Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 54
Filter
1.
J Virol ; 98(4): e0030824, 2024 Apr 16.
Article in English | MEDLINE | ID: mdl-38497663

ABSTRACT

Host antiviral proteins inhibit primate lentiviruses and other retroviruses by targeting many features of the viral life cycle. The lentiviral capsid protein and the assembled viral core are known to be inhibited through multiple, directly acting antiviral proteins. Several phenotypes, including those known as Lv1 through Lv5, have been described as cell type-specific blocks to infection against some but not all primate lentiviruses. Here we review important features of known capsid-targeting blocks to infection together with several blocks to infection for which the genes responsible for the inhibition still remain to be identified. We outline the features of these blocks as well as how current methodologies are now well suited to find these antiviral genes and solve these long-standing mysteries in the HIV and retrovirology fields.


Subject(s)
Capsid , Host-Pathogen Interactions , Lentivirus Infections , Lentivirus , Animals , Capsid/metabolism , Capsid Proteins/genetics , Capsid Proteins/metabolism , Lentivirus/metabolism , Lentivirus Infections/metabolism
2.
Neurochem Res ; 49(2): 388-401, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37847329

ABSTRACT

Endoplasmic reticulum (ER) dysfunction caused by excessive ER stress is a crucial mechanism underlying seizures-induced neuronal injury. Studies have shown that mitochondrial reactive oxygen species (ROS) are closely related to ER stress, and our previous study showed that activating transcription factor 5 (ATF5)-regulated mitochondrial unfolded protein response (mtUPR) modulated mitochondrial ROS generation in a hippocampal neuronal culture model of seizures. However, the effects of ATF5-regulated mtUPR on ER stress and the underlying mechanisms remain uncertain in epilepsy. In this study, ATF5 upregulation by lentivirus infection attenuated seizures-induced neuronal damage and apoptosis in a rat model of pilocarpine-induced epilepsy, whereas ATF5 downregulation by lentivirus infection had the opposite effects. ATF5 upregulation potentiated mtUPR by increasing the expression of mitochondrial chaperone heat shock protein 60 (HSP60) and caseinolytic protease proteolytic subunit (ClpP) and reducing mitochondrial ROS generation in pilocarpine-induced seizures in rats. Additionally, upregulation of ATF5 reduced the expression of glucose-regulated protein 78 (GRP78), protein kinase RNA-like endoplasmic reticulum kinase (PERK), activating transcription factor 4 (ATF4), and C/EBP homologous protein (CHOP), suggesting suppression of ER stress; Moreover, ATF5 upregulation attenuated apoptosis-related proteins such as B-cell lymphoma-2 (BCL2) downregulation, BCL2-associated X (BAX) and cleaved-caspase-3 upregulation. However, ATF5 downregulation exerted the opposite effects. Furthermore, pretreatment with the mitochondria-targeted antioxidant mito-TEMPO attenuated the harmful effects of ATF5 downregulation on ER stress and neuronal apoptosis by reducing mitochondrial ROS generation. Overall, our study suggested that ATF5-regulated mtUPR exerted neuroprotective effects against pilocarpine-induced seizures in rats and the underlying mechanisms might involve mitochondrial ROS-mediated ER stress.


Subject(s)
Epilepsy , Lentivirus Infections , Rats , Animals , Reactive Oxygen Species/metabolism , Pilocarpine/toxicity , Endoplasmic Reticulum Stress , Unfolded Protein Response , Apoptosis , Mitochondria/metabolism , Apoptosis Regulatory Proteins/metabolism , Epilepsy/chemically induced , Epilepsy/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Seizures/chemically induced , Seizures/metabolism , Neurons/metabolism , Lentivirus Infections/metabolism
3.
Exp Eye Res ; 236: 109654, 2023 11.
Article in English | MEDLINE | ID: mdl-37734427

