Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 903
Filter
1.
Viruses ; 16(4)2024 03 23.
Article in English | MEDLINE | ID: mdl-38675836

ABSTRACT

PYHIN proteins are only found in mammals and play key roles in the defense against bacterial and viral pathogens. The corresponding gene locus shows variable deletion and expansion ranging from 0 genes in bats, over 1 in cows, and 4 in humans to a maximum of 13 in mice. While initially thought to act as cytosolic immune sensors that recognize foreign DNA, increasing evidence suggests that PYHIN proteins also inhibit viral pathogens by more direct mechanisms. Here, we examined the ability of all 13 murine PYHIN proteins to inhibit HIV-1 and murine leukemia virus (MLV). We show that overexpression of p203, p204, p205, p208, p209, p210, p211, and p212 strongly inhibits production of infectious HIV-1; p202, p207, and p213 had no significant effects, while p206 and p214 showed intermediate phenotypes. The inhibitory effects on infectious HIV-1 production correlated significantly with the suppression of reporter gene expression by a proviral Moloney MLV-eGFP construct and HIV-1 and Friend MLV LTR luciferase reporter constructs. Altogether, our data show that the antiretroviral activity of PYHIN proteins is conserved between men and mice and further support the key role of nuclear PYHIN proteins in innate antiviral immunity.


Subject(s)
HIV-1 , Leukemia Virus, Murine , Phosphoproteins , Animals , Mice , Humans , HIV-1/immunology , HIV-1/genetics , Leukemia Virus, Murine/genetics , Leukemia Virus, Murine/immunology , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Nuclear Proteins/immunology , Virus Replication , Cell Line , Retroviridae Infections/immunology , Retroviridae Infections/virology
2.
PLoS Pathog ; 16(5): e1008605, 2020 05.
Article in English | MEDLINE | ID: mdl-32453763

ABSTRACT

As obligate parasites, viruses highjack, modify and repurpose the cellular machinery for their own replication. Viral proteins have, therefore, evolved biological functions, such as signalling potential, that alter host cell physiology in ways that are still incompletely understood. Retroviral envelope glycoproteins interact with several host proteins, extracellularly with their cellular receptor and anti-envelope antibodies, and intracellularly with proteins of the cytoskeleton or sorting, endocytosis and recirculation pathways. Here, we examined the impact of endogenous retroviral envelope glycoprotein expression and interaction with host proteins, particularly antibodies, on the cell, independently of retroviral infection. We found that in the commonly used C57BL/6 substrains of mice, where murine leukaemia virus (MLV) envelope glycoproteins are expressed by several endogenous MLV proviruses, the highest expressed MLV envelope glycoprotein is under the control of an immune-responsive cellular promoter, thus linking MLV envelope glycoprotein expression with immune activation. We further showed that antibody ligation induces extensive internalisation from the plasma membrane into endocytic compartments of MLV envelope glycoproteins, which are not normally subject to constitutive endocytosis. Importantly, antibody binding and internalisation of MLV envelope glycoproteins initiates signalling cascades in envelope-expressing murine lymphocytic cell lines, leading to cellular activation. Similar effects were observed by MLV envelope glycoprotein ligation by its cellular receptor mCAT-1, and by overexpression in human lymphocytic cells, where it required an intact tyrosine-based YXXΦ motif in the envelope glycoprotein cytoplasmic tail. Together, these results suggest that signalling potential is a general property of retroviral envelope glycoproteins and, therefore, a target for intervention.


Subject(s)
Antibodies, Viral/immunology , Calcium Channels/immunology , Cell Membrane/immunology , Endocytosis/immunology , Leukemia Virus, Murine/immunology , TRPV Cation Channels/immunology , Viral Envelope Proteins/immunology , Animals , Humans , Mice , Mice, Inbred BALB C
3.
Cell Host Microbe ; 24(6): 761-775.e6, 2018 12 12.
Article in English | MEDLINE | ID: mdl-30503508

ABSTRACT

TRIM5 is a RING domain E3 ubiquitin ligase with potent antiretroviral function. TRIM5 assembles into a hexagonal lattice on retroviral capsids, causing envelopment of the infectious core. Concomitantly, TRIM5 initiates innate immune signaling and orchestrates disassembly of the viral particle, yet how these antiviral responses are regulated by capsid recognition is unclear. We show that hexagonal assembly triggers N-terminal polyubiquitination of TRIM5 that collectively drives antiviral responses. In uninfected cells, N-terminal monoubiquitination triggers non-productive TRIM5 turnover. Upon TRIM5 assembly on virus, a trivalent RING arrangement allows elongation of N-terminally anchored K63-linked ubiquitin chains (N-K63-Ub). N-K63-Ub drives TRIM5 innate immune stimulation and proteasomal degradation. Inducing ubiquitination before TRIM5 assembly triggers premature degradation and ablates antiviral restriction. Conversely, driving N-K63 ubiquitination after TRIM5 assembly enhances innate immune signaling. Thus, the hexagonal geometry of TRIM5's antiviral lattice converts a capsid-binding protein into a multifunctional antiviral platform.


