Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 66
Filter
1.
PLoS Pathog ; 19(4): e1011339, 2023 04.
Article in English | MEDLINE | ID: mdl-37093892

ABSTRACT

Infection with viruses of animal origin pose a significant threat to human populations. Simian foamy viruses (SFVs) are frequently transmitted to humans, in which they establish a life-long infection, with the persistence of replication-competent virus. However, zoonotic SFVs do not induce severe disease nor are they transmitted between humans. Thus, SFVs represent a model of zoonotic retroviruses that lead to a chronic infection successfully controlled by the human immune system. We previously showed that infected humans develop potent neutralizing antibodies (nAbs). Within the viral envelope (Env), the surface protein (SU) carries a variable region that defines two genotypes, overlaps with the receptor binding domain (RBD), and is the exclusive target of nAbs. However, its antigenic determinants are not understood. Here, we characterized nAbs present in plasma samples from SFV-infected individuals living in Central Africa. Neutralization assays were carried out in the presence of recombinant SU that compete with SU at the surface of viral vector particles. We defined the regions targeted by the nAbs using mutant SU proteins modified at the glycosylation sites, RBD functional subregions, and genotype-specific sequences that present properties of B-cell epitopes. We observed that nAbs target conformational epitopes. We identified three major epitopic regions: the loops at the apex of the RBD, which likely mediate interactions between Env protomers to form Env trimers, a loop located in the vicinity of the heparan binding site, and a region proximal to the highly conserved glycosylation site N8. We provide information on how nAbs specific for each of the two viral genotypes target different epitopes. Two common immune escape mechanisms, sequence variation and glycan shielding, were not observed. We propose a model according to which the neutralization mechanisms rely on the nAbs to block the Env conformational change and/or interfere with binding to susceptible cells. As the SFV RBD is structurally different from known retroviral RBDs, our data provide fundamental knowledge on the structural basis for the inhibition of viruses by nAbs. Trial registration: The study was registered at www.clinicaltrials.gov: https://clinicaltrials.gov/ct2/show/NCT03225794/.


Subject(s)
Hominidae , Simian foamy virus , Animals , Humans , Simian foamy virus/genetics , Retroviridae , Antibodies, Neutralizing , Epitopes, B-Lymphocyte/genetics , HIV Antibodies
2.
Viruses ; 12(11)2020 11 02.
Article in English | MEDLINE | ID: mdl-33147813

ABSTRACT

In addition to regulatory or accessory proteins, some complex retroviruses gain a repertoire of micro-RNAs (miRNAs) to regulate and control virus-host interactions for efficient replication and spread. In particular, bovine and simian foamy viruses (BFV and SFV) have recently been shown to express a diverse set of RNA polymerase III-directed miRNAs, some with a unique primary miRNA double-hairpin, dumbbell-shaped structure not known in other viruses or organisms. While the mechanisms of expression and structural requirements have been studied, the functional importance of these miRNAs is still far from understood. Here, we describe the in silico identification of BFV miRNA targets and the subsequent experimental validation of bovine Ankyrin Repeat Domain 17 (ANKRD17) and Bax-interacting factor 1 (Bif1) target genes in vitro and, finally, the suppression of ANKRD17 downstream genes in the affected pathway. Deletion of the entire miRNA cassette in the non-coding part of the U3 region of the long terminal repeats attenuated replication of corresponding BFV mutants in bovine cells. This repression can be almost completely trans-complemented by the most abundant miRNA BF2-5p having the best scores for predicted and validated BFV miRNA target genes. Deletion of the miRNA cassette does not grossly affect particle release and overall particle composition.


Subject(s)
Host Microbial Interactions/genetics , MicroRNAs/genetics , Spumavirus/genetics , Virus Replication , Animals , Cattle , Cell Line , Computer Simulation , Host-Pathogen Interactions , Simian foamy virus/genetics , Spumavirus/physiology , Terminal Repeat Sequences
3.
Gene ; 734: 144382, 2020 Apr 15.
Article in English | MEDLINE | ID: mdl-31978513