ABSTRACT

SMAD3 downregulation is documented in transforming growth factor ß1 (TGF-ß1)-induced corneal fibroblasts differentiation to myofibroblasts ("fibroTOmyoDiff") or corneal wound healing. However, the exact regulatory mechanism of TGF-ß1/SMAD3 pathway in this context remains unclear. Here, we investigated the role and related mechanism of SMAD3 down-regulation in TGF-ß1-induced human corneal fibroTOmyoDiff. By detecting expression changes of SMAD family during this process, we demonstrated that SMAD3 protein expression was dramatically decreased in the process and the decrease occurred mainly in SMAD3 gene transcription. Furthermore, SMAD3 overexpression using lentivirus infection and knockdown using sgRNA lentivirus infection or siRNAs revealed that SMAD3 overexpression enhanced TGF-ß1-induced corneal fibroTOmyoDiff and vice versa. In addition, specific siRNAs and inhibitors targeting particular signaling pathway were used to figure out the intracellular signaling pathway regulating SMAD3, and the result showed that the decease of SMAD3 induced by TGF-ß1 stimulation in human corneal fibroblasts (HCFs) was strikingly prevented by SMAD4 knockdown or p38 signaling inhibitor SB203580 treatment. Collectively, these results demonstrate that, in TGF-ß1 induced corneal fibroTOmyoDiff, down-regulation of SMAD3 expression regulated by SMAD4 and p38 signaling pathways forms a negative feedback loop of TGFß signaling to avoid excessive activation of the signaling, which suggest that SMAD3 may be a key target for corneal fibrosis treatment.


Subject(s)
Lentivirus Infections , Transforming Growth Factor beta1 , Humans , Transforming Growth Factor beta1/metabolism , Myofibroblasts/metabolism , Smad3 Protein/metabolism , Feedback , RNA, Guide, CRISPR-Cas Systems , Cells, Cultured , Fibroblasts/metabolism , Lentivirus Infections/metabolism , Transforming Growth Factor beta/metabolism
4.
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
5.
BMC Vet Res ; 15(1): 424, 2019 Nov 27.
Article in English | MEDLINE | ID: mdl-31775763

ABSTRACT

BACKGROUND: The present study aimed to determine the expression of cytokines, which is associated with the immunological response of dairy goats against small ruminant lentivirus (SRLV). The study was conducted on 26 dairy goats in their second to sixth lactation, which were divided by breed and parity into two groups: SRLV naturally infected (N = 13) and non-infected (N = 13) animals. All goats in the study were asymptomatic. The milk and blood samples, which served as studied material were taken on days 7, 30, 120 and 240 of the lactation. The gene and protein expression of several cytokines was studied using Real-Time PCR and ELISA methods. RESULTS: INF-ß and INF-γ expression was down-regulated in the milk somatic cells (MSC) of SRLV-infected goats. However, an increased concentration of INF-ß was observed in the MSC in SRLV-infected goats, while INF-γ expression was not observed in both SRLV-infected and non-infected animals The SRLV-infected goats also displayed decreased expression of IL-1α, IL-1ß, IL-6 and INF-γ genes in the blood leukocytes,with IL-1α, IL-1ß and IL-6 protein levels also being decreased in the sera. TNF-α was the only gene that demonstrated increased expression in both the MSC and the blood of infected animals; however, no such overexpression was observed at the protein level. CONCLUSIONS: SRLV probably influences the immune system of infected animals by deregulating of the expression of cytokines. Further, epigenetic studies may clarify the mechanisms by which SRLV regulates the gene and protein expression of the host.


Subject(s)
Cytokines/metabolism , Goat Diseases/virology , Lentivirus Infections/veterinary , Milk/metabolism , Animals , Cytokines/genetics , Female , Gene Expression , Goat Diseases/blood , Goat Diseases/immunology , Goat Diseases/metabolism , Goats , Lactation , Lentivirus , Lentivirus Infections/metabolism , Leukocytes/metabolism , Milk/cytology
6.
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
7.
Vet Res ; 49(1): 113, 2018 Nov 13.
Article in English | MEDLINE | ID: mdl-30424807

ABSTRACT

The aim of the study was to analyze acute phase protein and cathelicidin gene responses to small ruminant lentivirus (SRLV) infection in goats. In uninfected goats, we found higher Cp and lower Fbγ mRNA levels in blood leucocytes (BL) than in milk somatic cells (MSC), as well as lower SAA, Hp, and CRP and higher Cp and AGP concentrations in blood serum than in milk. In SRLV-infected goats, we found higher Fbγ and MAP28 and lower Cp expression in MSC than in BL, and higher SAA, Hp, Fb, and MAP28 and lower AGP concentrations in milk than in blood serum. Higher SAA and Hp expressions in BL and Hp expression in MSC were found in SRLV-infected goats. In SRLV-infected goats, we observed a higher concentration of SAA in blood serum, while in milk, lower SAA, Cp, and MAP28 and higher MAP34 concentrations were observed. The expression profiles of the studied genes differed between BL/serum and MSC/milk. The elevated SAA concentration in blood serum was accompanied by a decreased concentration of SAA and Cp in the milk of infected goats. No differences in the expression of the other studied genes may mean that the SRLV has the ability to evade the immune system, continuing to replicate. The elevated concentration of SAA in blood serum may promote viral multiplication. This higher concentration of SAA in blood serum and simultaneous reduced concentration of SAA and Cp in milk may be additive indicators of this infection.