Subject(s)
Carrier Proteins/metabolism , Immunity, Innate/immunology , Retroviridae Infections/immunology , Ubiquitin-Protein Ligases/metabolism , Animals , Antiviral Restriction Factors , Capsid/chemistry , Capsid/metabolism , Carrier Proteins/genetics , HEK293 Cells , Humans , Leukemia Virus, Murine/enzymology , Leukemia Virus, Murine/genetics , Leukemia Virus, Murine/immunology , Mice , Mice, Inbred C57BL , Polysaccharides, Bacterial/chemistry , Polysaccharides, Bacterial/genetics , Polysaccharides, Bacterial/metabolism , Retroviridae Infections/metabolism , Retroviridae Infections/virology , THP-1 Cells , Tripartite Motif Proteins , Ubiquitin-Conjugating Enzymes/genetics , Ubiquitin-Conjugating Enzymes/metabolism , Ubiquitin-Protein Ligases/chemistry , Ubiquitin-Protein Ligases/genetics
4.
Annu Rev Virol ; 5(1): 323-340, 2018 09 29.
Article in English | MEDLINE | ID: mdl-30265629

ABSTRACT

SERINC genes encode for homologous multipass transmembrane proteins with unknown cellular function, despite being highly conserved across eukaryotes. Among the five SERINC genes found in humans, SERINC5 was shown to act as a powerful inhibitor of retroviruses. It is efficiently incorporated into virions and blocks the penetration of the viral core into target cells, by impairing the fusion process with a yet unclear mechanism. SERINC5 was also found to promote human immunodeficiency virus 1 (HIV-1) virion neutralization by antibodies, indicating a pleiotropic activity, which remains mostly unexplored. Counteracting factors have emerged independently in at least three retrovirus lineages, underscoring their fundamental importance during retrovirus evolution. Nef and S2 of primate and equine lentiviruses, and glycoGag of gammaretroviruses, act similarly by targeting SERINC5 to endosomes and excluding it from virions. Here, we discuss the features that distinguish SERINC5 from other known restriction factors, delineating a yet unique class of antiviral inhibitors.


Subject(s)
HIV/immunology , Immunity, Innate , Immunologic Factors/metabolism , Leukemia Virus, Murine/immunology , Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism , Animals , Humans , Mice
5.
Sci China Life Sci ; 61(8): 954-965, 2018 08.
Article in English | MEDLINE | ID: mdl-29705873

ABSTRACT

TRIM5α restricts retroviruses in a species-specific manner. Cyclophilin A was independently retrotransposed into the TRIM5 loci in different species, leading to the generation of antiviral TRIM5-cyclophilin A (TRIMCyp) proteins. Previously, we found that assam macaques express a TRIMCyp chimera (amTRIMCyp), along with a TRIM5α allelic protein (amTRIM5α). Herein, we investigated the antiviral activity of amTRIMCyp and amTRIM5α individually, as well as their interaction and joint effects. amTRIMCyp showed a divergent restriction pattern from amTRIM5α. Although both proteins potently restricted the replication of HIV-1, only amTRIM5α inhibited N-MLV. Remarkably, cellular anti-HIV-1 activity increased when amTRIMCyp and amTRIM5α were coexpressed, indicating a synergistic block of HIV-1 replication. Consistently, PMBCs from heterozygous amTRIM5α/TRIMCyp showed stronger resistance to HIV-1 infection than those from amTRIM5α/TRIM5α homozygotes. The anti-HIV-1 synergistic effect was dependent on the amTRIMCyp-amTRIM5α interaction. In contrast, amTRIMCyp completely abrogated the anti-N-MLV activity mediated by amTRIM5α, showing a dominant-negative effect, indicating that the generation of amTRIMCyp was involved in the trade-off between divergent restriction activities. Our results provide a new paradigm to study functional trade-offs mediated by allelic proteins, a theoretical basis for utilizing animal models with various TRIM5 alleles, as well as novel HIV-1 gene therapy strategies.


Subject(s)
HIV-1/immunology , Leukemia Virus, Murine/immunology , Macaca/immunology , Mutant Chimeric Proteins/immunology , Retroviridae Infections/immunology , Animals , Cats , Cell Line , Cyclophilin A/genetics , Cyclophilin A/immunology , Cyclophilin A/metabolism , Gene Expression/immunology , HEK293 Cells , HIV-1/physiology , Humans , Leukemia Virus, Murine/physiology , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/virology , Macaca/virology , Mice , Mutant Chimeric Proteins/genetics , Mutant Chimeric Proteins/metabolism , Proteins/genetics , Proteins/immunology , Proteins/metabolism , RNA Interference/immunology , Retroviridae Infections/virology , Ubiquitin-Protein Ligases
6.
J Immunol ; 199(4): 1372-1381, 2017 08 15.
Article in English | MEDLINE | ID: mdl-28687662

ABSTRACT

Accumulating evidence shows that innate immune responses are associated with extracellular nucleotides, particularly ATP. In this article, we demonstrate extensive protection of ATP/P2X7 signaling in a host against viral infection. Interestingly, we observed a significant increase in ATP as a danger signal in vesicular stomatitis virus (VSV)-infected cell supernatant and VSV-infected mice in an exocytosis- and pannexin channel-dependent manner. Furthermore, extracellular ATP reduces the replication of VSV, Newcastle disease virus, murine leukemia virus, and HSV in vivo and in vitro through the P2X7 receptor. Meanwhile, ATP significantly increases IFN-ß expression in a concentration- and time-dependent manner. Mechanistically, ATP facilitates IFN-ß secretion through P38/JNK/ATF-2 signaling pathways, which are crucial in promoting antiviral immunity. Taken together, these results demonstrate the protective role of extracellular ATP and P2X7 in viral infection and suggest a potential therapeutic role for ATP/P2X7 in viral diseases.