ABSTRACT

Japanese macaque (Macaca fuscata) is an indigenous Old World monkey (OWM) species that inhabits the Japanese archipelago. There are two subspecies of Japanese macaque: Yakushima macaque (M. f. yakui) which inhabits Yakushima Island exclusively, and Hondo macaque (M. f. fuscata) which inhabits the mainland of Japan. Yakushima macaque is considered to be branched off from a certain parental macaque group that had inhabited the mainland of Japan. However, the process of sub-speciation of the Yakushima macaque is still unclear at present. In this study, to gain new insight into the process of sub-speciation of Japanese macaque, we utilized the simian foamy virus (SFV) as a marker. SFVs are found in virtually all primates except humans and undergo species-specific cospeciation with the hosts. The phylogenetic analysis of conserved regions of the env gene in SFVs remarkably resembled that of the OWMs with high statistical confidence. The phylogenetic analyses also indicated that there are four (1-4) genotypes among Asian OWMs investigated. SFVs derived from Asian OWMs except Yakushima macaque were classified as genotypes 1-3, whereas SFVs isolated from all Yakushima macaques and one Hondo macaque were classified as genotype 4. Interestingly, genotype 4 was firstly branched off from the rest of the genotypes, which might indicate that the macaques infected with genotype 4 SFV were derived from the "older" population of Japanese macaques. The high prevalence of genotype 4 SFVs among Yakushima macaque might reflect the possibility that they are a descendant of the population settled earlier, which has been geographically isolated in Yakushima Island.


Subject(s)
Macaca fuscata/virology , Simian foamy virus/classification , Animals , Cells, Cultured , Genes, Viral , Genes, env , Integrases/genetics , Molecular Typing , Phylogeny , Simian foamy virus/genetics , Simian foamy virus/isolation & purification , Species Specificity
4.
Viruses ; 11(10)2019 10 10.
Article in English | MEDLINE | ID: mdl-31658739

ABSTRACT

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


Subject(s)
Leontopithecus/virology , Retroviridae Infections , Simian foamy virus , Animals , Animals, Wild/virology , Brazil/epidemiology , Genes, Viral , Monkey Diseases/virology , Phylogeny , Prevalence , Retroviridae Infections/epidemiology , Retroviridae Infections/prevention & control , Retroviridae Infections/transmission , Simian foamy virus/classification , Simian foamy virus/genetics , Simian foamy virus/isolation & purification
5.
Viruses ; 11(7)2019 07 03.
Article in English | MEDLINE | ID: mdl-31277268

ABSTRACT

Foamy viruses (FVs) are complex retroviruses present in many mammals, including nonhuman primates, where they are called simian foamy viruses (SFVs). SFVs can zoonotically infect humans, but very few complete SFV genomes are available, hampering the design of diagnostic assays. Gibbons are lesser apes widespread across Southeast Asia that can be infected with SFV, but only two partial SFV sequences are currently available. We used a metagenomics approach with next-generation sequencing of nucleic acid extracted from the cell culture of a blood specimen from a lesser ape, the pileated gibbon (Hylobates pileatus), to obtain the complete SFVhpi_SAM106 genome. We used Bayesian analysis to co-infer phylogenetic relationships and divergence dates. SFVhpi_SAM106 is ancestral to other ape SFVs with a divergence date of ~20.6 million years ago, reflecting ancient co-evolution of the host and SFVhpi_SAM106. Analysis of the complete SFVhpi_SAM106 genome shows that it has the same genetic architecture as other SFVs but has the longest recorded genome (13,885-nt) due to a longer long terminal repeat region (2,071 bp). The complete sequence of the SFVhpi_SAM106 genome fills an important knowledge gap in SFV genetics and will facilitate future studies of FV infection, transmission, and evolutionary history.


Subject(s)
Genome, Viral , Hylobates/virology , Monkey Diseases/virology , Retroviridae Infections/veterinary , Retroviridae Infections/virology , Simian foamy virus/genetics , Animals , Base Sequence , Bayes Theorem , Genes, Viral , Hominidae , Humans , Phylogeny , Recombination, Genetic , Sequence Alignment , Sequence Analysis , Simian foamy virus/classification , Terminal Repeat Sequences
7.
J Virol ; 93(11)2019 06 01.
Article in English | MEDLINE | ID: mdl-30894477

ABSTRACT

Cross-species transmission of simian foamy viruses (SFVs) from nonhuman primates (NHPs) to humans is currently ongoing. These zoonotic retroviruses establish lifelong persistent infection in their human hosts. SFV are apparently nonpathogenic in vivo, with ubiquitous in vitro tropism. Here, we aimed to identify envelope B-cell epitopes that are recognized following a zoonotic SFV infection. We screened a library of 169 peptides covering the external portion of the envelope from the prototype foamy virus (SFVpsc_huHSRV.13) for recognition by samples from 52 Central African hunters (16 uninfected and 36 infected with chimpanzee, gorilla, or Cercopithecus SFV). We demonstrate the specific recognition of peptide N96-V110 located in the leader peptide, gp18LP Forty-three variant peptides with truncations, alanine substitutions, or amino acid changes found in other SFV species were tested. We mapped the epitope between positions 98 and 108 and defined six amino acids essential for recognition. Most plasma samples from SFV-infected humans cross-reacted with sequences from apes and Old World monkey SFV species. The magnitude of binding to peptide N96-V110 was significantly higher for samples of individuals infected with a chimpanzee or gorilla SFV than those infected with a Cercopithecus SFV. In conclusion, we have been the first to define an immunodominant B-cell epitope recognized by humans following zoonotic SFV infection.IMPORTANCE Foamy viruses are the oldest known retroviruses and have been mostly described to be nonpathogenic in their natural animal hosts. SFVs can be transmitted to humans, in whom they establish persistent infection, like the simian lenti- and deltaviruses that led to the emergence of two major human pathogens, human immunodeficiency virus type 1 and human T-lymphotropic virus type 1. This is the first identification of an SFV-specific B-cell epitope recognized by human plasma samples. The immunodominant epitope lies in gp18LP, probably at the base of the envelope trimers. The NHP species the most genetically related to humans transmitted SFV strains that induced the strongest antibody responses. Importantly, this epitope is well conserved across SFV species that infect African and Asian NHPs.


