Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
Int J Mol Sci ; 25(4)2024 Feb 16.
Article in English | MEDLINE | ID: mdl-38397027

ABSTRACT

A feature of the SARS-CoV-2 Omicron subvariants BF.5 and BF.7 that recently circulated mainly in China and Japan was the high prevalence of the ORF7a: H47Y mutation, in which the 47th residue of ORF7a has been mutated from a histidine (H) to a tyrosine (Y). Here, we evaluated the effect of this mutation on the three main functions ascribed to the SARS-CoV-2 ORF7a protein. Our findings show that H47Y mutation impairs the ability of SARS-CoV-2 ORF7a to antagonize the type I interferon (IFN-I) response and to downregulate major histocompatibility complex I (MHC-I) cell surface levels, but had no effect in its anti-SERINC5 function. Overall, our results suggest that the H47Y mutation of ORF7a affects important functions of this protein, resulting in changes in virus pathogenesis.


Subject(s)
COVID-19 , Interferon Type I , Humans , SARS-CoV-2/genetics , SARS-CoV-2/metabolism , COVID-19/genetics , Interferon Type I/metabolism , Mutation , China
2.
PLoS Biol ; 17(2): e3000137, 2019 02.
Article in English | MEDLINE | ID: mdl-30726215

ABSTRACT

Tripartite motif (TRIM) proteins belong to a large family with many roles in host biology, including restricting virus infection. Here, we found that TRIM2, which has been implicated in cases of Charcot-Marie-Tooth disease (CMTD) in humans, acts by blocking hemorrhagic fever New World arenavirus (NWA) entry into cells. We show that Trim2-knockout mice, as well as primary fibroblasts from a CMTD patient with mutations in TRIM2, are more highly infected by the NWAs Junín and Tacaribe virus than wild-type mice or cells are. Using mice with different Trim2 gene deletions and TRIM2 mutant constructs, we demonstrate that its antiviral activity is uniquely independent of the RING domain encoding ubiquitin ligase activity. Finally, we show that one member of the TRIM2 interactome, signal regulatory protein α (SIRPA), a known inhibitor of phagocytosis, also restricts NWA infection and conversely that TRIM2 limits phagocytosis of apoptotic cells. In addition to demonstrating a novel antiviral mechanism for TRIM proteins, these studies suggest that the NWA entry and phagocytosis pathways overlap.


Subject(s)
Antigens, Differentiation/genetics , Arenaviruses, New World/genetics , Charcot-Marie-Tooth Disease/genetics , Host-Pathogen Interactions/genetics , Nuclear Proteins/genetics , Receptors, Immunologic/genetics , Animals , Antigens, Differentiation/immunology , Antigens, Differentiation/metabolism , Apoptosis , Arenaviruses, New World/growth & development , Arenaviruses, New World/pathogenicity , Brain/immunology , Brain/metabolism , Brain/virology , Cell Line, Tumor , Charcot-Marie-Tooth Disease/metabolism , Charcot-Marie-Tooth Disease/pathology , Chlorocebus aethiops , Fibroblasts/immunology , Fibroblasts/metabolism , Fibroblasts/virology , Gene Expression Regulation , HEK293 Cells , Host-Pathogen Interactions/immunology , Humans , Macrophages/immunology , Macrophages/metabolism , Macrophages/virology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/immunology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/immunology , Mitogen-Activated Protein Kinase 3/metabolism , Neurofilament Proteins/genetics , Neurofilament Proteins/immunology , Neurofilament Proteins/metabolism , Nuclear Proteins/immunology , Nuclear Proteins/metabolism , Osteoblasts/immunology , Osteoblasts/metabolism , Osteoblasts/virology , Primary Cell Culture , Receptors, Immunologic/immunology , Receptors, Immunologic/metabolism , Signal Transduction , Vero Cells , Virus Internalization
4.
J Virol ; 92(11)2018 06 01.
Article in English | MEDLINE | ID: mdl-29593034

