Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 88
Filter
1.
Bioengineered ; 12(2): 12498-12508, 2021 12.
Article in English | MEDLINE | ID: mdl-34927536

ABSTRACT

At present, the mechanism of siSCN9A in Vincristine (VCR)-induced neuropathic pain (NP) is still unclear. This study aimed to explore the analgesic mechanism of lentivirus-siSCN9A (LV-siSCN9A) infected neurons against NP. 40 male Sprague-Dawley (SD) rats were divided into a control group (injected with normal saline), a model group (VCR-induced NP model), a LV-SC group (NP model mice were injected with LV-SC-infected dorsal root ganglia (DRG) neuron cells under the microscope), and a LV-siSCN9A group (NP model mice were injected with LV-siSCN9A-infected DRG neuron cells under the microscope, with 10 rats in each group. The changes of mechanical withdrawal threshold (MWT) and thermal withdrawal latency (TWL) of rats in different groups were detected by behavior testing, the Nav1.7 changes in each group were detected by immunofluorescence double standard and Western-blot method. It was found that compared with the control group, the MWT and TWL of the rats in model group were significantly decreased (P < 0.05), and the expression levels of Nav1.7 messenger ribonucleic acid (mRNA) and proteins were significantly increased (P < 0.05). Compared with LV-SC group, the MWT and TWL of rats in LV-siSCN9A group were significantly increased (P < 0.05), the expression levels of Nav1.7 mRNA and proteins were significantly decreased (P < 0.05), and the CGRP expression of spinal dorsal horn was significantly decreased. It was concluded that the LV-siSCN9A infected neurons could play an analgesic role by down-regulating Nav1.7 expression induced by VCR in NP model.


Subject(s)
Lentivirus Infections/virology , Lentivirus/pathogenicity , Neuralgia/chemically induced , Neuralgia/virology , Neurons/drug effects , Neurons/virology , Vincristine/pharmacology , Analgesia/methods , Animals , Disease Models, Animal , Ganglia, Spinal/drug effects , Ganglia, Spinal/virology , Lentivirus Infections/genetics , Male , Mice , NAV1.7 Voltage-Gated Sodium Channel/genetics , Neuralgia/genetics , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley
2.
Viruses ; 13(7)2021 07 01.
Article in English | MEDLINE | ID: mdl-34372496

ABSTRACT

Maedi-visna virus (MVV) and caprine arthritis encephalitis virus (CAEV), referred to as small ruminant lentiviruses (SRLVs), belong to the genus Lentivirus of the Retroviridae family. SRLVs infect both sheep and goats, causing significant economic losses and animal welfare damage. Recent findings suggest an association between serological status and allelic variants of different genes such as TMEM154, TLR9, MYD88 and CCR5. The aim of this work was to investigate the role of specific polymorphisms of these genes in SRLVs infection in some sheep flocks in Italy. In addition to those already known, novel variants in the TMEM154 (P7H, I74V, I105V) gene were detected in this study. The risk of infection was determined finding an association between the serological status and polymorphisms P7H, E35K, N70I, I74V, I105V of TMEM154, R447Q, A462S and G520R in TLR9 gene, H176H* and K190K* in MYD88 genes, while no statistical association was observed for the 4-bp deletion of the CCR5 gene. Since no vaccines or treatments have been developed, a genetically based approach could be an innovative strategy to prevent and to control SRLVs infection. Our findings are an important starting point in order to define the genetic resistance profile towards SRLVs infection.


Subject(s)
Disease Resistance/genetics , Lentivirus Infections/genetics , Lentivirus Infections/veterinary , Lentivirus/genetics , Membrane Proteins/genetics , Myeloid Differentiation Factor 88/genetics , Polymorphism, Single Nucleotide , Receptors, CCR5/genetics , Toll-Like Receptor 9/genetics , Animals , Genetic Variation , Italy , Lentivirus/classification , Lentivirus Infections/immunology , Lentivirus Infections/prevention & control , Membrane Proteins/classification , Membrane Proteins/immunology , Risk Factors , Sheep , Sheep Diseases/genetics , Sheep Diseases/immunology , Sheep Diseases/virology
3.
Viruses ; 13(6)2021 06 07.
Article in English | MEDLINE | ID: mdl-34200141