Subject(s)
Acute-Phase Proteins/metabolism , Antimicrobial Cationic Peptides/metabolism , Goat Diseases/virology , Lentivirus Infections/veterinary , Leukocytes/metabolism , Milk/metabolism , Acute-Phase Proteins/genetics , Animals , Antimicrobial Cationic Peptides/genetics , Enzyme-Linked Immunosorbent Assay/veterinary , Goat Diseases/metabolism , Goats/metabolism , Goats/virology , Lentivirus Infections/metabolism , Lentivirus Infections/virology , Milk/cytology , Real-Time Polymerase Chain Reaction/veterinary , Cathelicidins
8.
J Neurosci ; 36(41): 10683-10695, 2016 10 12.
Article in English | MEDLINE | ID: mdl-27733618

ABSTRACT

HIV-1 infection of the brain causes the neurodegenerative syndrome HIV-associated neurocognitive disorders (HAND), for which there is no specific treatment. Herein, we investigated the actions of insulin using ex vivo and in vivo models of HAND. Increased neuroinflammatory gene expression was observed in brains from patients with HIV/AIDS. The insulin receptor was detected on both neurons and glia, but its expression was unaffected by HIV-1 infection. Insulin treatment of HIV-infected primary human microglia suppressed supernatant HIV-1 p24 levels, reduced CXCL10 and IL-6 transcript levels, and induced peroxisome proliferator-activated receptor gamma (PPAR-γ) expression. Insulin treatment of primary human neurons prevented HIV-1 Vpr-mediated cell process retraction and death. In feline immunodeficiency virus (FIV) infected cats, daily intranasal insulin treatment (20.0 IU/200 µl for 6 weeks) reduced CXCL10, IL-6, and FIV RNA detection in brain, although PPAR-γ in glia was increased compared with PBS-treated FIV+ control animals. These molecular changes were accompanied by diminished glial activation in cerebral cortex and white matter of insulin-treated FIV+ animals, with associated preservation of cortical neurons. Neuronal counts in parietal cortex, striatum, and hippocampus were higher in the FIV+/insulin-treated group compared with the FIV+/PBS-treated group. Moreover, intranasal insulin treatment improved neurobehavioral performance, including both memory and motor functions, in FIV+ animals. Therefore, insulin exerted ex vivo and in vivo antiviral, anti-inflammatory, and neuroprotective effects in models of HAND, representing a new therapeutic option for patients with inflammatory or infectious neurodegenerative disorders including HAND. SIGNIFICANCE STATEMENT: HIV-associated neurocognitive disorders (HAND) represent a spectrum disorder of neurocognitive dysfunctions resulting from HIV-1 infection. Although the exact mechanisms causing HAND are unknown, productive HIV-1 infection in the brain with associated neuroinflammation is a potential pathogenic mechanism resulting in neuronal damage and death. We report that, in HIV-infected microglia cultures, insulin treatment led to reduced viral replication and inflammatory gene expression. In addition, intranasal insulin treatment of experimentally feline immunodeficiency virus-infected animals resulted in improved motor and memory performances. We show that insulin restored expression of the nuclear receptor peroxisome proliferator-activated receptor gamma (PPAR-γ), which is suppressed by HIV-1 replication. Our findings indicate a unique function for insulin in improving neurological outcomes in lentiviral infections, implicating insulin as a therapeutic intervention for HAND.


Subject(s)
AIDS Dementia Complex/prevention & control , Hypoglycemic Agents/therapeutic use , Insulin/therapeutic use , Neuritis/prevention & control , Neurodegenerative Diseases/prevention & control , Neurons/pathology , Neuroprotective Agents/therapeutic use , Administration, Intranasal , Animals , Cats , Cell Death/drug effects , Female , HIV-1 , Human Immunodeficiency Virus Proteins/metabolism , Humans , Hypoglycemic Agents/administration & dosage , Immunodeficiency Virus, Feline , Insulin/administration & dosage , Lentivirus Infections/metabolism , Neuroglia/drug effects , Neuroglia/metabolism , Neurons/drug effects , Neurons/metabolism , Neuroprotective Agents/administration & dosage , Pregnancy , Receptor, Insulin/drug effects
9.
J Biol Chem ; 290(29): 17935-17945, 2015 Jul 17.
Article in English | MEDLINE | ID: mdl-26045556