Subject(s)
Simian foamy virus/immunology , Viral Envelope Proteins/immunology , Zoonoses/immunology , Adult , Animals , Antibodies, Viral/blood , Cameroon , Cercopithecus/virology , DNA, Viral/blood , Epitopes, B-Lymphocyte/genetics , Epitopes, B-Lymphocyte/immunology , Gabon , Gorilla gorilla/virology , Hominidae/immunology , Hominidae/virology , Humans , Male , Middle Aged , Pan troglodytes/virology , Retroviridae Infections/virology , Simian foamy virus/genetics , Spumavirus/genetics , Spumavirus/immunology , Viral Envelope Proteins/genetics , Zoonoses/genetics , Zoonoses/virology
8.
Arch Virol ; 163(9): 2507-2512, 2018 Sep.
Article in English | MEDLINE | ID: mdl-29860676

ABSTRACT

SFVmmu-DPZ9524 represents the third completely sequenced rhesus macaque simian foamy virus (SFV) isolate, alongside SFVmmu_K3T with a similar SFV-1-type env, and R289HybAGM with a SFV-2-like env. Sequence analysis demonstrates that, in gag and pol, SFVmmu-DPZ9524 is more closely related to R289HybAGM than to SFVmmu_K3T, which, outside of env, is more similar to a Japanese macaque isolate than to the other two rhesus macaque isolates SFVmmu-DPZ9524 and R289HybAGM. Further, we identify bel as another recombinant locus in R289HybAGM, confirming that recombination contributes to sequence diversity in SFV.


Subject(s)
DNA, Viral/genetics , Macaca mulatta/virology , Monkey Diseases/virology , Recombination, Genetic , Retroviridae Infections/veterinary , Simian foamy virus/genetics , Animals , Gene Expression , Gene Products, env/genetics , Gene Products, gag/genetics , Gene Products, pol/genetics , Phylogeny , Retroviridae Infections/virology , Sequence Analysis, DNA , Serogroup , Simian foamy virus/classification , Simian foamy virus/isolation & purification
9.
PLoS One ; 12(9): e0184251, 2017.
Article in English | MEDLINE | ID: mdl-28863180

ABSTRACT

Simian foamy viruses (SFVs) co-evolved with a wide range of Old World and New World primates (OWPs and NWPs, respectively) and occasionally transmit to humans. Previous studies of OWPs showed that the predominant site of SFV replication is the oral mucosa. However, very little is known about SFV viral loads (VLs) in the oral mucosa or blood of NWPs. NWPs have smaller body sizes, limiting collection of sufficient whole blood volumes to molecularly detect and quantify SFV. Our study evaluated the use of noninvasively collected buccal swabs to detect NWP SFV compared with detection in blood using a new NWP SFV quantitative PCR (qPCR) assay. Buccal and blood samples were collected from 107 captive NWPs in Brazil comprising eleven distinct genera at the Primate Center of Rio de Janeiro (n = 58) and at Fundação Jardim Zoológico da Cidade do Rio Janeiro (n = 49). NWP SFV western blot (WB) testing was performed on a subset of animals for comparison with PCR results. The qPCR assay was validated using distinct SFV polymerase sequences from seven NWP genera (Callithrix, Sapajus, Saimiri, Ateles, Alouatta, Cacajao and Pithecia). Assay sensitivity was 20 copies/106 cells, detectable in 90% of replicates. SFV DNA VLs were higher in buccal swabs (5 log copies/106 cells) compared to peripheral blood mononuclear cells (PBMCs) (3 log copies/106 cells). The qPCR assay was also more sensitive than nested PCR for detection of NWP SFV infection and identified an additional 27 SFV-infected monkeys of which 18 (90%) were WB-positive and three that were WB-negative. We show the utility of using both blood and buccal swabs and our new qPCR assay for detection and quantification of diverse NWP SFV, which will assist a better understanding of the epidemiology of SFV in NWPs and any potential zoonotic infection risk for humans exposed to NWPs.