ABSTRACT

The transmission of viruses from animal hosts into humans have led to the emergence of several diseases. Usually these cross-species transmissions are blocked by host restriction factors, which are proteins that can block virus replication at a specific step. In the natural virus host, the restriction factor activity is usually suppressed by a viral antagonist protein, but this is not the case for restriction factors from an unnatural host. However, due to ongoing viral evolution, sometimes the viral antagonist can evolve to suppress restriction factors in a new host, enabling cross-species transmission. Here we examine the classical case of this paradigm by reviewing research on APOBEC3 restriction factors and how they can suppress human immunodeficiency virus (HIV) and simian immunodeficiency virus (SIV). APOBEC3 enzymes are single-stranded DNA cytidine deaminases that can induce mutagenesis of proviral DNA by catalyzing the conversion of cytidine to promutagenic uridine on single-stranded viral (-)DNA if they escape the HIV/SIV antagonist protein, Vif. APOBEC3 degradation is induced by Vif through the proteasome pathway. SIV has been transmitted between Old World Monkeys and to hominids. Here we examine the adaptations that enabled such events and the ongoing impact of the APOBEC3-Vif interface on HIV in humans.


Subject(s)
APOBEC Deaminases/genetics , Host-Pathogen Interactions/genetics , Lentivirus Infections/genetics , Lentivirus Infections/transmission , Lentiviruses, Primate/physiology , Viral Zoonoses/transmission , Animals , Gene Products, vif/chemistry , Gene Products, vif/metabolism , HIV Infections/genetics , HIV Infections/transmission , HIV Infections/virology , HIV-1/physiology , Humans , Lentivirus Infections/virology , Protein Binding , Protein Isoforms , Structure-Activity Relationship , vif Gene Products, Human Immunodeficiency Virus/chemistry , vif Gene Products, Human Immunodeficiency Virus/metabolism
5.
Arch Virol ; 165(7): 1557-1567, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32356187

ABSTRACT

Caprine arthritis-encephalitis (CAE) is a chronic progressive infectious disease caused by caprine arthritis-encephalitis virus (CAEV) that seriously threatens the goat industry. Chronic infection and life-long multi-tissue inflammation are the typical features of the disease. Innate antiviral immunity is essential for the host defense system that rapidly recognizes and eliminates invading viruses. Interferon ß (IFN-ß) is important for innate immunity and regulates immunity against a broad spectrum of viruses. To investigate the details of the IFN-ß response to CAEV infection, the effects of six viral proteins and the molecular mechanisms by which they affect IFN-ß production were analyzed. Overexpression of DU and Vif promote virus proliferation and inhibit the production of IFN-ß. qRT-PCR and luciferase reporter assays showed that overexpression of Vif inhibits the expression of luciferase under the control of the ISRE, NF-κB or IFN-ß promoter but does not affect the expression of IFN-ß activated by IRF3, indicating that Vif negatively regulates IFN-ß production by affecting upstream signal transduction of IRF3. Amino acids 149-164 of Vif were found to be necessary for the inhibitory effect of IFN-ß production. Our results indicate that CAEV evades surveillance and clearance by intracellular innate immunity by downregulating IFN-ß production.


Subject(s)
Arthritis-Encephalitis Virus, Caprine/immunology , Gene Products, vif/immunology , Goat Diseases/immunology , Interferon-beta/immunology , Lentivirus Infections/veterinary , Animals , Arthritis-Encephalitis Virus, Caprine/genetics , Gene Products, vif/genetics , Goat Diseases/genetics , Goat Diseases/virology , Goats , Host-Pathogen Interactions , Immunity, Innate , Interferon-beta/genetics , Lentivirus Infections/genetics , Lentivirus Infections/immunology , Lentivirus Infections/virology , NF-kappa B/genetics , NF-kappa B/immunology
6.
J Dairy Sci ; 103(7): 6407-6411, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32331882

ABSTRACT

Caprine arthritis encephalitis (CAE) is a chronic disease caused by a retrovirus from the Lentivirus genus. No effective vaccines or treatments exist, and therefore genetic selection for CAE resistance might be a feasible alternative. To our best knowledge, no other studies have investigated the genetic architecture of CAE resistance in dairy goats. In this context, this study was designed to estimate genetic parameters for CAE infection in Alpine and Saanen goats using a Bayesian threshold model. A total of 542 adult goats (and >3-generation pedigree), which were group-housed in a population with high CAE prevalence, were tested based on a serological infection assessment test (negative = 1 or positive = 2) and used for this study. Genetic parameters were estimated using the BLUPF90 family programs. There was considerable genetic variability for CAE resistance, and pedigree-based heritability was significantly different from zero (0.026 < heritability < 0.128). Our findings indicate that the prevalence of CAE in goat herds can be reduced or eliminated through direct genetic selection for CAE resistance in addition to proper management strategies.