ABSTRACT

Sterile α motif (SAM) and histidine/aspartate (HD)-containing protein 1 (SAMHD1) restricts human/simian immunodeficiency virus infection in certain cell types and is counteracted by the virulence factor Vpx. Current evidence indicates that Vpx recruits SAMHD1 to the Cullin4-Ring Finger E3 ubiquitin ligase (CRL4) by facilitating an interaction between SAMHD1 and the substrate receptor DDB1- and Cullin4-associated factor 1 (DCAF1), thereby targeting SAMHD1 for proteasome-dependent down-regulation. Host-pathogen co-evolution and positive selection at the interfaces of host-pathogen complexes are associated with sequence divergence and varying functional consequences. Two alternative interaction interfaces are used by SAMHD1 and Vpx: the SAMHD1 N-terminal tail and the adjacent SAM domain or the C-terminal tail proceeding the HD domain are targeted by different Vpx variants in a unique fashion. In contrast, the C-terminal WD40 domain of DCAF1 interfaces similarly with the two above complexes. Comprehensive biochemical and structural biology approaches permitted us to delineate details of clade-specific recognition of SAMHD1 by lentiviral Vpx proteins. We show that not only the SAM domain but also the N-terminal tail engages in the DCAF1-Vpx interaction. Furthermore, we show that changing the single Ser-52 in human SAMHD1 to Phe, the residue found in SAMHD1 of Red-capped monkey and Mandrill, allows it to be recognized by Vpx proteins of simian viruses infecting those primate species, which normally does not target wild type human SAMHD1 for degradation.


Subject(s)
Host-Pathogen Interactions , Lentivirus Infections/metabolism , Lentivirus/physiology , Monomeric GTP-Binding Proteins/metabolism , Viral Regulatory and Accessory Proteins/metabolism , Virulence Factors/metabolism , Amino Acid Sequence , Animals , Crystallography, X-Ray , HEK293 Cells , Humans , Lentivirus Infections/virology , Molecular Docking Simulation , Molecular Sequence Data , Monomeric GTP-Binding Proteins/chemistry , Protein Structure, Tertiary , SAM Domain and HD Domain-Containing Protein 1 , Sequence Alignment , Viral Regulatory and Accessory Proteins/chemistry
10.
J Virol ; 88(11): 6268-80, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24672033

ABSTRACT

UNLABELLED: Epithelial barrier dysfunction during human immunodeficiency virus (HIV) infection has largely been attributed to the rapid and severe depletion of CD4(+) T cells in the gastrointestinal (GI) tract. Although it is known that changes in mucosal gene expression contribute to intestinal enteropathy, the role of small noncoding RNAs, specifically microRNA (miRNA), has not been investigated. Using the simian immunodeficiency virus (SIV)-infected nonhuman primate model of HIV pathogenesis, we investigated the effect of viral infection on miRNA expression in intestinal mucosa. SIV infection led to a striking decrease in the expression of mucosal miRNA compared to that in uninfected controls. This decrease coincided with an increase in 5'-3'-exoribonuclease 2 protein and alterations in DICER1 and Argonaute 2 expression. Targets of depleted miRNA belonged to molecular pathways involved in epithelial proliferation, differentiation, and immune response. Decreased expression of several miRNA involved in maintaining epithelial homeostasis in the gut was localized to the proliferative crypt region of the intestinal epithelium. Our findings suggest that SIV-induced decreased expression of miRNA involved in epithelial homeostasis, disrupted expression of miRNA biogenesis machinery, and increased expression of XRN2 are involved in the development of epithelial barrier dysfunction and gastroenteropathy. IMPORTANCE: MicroRNA (miRNA) regulate the development and function of intestinal epithelial cells, and many viruses disrupt normal host miRNA expression. In this study, we demonstrate that SIV and HIV disrupt expression of miRNA in the small intestine during infection. The depletion of several key miRNA is localized to the proliferative crypt region of the gut epithelium. These miRNA are known to control expression of genes involved in inflammation, cell death, and epithelial maturation. Our data indicate that this disruption might be caused by altered expression of miRNA biogenesis machinery during infection. These findings suggest that the disruption of miRNA in the small intestine likely plays a role in intestinal enteropathy during HIV infection.