Subject(s)
Leukocytes, Mononuclear/virology , Primates/virology , Retroviridae Infections/diagnosis , Simian foamy virus/genetics , Specimen Handling/methods , Animals , Brazil , DNA, Viral/genetics , Humans , Monkey Diseases/diagnosis , Monkey Diseases/virology , Mouth Mucosa/virology , Phylogeny , Plasmids/metabolism , Polymerase Chain Reaction , Real-Time Polymerase Chain Reaction , Retroviridae Infections/veterinary , Sensitivity and Specificity , Species Specificity , Viral Tropism , Zoonoses/virology
10.
PLoS One ; 12(9): e0184502, 2017.
Article in English | MEDLINE | ID: mdl-28931021

ABSTRACT

Simian foamy viruses (SFVs) are retroviruses present in nearly all nonhuman primates (NHPs), including Old World primates (OWP) and New World primates (NWP). While all confirmed human infections with SFV are from zoonotic transmissions originating from OWP, little is known about the zoonotic transmission potential of NWP SFV. We conducted a longitudinal, prospective study of 56 workers occupationally exposed to NWP in Brazil. Plasma from these workers was tested using Western blot (WB) assays containing NWP SFV antigens. Genomic DNA from blood and buccal swabs was analyzed for the presence of proviral SFV sequences by three nested PCR tests and a new quantitative PCR assay. Exposure histories were obtained and analyzed for associations with possible SFV infection. Ten persons (18%) tested seropositive and two persons were seroindeterminate (3.6%) for NWP SFV. Six persons had seroreactivity over 2-3 years suggestive of persistent infection. All SFV NWP WB-positive workers reported at least one incident involving NWP, including six reporting NWP bites. NWP SFV viral DNA was not detected in the blood or buccal swabs from all 12 NWP SFV seroreactive workers. We also found evidence of SFV seroreversion in three workers suggestive of possible clearance of infection. Our findings suggest that NWP SFV can be transmitted to occupationally-exposed humans and can elicit specific humoral immune responses but infection remains well-controlled resulting in latent infection and may occasionally clear.


Subject(s)
Retroviridae Infections/diagnosis , Simian foamy virus/genetics , Zoonoses/diagnosis , Adult , Animals , Antigens, Viral/immunology , Antigens, Viral/metabolism , Brazil , DNA, Viral/blood , DNA, Viral/metabolism , Female , Humans , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/virology , Longitudinal Studies , Male , Middle Aged , Mouth Mucosa/virology , Polymerase Chain Reaction , Primates , Prospective Studies , Retroviridae Infections/transmission , Retroviridae Infections/virology , Risk , Simian foamy virus/isolation & purification , Zoonoses/virology
11.
Infect Genet Evol ; 51: 54-66, 2017 07.
Article in English | MEDLINE | ID: mdl-28274887

ABSTRACT

Simian foamy viruses (SFVs) infect most nonhuman primate species and appears to co-evolve with its hosts. This co-evolutionary signal is particularly strong among great apes, including orangutans (genus Pongo). Previous studies have identified three distinct orangutan SFV clades. The first of these three clades is composed of SFV from P. abelii from Sumatra, the second consists of SFV from P. pygmaeus from Borneo, while the third clade is mixed, comprising an SFV strain found in both species of orangutan. The existence of the mixed clade has been attributed to an expansion of P. pygmaeus into Sumatra following the Mount Toba super-volcanic eruption about 73,000years ago. Divergence dating, however, has yet to be performed to establish a temporal association with the Toba eruption. Here, we use a Bayesian framework and a relaxed molecular clock model with fossil calibrations to test the Toba hypothesis and to gain a more complete understanding of the evolutionary history of orangutan SFV. As with previous studies, our results show a similar three-clade orangutan SFV phylogeny, along with strong statistical support for SFV-host co-evolution in orangutans. Using Bayesian inference, we date the origin of orangutan SFV to >4.7 million years ago (mya), while the mixed species clade dates to approximately 1.7mya, >1.6 million years older than the Toba super-eruption. These results, combined with fossil and paleogeographic evidence, suggest that the origin of SFV in Sumatran and Bornean orangutans, including the mixed species clade, likely occurred on the mainland of Indo-China during the Late Pliocene and Calabrian stage of the Pleistocene, respectively.