ABSTRACT

The apolipoprotein B editing complex 3 (APOBEC3) proteins are potent retroviral restriction factors that are under strong positive selection, both in terms of gene copy number and sequence diversity. A common feature of all the members of the APOBEC3 family is the presence of one or two cytidine deamination domains, essential for cytidine deamination of retroviral reverse transcripts as well as packaging into virions. Several studies have indicated that human and mouse APOBEC3 proteins restrict retrovirus infection via cytidine deaminase (CD)-dependent and -independent means. To understand the relative contribution of CD-independent restriction in vivo, we created strains of transgenic mice on an APOBEC3 knockout background that express a deaminase-dead mouse APOBEC3 due to point mutations in both CD domains (E73Q/E253Q). Here, we show that the CD-dead APOBEC3 can restrict murine retroviruses in vivo Moreover, unlike the wild-type protein, the mutant APOBEC3 is not packaged into virions but acts only as a cell-intrinsic restriction factor that blocks reverse transcription by incoming viruses. Finally, we show that wild-type and CD-dead mouse APOBEC3 can bind to murine leukemia virus (MLV) reverse transcriptase. Our findings suggest that the mouse APOBEC3 cytidine deaminase activity is not required for retrovirus restriction.IMPORTANCE APOBEC3 proteins are important host cellular restriction factors essential for restricting retrovirus infection by causing mutations in the virus genome and by blocking reverse transcription. While both methods of restriction function in vitro, little is known about their role during in vivo infection. By developing transgenic mice with mutations in the cytidine deamination domains needed for enzymatic activity and interaction with viral RNA, we show that APOBEC3 proteins can still restrict in vivo infection by interacting with reverse transcriptase and blocking its activity. These studies demonstrate that APOBEC3 proteins have evolved multiple means for blocking retrovirus infection and that all of these means function in vivo.


Subject(s)
Cytidine Deaminase/genetics , Leukemia Virus, Murine/genetics , Retroviridae Infections/prevention & control , Reverse Transcriptase Inhibitors/metabolism , Reverse Transcription/genetics , Animals , Cell Line , Cytidine Deaminase/metabolism , Deamination/genetics , HEK293 Cells , Humans , Leukemia Virus, Murine/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , RNA, Viral/genetics , RNA-Directed DNA Polymerase/genetics
5.
J Virol ; 90(17): 8005-12, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27356895

ABSTRACT

UNLABELLED: APOBEC3 knockout and human APOBEC3A and -3G transgenic mice were tested for their ability to be infected by the herpesviruses herpes simplex virus 1 and murine herpesvirus 68 and the parvovirus minute virus of mice (MVM). Knockout, APOBEC3A and APOBEC3G transgenic, and wild-type mice were equally infected by the herpesviruses, while APOBEC3A but not mouse APOBEC3 conferred resistance to MVM. No viruses showed evidence of cytidine deamination by mouse or human APOBEC3s. These data suggest that in vitro studies implicating APOBEC3 proteins in virus resistance may not reflect their role in vivo IMPORTANCE: It is well established that APOBEC3 proteins in different species are a critical component of the host antiretroviral defense. Whether these proteins also function to inhibit other viruses is not clear. There have been a number of in vitro studies suggesting that different APOBEC3 proteins restrict herpesviruses and parvoviruses, among others, but whether they also work in vivo has not been demonstrated. Our studies looking at the role of mouse and human APOBEC3 proteins in transgenic and knockout mouse models of viral infection suggest that these restriction factors are not broadly antiviral and demonstrate the importance of testing their activity in vivo.


Subject(s)
APOBEC-3G Deaminase/metabolism , Cytidine Deaminase/metabolism , Herpesviridae Infections/immunology , Parvoviridae Infections/immunology , Proteins/metabolism , Animals , Disease Models, Animal , Disease Resistance , Herpesvirus 1, Human/immunology , Mice , Mice, Knockout , Mice, Transgenic , Minute Virus of Mice/immunology , Rhadinovirus/immunology
6.
J Immunol ; 195(10): 4565-70, 2015 Nov 15.
Article in English | MEDLINE | ID: mdl-26546688

ABSTRACT

Apolipoprotein B editing complex 3 family members are cytidine deaminases that play important roles in intrinsic responses to infection by retroviruses and have been implicated in the control of other viruses, such as parvoviruses, herpesviruses, papillomaviruses, hepatitis B virus, and retrotransposons. Although their direct effect on modification of viral DNA has been clearly demonstrated, whether they play additional roles in innate and adaptive immunity to viruses is less clear. We review the data regarding the various steps in the innate and adaptive immune response to virus infection in which apolipoprotein B editing complex 3 proteins have been implicated.


Subject(s)
Cytosine Deaminase/immunology , DNA, Viral/immunology , HIV Infections/immunology , HIV-1/immunology , Virus Diseases/immunology , APOBEC Deaminases , Adaptive Immunity/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cytidine Deaminase , Cytosine Deaminase/genetics , DNA, Viral/genetics , HIV Infections/virology , Hepatitis B/immunology , Herpesviridae Infections/immunology , Herpesviridae Infections/virology , Humans , Papillomavirus Infections/immunology , Parvoviridae Infections/immunology , Parvoviridae Infections/virology
7.
Retrovirology ; 13(1): 45, 2016 06 30.
Article in English | MEDLINE | ID: mdl-27363431

ABSTRACT

Humans encode seven APOBEC3 proteins (A-H), with A3G, 3F and 3H as the major factors restricting HIV-1 replication. HIV-1, however, encodes Vif, which counteracts A3 proteins by chaperoning them to the proteasome where they are degraded. Vif polymorphisms found in HIV-1s isolated from infected patients have varying anti-A3G potency when assayed in vitro, but there are few studies demonstrating this in in vivo models. Here, we created Friend murine leukemia viruses encoding vif alleles that were previously shown to differentially neutralize A3G in cell culture or that were originally found in primary HIV-1 isolates. Infection of transgenic mice expressing different levels of human A3G showed that these naturally occurring Vif variants differed in their ability to counteract A3G during in vivo infection, although the effects on viral replication were not identical to those seen in cultured cells. We also found that the polymorphic Vifs that attenuated viral replication reverted to wild type only in A3G transgenic mice. Finally, we found that the level of A3G-mediated deamination was inversely correlated with the level of viral replication. This animal model should be useful for studying the functional significance of naturally occurring vif polymorphisms, as well as viral evolution in the presence of A3G.


Subject(s)
APOBEC-3G Deaminase/metabolism , HIV Infections/virology , HIV-1/genetics , Polymorphism, Genetic , vif Gene Products, Human Immunodeficiency Virus/genetics , APOBEC-3G Deaminase/genetics , Alleles , Animals , Disease Models, Animal , Friend murine leukemia virus/genetics , Friend murine leukemia virus/physiology , Humans , Mice , Mice, Transgenic , Mutation , Virus Replication
8.
PLoS Pathog ; 10(5): e1004145, 2014 May.
Article in English | MEDLINE | ID: mdl-24851906

ABSTRACT

The apolipoprotein B editing complex 3 (A3) cytidine deaminases are among the most highly evolutionarily selected retroviral restriction factors, both in terms of gene copy number and sequence diversity. Primate genomes encode seven A3 genes, and while A3F and 3G are widely recognized as important in the restriction of HIV, the role of the other genes, particularly A3A, is not as clear. Indeed, since human cells can express multiple A3 genes, and because of the lack of an experimentally tractable model, it is difficult to dissect the individual contribution of each gene to virus restriction in vivo. To overcome this problem, we generated human A3A and A3G transgenic mice on a mouse A3 knockout background. Using these mice, we demonstrate that both A3A and A3G restrict infection by murine retroviruses but by different mechanisms: A3G was packaged into virions and caused extensive deamination of the retrovirus genomes while A3A was not packaged and instead restricted infection when expressed in target cells. Additionally, we show that a murine leukemia virus engineered to express HIV Vif overcame the A3G-mediated restriction, thereby creating a novel model for studying the interaction between these proteins. We have thus developed an in vivo system for understanding how human A3 proteins use different modes of restriction, as well as a means for testing therapies that disrupt HIV Vif-A3G interactions.


Subject(s)
Cytidine Deaminase/physiology , Proteins/physiology , Retroviridae Infections/genetics , Retroviridae Infections/virology , Viral Load/genetics , APOBEC-3G Deaminase , Animals , Cells, Cultured , HIV-1/physiology , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , NIH 3T3 Cells , Retroviridae/physiology , Virus Assembly/genetics , Virus Internalization , Virus Replication/genetics , vif Gene Products, Human Immunodeficiency Virus/genetics
9.
Proc Natl Acad Sci U S A ; 110(22): 9078-83, 2013 May 28.
Article in English | MEDLINE | ID: mdl-23671100

ABSTRACT

Pathogenic retroviruses have evolved multiple means for evading host restriction factors such as apolipoprotein B editing complex (APOBEC3) proteins. Here, we show that murine leukemia virus (MLV) has a unique means of counteracting APOBEC3 and other cytosolic sensors of viral nucleic acid. Using virus isolated from infected WT and APOBEC3 KO mice, we demonstrate that the MLV glycosylated Gag protein (glyco-Gag) enhances viral core stability. Moreover, in vitro endogenous reverse transcription reactions of the glyco-Gag mutant virus were substantially inhibited compared with WT virus, but only in the presence of APOBEC3. Thus, glyco-Gag rendered the reverse transcription complex in the viral core resistant to APOBEC3. Glyco-Gag in the virion also rendered MLV resistant to other cytosolic sensors of viral reverse transcription products in newly infected cells. Strikingly, glyco-Gag mutant virus reverted to glyco-Gag-containing virus only in WT and not APOBEC3 KO mice, indicating that counteracting APOBEC3 is the major function of glyco-Gag. Thus, in contrast to the HIV viral infectivity factor protein, which prevents APOBEC3 packaging in the virion, the MLV glyco-Gag protein uses a unique mechanism to counteract the antiviral action of APOBEC3 in vivo--namely, protecting the reverse transcription complex in viral cores from APOBEC3. These data suggest that capsid integrity may play a critical role in virus resistance to intrinsic cellular antiviral resistance factors that act at the early stages of infection.


Subject(s)
Cytidine Deaminase/antagonists & inhibitors , Gene Products, gag/metabolism , Host-Pathogen Interactions/physiology , Leukemia Virus, Murine/physiology , Reverse Transcription/physiology , Animals , Blotting, Western , Cytidine Deaminase/genetics , Cytidine Deaminase/metabolism , DNA Primers/genetics , Gene Products, gag/pharmacology , Glycosylation , Leukemia Virus, Murine/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , NIH 3T3 Cells , Reverse Transcriptase Polymerase Chain Reaction
10.
bioRxiv ; 2023 Sep 19.
Article in English | MEDLINE | ID: mdl-37786722

ABSTRACT

Membrane-associated RING-CH (MARCH) 2 protein is a member of the MARCH protein family of RING-CH finger E3 ubiquitin ligases that have important functions in regulating the levels of proteins found on the cell surface. MARCH1, 2 and 8 inhibit HIV-1 infection by preventing the incorporation of the envelope glycoproteins in nascent virions. However, a better understanding on the mechanism utilized by MARCH proteins to restrict HIV-1 is needed. In this report, we identify an amino acid in human MARCH2, that is absent in mouse MARCH2, critical for its antiretroviral function. Moreover, we map the domains of human MARCH2 critical for restricting as well as binding to the HIV-1 envelope glycoproteins. Our findings reveal important new aspects of the antiviral mechanism utilized by human MARCH2 to restrict HIV-1 that have potential implications to all MARCH proteins with antiviral functions.

11.
J Virol ; 85(14): 7177-85, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21561911

ABSTRACT

Cellular apoptosis induced by viral genes can play a critical role in determining virulence as well as viral persistence. This form of cell death has been of interest with respect to Theiler's murine encephalomyelitis virus (TMEV) because the GDVII strain and members of the GDVII subgroup are highly neurovirulent, while the DA strain and members of the TO subgroup induce a chronic progressive inflammatory demyelination with persistence of the virus in the central nervous system. The TMEV L protein has been identified as important in the pathogenesis of Theiler's virus-induced demyelinating disease (TMEV-IDD). We now show that DA L is apoptotic following transfection of L expression constructs or following DA virus infection of HeLa cells; the apoptotic activity depends on the presence of the serine/threonine domain of L, especially a serine at amino acid 57. In contrast, GDVII L has little apoptotic activity following transfection of L expression constructs in HeLa cells and is antiapoptotic following GDVII infection of HeLa cells. Of note, both DA and GDVII L cleave caspase-3 in BHK-21 cells, although neither implements the full apoptotic machinery in this cell type as manifested by the induction of terminal deoxynucleotidyltransferase-mediated dUTP-biotin nick end labeling (TUNEL) staining. The differences in apoptotic activities of DA and GDVII L in varied cell types may play an important role in TMEV subgroup-specific disease phenotypes.


Subject(s)
Apoptosis/physiology , Theilovirus/physiology , Viral Proteins/physiology , Amino Acid Sequence , Base Sequence , DNA Primers , HeLa Cells , Humans , Molecular Sequence Data , Sequence Homology, Amino Acid , Viral Proteins/chemistry
12.
Nat Commun ; 13(1): 2935, 2022 05 26.
Article in English | MEDLINE | ID: mdl-35618710

ABSTRACT

Serine Incorporator 5 (SERINC5), a cellular multipass transmembrane protein that is involved in sphingolipid and phosphatydilserine biogenesis, potently restricts a number of retroviruses, including Human Immunodeficiency Virus (HIV). SERINC5 is incorporated in the budding virions leading to the inhibition of virus infectivity. In turn, retroviruses, including HIV, encode factors that counteract the antiviral effect of SERINC5. While SERINC5 has been well studied in retroviruses, little is known about its role in other viral families. Due to the paucity of information regarding host factors targeting Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2), we evaluated the effect of SERINC proteins on SARS-CoV-2 infection. Here, we show SERINC5 inhibits SARS-CoV-2 entry by blocking virus-cell fusion, and SARS-CoV-2 ORF7a counteracts the antiviral effect of SERINC5 by blocking the incorporation of over expressed SERINC5 in budding virions.


Subject(s)
COVID-19 , HIV Infections , Antiviral Agents/pharmacology , Humans , Membrane Proteins , SARS-CoV-2 , Virion/physiology
13.
J Virol ; 84(18): 9181-9, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20610716

ABSTRACT

The DA strain of Theiler's murine encephalomyelitis virus (TMEV), a member of the Cardiovirus genus of the family Picornaviridae, causes persistent infection in susceptible mice, associated with restricted expression of viral proteins, and induces a demyelinating disease of the central nervous system. DA-induced demyelinating disease serves as a model of human multiple sclerosis because of similarities in pathology and because host immune responses contribute to pathogenesis in both disorders. In contrast, the GDVII strain of TMEV causes acute lethal encephalitis with no virus persistence. Cardiovirus L is a multifunctional protein that blocks beta interferon (IFN-beta) gene transcription. We show that both DA L and GDVII L disrupt IFN-beta gene transcription induction by IFN regulatory factor 3 (IRF-3) but do so at different points in the signaling pathway. DA L blocks IFN-beta gene transcription downstream of mitochondrial antiviral signaling protein (MAVS) but upstream of IRF-3 activation, while GDVII L acts downstream of IRF-3 activation. Both DA L and GDVII L block IFN-beta gene transcription in infected mice; however, IFN-beta mRNA is expressed at low levels in the central nervous systems of mice persistently infected with DA. The particular level of IFN-beta mRNA expression set by DA L as well as other factors in the IRF-3 pathway may play a role in virus persistence, inflammation, and the restricted expression of viral proteins during the late stage of demyelinating disease.


Subject(s)
Immune Evasion , Interferon-beta/antagonists & inhibitors , Interferon-beta/immunology , Theilovirus/immunology , Theilovirus/pathogenicity , Animals , Central Nervous System/immunology , Central Nervous System/pathology , Central Nervous System/virology , Mice , Signal Transduction , Viral Proteins/immunology
14.
J Virol ; 84(3): 1348-54, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19923182

ABSTRACT

The DA strain and other members of the TO subgroup of Theiler's murine encephalomyelitis virus (TMEV) induce a persistent central nervous system infection associated with an inflammatory white matter demyelinating disease. TO subgroup strains synthesize an 18-kDa protein, L*, out of frame with the polyprotein from an initiation codon 13 nucleotides downstream from the polyprotein's AUG codon. We previously generated a mutant virus from our infectious DA full-length clone that has a change of the L* AUG codon to ACG (with no change in the polyprotein's amino acid sequence). Studies of this mutant virus showed that L* was key to the TO subgroup phenotype because the mutant had a decreased ability to persist and demyelinate. This work was initially called into question because a similar mutant derived from a different full-length DA infectious clone persisted and demyelinated similarly to wild-type DA virus (O. van Eyll and T. Michiels, J. Virol. 74:9071-9077, 2000). We now report that (i) the sequence of the L* coding region differs in the two infectious clones, resulting in a Ser or Leu as the predicted amino acid at position 93 of L* (with no change in the polyprotein's amino acid sequence), (ii) the difference in this amino acid is key to the phenotypic differences between the two mutants, and (iii) the change in amino acid 93 may affect L* phosphorylation. It is of interest that this amino acid only appears critical in determining the virus phenotype when L* is present in a significantly reduced amount (i.e., following translation from an ACG initiating codon).


Subject(s)
Demyelinating Diseases/virology , Theilovirus/physiology , Viral Proteins/physiology , Animals , Base Sequence , Blotting, Western , Cell Line , Codon , Cricetinae , DNA Primers , Mice , Theilovirus/chemistry , Viral Proteins/chemistry , Viral Proteins/genetics
15.
mBio ; 12(2)2021 03 02.
Article in English | MEDLINE | ID: mdl-33653895

ABSTRACT

The membrane-associated RING-CH (MARCH) proteins belong to a family of E3 ubiquitin ligases, whose main function is to remove transmembrane proteins from the plasma membrane. Recent work has shown that the human MARCH1, 2, and 8 are antiretroviral factors that target the human immunodeficiency virus type 1 (HIV-1) envelope glycoproteins by reducing their incorporation in the budding virions. Nevertheless, the dearth of information regarding the antiviral mechanism of this family of proteins necessitates further examination. In this study, using both the human MARCH proteins and their mouse homologues, we provide a comprehensive analysis of the antiretroviral mechanism of this family of proteins. Moreover, we show that human MARCH proteins restrict to various degrees the envelope glycoproteins of a diverse number of viruses. This report sheds light on the important antiviral function of MARCH proteins and their significance in cell intrinsic immunity.IMPORTANCE This study examines the mechanism utilized by different MARCH proteins to restrict retrovirus infection. MARCH proteins block the incorporation of envelope glycoproteins to the budding virions. In this report, by comparing the human and mouse MARCH genes and using murine leukemia virus (MLV) and HIV-1, we identify differences in the mechanism of restriction among MARCH proteins. Furthermore, we perform a comprehensive analysis on a number of envelope glycoproteins and show that MARCH proteins have broad antiviral functions.