Subject(s)
HIV , Intestinal Mucosa/metabolism , Intestinal Mucosa/physiopathology , Lentivirus Infections/metabolism , MicroRNAs/metabolism , Simian Immunodeficiency Virus , Adult , Animals , Base Sequence , CD4-Positive T-Lymphocytes/immunology , Computational Biology , Densitometry , Flow Cytometry , Humans , Intestinal Mucosa/immunology , Laser Capture Microdissection , Lentivirus Infections/physiopathology , Macaca mulatta , Male , Microarray Analysis , Middle Aged , Molecular Sequence Data , Real-Time Polymerase Chain Reaction , Sequence Analysis, RNA , Viral Load
11.
J Virol ; 88(6): 3255-72, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24390322

ABSTRACT

UNLABELLED: BST2/tetherin inhibits the release of enveloped viruses from cells. Primate lentiviruses have evolved specific antagonists (Vpu, Nef, and Env). Here we characterized tetherin proteins of species representing both branches of the order Carnivora. Comparison of tiger and cat (Feliformia) to dog and ferret (Caniformia) genes demonstrated that the tiger and cat share a start codon mutation that truncated most of the tetherin cytoplasmic tail early in the Feliformia lineage (19 of 27 amino acids, including the dual tyrosine motif). Alpha interferon (IFN-α) induced tetherin and blocked feline immunodeficiency virus (FIV) replication in lymphoid and nonlymphoid feline cells. Budding of bald FIV and HIV particles was blocked by carnivore tetherins. However, infectious FIV particles were resistant, and spreading FIV replication was uninhibited. Antagonism mapped to the envelope glycoprotein (Env), which rescued FIV from carnivore tetherin restriction when expressed in trans but, in contrast to known antagonists, did not rescue noncognate particles. Also unlike the primate lentiviral antagonists, but similar to the Ebola virus glycoprotein, FIV Env did not reduce intracellular or cell surface tetherin levels. Furthermore, FIV-enveloped FIV particles actually required tetherin for optimal release from cells. The results show that FIV Envs mediate a distinctive tetherin evasion. Well adapted to a phylogenetically ancient tetherin tail truncation in the Felidae, it requires functional virion incorporation of Env, and it shields the budding particle without downregulating plasma membrane tetherin. Moreover, FIV has evolved dependence on this protein: particles containing FIV Env need tetherin for optimal release from the cell, while Env(-) particles do not. IMPORTANCE: HIV-1 antagonizes the restriction factor tetherin with the accessory protein Vpu, while HIV-2 and the filovirus Ebola use their envelope (Env) glycoproteins for this purpose. It turns out that the FIV tetherin antagonist is also its Env protein, but the mechanism is distinctive. Unlike other tetherin antagonists, FIV Env cannot act in trans to rescue vpu-deficient HIV-1. It must be incorporated specifically into FIV virions to be active. Also unlike other retroviral antagonists, but similar to Ebola virus Env, it does not act by downregulating or degrading tetherin. FIV Env might exclude tetherin locally or direct assembly to tetherin-negative membrane domains. Other distinctive features are apparent, including evidence that this virus evolved an equilibrium in which tetherin is both restriction factor and cofactor, as FIV requires tetherin for optimal particle release.


Subject(s)
Antigens, CD/metabolism , Cat Diseases/metabolism , Immunodeficiency Virus, Feline/metabolism , Lentivirus Infections/veterinary , Viral Envelope Proteins/metabolism , Virion/metabolism , Amino Acid Sequence , Animals , Antigens, CD/chemistry , Antigens, CD/genetics , Cat Diseases/genetics , Cat Diseases/virology , Cats , Dogs , Immunodeficiency Virus, Feline/chemistry , Immunodeficiency Virus, Feline/genetics , Lentivirus Infections/genetics , Lentivirus Infections/metabolism , Lentivirus Infections/virology , Molecular Sequence Data , Sequence Alignment , Viral Envelope Proteins/genetics , Virion/genetics
12.
J Biol Chem ; 287(27): 22882-8, 2012 Jun 29.
Article in English | MEDLINE | ID: mdl-22514281

ABSTRACT

Zinc-finger antiviral protein (ZAP) is a host factor that specifically inhibits the replication of certain viruses, including HIV-1, Ebola virus, and Sindbis virus. ZAP binds directly to specific viral mRNAs and recruits cellular mRNA degradation machinery to degrade the target RNA. ZAP has also been suggested to repress translation of the target mRNA. In this study, we report that ZAP is phosphorylated by glycogen synthase kinase 3ß (GSK3ß). GSK3ß sequentially phosphorylated Ser-270, Ser-266, Ser-262, and Ser-257 of rat ZAP. Inhibition of GSK3ß by inhibitor SB216763 or down-regulation of GSK3ß by RNAi reduced the antiviral activity of ZAP. These results indicate that phosphorylation of ZAP by GSK3ß modulates ZAP activity.