Subject(s)
Adenosine Triphosphate/metabolism , Interferon-beta/biosynthesis , Receptors, Purinergic P2X7/metabolism , Vesicular Stomatitis/immunology , Vesicular stomatitis Indiana virus/physiology , Adenosine Triphosphate/pharmacology , Animals , Immunity, Innate , Interferon-beta/genetics , Interferon-beta/immunology , Leukemia Virus, Murine/drug effects , Leukemia Virus, Murine/immunology , Luminescent Measurements , Mice , Newcastle disease virus/drug effects , Newcastle disease virus/immunology , RAW 264.7 Cells , Receptors, Purinergic P2X7/immunology , Signal Transduction , Simplexvirus/drug effects , Simplexvirus/immunology , Vesicular Stomatitis/virology , Vesicular stomatitis Indiana virus/drug effects , Vesicular stomatitis Indiana virus/immunology , Virus Replication/drug effects
7.
Biochim Biophys Acta Mol Basis Dis ; 1863(10 Pt B): 2594-2600, 2017 10.
Article in English | MEDLINE | ID: mdl-27816520

ABSTRACT

Active participation of endogenous retroviruses (ERVs) in disease processes has been exemplified by the finding that the HERV (human ERV)-W envelope protein is involved in the pathogenesis of multiple sclerosis, an autoimmune disease. We also demonstrated that injury-elicited stressors alter the expression of murine ERVs (MuERVs), both murine leukemia virus-type and mouse mammary tumor virus (MMTV)-type (MMTV-MuERV). In this study, to evaluate MMTV-MuERVs' responses to stress (e.g., injury, infection)-elicited systemic glucocorticoid (GC) levels, we examined the GC-stress response of 64 MMTV-MuERV promoters isolated from the genomes of 23 mouse strains. All 64 promoters responded to treatment with a synthetic GC, dexamethasone (DEX), at a wide range from a 0.6- to 85.7-fold increase in reporter activity compared to no treatment. An analysis of the 10 lowest and 10 highest DEX responders revealed specific promoter elements exclusively present in either the three lowest or the two highest responders. Each promoter had a unique profile of transcription regulatory elements and the glucocorticoid response element (GRE) was identified in all promoters with the number of GREs ranging from 2 to 7. The three lowest DEX responders were the only promoters with two GREs. The findings from this study suggest that certain MMTV-MuERVs are more responsive to stress-elicited systemic GC elevation compared to the others. The mouse strain-specific genomic MMTV-MuERV profiles and individual MMTV-MuERVs' differential responses to GC-stress might explain, at least in part, the variable inflammatory responses to injury and/or infection, often observed among different mouse strains. This article is part of a Special Issue entitled: Immune and Metabolic Alterations in Trauma and Sepsis edited by Dr. Raghavan Raju.


Subject(s)
Dexamethasone/pharmacology , Endogenous Retroviruses/immunology , Glucocorticoids/pharmacology , Leukemia Virus, Murine/immunology , Mammary Tumor Virus, Mouse/immunology , Stress, Physiological , Animals , Endogenous Retroviruses/genetics , Leukemia Virus, Murine/genetics , Mammary Tumor Virus, Mouse/genetics , Mice , Response Elements/immunology , Species Specificity , Stress, Physiological/drug effects , Stress, Physiological/immunology
8.
J Immunol ; 197(9): 3628-3638, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27647833

ABSTRACT

Elucidation of the immune requirements for control or elimination of retroviral infection remains an important aim. We studied the induction of adaptive immunity to neonatal infection with a murine retrovirus, under conditions leading to immunological tolerance. We found that the absence of either maternal or offspring adaptive immunity permitted efficient vertical transmission of the retrovirus. Maternal immunodeficiency allowed the retrovirus to induce central Th cell tolerance in the infected offspring. In turn, this compromised the offspring's ability to mount a protective Th cell-dependent B cell response. However, in contrast to T cells, offspring B cells were not centrally tolerized and retained their ability to respond to the infection when provided with T cell help. Thus, escape of retrovirus-specific B cells from deletional tolerance offers the opportunity to induce protective retroviral immunity by restoration of retrovirus-specific T cell help, suggesting similar T cell immunotherapies for persistent viral infections.


Subject(s)
Adoptive Transfer , B-Lymphocytes/immunology , Infectious Disease Transmission, Vertical/prevention & control , Leukemia Virus, Murine/immunology , Leukemia, Experimental/prevention & control , Retroviridae Infections/prevention & control , T-Lymphocytes/immunology , Tumor Virus Infections/prevention & control , Animals , Animals, Newborn , B-Lymphocytes/transplantation , B-Lymphocytes/virology , Cells, Cultured , Central Tolerance , Female , Leukemia, Experimental/immunology , Male , Maternal Exposure/adverse effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Pregnancy , Retroviridae Infections/immunology , Retroviridae Infections/transmission , T-Lymphocytes/transplantation , T-Lymphocytes/virology , Tumor Virus Infections/immunology , Tumor Virus Infections/transmission
9.
Immunology ; 146(1): 33-49, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25959091

ABSTRACT

In the Sp6 mouse plasmacytoma model, a whole-cell vaccination with Sp6 cells expressing de novo B7-1 (Sp6/B7) induced anatomically localized and cytotoxic T cell (CTL)-mediated protection against wild-type (WT) Sp6. Both WT Sp6 and Sp6/B7 showed down-regulated expression of MHC H-2 L(d). Increase of H-2 L(d) expression by cDNA transfection (Sp6/B7/L(d)) raised tumour immune protection and shifted most CTL responses towards H-2 L(d)-restricted antigenic epitopes. The tumour-protective responses were not specific for the H-2 L(d)-restricted immunodominant AH1 epitope of the gp70 common mouse tumour antigen, although WT Sp6 and transfectants were able to present it to specific T cells in vitro. Gp70 transcripts, absent in secondary lymphoid organs of naive mice, were detected in immunized mice as well as in splenocytes from naive mice incubated in vitro with supernatants of CTL-lysed Sp6 cell cultures, containing damage-associated molecular patterns (DAMPs). It has been shown that Toll-like receptor triggering induces gp70 expression. Damage-associated molecular patterns are released by CTL-mediated killing of Sp6/B7-Sp6/B7/L(d) cells migrated to draining lymph nodes during immunization and may activate gp70 expression and presentation in most resident antigen-presenting cells. The same could also apply for Mus musculus endogenous ecotropic murine leukaemia virus 1 particles present in Sp6-cytosol, discharged by dying cells and superinfecting antigen-presenting cells. The outcome of such a massive gp70 cross-presentation would probably be tolerogenic for the high-affinity AH1-gp70-specific CTL clones. In this scenario, autologous whole-tumour-cell vaccines rescue tumour-specific immunoprotection by amplification of subdominant tumour antigen responses when those against the immune dominant antigens are lost.


Subject(s)
Antigens, Neoplasm/immunology , Cancer Vaccines/immunology , H-2 Antigens/immunology , Plasmacytoma/immunology , Animals , Antigens, Neoplasm/biosynthesis , CD4-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Cell Movement/immunology , Female , H-2 Antigens/biosynthesis , H-2 Antigens/genetics , HMGB1 Protein/metabolism , Leukemia Virus, Murine/immunology , Lymph Nodes/cytology , Lymph Nodes/immunology , Lymphocyte Depletion , Lymphocyte Subsets/immunology , Male , Mice , Mice, Inbred BALB C , Plasmacytoma/therapy , T-Lymphocytes, Cytotoxic/immunology , Vaccination
10.
Cell Host Microbe ; 17(4): 423-5, 2015 Apr 08.
Article in English | MEDLINE | ID: mdl-25856751

ABSTRACT

Viruses possess elaborate defensive mechanisms to evade host innate immune responses. In this issue of Cell Host & Microbe, Stavrou et al. (2015) reveal how the murine leukemia virus uses a sugar-protein shield to protect from inevitable destruction by cellular innate immune factors including the APOBEC3 DNA mutating enzyme.


Subject(s)
Cytidine Deaminase/metabolism , DNA, Viral/metabolism , Interferon-beta/metabolism , Leukemia Virus, Murine/immunology , Macrophages/immunology , Animals
11.
Cell Host Microbe ; 17(4): 478-88, 2015 Apr 08.
Article in English | MEDLINE | ID: mdl-25816774

ABSTRACT

Intrinsic restriction factors and viral nucleic acid sensors are important for the anti-viral response. Here, we show how upstream sensing of retroviral reverse transcripts integrates with the downstream effector APOBEC3, an IFN-induced cytidine deaminase that introduces lethal mutations during retroviral reverse transcription. Using a murine leukemia virus (MLV) variant with an unstable capsid that induces a strong IFNß antiviral response, we identify three sensors, IFI203, DDX41, and cGAS, required for MLV nucleic acid recognition. These sensors then signal using the adaptor STING, leading to increased production of IFNß and other targets downstream of the transcription factor IRF3. Using knockout and mutant mice, we show that APOBEC3 limits the levels of reverse transcripts that trigger cytosolic sensing, and that nucleic acid sensing in vivo increases expression of IFN-regulated restriction factors like APOBEC3 that in turn reduce viral load. These studies underscore the importance of the multiple layers of protection afforded by host factors.


Subject(s)
Cytidine Deaminase/metabolism , DNA, Viral/metabolism , Interferon-beta/metabolism , Leukemia Virus, Murine/immunology , Macrophages/immunology , Animals , Cell Line , DEAD-box RNA Helicases/metabolism , Leukemia Virus, Murine/physiology , Macrophages/virology , Membrane Proteins/metabolism , Mice, Inbred C57BL , Mice, Knockout , Nuclear Proteins/metabolism , Nucleotidyltransferases/metabolism , Reverse Transcription , Signal Transduction
12.
J Virol ; 89(4): 2342-57, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25505062

ABSTRACT

UNLABELLED: Retroviruses are pathogens with rapid infection cycles that can be a source of disease, genome instability, and tumor development in their hosts. Host intrinsic restriction factors, such as APOBEC3 (A3) proteins, are constitutively expressed and dedicated to interfering with the replication cycle of retroviruses. To survive, propagate, and persist, retroviruses must counteract these restriction factors, often by way of virus genome-encoded accessory proteins. Glycosylated Gag, also called glycosylated Pr80 Gag (gPr80), is a gammaretrovirus genome-encoded protein that inhibits the antiretroviral activity of mouse A3 (mA3). Here we show that gPr80 exerts two distinct inhibitory effects on mA3: one that antagonizes deamination-independent restriction and another one that inhibits its deaminase activity. More specifically, we find that the number of N-glycosylated residues in gPr80 inversely correlates with the sensitivity of a gammaretrovirus to deamination by mouse A3 and also, surprisingly, by human A3G. Finally, our work highlights that retroviruses which have successfully integrated into the mouse germ line generally express a gPr80 with fewer glycosylated sites than exogenous retroviruses. This observation supports the suggestion that modulation of A3 deamination intensity could be a desirable attribute for retroviruses to increase genetic diversification and avoid immune detection. Overall, we present here the first description of how gammaretroviruses employ posttranslational modification to antagonize and modulate the activity of a host genome-encoded retroviral restriction factor. IMPORTANCE: APOBEC3 proteins are host factors that have a major role in protecting humans and other mammals against retroviruses. These enzymes hinder their replication and intensely mutate their DNA, thereby inactivating viral progeny and the spread of infection. Here we describe a newly recognized way in which some retroviruses protect themselves against the mutator activity of APOBEC3 proteins. We show that gammaretroviruses expressing an accessory protein called glycosylated Gag, or gPr80, use the host's posttranslational machinery and, more specifically, N-linked glycosylation as a way to modulate their sensitivity to mutations by APOBEC3 proteins. By carefully controlling the amount of mutations caused by APOBEC3 proteins, gammaretroviruses can find a balance that helps them evolve and persist.


Subject(s)
Cytidine Deaminase/antagonists & inhibitors , Gene Products, gag/metabolism , Leukemia Virus, Murine/immunology , Protein Processing, Post-Translational , APOBEC Deaminases , Animals , Cell Line , Cytosine Deaminase/antagonists & inhibitors , Deamination , Glycosylation , Humans , Leukemia Virus, Murine/physiology , Mice, Inbred C57BL , Mice, Knockout
13.
J Virol ; 89(1): 155-64, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25320301

ABSTRACT

UNLABELLED: Interleukin-1 beta (IL-1ß) is an inflammatory cytokine that is secreted in response to inflammasome activation by innate microbe-sensing pathways. Although some retroviruses can trigger IL-1ß secretion through the DNA-sensing molecule IFI16, the effect of IL-1ß on the course of infection is unknown. To test whether IL-1ß secretion affects retroviral replication in vivo, I constructed a novel murine leukemia virus strain (FMLV-IL-1ß) that encodes the mature form of IL-1ß. This virus replicated with kinetics similar to that of wild-type virus in tissue culture but caused a dramatically more aggressive infection of both C57BL/6 and BALB/c mice. By 7 days postinfection (PI), mice infected with FMLV-IL-1ß exhibited splenomegaly and viral loads 300-fold higher than those in mice infected with wild-type FMLV. Furthermore, the enlarged spleens of FMLV-IL-1ß-infected mice correlated with a large expansion of Gr-1(+) CD11b(+) myeloid-derived suppressor cells, as well as elevated levels of immune activation. Although FMLV-IL-1ß infection was controlled by C57BL/6 mice by 14 days p.i., FMLV-IL-1ß was able to establish a significant persistent infection and immune activation in BALB/c mice. These results demonstrate that IL-1ß secretion is a powerful positive regulator of retroviral infection and that FMLV-IL-1ß represents a new model of proinflammatory retroviral infection. IMPORTANCE: Interleukin-1 beta (IL-1ß) is an inflammatory cytokine released in response to activation of innate pathogen-sensing pathways during microbial infection. To examine the potential impact of IL-1ß on retroviral replication in vivo, I constructed a novel mouse retrovirus strain (FMLV-IL-1ß) that encodes IL-1ß and promotes abundant IL-1ß secretion from infected cells. This virus replicates with normal kinetics in cultured cells but displays a dramatically enhanced ability to replicate in mice and caused persistent infection and immune activation in the BALB/c strain of mice. These results establish IL-1ß as a positive regulator of retroviral replication and suggest that targeting this pathway may have therapeutic benefits in infections with proinflammatory retroviruses. This virus can also be used to further study the impact of inflammatory pathways on retroviral infection.


Subject(s)
Interleukin-1beta/metabolism , Leukemia Virus, Murine/immunology , Leukemia Virus, Murine/pathogenicity , Leukemia, Experimental/pathology , Retroviridae Infections/pathology , Tumor Virus Infections/pathology , Animals , Host-Pathogen Interactions , Interleukin-1beta/genetics , Leukemia Virus, Murine/genetics , Leukemia Virus, Murine/physiology , Leukemia, Experimental/virology , Mice, Inbred BALB C , Mice, Inbred C57BL , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Retroviridae Infections/virology , Splenomegaly/pathology , Tumor Virus Infections/virology , Viral Load , Virulence , Virus Replication
14.
J Virol ; 88(10): 5661-76, 2014 May.
Article in English | MEDLINE | ID: mdl-24600008

ABSTRACT

UNLABELLED: The tripartite motif (TRIM) family of proteins includes the TRIM5α antiretroviral restriction factor. TRIM5α from many Old World and some New World monkeys can restrict the human immunodeficiency virus type 1 (HIV-1), while human TRIM5α restricts N-tropic murine leukemia virus (N-MLV). TRIM5α forms highly dynamic cytoplasmic bodies (CBs) that associate with and translocate on microtubules. However, the functional involvement of microtubules or other cytoskeleton-associated factors in the viral restriction process had not been shown. Here, we demonstrate the dependency of TRIM5α-mediated restriction on microtubule-mediated transport. Pharmacological disruption of the microtubule network using nocodazole or disabling it using paclitaxel (originally named taxol) decreased the restriction of N-MLV and HIV-1 by human or simian alleles of TRIM5α, respectively. In addition, pharmacological inhibition of dynein motor complexes using erythro-9-(2-hydroxy-3-nonyl)adenine (EHNA) and small interfering RNA-mediated depletion of the dynein heavy chain (DHC) similarly decreased TRIM5α-mediated restriction. The loss in restriction resulting from either the disassembly of microtubules or the disruption of dynein motor activity was seen for both endogenous and overexpressed TRIM5α and was not due to differences in protein stability or cell viability. Both nocodazole treatment and DHC depletion interfered with the dynamics of TRIM5α CBs, increasing their size and altering their intracellular localization. In addition, nocodazole, paclitaxel, and DHC depletion were all found to increase the stability of HIV-1 cores in infected cells, providing an alternative explanation for the decreased restriction. In conclusion, association with microtubules and the translocation activity of dynein motor complexes are required to achieve efficient restriction by TRIM5α. IMPORTANCE: The primate innate cellular defenses against infection by retroviruses include a protein named TRIM5α, belonging to the family of restriction factors. TRIM5α is present in the cytoplasm, where it can intercept incoming retroviruses shortly after their entry. How TRIM5α manages to be present at the appropriate subcytoplasmic location to interact with its target is unknown. We hypothesized that TRIM5α, either as a soluble protein or a high-molecular-weight complex (the cytoplasmic body), is transported within the cytoplasm by a molecular motor called the dynein complex, which is known to interact with and move along microtubules. Our results show that destructuring microtubules or crippling their function decreased the capacity of human or simian TRIM5α to restrict their retroviral targets. Inhibiting dynein motor activity, or reducing the expression of a key component of this complex, similarly affected TRIM5α-mediated restriction. Thus, we have identified specific cytoskeleton structures involved in innate antiretroviral defenses.


Subject(s)
Carrier Proteins/metabolism , Dyneins/metabolism , HIV-1/immunology , Leukemia Virus, Murine/immunology , Microtubules/metabolism , Animals , Antiviral Restriction Factors , Biological Transport , Cell Line , Humans , Macaca mulatta , Proteins/metabolism , Tripartite Motif Proteins , Ubiquitin-Protein Ligases
15.
J Virol Methods ; 200: 47-53, 2014 May.
Article in English | MEDLINE | ID: mdl-24556162

ABSTRACT

Many monoclonal antibodies (MAbs) reactive with various proteins of murine leukemia viruses (MuLVs) have been developed. In this report two additional MAbs with differing and unusual specificities are described. MAb 573 is reactive with the envelope protein of all MuLVs tested including viruses in the ecotropic, xenotropic, polytropic and amphotropic classes. Notably, MAb 573 is one of only two reported MAbs that react with the envelope protein of amphotropic MuLVs. This MAb appears to recognize a conformational epitope within the envelope protein, as it reacts strongly with live virus and live infected cells, but does not react with formalin-fixed or alcohol-fixed infected cells or denatured viral envelope protein in immunoblots. In contrast, Mab 538 reacts only with an epitope unique to the envelope protein of the Moloney (Mo-) strain of MuLV, a prototypic ecotropic MuLV that is the basis for many retroviral tools used in molecular biology. MAb 538 can react with live cells and viruses, or detergent denatured or fixed envelope protein. The derivation of these antibodies as well as their characterization with regard to their isotype, range of reactivity with different MuLVs and utility in different immunological procedures are described in this study.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/isolation & purification , Antibodies, Viral/immunology , Antibodies, Viral/isolation & purification , Cross Reactions , Leukemia Virus, Murine/immunology , Viral Envelope Proteins/immunology , Animals , Mice, Inbred BALB C
16.
J Virol ; 88(7): 3850-60, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24453360