Subject(s)
Arthritis-Encephalitis Virus, Caprine , Genetic Predisposition to Disease , Goat Diseases/virology , Lentivirus Infections/veterinary , Animals , Bayes Theorem , Goat Diseases/epidemiology , Goats , Lentivirus Infections/genetics , Lentivirus Infections/virology
7.
Viruses ; 11(9)2019 09 07.
Article in English | MEDLINE | ID: mdl-31500260

ABSTRACT

Feline immunodeficiency virus (FIV) is a naturally occurring T-cell tropic lentiviral disease of felids with many similarities to HIV/AIDS in humans. Similar to primate lentiviral-host interactions, feline APOBEC3 (A3) has been shown to inhibit FIV infection in a host-specific manner and feline A3 degradation is mediated by FIV Vif. Further, infection of felids with non-native FIV strains results in restricted viral replication in both experimental and naturally occurring infections. However, the link between molecular A3-Vif interactions and A3 biological activity during FIV infection has not been well characterized. We thus examined expression of the feline A3 genes A3Z2, A3Z3 and A3Z2-Z3 during experimental infection of domestic cats with host-adapted domestic cat FIV (referred to as FIV) and non-adapted Puma concolor FIV (referred to as puma lentivirus, PLV). We determined A3 expression in different tissues and blood cells from uninfected, FIV-infected, PLV-infected and FIV/PLV co-infected cats; and in purified blood cell subpopulations from FIV-infected and uninfected cats. Additionally, we evaluated regulation of A3 expression by cytokines, mitogens, and FIV infection in cultured cells. In all feline cells and tissues studied, there was a striking difference in expression between the A3 genes which encode FIV inhibitors, with A3Z3 mRNA abundance exceeding that of A3Z2-Z3 by 300-fold or more. Interferon-alpha treatment of cat T cells resulted in upregulation of A3 expression, while treatment with interferon-gamma enhanced expression in cat cell lines. In cats, secondary lymphoid organs and peripheral blood mononuclear cells (PBMC) had the highest basal A3 expression levels and A3 genes were differentially expressed among blood T cells, B cells, and monocytes. Acute FIV and PLV infection of cats, and FIV infection of primary PBMC resulted in no detectable change in A3 expression with the exception of significantly elevated A3 expression in the thymus, the site of highest FIV replication. We conclude that cat A3 expression is regulated by cytokine treatment but, by and large, lentiviral infection did not appear to alter expression. Differences in A3 expression in different blood cell subsets did not appear to impact FIV viral replication kinetics within these cells. Furthermore, the relative abundance of A3Z3 mRNA compared to A3Z2-Z3 suggests that A3Z3 may be the major active anti-lentiviral APOBEC3 gene product in domestic cats.


Subject(s)
Cytosine Deaminase/immunology , Feline Acquired Immunodeficiency Syndrome/enzymology , Immunodeficiency Virus, Feline/physiology , Lentivirus Infections/veterinary , Animals , B-Lymphocytes/immunology , Cats , Cytosine Deaminase/genetics , Feline Acquired Immunodeficiency Syndrome/genetics , Feline Acquired Immunodeficiency Syndrome/immunology , Feline Acquired Immunodeficiency Syndrome/virology , Host-Pathogen Interactions , Immunodeficiency Virus, Feline/genetics , Lentivirus Infections/enzymology , Lentivirus Infections/genetics , Lentivirus Infections/immunology , T-Lymphocytes/immunology , Virus Replication
8.
BMC Vet Res ; 15(1): 230, 2019 Jul 06.
Article in English | MEDLINE | ID: mdl-31277645

ABSTRACT

BACKGROUND: The small ruminant lentiviruses (SRLVs) are a heterogeneous group of viruses that includes caprine arthritis encephalitis virus (CAEV) and Maedi-Visna virus (MVV). SRLVs affect the production and welfare of sheep and goats worldwide. There is currently no effective treatment. Their high mutation rate precludes vaccine development, making innovative control measures necessary. A variant of the chemokine (C-C motif) receptor 5 (CCR5) gene is reportedly involved in resistance to human immunodeficiency (HIV) infection in humans and to SRLV in sheep. The aim of this study was to analyse the genetic structure and variability of the CCR5 gene in goats and to carry out a cross-sectional study to investigate the role of CCR5 genetic variants in controlling susceptibility/resistance to CAEV. RESULTS: The variant g.1059 T located in the promoter region revealed an interesting association with high proviral loads (a 2.8-fold increased risk). A possible explanation could be an alteration of the transcriptional level. Overexpression of the CCR5 receptor on the cell surface may increase virus internalization and proviral load as a consequence. CONCLUSIONS: Our findings could be advantageously used to reduce the susceptibility of goat herds to CAEV by negatively selecting animals carrying the g.1059 T mutation. Eliminating animals predisposed to high proviral loads could also limit the development of clinical signs and the spread of the virus, since these animals are also highly efficient in shedding the virus.