Subject(s)
Carrier Proteins/metabolism , Glycogen Synthase Kinase 3/metabolism , Lentivirus Infections/metabolism , Lentivirus/genetics , Animals , Carrier Proteins/genetics , Gene Library , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3/genetics , Glycogen Synthase Kinase 3 beta , HEK293 Cells , Humans , Indoles/pharmacology , Lentivirus Infections/genetics , Lentivirus Infections/immunology , Maleimides/pharmacology , Phosphorylation/physiology , RNA, Small Interfering/genetics , RNA, Viral/metabolism , RNA-Binding Proteins , Rats , Serine/metabolism , Threonine/metabolism
13.
J Virol ; 86(22): 12053-68, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22933280

ABSTRACT

Productive replication of human immunodeficiency virus type 1 (HIV-1) occurs efficiently only in humans. The posttranscriptional stages of the HIV-1 life cycle proceed poorly in mouse cells, with a resulting defect in viral assembly and release. Previous work has shown that the presence of human chromosome 2 increases HIV-1 production in mouse cells. Recent studies have shown that human chromosome region maintenance 1 (hCRM1) stimulates Gag release from rodent cells. Here we report that expressions of hCRM1 in murine cells resulted in marked increases in the production of infectious HIV-1 and feline immunodeficiency virus (FIV). HIV-1 production was also increased by hSRp40, and a combination of hCRM1 and hSRp40 resulted in a more-than-additive effect on HIV-1 release. In contrast, the overexpression of mouse CRM1 (mCRM1) minimally affected HIV-1 and FIV production and did not antagonize hCRM1. In the presence of hCRM1 there were large increases in the amounts of released capsid, which paralleled the increases in the infectious titers. Consistent with this finding, the ratios of unspliced to spliced HIV-1 mRNAs in mouse cells expressing hCRM1 and SRp40 became similar to those of human cells. Furthermore, imaging of intron-containing FIV RNA showed that hCRM1 increased RNA export to the cytoplasm.By testing chimeras between mCRM1 and hCRM1 and comparing those sequences to feline CRM1, we mapped the functional domain to HEAT (Huntingtin, elongation factor 3, protein phosphatase 2A, and the yeast kinase TOR1) repeats 4A to 9A and a triple point mutant in repeat 9A, which showed a loss of function. Structural analysis suggested that this region of hCRM1 may serve as a binding site for viral or cellular factors to facilitate lentiviral RNA nuclear export.


Subject(s)
HIV Infections/metabolism , HIV/metabolism , Immunodeficiency Virus, Feline/metabolism , Karyopherins/physiology , Lentivirus Infections/metabolism , Receptors, Cytoplasmic and Nuclear/physiology , Active Transport, Cell Nucleus , Alleles , Animals , Cell Cycle Proteins/metabolism , Cytoplasm/metabolism , HeLa Cells , Humans , Introns , Karyopherins/metabolism , Mice , Molecular Conformation , Plasmids/metabolism , RNA, Viral/metabolism , RNA-Binding Proteins/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Repressor Proteins/metabolism , Serine-Arginine Splicing Factors , Transfection , Exportin 1 Protein
15.
J Immunol ; 185(11): 6876-82, 2010 Dec 01.
Article in English | MEDLINE | ID: mdl-21041724

ABSTRACT

Interactions between the Fc segment of IgG and FcγRs on a variety of cells are likely to play an important role in the anti-HIV activity of Abs. Because the nature of the glycan structure on the Fc domain is a critical determinant of Fc-FcγR binding, proper Fc glycosylation may contribute to Ab-mediated protection. We have generated five different glycoforms of the broadly HIV-1-neutralizing mAb 2G12 in wild-type and glycoengineered plants and Chinese hamster ovary cells. Plant-derived 2G12 exhibited highly homogeneous glycosylation profiles with a single dominant N-glycan species. Using flow cytometry with FcγR-expressing cell lines, all 2G12 glycoforms demonstrated similar binding to FcγRI, FcγRIIa, and FcγRIIb. In contrast, two glycoforms derived from glycoengineered plants that lack plant-specific xylose and core α1,3-fucose, and instead carry human-like glycosylation with great uniformity, showed significantly enhanced binding to FcγRIIIa compared with Chinese hamster ovary or wild-type plant-derived 2G12. Using surface plasmon resonance, we show that binding of 2G12 to FcγRIIIa is markedly affected by core fucose, irrespective of its plant-specific α1,3 or mammalian-type α1,6 linkage. Consistent with this finding, 2G12 glycoforms lacking core fucose (and xylose) mediated higher antiviral activity against HIV-1 or simian immunodeficiency virus as measured by Ab-dependent cell-mediated virus inhibition. This is, to our knowledge, the first demonstration that specific alterations of Fc glycosylation can improve antiviral activity. Such alterations may result in better immunotherapeutic reagents. Moreover, biasing vaccine-induced immune responses toward optimal Fc glycosylation patterns could result in improved vaccine efficacy.