ABSTRACT

UNLABELLED: Many murine leukemia viruses (MLVs) are partially resistant to restriction by mouse APOBEC3 (mA3) and essentially fully resistant to induction of G-to-A mutations by mA3. In contrast, Vif-deficient HIV-1 (ΔVif HIV-1) is profoundly restricted by mA3, and the restriction includes high levels of G-to-A mutation. Human APOBEC3G (hA3G), unlike mA3, is fully active against MLVs. We produced a glutathione S-transferase-mA3 fusion protein in insect cells and demonstrated that it possesses cytidine deaminase activity, as expected. This activity is localized within the N-terminal domain of this 2-domain protein; the C-terminal domain is enzymatically inactive but required for mA3 encapsidation into retrovirus particles. We found that a specific arginine residue and several aromatic residues, as well as the zinc-coordinating cysteines in the C-terminal domain, are necessary for mA3 packaging; a structural model of this domain suggests that these residues line a potential nucleic acid-binding interface. Mutation of a few potential phosphorylation sites in mA3 drastically reduces its antiviral activity by impairing either deaminase activity or its encapsidation. mA3 deaminates short single-stranded DNA oligonucleotides preferentially toward their 3' ends, whereas hA3G exhibits the opposite polarity. However, when packaged into infectious ΔVif HIV-1 virions, both mA3 and hA3G preferentially induce deaminations toward the 5' end of minus-strand viral DNA, presumably because of the sequence of events during reverse transcription in vivo. Despite the fact that mA3 in MLV particles does not induce detectable deaminations upon infection, its deaminase activity is easily detected in virus lysates. We still do not understand how MLV resists mA3-induced G-to-A mutation. IMPORTANCE: One way that mammalian cells defend themselves against infection by retroviruses is with APOBEC3 proteins. These proteins convert cytidine bases to uridine bases in retroviral DNA. However, mouse APOBEC3 protein blocks infection by murine leukemia viruses without catalyzing this base change, and the mechanism of inhibition is not understood in this case. We have produced recombinant mouse APOBEC3 protein for the first time and characterized it here in a number of ways. Our mutational studies shed light on the mechanism by which mouse APOBEC3 protein is incorporated into retrovirus particles. While mouse APOBEC3 does not catalyze base changes in murine leukemia virus DNA, it can be recovered from these virus particles in enzymatically active form; it is still not clear why it fails to induce base changes when these viruses infect new cells.


Subject(s)
Cytidine Deaminase/metabolism , HIV-1/immunology , Leukemia Virus, Murine/immunology , Animals , Cell Line , Cytidine Deaminase/genetics , DNA Mutational Analysis , Glutathione Transferase/genetics , Glutathione Transferase/metabolism , HIV-1/physiology , Insecta , Leukemia Virus, Murine/physiology , Mice , Phosphorylation , Protein Processing, Post-Translational , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Virus Assembly
17.
J Virol Methods ; 193(2): 332-6, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23810854

ABSTRACT

Retroviral vectors derived from the murine leukemia virus (MuLV) are widely used as the starting material in the development of vectors for gene therapy and critical in answering questions relating to viral pathogenesis. The p30 capsid (CA) is the major viral core protein and an internal group antigen in MuLV. In this study, an enzyme-linked immunosorbent assay (ELISA) was developed for quantitation of MuLV infectious particles with p30 CA core antigen protein. The ELISA was developed using several goat-polyclonal serum against MuLV p30 generated by the NCI as primary antibody and a rat-monoclonal antibody to CA available from ATCC. The MuLV p30 CA antigen was standardized against recombinant MuLV p30 CA expressed from bacteria. The assay is sensitive, accurate and linear within a defined concentration range of CA. Comparison with different MuLV quantitative methods including reporter gene transfer, reverse transcriptase activity assay, and viral RNA quantitative PCR, showed this ELISA protocol to be highly quantifiable within defined ranges, which can be correlated with infectious viral titer.


Subject(s)
Antibodies, Viral , Capsid Proteins/analysis , Leukemia Virus, Murine/isolation & purification , Viral Load/methods , Animals , Capsid Proteins/immunology , Enzyme-Linked Immunosorbent Assay/methods , Goats , Leukemia Virus, Murine/immunology , Sensitivity and Specificity
18.
J Virol ; 87(16): 9271-8, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23785198

ABSTRACT

The host restriction factors TRIM5α and TRIMCyp potently inhibit retrovirus infection by binding to the incoming retrovirus capsid. TRIM5 proteins are dimeric, and their association with the viral capsid appears to be enhanced by avidity effects owing to formation of higher-order oligomeric complexes. We examined the stoichiometric requirement for TRIM5 functional recognition by quantifying the efficiencies of restriction of HIV-1 and murine leukemia virus (MLV) particles containing various proportions of restriction-sensitive and -insensitive CA subunits. Both TRIMCyp and TRIM5α inhibited infection of retrovirus particles containing as little as 25% of the restriction-sensitive CA protein. Accordingly, we also observed efficient binding of TRIMCyp in vitro to capsid assemblies containing as little as one-fourth wild-type CA protein. Paradoxically, the ability of HIV-1 particles to abrogate TRIMCyp restriction in trans was more strongly dependent on the fraction of wild-type CA than was restriction of infection. Collectively, our results indicate that TRIM5 restriction factors bind to retroviral capsids in a highly cooperative manner and suggest that TRIM5 can engage a capsid lattice containing a minimum of three or fewer recognizable subunits per hexamer. Our study supports a model in which localized binding of TRIM5 to the viral capsid nucleates rapid polymerization of a TRIM5 lattice on the capsid surface.


Subject(s)
Capsid/immunology , Carrier Proteins/immunology , HIV-1/immunology , Leukemia Virus, Murine/immunology , Animals , Antiviral Restriction Factors , Capsid/metabolism , Carrier Proteins/metabolism , Cell Line , Humans , Protein Binding , Protein Multimerization , Tripartite Motif Proteins , Ubiquitin-Protein Ligases
19.
J Neuroimmunol ; 261(1-2): 37-43, 2013 Aug 15.
Article in English | MEDLINE | ID: mdl-23726765

ABSTRACT

B6 mice infected with LP-BM5 develop severe immunodeficiency (termed murine acquired immunodeficiency syndrome (MAIDS)) and peripheral neuropathy. To determine whether microglial CD40 is involved in LP-BM5-induced peripheral neuropathy, B6-CD40 knockout (KO) mice and B6-CD40 KO mice adoptively transferred either total leukocytes or B cells were examined for behavioral sensitivity, tissue viral loads, cytokine responses, and the development of MAIDS. All three CD40 KO groups developed MAIDS, the severity of which was correlated with peripheral cytokine responses. CD40 KO mice displayed significantly reduced mechanical hypersensitivity post-infection compared to wild-type mice regardless of cell transfer. These findings support microglial CD40 involvement in LP-BM5-induced peripheral neuropathy.


Subject(s)
CD40 Antigens/immunology , Leukemia Virus, Murine/immunology , Microglia/immunology , Murine Acquired Immunodeficiency Syndrome/immunology , Peripheral Nervous System Diseases/metabolism , Retroviridae/immunology , Animals , B-Lymphocytes/immunology , B-Lymphocytes/pathology , B-Lymphocytes/virology , CD40 Antigens/deficiency , Leukocytes/immunology , Leukocytes/pathology , Leukocytes/virology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microglia/pathology , Microglia/virology , Murine Acquired Immunodeficiency Syndrome/pathology , Murine Acquired Immunodeficiency Syndrome/virology , Peripheral Nervous System Diseases/immunology , Peripheral Nervous System Diseases/virology , Random Allocation
20.
J Virol ; 87(1): 257-72, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23077300

ABSTRACT

Members of the tripartite interaction motif (TRIM) family of E3 ligases are emerging as critical regulators of innate immunity. To identify new regulators, we carried out a screen of 43 human TRIM proteins for the ability to activate NF-κB, AP-1, and interferon, hallmarks of many innate immune signaling pathways. We identified 16 TRIM proteins that induced NF-κB and/or AP-1. We found that one of these, TRIM62, functions in the TRIF branch of the TLR4 signaling pathway. Knockdown of TRIM62 in primary macrophages led to a defect in TRIF-mediated late NF-κB, AP-1, and interferon production after lipopolysaccharide challenge. We also discovered a role for TRIM15 in the RIG-I-mediated interferon pathway upstream of MAVS. Knockdown of TRIM15 limited virus/RIG-I ligand-induced interferon production and enhanced vesicular stomatitis virus replication. In addition, most TRIM proteins previously identified to inhibit murine leukemia virus (MLV) demonstrated an ability to induce NF-κB/AP-1. Interfering with the NF-κB and AP-1 signaling induced by the antiretroviral TRIM1 and TRIM62 proteins rescued MLV release. In contrast, human immunodeficiency virus type 1 (HIV-1) gene expression was increased by TRIM proteins that induce NF-κB. HIV-1 resistance to inflammatory TRIM proteins mapped to the NF-κB sites in the HIV-1 long terminal repeat (LTR) U3 and could be transferred to MLV. Thus, our work identifies new TRIM proteins involved in innate immune signaling and reinforces the striking ability of HIV-1 to exploit innate immune signaling for the purpose of viral replication.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , HIV-1/immunology , Immunity, Innate , Leukemia Virus, Murine/immunology , Membrane Proteins/metabolism , Signal Transduction , Vesiculovirus/immunology , Adaptor Proteins, Signal Transducing/genetics , Gene Knockdown Techniques , Humans , Interferons/metabolism , Membrane Proteins/genetics , NF-kappa B/metabolism , Transcription Factor AP-1/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...