Subject(s)
Goat Diseases/genetics , Goat Diseases/virology , Lentivirus Infections/veterinary , Receptors, CCR5/genetics , Animals , Arthritis-Encephalitis Virus, Caprine , Cross-Sectional Studies , Genetic Predisposition to Disease , Goats , Lentivirus Infections/genetics , Polymorphism, Single Nucleotide , Promoter Regions, Genetic , Proviruses , Sequence Analysis, DNA
9.
Biomed Res Int ; 2019: 4279573, 2019.
Article in English | MEDLINE | ID: mdl-31214614

ABSTRACT

Small ruminant lentiviruses (SRLVs) belong to the genus Lentivirus in the Retroviridae family. There are five genotypes (A, B, C, D, and E), where genotypes A and B have a global distribution and genotypes C, D, and E are limited to Europe. The presence of SRLV has been confirmed in Mexico, with genotype B detected in the central region of the country. We examined the presence of SRLVs and genotype prevalence in 1014 sheep and 1383 goats from 12 Mexican states. Using a commercial competitive ELISA (cELISA) test, we detected SRLV antibodies in 107 sheep (10.55%) and 466 goats (33.69%). We used an endpoint PCR to amplify the LTR region on seropositive animals. A total of 50 sheep and 75 goats tested positive via PCR. Positive amplicons from 11 sheep and 17 goats from ten Mexican States were cloned and sequenced. With the LTR sequence data obtained in this study, a phylogenetic analysis was performed; we also constructed a phylogenetic tree using the obtained sequences and GenBank's available sequences. All studied sequences were associated with genotype B, specifically with the FESC-752 isolate previously identified in Mexico. Highly conserved transcription factor binding sites were observed in analyzed alignments, such as AML (vis), AP-4, and TATA box. However, we identified nucleotide differences at site AP-1 that suggest function loss. Our study found that ovine and caprine genotype B SRLVs are widely distributed in Mexico; a highly conserved LTR region among the sequences evaluated in this study was also found.


Subject(s)
Genotype , Goat Diseases , Goats/virology , Lentivirus Infections/genetics , Lentivirus/genetics , Sheep Diseases , Sheep/virology , Terminal Repeat Sequences , Animals , Female , Goat Diseases/genetics , Goat Diseases/virology , Male , Mexico , Sheep Diseases/genetics , Sheep Diseases/virology
10.
BMC Genomics ; 20(1): 62, 2019 Jan 18.
Article in English | MEDLINE | ID: mdl-30658565

ABSTRACT

BACKGROUND: MicroRNAs (miRNAs) are short endogenous, single-stranded, noncoding small RNA molecules of approximately 22 nucleotides in length. They regulate gene expression posttranscriptionally by silencing mRNA expression, thus orchestrating many physiological processes. The Small Ruminant Lentiviruses (SRLV) group includes the Visna Maedi Virus (VMV) and Caprine Arthritis Encephalitis (CAEV) viruses, which cause a disease in sheep and goats characterized by pneumonia, mastitis, arthritis and encephalitis. Their main target cells are from the monocyte/macrophage lineage. To date, there are no studies on the role of miRNAs in this viral disease. RESULTS: Using RNA-seq technology and bioinformatics analysis, the expression levels of miRNAs during different clinical stages of infection were studied. A total of 212 miRNAs were identified, of which 46 were conserved sequences in other species but found for the first time in sheep, and 12 were completely novel. Differential expression analysis comparing the uninfected and seropositive groups showed changes in several miRNAs; however, no significant differences were detected between seropositive asymptomatic and diseased sheep. The robust increase in the expression level of oar-miR-21 is consistent with its increased expression in other viral diseases. Furthermore, the target prediction of the dysregulated miRNAs revealed that they control genes involved in proliferation-related signalling pathways, such as the PI3K-Akt, AMPK and ErbB pathways. CONCLUSIONS: To the best of our knowledge, this is the first study reporting miRNA profiling in sheep in response to SRLV infection. The known functions of oar-miR-21 as a regulator of inflammation and proliferation appear to be a possible cause of the lesions caused in the sheep's lungs. This miRNA could be an indicator for the severity of the lung lesions, or a putative target for therapeutic intervention.


Subject(s)
Lentivirus Infections/veterinary , Lung/metabolism , MicroRNAs/genetics , Sequence Analysis, RNA/methods , Sheep Diseases/genetics , Animals , Arthritis-Encephalitis Virus, Caprine/physiology , Cluster Analysis , Female , Gene Expression Profiling , Gene Regulatory Networks , Host-Pathogen Interactions , Lentivirus Infections/genetics , Lentivirus Infections/virology , Lung/pathology , Lung/virology , Sheep , Sheep Diseases/virology , Visna-maedi virus/physiology
11.
Nat Commun ; 9(1): 5193, 2018 12 05.
Article in English | MEDLINE | ID: mdl-30518925

ABSTRACT

Immunosuppression is a hallmark of tumor progression, and treatments that inhibit or deplete monocytic myeloid-derived suppressive cells could promote anti-tumor immunity. c-FLIP is a central regulator of caspase-8-mediated apoptosis and necroptosis. Here we show that low-dose cytotoxic chemotherapy agents cause apoptosis linked to c-FLIP down-regulation selectively in monocytes. Enforced expression of c-FLIP or viral FLIP rescues monocytes from cytotoxicity and concurrently induces potent immunosuppressive activity, in T cell cultures and in vivo models of tumor progression and immunotherapy. FLIP-transduced human blood monocytes can suppress graft versus host disease. Neither expression of FLIP in granulocytes nor expression of other anti-apoptotic genes in monocytes conferred immunosuppression, suggesting that FLIP effects on immunosuppression are specific to monocytic lineage and distinct from death inhibition. Mechanistically, FLIP controls a broad transcriptional program, partially by NF-κB activation. Therefore, modulation of FLIP in monocytes offers a means to elicit or block immunosuppressive myeloid cells.


Subject(s)
CASP8 and FADD-Like Apoptosis Regulating Protein/immunology , Lentivirus Infections/immunology , Monocytes/immunology , NF-kappa B/immunology , Apoptosis , CASP8 and FADD-Like Apoptosis Regulating Protein/genetics , Cells, Cultured , Humans , Immunosuppression Therapy , Lentivirus/physiology , Lentivirus Infections/genetics , Lentivirus Infections/physiopathology , Lentivirus Infections/virology , Myeloid Cells/immunology , NF-kappa B/genetics
12.
Viruses ; 10(8)2018 08 17.
Article in English | MEDLINE | ID: mdl-30126090

ABSTRACT

Lentiviruses are infectious agents of a number of animal species, including sheep, goats, horses, monkeys, cows, and cats, in addition to humans. As in the human case, the host immune response fails to control the establishment of chronic persistent infection that finally leads to a specific disease development. Despite intensive research on the development of lentivirus vaccines, it is still not clear which immune responses can protect against infection. Viral mutations resulting in escape from T-cell or antibody-mediated responses are the basis of the immune failure to control the infection. The innate immune response provides the first line of defense against viral infections in an antigen-independent manner. Antiviral innate responses are conducted by dendritic cells, macrophages, and natural killer cells, often targeted by lentiviruses, and intrinsic antiviral mechanisms exerted by all cells. Intrinsic responses depend on the recognition of the viral pathogen-associated molecular patterns (PAMPs) by pathogen recognition receptors (PRRs), and the signaling cascades leading to an antiviral state by inducing the expression of antiviral proteins, including restriction factors. This review describes the latest advances on innate immunity related to the infection by animal lentiviruses, centered on small ruminant lentiviruses (SRLV), equine infectious anemia virus (EIAV), and feline (FIV) and bovine immunodeficiency viruses (BIV), specifically focusing on the antiviral role of the major restriction factors described thus far.


Subject(s)
Gene Expression Regulation/immunology , Immunity, Innate , Interferon Regulatory Factors/immunology , Lentivirus Infections/immunology , Receptors, Pattern Recognition/immunology , Animals , Cats , Cattle , Dendritic Cells/immunology , Dendritic Cells/virology , Goats , Horses , Immunodeficiency Virus, Bovine/immunology , Immunodeficiency Virus, Bovine/pathogenicity , Immunodeficiency Virus, Feline/immunology , Immunodeficiency Virus, Feline/pathogenicity , Infectious Anemia Virus, Equine/immunology , Infectious Anemia Virus, Equine/pathogenicity , Interferon Regulatory Factors/genetics , Killer Cells, Natural/immunology , Killer Cells, Natural/virology , Lentivirus Infections/genetics , Lentivirus Infections/virology , Macrophages/immunology , Macrophages/virology , Pathogen-Associated Molecular Pattern Molecules/immunology , Receptors, Pattern Recognition/genetics , Sheep , T-Lymphocytes/immunology , T-Lymphocytes/virology
13.
Retrovirology ; 15(1): 26, 2018 03 20.
Article in English | MEDLINE | ID: mdl-29554922