Subject(s)
Anti-HIV Agents/pharmacology , Antibodies, Monoclonal/pharmacology , HIV-1/immunology , Immunoglobulin Fc Fragments/metabolism , Receptors, IgG/metabolism , AIDS Vaccines/immunology , Animals , Anti-HIV Agents/metabolism , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/metabolism , Antibody-Dependent Cell Cytotoxicity/immunology , Binding Sites, Antibody/immunology , Broadly Neutralizing Antibodies , CHO Cells , Cell Line , Cricetinae , Cricetulus , Glycosylation , HIV Antibodies , HIV-1/genetics , HIV-1/growth & development , Humans , Lentivirus Infections/immunology , Lentivirus Infections/metabolism , Lentivirus Infections/prevention & control , Neutralization Tests/methods , Protein Binding/immunology , Receptors, IgG/biosynthesis , Receptors, IgG/genetics , Simian Immunodeficiency Virus/genetics , Simian Immunodeficiency Virus/growth & development , Simian Immunodeficiency Virus/immunology , Nicotiana/genetics , Nicotiana/immunology
16.
Proc Natl Acad Sci U S A ; 106(19): 8015-20, 2009 May 12.
Article in English | MEDLINE | ID: mdl-19416840

ABSTRACT

We and others have reported that the vast majority of virus-producing CD4(+) T cells during the acute infection of rhesus macaques with simian immunodeficiency virus (SIV) or CXCR4 (X4)-using simian/human immunodeficiency viruses (SHIVs) exhibited a nonactivated phenotype. These findings have been extended to show that resting CD4(+) T lymphocytes collected from SIV- or X4-SHIV-infected animals during the first 10 days of infection continue to release virus ex vivo. Furthermore, we observed high frequencies of integrated viral DNA (up to 5.1 x 10(4) DNA copies per 10(5) cells) in circulating resting CD4(+) T cells during the first 10 days of the infection. Integration of SIV DNA was detected only in memory CD4(+) T cells and SHIVs preferentially integrated into resting naïve CD4(+) T cells. Taken together, these results show that during the acute infection large numbers of resting CD4(+) T cells carry integrated nonhuman primate lentiviral DNA and are the major source of progeny virions irrespective of coreceptor usage. Prompt and sustained interventions are therefore required to block the rapid systemic dissemination of virus and prevent an otherwise fatal clinical outcome.


Subject(s)
CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/virology , DNA, Viral/metabolism , Lentivirus Infections/blood , Lentivirus Infections/virology , Animals , Calibration , Immunologic Memory , Immunophenotyping , Lentivirus Infections/metabolism , Macaca mulatta , Models, Biological , Phenotype , Simian Acquired Immunodeficiency Syndrome/blood , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/genetics , Treatment Outcome , Virus Replication
17.
Eur J Med Res ; 27(1): 5, 2022 Jan 13.
Article in English | MEDLINE | ID: mdl-35022077

ABSTRACT

BACKGROUND: Intervertebral disc degeneration (IDD) is a natural progression of age-related processes. Associated with IDD, degenerative disc disease (DDD) is a pathologic condition implicated as a major cause of chronic lower back pain, which can have a severe impact on the quality of life of patients. As degeneration progression is associated with elevated levels of inflammatory cytokines, enhanced aggrecan and collagen degradation, and changes in the disc cell phenotype. The purpose of this study was to investigate the biological and cytological characteristics of rabbit nucleus pulposus mesenchymal stem cells (NPMSCs)-a key factor in IDD-and to determine the effect of the growth and differentiation factor-5 (GDF5) on the differentiation of rabbit NPMSCs transduced with a lentivirus vector. METHODS: An in vitro culture model of rabbit NPMSCs was established and NPMSCs were identified by flow cytometry (FCM) and quantitative real-time PCR (qRT-PCR). Subsequently, NPMSCs were randomly divided into three groups: a transfection group (the lentiviral vector carrying GDF5 gene used to transfect NPMSCs); a control virus group (the NPMSCs transfected with an ordinary lentiviral vector); and a normal group (the NPMSCs alone). FCM, qRT-PCR, and western blot (WB) were used to detect the changes in NPMSCs. RESULTS: The GDF5-transfected NPMSCs displayed an elongated shape, with decreased cell density, and significantly increased GDF5 positivity rate in the transfected group compared to the other two groups (P < 0.01). The mRNA levels of Krt8, Krt18, and Krt19 in the transfected group were significantly higher in comparison with the other two groups (P < 0.01), and the WB results were consistent with that of qRT-PCR. CONCLUSIONS: GDF5 could induce the differentiation of NPMSCs. The lentiviral vector carrying the GDF5 gene could be integrated into the chromosome genome of NPMSCs and promoted differentiation of NPMSCs into nucleus pulposus cells. Our findings advance the development of feasible and effective therapies for IDD.