Subject(s)
Genes, Viral , Genome, Viral , Host-Pathogen Interactions/genetics , Pongo/virology , Retroviridae Infections/veterinary , Simian foamy virus/genetics , Animals , Bayes Theorem , Biological Coevolution , Borneo/epidemiology , DNA, Mitochondrial/genetics , Electron Transport Complex IV/genetics , Fossils , Gene Expression , History, Ancient , Indonesia/epidemiology , Pongo/classification , Pongo/genetics , Retroviridae Infections/epidemiology , Retroviridae Infections/history , Retroviridae Infections/virology , Simian foamy virus/classification , Volcanic Eruptions/history
12.
PLoS One ; 11(6): e0157709, 2016.
Article in English | MEDLINE | ID: mdl-27310836

ABSTRACT

Zoonotic transmission of simian retroviruses in West-Central Africa occurring in primate hunters has resulted in pandemic spread of human immunodeficiency viruses (HIVs) and human T-lymphotropic viruses (HTLVs). While simian foamy virus (SFV) and simian T- lymphotropic virus (STLV)-like infection were reported in healthy persons exposed to nonhuman primates (NHPs) in West-Central Africa, less is known about the distribution of these viruses in Western Africa and in hospitalized populations. We serologically screened for SFV and STLV infection using 1,529 specimens collected between 1985 and 1997 from Côte d'Ivoire patients with high HIV prevalence. PCR amplification and analysis of SFV, STLV, and HIV/SIV sequences from PBMCs was used to investigate possible simian origin of infection. We confirmed SFV antibodies in three persons (0.2%), two of whom were HIV-1-infected. SFV polymerase (pol) and LTR sequences were detected in PBMC DNA available for one HIV-infected person. Phylogenetic comparisons with new SFV sequences from African guenons showed infection likely originated from a Chlorocebus sabaeus monkey endemic to Côte d'Ivoire. 4.6% of persons were HTLV seropositive and PCR testing of PBMCs from 15 HTLV seroreactive persons identified nine with HTLV-1 and one with HTLV-2 LTR sequences. Phylogenetic analysis showed that two persons had STLV-1-like infections, seven were HTLV-1, and one was an HTLV-2 infection. 310/858 (53%), 8/858 (0.93%), and 18/858 (2.1%) were HIV-1, HIV-2, and HIV-positive but undifferentiated by serology, respectively. No SIV sequences were found in persons with HIV-2 antibodies (n = 1) or with undifferentiated HIV results (n = 7). We document SFV, STLV-1-like, and dual SFV/HIV infection in Côte d'Ivoire expanding the geographic range for zoonotic simian retrovirus transmission to West Africa. These findings highlight the need to define the public health consequences of these infections. Studying dual HIV-1/SFV infections in immunocompromised populations may provide a new opportunity to better understand SFV pathogenicity and transmissibility in humans.


Subject(s)
Deltaretrovirus Infections/diagnosis , HIV Infections/diagnosis , HIV-1/isolation & purification , Retroviridae Infections/diagnosis , Simian foamy virus/isolation & purification , Animals , Antibodies, Viral/blood , Chlorocebus aethiops , Coinfection , Cote d'Ivoire/epidemiology , DNA, Viral/genetics , Deltaretrovirus Infections/epidemiology , Deltaretrovirus Infections/virology , HIV Infections/epidemiology , HIV Infections/virology , HIV-1/classification , HIV-1/genetics , HIV-2/classification , HIV-2/genetics , HIV-2/isolation & purification , Human T-lymphotropic virus 1/classification , Human T-lymphotropic virus 1/genetics , Human T-lymphotropic virus 1/isolation & purification , Human T-lymphotropic virus 2/classification , Human T-lymphotropic virus 2/genetics , Human T-lymphotropic virus 2/isolation & purification , Humans , Leukocytes, Mononuclear/virology , Monkey Diseases/diagnosis , Monkey Diseases/epidemiology , Monkey Diseases/virology , Phylogeny , Retroviridae Infections/epidemiology , Retroviridae Infections/virology , Simian foamy virus/classification , Simian foamy virus/genetics
13.
Retrovirology ; 12: 94, 2015 Nov 14.
Article in English | MEDLINE | ID: mdl-26576961