ABSTRACT

The InterFeron Induced TransMembrane (IFITM) proteins are interferon stimulated genes that restrict many viruses, including HIV-1. SAMHD1 is another restriction factor blocking replication of HIV-1 and other viruses. Some lentiviruses evolved Vpx/Vpr proteins to degrade SAMHD1. However, this viral antagonism can be perturbed by host mechanisms: a recent study showed that in interferon (IFN) treated THP1 cells, Vpx is unable to degrade SAMHD1. In the present work, we designed an Interferon Stimulated Genes (ISGs)-targeted CRISPR knockout screen in order to identify ISGs regulating this phenotype. We found that IFITM proteins contribute to the IFNα-mediated protection of SAMHD1 by blocking VSV-G-mediated entry of the lentiviral particles delivering Vpx. Consistent with this, IFNα treatment and IFITM expression had no effect when the A-MLV envelope was used for pseudotyping. Using an assay measuring viral entry, we show that IFNα and IFITMs directly block the delivery of Vpx into cells by inhibiting VSV-G viral fusion. Strikingly, the VSV-G envelope was significantly more sensitive to this IFNα entry block and to IFITMs than HIV-1's natural envelope. This highlights important differences between VSV-G pseudotyped and wild-type HIV-1, in particular relative to the pathways they use for viral entry, suggesting that HIV-1 may have evolved to escape restriction factors blocking entry.


Subject(s)
Clustered Regularly Interspaced Short Palindromic Repeats , Host-Pathogen Interactions , Lentivirus Infections/metabolism , Lentivirus Infections/virology , Lentivirus/physiology , Membrane Proteins/metabolism , SAM Domain and HD Domain-Containing Protein 1/metabolism , Cell Line , Gene Knockout Techniques , HIV-1/physiology , Humans , Interferons/pharmacology , Lentivirus Infections/genetics , Membrane Proteins/genetics , Phenotype , Proteolysis/drug effects , Viral Regulatory and Accessory Proteins/metabolism , Virus Internalization
14.
Virology ; 515: 158-164, 2018 02.
Article in English | MEDLINE | ID: mdl-29289827

ABSTRACT

The bovine immunodeficiency virus (BIV) Rev shuttling protein contains nuclear/nucleolar localization signals and nuclear import/export mechanisms that are novel among lentivirus Rev proteins. Several viral proteins localize to the nucleolus, which may play a role in processes that are essential to the outcome of viral replication. Although BIV Rev localizes to the nucleoli of transfected/infected cells and colocalizes with one of its major proteins, nucleophosmin (NPM1, also known as B23), the role of the nucleolus and B23 in BIV replication remains to be determined. Here, we demonstrate for the first time that BIV Rev interacts with nucleolar phosphoprotein B23 in cells. Using small interfering RNA (siRNA) technology, we show that depletion of B23 expression inhibits virus production by BIV-infected cells, indicating that B23 plays an important role in BIV replication. The interaction between Rev and B23 may represent a potential new target for the development of antiviral drugs against lentiviruses.


Subject(s)
Cattle Diseases/metabolism , Gene Products, rev/metabolism , Immunodeficiency Virus, Bovine/physiology , Lentivirus Infections/veterinary , Nuclear Proteins/metabolism , Virus Replication , Animals , Cattle , Cattle Diseases/genetics , Cattle Diseases/virology , Cell Nucleolus/metabolism , Cell Nucleolus/virology , Gene Products, rev/genetics , Immunodeficiency Virus, Bovine/genetics , Lentivirus Infections/genetics , Lentivirus Infections/metabolism , Lentivirus Infections/virology , Nuclear Proteins/genetics , Nucleophosmin
15.
Mol Ther ; 26(2): 634-647, 2018 02 07.
Article in English | MEDLINE | ID: mdl-29221804

ABSTRACT

Efficient transduction tools are a hallmark for both research and therapy development. Here, we introduce new insights into the generation of lentiviral vectors with improved performance by utilizing producer cells with increased production rates of extracellular vesicles through CD9 overexpression. Most human cells secrete small vesicles from their surface (microvesicles) or intraluminal endosome-derived membranes (exosomes). In particular, enhanced levels of the tetraspanin CD9 result in significantly increased numbers of extracellular vesicles with exosome-like features that were secreted from four different human cell lines. Intriguingly, exosomes and their biogenesis route display similarities to lentivirus and we examined the impact of CD9 expression on release and infectivity of recombinant lentiviral vectors. Although the titers of released viral particles were not increased upon production in high CD9 cells, we observed improved performance in terms of both speed and efficiency of lentiviral gene delivery into numerous human cell lines, including HEK293, HeLa, SH-SY5Y, as well as B and T lymphocytes. Here, we demonstrate that enhanced CD9 enables lentiviral transduction in the absence of any pseudotyping viral glycoprotein or fusogenic molecule. Our findings indicate an important role of CD9 for lentiviral vector and exosome biogenesis and point out a remarkable function of this tetraspanin in membrane fusion, viral infectivity, and exosome-mediated horizontal information transfer.


Subject(s)
Exosomes/metabolism , Lentivirus Infections/metabolism , Lentivirus Infections/virology , Lentivirus/physiology , Tetraspanin 29/metabolism , Biomarkers , Cell Line , Extracellular Vesicles/metabolism , Gene Expression , Host-Pathogen Interactions , Humans , Lentivirus Infections/genetics , Tetraspanin 29/genetics , Viral Envelope Proteins/metabolism
17.
PLoS Pathog ; 12(10): e1005865, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27732658

ABSTRACT

Humans express seven human APOBEC3 proteins, which can inhibit viruses and endogenous retroelements through cytidine deaminase activity. The seven paralogs differ in the potency of their antiviral effects, as well as in their antiviral targets. One APOBEC3, APOBEC3C, is exceptional as it has been found to only weakly block viruses and endogenous retroelements compared to other APOBEC3s. However, our positive selection analyses suggest that APOBEC3C has played a role in pathogen defense during primate evolution. Here, we describe a single nucleotide polymorphism in human APOBEC3C, a change from serine to isoleucine at position 188 (I188) that confers potent antiviral activity against HIV-1. The gain-of-function APOBEC3C SNP results in increased enzymatic activity and hypermutation of target sequences when tested in vitro, and correlates with increased dimerization of the protein. The I188 is widely distributed in human African populations, and is the ancestral primate allele, but is not found in chimpanzees or gorillas. Thus, while other hominids have lost activity of this antiviral gene, it has been maintained, or re-acquired, as a more active antiviral gene in a subset of humans. Taken together, our results suggest that APOBEC3C is in fact involved in protecting hosts from lentiviruses.


Subject(s)
Cytidine Deaminase/genetics , Genetic Predisposition to Disease/genetics , Lentivirus Infections/genetics , Polymorphism, Single Nucleotide , Animals , HIV Infections/genetics , Humans , Polymerase Chain Reaction , Primates
18.
J Virol ; 90(15): 6724-6737, 2016 08 01.
Article in English | MEDLINE | ID: mdl-27170760

ABSTRACT

UNLABELLED: Nonhuman primates (NHPs) are a historically important source of zoonotic viruses and are a gold-standard model for research on many human pathogens. However, with the exception of simian immunodeficiency virus (SIV) (family Retroviridae), the blood-borne viruses harbored by these animals in the wild remain incompletely characterized. Here, we report the discovery and characterization of two novel simian pegiviruses (family Flaviviridae) and two novel simian arteriviruses (family Arteriviridae) in wild African green monkeys from Zambia (malbroucks [Chlorocebus cynosuros]) and South Africa (vervet monkeys [Chlorocebus pygerythrus]). We examine several aspects of infection, including viral load, genetic diversity, evolution, and geographic distribution, as well as host factors such as age, sex, and plasma cytokines. In combination with previous efforts to characterize blood-borne RNA viruses in wild primates across sub-Saharan Africa, these discoveries demonstrate that in addition to SIV, simian pegiviruses and simian arteriviruses are widespread and prevalent among many African cercopithecoid (i.e., Old World) monkeys. IMPORTANCE: Primates are an important source of viruses that infect humans and serve as an important laboratory model of human virus infection. Here, we discover two new viruses in African green monkeys from Zambia and South Africa. In combination with previous virus discovery efforts, this finding suggests that these virus types are widespread among African monkeys. Our analysis suggests that one of these virus types, the simian arteriviruses, may have the potential to jump between different primate species and cause disease. In contrast, the other virus type, the pegiviruses, are thought to reduce the disease caused by human immunodeficiency virus (HIV) in humans. However, we did not observe a similar protective effect in SIV-infected African monkeys coinfected with pegiviruses, possibly because SIV causes little to no disease in these hosts.


Subject(s)
Arterivirus Infections/epidemiology , Biological Evolution , Flaviviridae Infections/epidemiology , Genetic Variation , Lentivirus Infections/epidemiology , Viral Load , Africa/epidemiology , Animals , Animals, Wild , Arterivirus/genetics , Arterivirus/pathogenicity , Arterivirus Infections/genetics , Arterivirus Infections/virology , Flaviviridae/genetics , Flaviviridae/pathogenicity , Flaviviridae Infections/genetics , Flaviviridae Infections/virology , Genome, Viral , Haplorhini , Humans , Lentivirus/genetics , Lentivirus/pathogenicity , Lentivirus Infections/genetics , Lentivirus Infections/virology , Phylogeny , Prevalence
19.
PLoS Pathog ; 11(12): e1005304, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26658285

ABSTRACT

Viruses impose diverse and dynamic challenges on host defenses. Diversifying selection of codons and gene copy number variation are two hallmarks of genetic innovation in antiviral genes engaged in host-virus genetic conflicts. The myxovirus resistance (Mx) genes encode interferon-inducible GTPases that constitute a major arm of the cell-autonomous defense against viral infection. Unlike the broad antiviral activity of MxA, primate MxB was recently shown to specifically inhibit lentiviruses including HIV-1. We carried out detailed evolutionary analyses to investigate whether genetic conflict with lentiviruses has shaped MxB evolution in primates. We found strong evidence for diversifying selection in the MxB N-terminal tail, which contains molecular determinants of MxB anti-lentivirus specificity. However, we found no overlap between previously-mapped residues that dictate lentiviral restriction and those that have evolved under diversifying selection. Instead, our findings are consistent with MxB having a long-standing and important role in the interferon response to viral infection against a broader range of pathogens than is currently appreciated. Despite its critical role in host innate immunity, we also uncovered multiple functional losses of MxB during mammalian evolution, either by pseudogenization or by gene conversion from MxA genes. Thus, although the majority of mammalian genomes encode two Mx genes, this apparent stasis masks the dramatic effects that recombination and diversifying selection have played in shaping the evolutionary history of Mx genes. Discrepancies between our study and previous publications highlight the need to account for recombination in analyses of positive selection, as well as the importance of using sequence datasets with appropriate depth of divergence. Our study also illustrates that evolutionary analyses of antiviral gene families are critical towards understanding molecular principles that govern host-virus interactions and species-specific susceptibility to viral infection.


Subject(s)
Evolution, Molecular , Host-Parasite Interactions/genetics , Lentivirus Infections/genetics , Myxovirus Resistance Proteins/genetics , Amino Acid Sequence , Animals , DNA Copy Number Variations , Haplorhini , Humans , Immunity, Innate/genetics , Molecular Sequence Data , Phylogeny , Reverse Transcriptase Polymerase Chain Reaction
20.
PLoS Pathog ; 11(9): e1005149, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26394054

ABSTRACT

Cross-species transmissions of viruses from animals to humans are at the origin of major human pathogenic viruses. While the role of ecological and epidemiological factors in the emergence of new pathogens is well documented, the importance of host factors is often unknown. Chimpanzees are the closest relatives of humans and the animal reservoir at the origin of the human AIDS pandemic. However, despite being regularly exposed to monkey lentiviruses through hunting, chimpanzees are naturally infected by only a single simian immunodeficiency virus, SIVcpz. Here, we asked why chimpanzees appear to be protected against the successful emergence of other SIVs. In particular, we investigated the role of the chimpanzee APOBEC3 genes in providing a barrier to infection by most monkey lentiviruses. We found that most SIV Vifs, including Vif from SIVwrc infecting western-red colobus, the chimpanzee's main monkey prey in West Africa, could not antagonize chimpanzee APOBEC3G. Moreover, chimpanzee APOBEC3D, as well as APOBEC3F and APOBEC3H, provided additional protection against SIV Vif antagonism. Consequently, lentiviral replication in primary chimpanzee CD4(+) T cells was dependent on the presence of a lentiviral vif gene that could antagonize chimpanzee APOBEC3s. Finally, by identifying and functionally characterizing several APOBEC3 gene polymorphisms in both common chimpanzees and bonobos, we found that these ape populations encode APOBEC3 proteins that are uniformly resistant to antagonism by monkey lentiviruses.


Subject(s)
Cytidine Deaminase/genetics , Lentivirus Infections/genetics , Pan troglodytes/immunology , Pan troglodytes/virology , Simian Immunodeficiency Virus/genetics , Animals , Blotting, Western , CD4-Positive T-Lymphocytes/immunology , Cytidine Deaminase/immunology , Genes, vif/genetics , Haplorhini , Lentivirus/genetics , Lentivirus Infections/immunology , Molecular Sequence Data , Phylogeny , Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...