Subject(s)
Gene Expression Regulation , Growth Differentiation Factor 5/genetics , Lentivirus Infections/virology , Lentivirus , Mesenchymal Stem Cells/cytology , Nucleus Pulposus/metabolism , Animals , Cell Differentiation , Cells, Cultured , Disease Models, Animal , Growth Differentiation Factor 5/biosynthesis , Lentivirus Infections/metabolism , Lentivirus Infections/pathology , Mesenchymal Stem Cells/virology , Nucleus Pulposus/pathology , Nucleus Pulposus/virology , Rabbits
19.
Virol J ; 7: 134, 2010 Jun 23.
Article in English | MEDLINE | ID: mdl-20569475

ABSTRACT

Bovine ISG15 (bISG15) is an interferon inducible ubiquitin-like protein that is responsible for the establishment of early pregnancy in ruminant, understanding the properties of bISG15 capable of being inducible in fetal bovine lung (FBL) cells upon infection of bovine immunodeficiency virus (BIV) is of significant importance. In this study, we investigated the expression of bISG15 in poly I:C treated FBL cells. The increased expression of bISG15 was observed, and the inhibition of BIV replication was also detected in FBL cells. Elimination of bISG15 expression by small interfering RNA reversed the bISG15 mediated inhibition of BIV replication. These findings demonstrate that bISG15 plays an important role in inhibition of the BIV replication in FBL cells. Furthermore, real-time PCR and western blot assay revealed that bISG15's expression can also be induced in BIV infected FBL cells. Taken together, bISG15 is an antiviral and inducible protein in BIV infected FBL cells.


Subject(s)
Antiviral Agents/metabolism , Cattle Diseases/metabolism , Gene Expression Regulation , Immunodeficiency Virus, Bovine/physiology , Lentivirus Infections/veterinary , Lung/metabolism , Ubiquitins/metabolism , Animals , Cattle , Cattle Diseases/genetics , Cattle Diseases/virology , Gene Expression Regulation, Viral , Immunodeficiency Virus, Bovine/genetics , Lentivirus Infections/genetics , Lentivirus Infections/metabolism , Lentivirus Infections/virology , Lung/virology , Ubiquitins/genetics , Virus Replication
20.
Viruses ; 12(6)2020 05 27.
Article in English | MEDLINE | ID: mdl-32471198

ABSTRACT

Mammals have developed clever adaptive and innate immune defense mechanisms to protect against invading bacterial and viral pathogens. Human innate immunity is continuously evolving to expand the repertoire of restriction factors and one such family of intrinsic restriction factors is the APOBEC3 (A3) family of cytidine deaminases. The coordinated expression of seven members of the A3 family of cytidine deaminases provides intrinsic immunity against numerous foreign infectious agents and protects the host from exogenous retroviruses and endogenous retroelements. Four members of the A3 proteins-A3G, A3F, A3H, and A3D-restrict HIV-1 in the absence of virion infectivity factor (Vif); their incorporation into progeny virions is a prerequisite for cytidine deaminase-dependent and -independent activities that inhibit viral replication in the host target cell. HIV-1 encodes Vif, an accessory protein that antagonizes A3 proteins by targeting them for polyubiquitination and subsequent proteasomal degradation in the virus producing cells. In this review, we summarize our current understanding of the role of human A3 proteins as barriers against HIV-1 infection, how Vif overcomes their antiviral activity, and highlight recent structural and functional insights into A3-mediated restriction of lentiviruses.


Subject(s)
APOBEC Deaminases/physiology , Lentivirus Infections/immunology , Lentivirus/immunology , APOBEC Deaminases/chemistry , Animals , Base Sequence , Humans , Lentivirus/metabolism , Lentivirus Infections/metabolism , Protein Structure, Tertiary , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL