovirus (XMRV) is a recently discovered gammaretrovirus that was originally detected in prostate tumors. However, a causal relationship between XMRV and prostate cancer remains controversial due to conflicting reports on its etiologic occurrence. Even though gammaretroviruses are known to induce cancer in animals, a mechanism for XMRV-induced carcinogenesis remains unknown. Several mechanisms including insertional mutagenesis, proinflammatory effects, oncogenic viral proteins, immune suppression, and altered epithelial/stromal interactions have been proposed for a role of XMRV in prostate cancer. However, biochemical data supporting any of these mechanisms are lacking. Therefore, our aim was to evaluate a potential role of XMRV in prostate carcinogenesis. METHODS: Growth kinetics of prostate cancer cells are conducted by MTT assay. In vitro transformation and invasion was carried out by soft agar colony formation, and Matrigel cell invasion assay, respectively. p27(Kip1) expression was determined by Western blot and MMP activation was evaluated by gelatin-zymography. Up-regulation of miR221 and miR222 expression was examined by real-time PCR. RESULTS: We demonstrate that XMRV infection can accelerate cellular proliferation, enhance transformation, and increase invasiveness of slow growing prostate cancer cells. The molecular basis of these viral induced activities is mediated by the downregulation of cyclin/cyclin dependent kinase inhibitor p27(Kip1) . Downstream analyses illustrated that XMRV infection upregulates miR221 and miR222 expression that target p27(Kip1) mRNA. CONCLUSIONS: We propose that downregulation of p27(Kip1) by XMRV infection facilitates transition of G1 to S, thereby accelerates growth of prostate cancer cells. Our findings implicate that if XMRV is present in humans, then under appropriate cellular microenvironment it may serve as a cofactor to promote cancer progression in the prostate.


Subject(s)
Adenocarcinoma/pathology , Cell Proliferation , Cell Transformation, Neoplastic/pathology , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Down-Regulation/physiology , Prostatic Neoplasms/pathology , Xenotropic murine leukemia virus-related virus/physiology , Adenocarcinoma/physiopathology , Adenocarcinoma/virology , Cell Line, Tumor , Disease Progression , Humans , Male , Metalloproteases/metabolism , MicroRNAs/metabolism , Neoplasm Invasiveness , Prostatic Neoplasms/physiopathology , Prostatic Neoplasms/virology , RNA, Messenger/metabolism , Up-Regulation
9.
Retrovirology ; 9: 58, 2012 Jul 24.
Article in English | MEDLINE | ID: mdl-22828015

ABSTRACT

BACKGROUND: One of the unique features of gammaretroviruses is that they contain an additional extended form of Gag, glyco-gag, which initiates in the leader sequence. MuLV glyco-gag, gPr80Gag, promotes retrovirus replication and disease progression. Although virtually all infectious MuLVs encode glyco-gag, XMRV (xenotropic murine leukemia virus-related virus) lacks the classical gPr80Gag sequence. We examined XMRV to determine if its leader sequence contains glyco-gag activity, whether the presence of conventional gPr80Gag affects replication of XMRV, and we describe the evolution of glyco-gag-deficient MuLVs in Mus. RESULTS: We introduced several mutations disrupting two putative but noncanonical glyco-gag proteins in the leader sequence region in XMRV and found that those mutations did not affect virus release nor susceptibility to the antiviral activity of hA3G (human APOBEC3G). A chimeric XMRV encoding the Moloney MuLV (M-MuLV) leader sequence (MXMRV) demonstrated that M-MuLV glyco-gag facilitated MXMRV release and increased infectivity. Infectivity assays with several cell lines showed that glyco-gag increases XMRV infectivity in all cell lines tested, but the level of this increase varies in different cell lines. Because MuLV glyco-gag counteracts mouse APOBEC3, we investigated whether M-MuLV glyco-gag enhances XMRV infection by counteracting human APOBEC3. Comparison of hAPOBEC3 isoforms expressed in different cell lines indicated that hA3B was the most likely candidate for a restrictive hA3. However over-expression of hA3B showed no enhanced restriction of infection by XMRV compared to MXMRV. Endogenous MuLVs in the sequenced mouse genome were screened for canonical glyco-gag, which was identified in two clades of xenotropic MuLVs (X-MuLVs) and ecotropic MuLVs, but not in other X-MuLVs or in any polytropic MuLVs. CONCLUSIONS: M-MuLV glyco-gag facilitates XMRV replication, and the leader sequence region in XMRV does not encode proteins equivalent to M-MuLV glyco-gag. The fact that the ability of glyco-gag to enhance XMRV infection varies in different cell lines suggests a glyco-gag sensitive restrictive factor that further reduces XMRV infectivity. The M-MuLV glyco-gag enhancement for XMRV replication is through a hAPOBEC3 independent mechanism. The absence of glyco-gag in MuLVs carried by western European mice suggests that loss of this sequence is a relatively recent event with limited subspecies distribution.


Subject(s)
Cytosine Deaminase/metabolism , Gene Products, gag/metabolism , Glycoproteins/metabolism , Moloney murine leukemia virus/metabolism , Virus Replication , Xenotropic murine leukemia virus-related virus/physiology , APOBEC Deaminases , Amino Acid Sequence , Animals , Base Sequence , Cytidine Deaminase , Cytosine Deaminase/antagonists & inhibitors , Cytosine Deaminase/genetics , Evolution, Molecular , Gene Products, gag/classification , Gene Products, gag/genetics , Genome, Viral , Glycoproteins/genetics , Glycosylation , HEK293 Cells , HeLa Cells , Hep G2 Cells , Humans , Isoenzymes/genetics , Isoenzymes/metabolism , Mice , Molecular Sequence Data , Moloney murine leukemia virus/genetics , Mutagenesis, Site-Directed , Mutation , Phylogeny , Rats , Virus Release , Xenotropic murine leukemia virus-related virus/genetics , Xenotropic murine leukemia virus-related virus/metabolism
10.
Biochem Biophys Res Commun ; 428(1): 17-23, 2012 Nov 09.
Article in English | MEDLINE | ID: mdl-23047007

ABSTRACT

BST-2 (bone marrow stromal cell antigen 2) is an interferon-inducible protein that inhibits the release of a variety of enveloped viruses by tethering viral particles to the cell surface. Xenotropic murine leukemia virus-related virus (XMRV) is a gamma-retrovirus that was derived from the recombination of two endogenous murine leukemia viruses during the production of a prostate cell line in mice. In this study, we observed that XMRV was highly sensitive to the inhibition by human BST-2. We were able to determine the structural domains of BST-2 that are essential to restrict XMRV, including the transmembrane domain, the coiled-coil ectodomain, the C-terminal glycosylphosphatidylinositol (GPI) anchor, the two putative N-linked glycosylation sites, and the three extracellular cysteine residues. Protease treatment effectively released XMRV particles into the supernatant, supporting the notion that BST-2 tethered nascent particles to the cell surface. These data suggest that BST-2 poses a strong restriction toward XMRV production.


Subject(s)
Antigens, CD/metabolism , Retroviridae Infections/metabolism , Retroviridae Infections/virology , Virus Replication , Xenotropic murine leukemia virus-related virus/physiology , Animals , Antigens, CD/genetics , Chlorocebus aethiops , GPI-Linked Proteins/genetics , GPI-Linked Proteins/metabolism , HEK293 Cells , HeLa Cells , Humans , Protein Structure, Tertiary , Vero Cells
11.
J Virol ; 85(14): 7195-202, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21543496

ABSTRACT

Chronic fatigue syndrome (CFS) is a multisystem disorder characterized by prolonged and severe fatigue that is not relieved by rest. Attempts to treat CFS have been largely ineffective primarily because the etiology of the disorder is unknown. Recently, CFS has been associated with xenotropic murine leukemia virus-related virus (XMRV) as well as other murine leukemia virus (MLV)-related viruses, though not all studies have found these associations. We collected blood samples from 100 CFS patients and 200 self-reported healthy volunteers from the same geographical area. We analyzed these in a blind manner using molecular, serological, and viral replication assays. We also analyzed samples from patients in the original study that reported XMRV in CFS patients. We did not find XMRV or related MLVs either as viral sequences or infectious viruses, nor did we find antibodies to these viruses in any of the patient samples, including those from the original study. We show that at least some of the discrepancy with previous studies is due to the presence of trace amounts of mouse DNA in the Taq polymerase enzymes used in these previous studies. Our findings do not support an association between CFS and MLV-related viruses, including XMRV, and the off-label use of antiretrovirals for the treatment of CFS does not seem justified at present.


Subject(s)
Fatigue Syndrome, Chronic/virology , Xenotropic murine leukemia virus-related virus/isolation & purification , Adult , Base Sequence , Blotting, Western , DNA Primers , Enzyme-Linked Immunosorbent Assay , Female , Humans , Polymerase Chain Reaction , Virus Replication , Xenotropic murine leukemia virus-related virus/physiology
12.
J Virol ; 85(7): 3179-86, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21270144

ABSTRACT

Xenotropic murine leukemia virus-related virus (XMRV) is a gammaretrovirus linked to prostate carcinoma and chronic fatigue syndrome. Here we report that NF-κB activation can markedly increase XMRV production. The inflammatory cytokine tumor necrosis factor alpha (TNF-α), which activates NF-κB, significantly augmented viral Gag protein production in XMRV-infected cells. Reporter assays showed that TNF-α and Epstein-Barr virus (EBV) latent membrane protein 1 (LMP1), an intrinsic NF-κB activator, increased long terminal repeat (LTR)-dependent XMRV transcription. We identified two NF-κB binding sites (designated κB-1 and κB-2) in the LTR U3 region of XMRV and demonstrated that both sites bind to the NF-κB component p65/RelA. Mutation of the κB-1 site, but not the κB-2 site, impaired responsiveness to TNF-α and LMP1 in reporter assays. A mutant XMRV with a mutation at the κB-1 site replicated significantly less efficiently than the wild-type XMRV in the prostate carcinoma LNCaP, DU145, and PC-3 cell lines, HEK293 cells, the EBV-immortalized cell line IB4, and the Burkitt's lymphoma cell line BJAB. These results demonstrate that TNF-α and EBV LMP1 enhance XMRV replication in prostate carcinoma and B-lineage cells through the κB-1 site in the XMRV LTR, suggesting that inflammation, EBV infection, and other conditions leading to NF-κB activation may promote XMRV spread in humans.


Subject(s)
B-Lymphocytes/virology , Carcinoma/virology , NF-kappa B/metabolism , Prostatic Neoplasms/virology , Transcription, Genetic , Virus Replication , Xenotropic murine leukemia virus-related virus/physiology , Binding Sites , Cell Line, Tumor , Gene Products, gag/biosynthesis , Humans , Male , Protein Binding , Terminal Repeat Sequences/genetics , Transcription Factor RelA/metabolism , Tumor Necrosis Factor-alpha/metabolism
13.
Transfusion ; 52(2): 317-25, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22212105

ABSTRACT

BACKGROUND: Although recent data have brought into question the association between xenotropic murine leukemia virus-related virus (XMRV) and chronic fatigue syndrome, one group has reported evidence of human infection with distinct polytropic murine leukemia viruses (MLVs). Occult retroviral infection among humans poses a significant public health risk should it be introduced into the blood supply. To explore the possibility of cross-species transmission of MLVs to humans, we sought molecular and serologic evidence of XRMV/MLV infection among a cohort of animal workers highly exposed to mice. STUDY DESIGN AND METHODS: Before the commencement of the study, the laboratory and equipment were demonstrated to be free of XMRV/MLV DNA sequences. DNA extracted from 43 animal workers was tested using nested polymerase chain reaction (PCR) with published primer sets, targeting regions of XMRV and MLV gag. Negative controls were assayed in a 1:1 ratio with specimens. Serum specimens were tested using a validated immunoblot assay containing cross-reactive XMRV antigens. RESULTS: Initial molecular assays demonstrated that the physical space and laboratory equipment were free of MLV and XMRV DNA sequences. Nested PCR assays using multiple primer sets successfully amplified XMRV and MLV sequences from positive controls with high sensitivity. A single, nonreproducible, false-positive result from one specimen was shown to be the result of subsequent contamination. Immunoblotting of all subjects' sera failed to demonstrate any evidence of seroreactivity to XMRV proteins. CONCLUSIONS: There was no evidence of human infection with XMRV/MLV among a cohort of individuals highly exposed to mice. These data suggest that the likelihood of cross-species transmission events of MLV from mice to humans is low.


Subject(s)
Animal Technicians/statistics & numerical data , Cross Infection/epidemiology , Retroviridae Infections/epidemiology , Retroviridae Infections/transmission , Xenotropic murine leukemia virus-related virus , Animals , Antibodies, Viral/blood , Cells, Cultured , Cross Infection/etiology , Humans , Laboratory Personnel/statistics & numerical data , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Middle Aged , Occupational Exposure/statistics & numerical data , Retroviridae Infections/virology , Species Specificity , Validation Studies as Topic , Xenotropic murine leukemia virus-related virus/physiology
14.
Transfusion ; 52(2): 298-306, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22098340

ABSTRACT

BACKGROUND: When xenotropic murine leukemia virus-related virus (XMRV) was first reported in association with chronic fatigue syndrome, it was suggested that it might offer a risk to blood safety. Thus, the prevalence of the virus among blood donors and, if present, its transmissibility by transfusion need to be defined. STUDY DESIGN AND METHODS: Two populations of routine blood donor samples (1435 and 13,399) were obtained for prevalence evaluations; samples from a linked donor-recipient repository were also evaluated. Samples were tested for the presence of antibodies to XMRV-related recombinant antigens and/or for XMRV RNA, using validated, high-throughput systems. RESULTS: The presence of antibodies to XMRV could not be confirmed among a total of 17,249 blood donors or recipients (0%; 95% confidence interval [CI], 0%-0.017%); 1763 tested samples were nonreactive for XMRV RNA (0%; 95% CI, 0%-0.17%). Evidence of infection was absent from 109 recipients and 830 evaluable blood samples tested after transfusion of a total of 3741 blood components. CONCLUSIONS: XMRV and related murine leukemia virus (MLV) markers are not present among a large population of blood donors and evidence of transfusion transmission could not be detected. Thus, these viruses do not currently pose a threat to blood recipient safety and further actions relating to XMRV and MLV are not justified.


Subject(s)
Blood Safety , Retroviridae Infections/blood , Retroviridae Infections/transmission , Xenotropic murine leukemia virus-related virus/physiology , Adolescent , Adult , Blood Donors/statistics & numerical data , Blood Safety/methods , Blood Specimen Collection/methods , Blood Specimen Collection/standards , Blood Specimen Collection/statistics & numerical data , Fatigue Syndrome, Chronic/blood , Fatigue Syndrome, Chronic/epidemiology , Fatigue Syndrome, Chronic/etiology , Fatigue Syndrome, Chronic/virology , Female , Humans , Male , Middle Aged , RNA, Viral/blood , RNA, Viral/isolation & purification , Retroviridae Infections/epidemiology , Retroviridae Infections/virology , Risk Factors , Serologic Tests , Transplantation/physiology , Transplantation/statistics & numerical data , Xenotropic murine leukemia virus-related virus/genetics , Xenotropic murine leukemia virus-related virus/isolation & purification
15.
Rev Med Virol ; 21(1): 3-17, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21294212

ABSTRACT

The recent discovery of xenotropic murine leukaemia virus-related virus (XMRV) in prostate cancer tissues and in the blood of individuals suffering from chronic fatigue syndrome has attracted considerable interest. However, the relevance and significance of XMRV to human disease remain unclear, since the association has not been confirmed in other studies. XMRV is the first gammaretrovirus to be found in humans. XMRV and murine leukaemia viruses share similar structures and genomic organisation. Human restriction factors such as APOBEC3 or tetherin inhibit XMRV replication. Although XMRV induces low rates of transformation in cell culture, it might be able to induce cancer by low-frequency insertional activation of oncogenes or through the generation of highly active transforming viruses. A preference for regulatory regions of transcriptional active genes has been observed after a genomic-wide analysis of XMRV integration sites. Genes related to carcinogenesis and androgen signalling have been identified in the vicinity of integration sites. The XMRV genome contains a glucocorticoid responsive element, and androgens could modulate viral replication in the prostate. Evidence supporting the involvement of XMRV in chronic fatigue syndrome is still very weak, and needs further confirmation and validation. Currently approved anti-retroviral drugs such as zidovudine, tenofovir and raltegravir are efficient inhibitors of XMRV replication in vitro. These drugs might be useful to treat XMRV infection in humans. The identification of XMRV has potentially serious health implications for the implementation of novel techniques including gene therapy or xenotransplantation, while raising concerns on the need for screening donated blood to prevent transmission through transfusion.


Subject(s)
Fatigue Syndrome, Chronic/virology , Prostatic Neoplasms/virology , Xenotropic murine leukemia virus-related virus/physiology , Animals , Fatigue Syndrome, Chronic/genetics , Fatigue Syndrome, Chronic/metabolism , Female , Gene Expression Regulation, Viral , Humans , Male , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Xenotropic murine leukemia virus-related virus/genetics
16.
J Autoimmun ; 37(4): 311-8, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21982749

ABSTRACT

The envelope glycoprotein gp70 of endogenous retroviruses implicated in murine lupus nephritis is secreted by hepatocytes and its expression is controlled by Sgp3 (serum gp70 production 3) and Sgp4 loci derived from lupus-prone mice. Among three different endogenous retroviruses (ecotropic, xenotropic and polytropic), xenotropic viruses are considered to be the major source of serum gp70. Although the abundance of xenotropic viral gp70 RNA in livers was up-regulated by the presence of these two Sgp loci, it has not yet been clear whether Sgp3 and Sgp4 regulate the expression of a fraction or multiple xenotropic viruses present in mouse genome. To address this question, we determined the genetic origin of xenotropic viral sequences expressed in wild-type and two different Sgp congenic C57BL/6 mice. Among 14 xenotropic proviruses present in the C57BL/6 genome, only two proviruses (Xmv10 and Xmv14) were actively transcribed in wild-type C57BL/6 mice. In contrast, Sgp3 enhanced the transcription of Xmv10 and induced the transcription of three additional xenotropic viruses (Xmv15, Xmv17 and Xmv18), while Sgp4 induced the expression of a different xenotropic virus (Xmv13). Notably, stimulation of TLR7 in Sgp3 congenic C57BL/6 mice led to a highly enhanced expression of potentially replication-competent Xmv18. These results indicated that Sgp3 and Sgp4 independently regulated the transcription of distinct and restricted sets of xenotropic viruses in trans, thereby promoting the production of nephritogenic gp70 autoantigens. Furthermore, the induced expression of potentially replication-competent xenotropic viruses by Sgp3 may contribute to the development of autoimmune responses against gp70 through the activation of TLR7.


Subject(s)
Glycoproteins/metabolism , Lupus Nephritis/genetics , Molecular Chaperones/metabolism , Retroviridae Infections/genetics , Viral Envelope Proteins/metabolism , Xenotropic murine leukemia virus-related virus/physiology , Animals , Autoantibodies/blood , Gene Expression Regulation, Viral/immunology , Gene Products, env/blood , Gene Products, env/genetics , Gene Products, env/metabolism , Glycoproteins/genetics , Lupus Nephritis/etiology , Lupus Nephritis/immunology , Lupus Nephritis/virology , Male , Mice , Mice, Congenic , Mice, Inbred C57BL , Mice, Inbred NZB , Molecular Chaperones/genetics , Molecular Chaperones/immunology , Retroviridae Infections/complications , Retroviridae Infections/immunology , Retroviridae Infections/virology , Transcriptional Activation/immunology , Viral Envelope Proteins/genetics , Viral Envelope Proteins/immunology , Xenotropic murine leukemia virus-related virus/pathogenicity
17.
Virol J ; 8: 531, 2011 Dec 12.
Article in English | MEDLINE | ID: mdl-22152111

ABSTRACT

BACKGROUND: Xenotropic murine leukemia virus (MLV)-related virus (XMRV) is a gammaretrovirus that was discovered in prostate cancer tissues. Recently, it has been proposed that XMRV is a laboratory contaminant and may have originated via a rare recombination event. Host restriction factor APOBEC3G (A3G) has been reported to severely restrict XMRV replication in human peripheral blood mononuclear cells. Interestingly, XMRV infects and replicates efficiently in prostate cancer cells of epithelial origin. It has been proposed that due to lack off or very low levels of A3G protein XMRV is able to productively replicate in these cells. FINDINGS: This report builds on and challenges the published data on the absence of A3G protein in prostate epithelial cells lines. We demonstrate the presence of A3G in prostate epithelial cell lines (LNCaP and DU145) by western blot and mass spectrometry. We believe the discrepancy in A3G detection is may be due to selection and sensitivity of A3G antibodies employed in the prior studies. Our results also indicate that XMRV produced from A3G expressing LNCaP cells can infect and replicate in target cells. Most importantly our data reveal downregulation of A3G in XMRV infected LNCaP and DU145 cells. CONCLUSIONS: We propose that XMRV replicates efficiently in prostate epithelial cells by downregulating A3G expression. Given that XMRV lacks accessory proteins such as HIV-1 Vif that are known to counteract A3G function in human cells, our data suggest a novel mechanism by which retroviruses can counteract the antiviral effects of A3G proteins.


Subject(s)
Cytidine Deaminase/genetics , Prostate/metabolism , Xenotropic murine leukemia virus-related virus/physiology , APOBEC-3G Deaminase , Amino Acid Sequence , Animals , Blotting, Western , Cell Line, Tumor , Cytidine Deaminase/metabolism , Down-Regulation , Electrophoresis, Polyacrylamide Gel , Epithelial Cells , Gene Expression , Host-Pathogen Interactions , Humans , Male , Mass Spectrometry , Mice , Molecular Sequence Data , Prostate/cytology , Prostate/virology , Prostatic Neoplasms , Virus Replication
18.
Virol J ; 8: 423, 2011 Sep 06.
Article in English | MEDLINE | ID: mdl-21896167

ABSTRACT

BACKGROUND: XMRV is a gammaretrovirus first identified in prostate tissues of Prostate Cancer (PC) patients and later in the blood cells of patients with Chronic Fatigue Syndrome (CFS). Although XMRV is thought to use XPR1 for cell entry, it infects A549 cells that do not express XPR1, suggesting usage of other receptors or co-receptors. METHODS: To study the usage of different receptors and co- receptors that could play a role in XMRV infection of lymphoid cells and GHOST (GFP- Human osteosarcoma) cells expressing CD4 along with different chemokine receptors including CCR1, CCR2, etc., were infected with XMRV. Culture supernatants and cells were tested for XMRV replication using real time quantitative PCR. RESULTS: Infection and replication of XMRV was seen in a variety of GHOST cells, LNCaP, DU145, A549 and Caski cell lines. The levels of XMRV replication varied in different cell lines showing differential replication in different cell lines. However, replication in A549 which lacks XPR1 expression was relatively higher than DU145 but lower than, LNCaP. XMRV replication varied in GHOST cell lines expressing CD4 and each of the co- receptors CCR1-CCR8 and bob. There was significant replication of XMRV in CCR3 and Bonzo although it is much lower when compared to DU145, A549 and LNCaP. CONCLUSION: XMRV replication was observed in GHOST cells that express CD4 and each of the chemokine receptors ranging from CCR1- CCR8 and BOB suggesting that infectivity in hematopoietic cells could be mediated by use of these receptors.


Subject(s)
Bone Neoplasms/virology , Osteosarcoma/virology , Receptors, Chemokine/metabolism , Receptors, G-Protein-Coupled/metabolism , Receptors, Peptide/metabolism , Receptors, Virus/metabolism , Virus Replication , Xenotropic murine leukemia virus-related virus/physiology , Bone Neoplasms/genetics , Bone Neoplasms/metabolism , Bone Neoplasms/pathology , CD4 Antigens/biosynthesis , Cell Line , Fatigue Syndrome, Chronic/genetics , Fatigue Syndrome, Chronic/metabolism , Fatigue Syndrome, Chronic/virology , Gene Expression , Humans , Male , Organ Specificity , Osteosarcoma/genetics , Osteosarcoma/metabolism , Osteosarcoma/pathology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/virology , Real-Time Polymerase Chain Reaction , Receptors, Chemokine/genetics , Receptors, G-Protein-Coupled/genetics , Receptors, Peptide/genetics , Receptors, Virus/genetics , Xenotropic and Polytropic Retrovirus Receptor
19.
Retrovirology ; 7: 113, 2010 Dec 22.
Article in English | MEDLINE | ID: mdl-21176195

ABSTRACT

The 1st International Workshop on Xenotropic Murine Leukemia Virus-Related Retrovirus (XMRV), co-sponsored by the National Institutes of Health, The Department of Health and Human Services and Abbott Diagnostics, was convened on September 7/8, 2010 on the NIH campus, Bethesda, MD. Attracting an international audience of over 200 participants, the 2-day event combined a series of plenary talks with updates on different aspects of XMRV research, addressing basic gammaretrovirus biology, host response, association of XMRV with chronic fatigue syndrome and prostate cancer, assay development and epidemiology. The current status of XMRV research, concerns among the scientific community and suggestions for future actions are summarized in this meeting report.


Subject(s)
Retroviridae Infections/virology , Retroviridae/physiology , Xenotropic murine leukemia virus-related virus/physiology , Animals , Fatigue Syndrome, Chronic/genetics , Fatigue Syndrome, Chronic/metabolism , Fatigue Syndrome, Chronic/virology , Female , Host-Pathogen Interactions , Humans , Male , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/virology , Retroviridae/genetics , Retroviridae/isolation & purification , Retroviridae Infections/diagnosis , Retroviridae Infections/genetics , Retroviridae Infections/metabolism , Xenotropic murine leukemia virus-related virus/genetics , Xenotropic murine leukemia virus-related virus/isolation & purification
SELECTION OF CITATIONS
SEARCH DETAIL