ABSTRACT

BACKGROUND: While simian foamy viruses have co-evolved with their primate hosts for millennia, most scientific studies have focused on understanding infection in Old World primates with little knowledge available on the epidemiology and natural history of SFV infection in New World primates (NWPs). To better understand the geographic and species distribution and evolutionary history of SFV in NWPs we extend our previous studies in Brazil by screening 15 genera consisting of 29 NWP species (140 monkeys total), including five genera (Brachyteles, Cacajao, Callimico, Mico, and Pithecia) not previously analyzed. Monkey blood specimens were tested using a combination of both serology and PCR to more accurately estimate prevalence and investigate transmission patterns. Sequences were phylogenetically analyzed to infer SFV and host evolutionary histories. RESULTS: The overall serologic and molecular prevalences were 42.8 and 33.6 %, respectively, with a combined assay prevalence of 55.8 %. Discordant serology and PCR results were observed for 28.5 % of the samples, indicating that both methods are currently necessary for estimating NWP SFV prevalence. SFV prevalence in sexually mature NWPs with a positive result in any of the WB or PCR assays was 51/107 (47.7 %) compared to 20/33 (61 %) for immature animals. Epidemiological analyses revealed an increase in SFV prevalence with age in captive Cebus monkeys. Phylogenetic analysis identified novel SFVs in Cacajao, Leontopithecus, and Chiropotes species that had 6-37 % nucleotide divergence to other NWP SFV. Comparison of host and SFV phylogenies showed an overall cospeciation evolutionary history with rare ancient and contemporaneous host-switching for Saimiri and Leontopithecus and Cebus xanthosternos, respectively. CONCLUSIONS: We identified novel SFV in four neotropical monkey genera in Brazil and demonstrate that SFV prevalence increases with age in Cebus monkeys. Importantly, our test results suggest that both molecular and serological screening are currently required to accurately determine infection with NWP SFV. Our study significantly expands knowledge of the epidemiology and natural history of NWP SFVs. The tools and information provided in our study will facilitate further investigation of SFV in NWPs and the potential for zoonotic infection with these viruses.


Subject(s)
Monkey Diseases , Platyrrhini , Retroviridae Infections/veterinary , Simian foamy virus/classification , Simian foamy virus/genetics , Age Factors , Animals , Brazil/epidemiology , Humans , Monkey Diseases/epidemiology , Monkey Diseases/virology , Phylogeny , Polymerase Chain Reaction , Prevalence , Retroviridae Infections/epidemiology , Retroviridae Infections/transmission , Retroviridae Infections/virology , Simian foamy virus/isolation & purification , Zoonoses/transmission , Zoonoses/virology
14.
Retrovirology ; 12: 89, 2015 Oct 29.
Article in English | MEDLINE | ID: mdl-26514626

ABSTRACT

BACKGROUND: Although simian foamy viruses (SFV) are the only exogenous retroviruses to infect New World monkeys (NWMs), little is known about their evolutionary history and epidemiology. Previous reports show distinct SFVs among NWMs but were limited to small numbers of captive or wild monkeys from five (Cebus, Saimiri, Ateles, Alouatta, and Callithrix) of the 15 NWM genera. Other studies also used only PCR testing or serological assays with limited validation and may have missed infection in some species. We developed and validated new serological and PCR assays to determine the prevalence of SFV in blood specimens from a large number of captive NWMs in the US (n = 274) and in captive and wild-caught NWMs (n = 236) in Peruvian zoos, rescue centers, and illegal trade markets. Phylogenetic and co-speciation reconciliation analyses of new SFV polymerase (pol) and host mitochondrial cytochrome B sequences, were performed to infer SFV and host co-evolutionary histories. RESULTS: 124/274 (45.2 %) of NWMs captive in the US and 59/157 (37.5 %) of captive and wild-caught NWMs in Peru were SFV WB-positive representing 11 different genera (Alouatta, Aotus, Ateles, Cacajao, Callithrix, Cebus, Lagothrix, Leontopithecus, Pithecia, Saguinus and Saimiri). Seroprevalences were lower at rescue centers (10/53, 18.9 %) compared to zoos (46/97, 47.4 %) and illegal trade markets (3/7, 8/19, 42.9 %) in Peru. Analyses showed that the trees of NWM hosts and SFVs have remarkably similar topologies at the level of species and sub-populations suggestive of co-speciation. Phylogenetic reconciliation confirmed 12 co-speciation events (p < 0.002) which was further supported by obtaining highly similar divergence dates for SFV and host genera and correlated SFV-host branch times. However, four ancient cross-genus transmission events were also inferred for Pitheciinae to Atelidae, Cacajao to ancestral Callithrix or Cebus monkeys, between Callithrix and Cebus monkeys, and Lagothrix to Alouatta. CONCLUSIONS: We demonstrate a broad distribution and stable co-speciation history of SFV in NWMs at the species level. Additional studies are necessary to further explore the epidemiology and natural history of SFV infection of NWMs and to determine the zoonotic potential for persons exposed to infected monkeys in captivity and in the wild.


Subject(s)
Monkey Diseases/epidemiology , Platyrrhini/virology , Primates/virology , Retroviridae Infections/veterinary , Simian foamy virus/genetics , Simian foamy virus/isolation & purification , Animals , Biological Evolution , Humans , Monkey Diseases/virology , Peru/epidemiology , Phylogeny , Polymerase Chain Reaction , Retroviridae Infections/blood , Retroviridae Infections/epidemiology , Sensitivity and Specificity , Seroepidemiologic Studies , Serologic Tests
15.
J Virol ; 89(24): 12480-91, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26446599

ABSTRACT

UNLABELLED: Simian foamy virus (SFV) is a ubiquitous retrovirus in nonhuman primates (NHPs) that can be transmitted to humans, mostly through severe bites. In the past few years, our laboratory has identified more than 50 hunters from central Africa infected with zoonotic SFVs. Analysis of the complete sequences of five SFVs obtained from these individuals revealed that env was the most variable gene. Furthermore, recombinant SFV strains, some of which involve sequences in the env gene, were recently identified. Here, we investigated the variability of the env genes of zoonotic SFV strains and searched for possible recombinants. We sequenced the complete env gene or its surface glycoprotein region (SU) from DNA amplified from the blood of (i) a series of 40 individuals from Cameroon or Gabon infected with a gorilla or chimpanzee foamy virus (FV) strain and (ii) 1 gorilla and 3 infected chimpanzees living in the same areas as these hunters. Phylogenetic analyses revealed the existence of two env variants among both the gorilla and chimpanzee FV strains that were present in zoonotic and NHP strains. These variants differ greatly (>30% variability) in a 753-bp-long region located in the receptor-binding domain of SU, whereas the rest of the gene is very conserved. Although the organizations of the Env protein sequences are similar, the potential glycosylation patterns differ between variants. Analysis of recombination suggests that the variants emerged through recombination between different strains, although all parental strains could not be identified. IMPORTANCE: SFV infection in humans is a great example of a zoonotic retroviral infection that has not spread among human populations, in contrast to human immunodeficiency viruses (HIVs) and human T-lymphotropic viruses (HTLVs). Recombination was a major mechanism leading to the emergence of HIV. Here, we show that two SFV molecular envelope gene variants circulate among ape populations in Central Africa and that both can be transmitted to humans. These variants differ greatly in the SU region that corresponds to the part of the Env protein in contact with the environment. These variants may have emerged through recombination between SFV strains infecting different NHP species.


Subject(s)
Gene Products, env/genetics , Phylogeny , Recombination, Genetic , Retroviridae Infections/genetics , Simian foamy virus/genetics , Animals , Ape Diseases/epidemiology , Ape Diseases/genetics , Ape Diseases/virology , Cameroon , Gabon , Gorilla gorilla , Humans , Pan troglodytes , Retroviridae Infections/epidemiology , Retroviridae Infections/veterinary
16.
Virus Res ; 208: 1-6, 2015 Oct 02.
Article in English | MEDLINE | ID: mdl-26047587

ABSTRACT

Foamy viruses infect a wide range of placental mammals, including primates. However, despite of great diversity of New World primates, only three strains of neotropical simian foamy viruses (SFV) have been described. Only after 40 years since serological characterization, the complete sequence of an SFVcap strain infecting a family of six capuchin monkeys (Sapajus xanthosternos) was obtained. Co-culture of primate peripheral blood mononuclear cells with Cf2Th canine cells was established and monitored for the appearance of cytopathic effects, PCR amplification of integrated SFV proviral genome and viral reverse transcriptase activity. The novel SFVcap was fully sequenced through a next-generation sequencing protocol. Phylogenetic analysis of the complete genome grouped SFVcap and SFVmar, both infecting primate species of the Cebidae family with a genetic similarity of approximately 85%. Similar ORF sizes were observed among SFV from neotropical primates, and env and pol genes were the most conserved. Neotropical SFV presented the smallest LTRs among exogenous mammalians. The novel SFVcap strain provides a valuable research tool for the FV community.


Subject(s)
Genome, Viral , Monkey Diseases/virology , Retroviridae Infections/veterinary , Simian foamy virus/isolation & purification , Animals , Brazil , Cebus/virology , Dogs , Molecular Sequence Data , Phylogeny , Retroviridae Infections/virology , Simian foamy virus/classification , Simian foamy virus/genetics
17.
J Virol ; 89(14): 7414-6, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25926651

ABSTRACT

Simian foamy viruses (SVF) are ubiquitous in nonhuman primates (NHP). SFV can be zoonotically transmitted to humans who either work with or live commensally with NHP. We analyzed the blood of 45 Bangladeshi performing monkey owners (an ethnic group called the Bedey) for SFV infection. Surprisingly, a PCR assay failed to detect SFV infection in any of these participants. This is in contrast to our previously reported infection rate of about 5% among Bangladeshi villagers.


Subject(s)
Retroviridae Infections/epidemiology , Simian foamy virus/isolation & purification , Transients and Migrants , Zoonoses/epidemiology , Animals , Bangladesh/epidemiology , Female , Humans , Macaca , Male , Polymerase Chain Reaction , RNA, Viral/blood , Simian foamy virus/genetics
18.
Curr Opin Virol ; 10: 47-55, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25698621

ABSTRACT

Most viral pathogens that have emerged in humans have originated from various animal species. Emergence is a multistep process involving an initial spill-over of the infectious agent into single individuals and its subsequent dissemination into the human population. Similar to simian immunodeficiency viruses and simian T lymphotropic viruses, simian foamy viruses (SFV) are retroviruses that are widespread among non-human primates and can be transmitted to humans, giving rise to a persistent infection, which seems to be controlled in the case of SFV. In this review, we present current data on the discovery, cross-species transmission, and molecular evolution of SFV in human populations initially infected and thus at risk for zoonotic emergence.


Subject(s)
Retroviridae Infections/immunology , Retroviridae Infections/transmission , Simian foamy virus/genetics , Simian foamy virus/immunology , Animals , DNA, Viral/genetics , Evolution, Molecular , Genome, Viral , Humans , Primate Diseases/virology , Primates , Retroviridae Infections/veterinary , Retroviridae Infections/virology , Simian foamy virus/isolation & purification , Simian foamy virus/physiology , Simian foamy virus/ultrastructure , Viral Structural Proteins/genetics , Viral Structural Proteins/metabolism , Viral Tropism , Zoonoses
19.
Gene ; 548(1): 149-54, 2014 Sep 10.
Article in English | MEDLINE | ID: mdl-25017058

ABSTRACT

Foamy viruses belong to the genus Spumavirus of the family Retroviridae and have been isolated from many mammalian species. It was reported that simian foamy viruses (SFVs) have co-evolved with host species. In this study, we isolated four strains (WK1, WK2, AR1 and AR2) of SFV (named SFVjm) from Japanese macaques (Macaca fuscata) in main island Honshu of Japan. We constructed an infectious molecular clone of SFVjm strain WK1, termed pJM356. The virus derived from the clone replicated and induced syncytia in human (human embryonic kidney 293T cells), African green monkey (Vero cells) and mouse cell lines (Mus dunni tail fibroblast cells). Phylogenetic analysis also revealed that these four SFVjm strains formed two distinct SFVjm clusters. SFVjm strains WK1 and WK2 and SFV isolated from Taiwanese macaques (Macaca cyclopis) formed one cluster, whereas strains AR1 and AR2 formed the other cluster with SFV isolated from a rhesus macaque (Macaca mulatta).


Subject(s)
Phylogeny , Simian foamy virus/genetics , Simian foamy virus/pathogenicity , Animals , Chlorocebus aethiops , DNA, Mitochondrial , HEK293 Cells/virology , Humans , Japan , Macaca/virology , Mice , Molecular Sequence Data , Promoter Regions, Genetic , Sequence Homology, Amino Acid , Simian foamy virus/isolation & purification , Spumavirus/genetics , Spumavirus/isolation & purification , Vero Cells , Viral Proteins/genetics
20.
ScientificWorldJournal ; 2014: 487969, 2014.
Article in English | MEDLINE | ID: mdl-25009830

ABSTRACT

Retroviral vectors are potent tools for gene delivery and various biomedical applications. To accomplish a gene transfer task successfully, retroviral vectors must effectively transduce diverse cell cultures at different phases of a cell cycle. However, very promising retroviral vectors based on the foamy viral (FV) backbone lack the capacity to efficiently transduce quiescent cells. It is hypothesized that this phenomenon might be explained as the inability of foamy viruses to form a pre-integration complex (PIC) with nuclear import activity in growth-arrested cells, which is the characteristic for lentiviruses (HIV-1). In this process, the HIV-1 central polypurine tract (cPPT) serves as a primer for plus-strand synthesis to produce a "flap" element and is believed to be crucial for the subsequent double-stranded cDNA formation of all retroviral RNA genomes. In this study, the effects of the lentiviral cPPT element on the FV transduction potential in dividing and growth-arrested (G1/S phase) adenocarcinomic human alveolar basal epithelial (A549) cells are investigated by experimental and theoretical methods. The results indicated that the HIV-1 cPPT element in a foamy viral vector background will lead to a significant reduction of the FV transduction and viral titre in growth-arrested cells due to the absence of PICs with nuclear import activity.


Subject(s)
Cell Division/physiology , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , Growth Inhibitors/administration & dosage , HIV-1/genetics , Simian foamy virus/genetics , Cell Division/drug effects , Cell Line, Tumor , Forecasting , Humans , Lentivirus/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...