Subject(s)
Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Viral Envelope Proteins/metabolism , Animals , HEK293 Cells , HIV-1/metabolism , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred BALB C , NIH 3T3 Cells , Ubiquitin-Protein Ligases/classification , Virus Assembly
16.
Antiviral Res ; 195: 105183, 2021 11.
Article in English | MEDLINE | ID: mdl-34626674

ABSTRACT

The likelihood of continued circulation of COVID-19 and its variants, and novel coronaviruses due to future zoonotic transmissions, combined with the current paucity of coronavirus antivirals, emphasize the need for improved screening in developing effective antivirals for the treatment of infection by SARS-CoV-2 (CoV2) and other coronaviruses. Here we report the development of a live-cell based assay for evaluating the intracellular function of the critical, highly-conserved CoV2 target, the Main 3C-like protease (Mpro). This assay is based on expression of native wild-type mature CoV2 Mpro, the function of which is quantitatively evaluated in living cells through cleavage of a biosensor leading to loss of fluorescence. Evaluation does not require cell harvesting, allowing for multiple measurements from the same cells facilitating quantification of Mpro inhibition, as well as recovery of function upon removal of inhibitory drugs. The pan-coronavirus Mpro inhibitor, GC376, was utilized in this assay and effective inhibition of intracellular CoV2 Mpro was found to be consistent with levels required to inhibit CoV2 infection of human lung cells. We demonstrate that GC376 is an effective inhibitor of intracellular CoV2 Mpro at low micromolar levels, while other predicted Mpro inhibitors, bepridil and alverine, are not. Results indicate this system can provide a highly effective high-throughput coronavirus Mpro screening system.


Subject(s)
Biosensing Techniques , Coronavirus 3C Proteases/antagonists & inhibitors , Protease Inhibitors/pharmacology , Pyrrolidines/pharmacology , SARS-CoV-2/enzymology , Sulfonic Acids/pharmacology , Drug Evaluation, Preclinical , Fluorescence , HEK293 Cells , Humans
17.
mBio ; 11(4)2020 07 14.
Article in English | MEDLINE | ID: mdl-32665269

ABSTRACT

The serine incorporator (SERINC) proteins are multipass transmembrane proteins that affect sphingolipid and phosphatidylserine synthesis. Human SERINC5 and SERINC3 were recently shown to possess antiretroviral activity for a number of retroviruses, including human immunodeficiency virus (HIV), murine leukemia virus (MLV), and equine infectious anemia virus (EIAV). In the case of MLV, the glycosylated Gag (glyco-Gag) protein was shown to counteract SERINC5-mediated restriction in in vitro experiments and the viral envelope was found to determine virion sensitivity or resistance to SERINC5. However, nothing is known about the in vivo function of SERINC5. Antiretroviral function of a host factor in vitro is not always associated with antiretroviral function in vivo Using SERINC5-/- mice that we had generated, we showed that mouse SERINC5 (mSERINC5) restriction of MLV infection in vivo is influenced not only by glyco-Gag but also by the retroviral envelope. Finally, we also examined the in vivo function of the other SERINC gene with known antiretroviral functions, SERINC3. By using SERINC3-/- mice, we found that the murine homologue, mSERINC3, had no antiretroviral role either in vivo or in vitro To our knowledge, this report provides the first data showing that SERINC5 restricts retrovirus infection in vivo and that restriction of retrovirus infectivity in vivo is dependent on the presence of both glyco-Gag and the viral envelope.IMPORTANCE This study examined for the first time the in vivo function of the serine incorporator (SERINC) proteins during retrovirus infection. SERINC3 and SERINC5 (SERINC3/5) restrict a number of retroviruses, including human immunodeficiency virus 1 (HIV-1) and murine leukemia virus (MLV), by blocking their entry into cells. Nevertheless, HIV-1 and MLV encode factors, Nef and glycosylated Gag, respectively, that counteract SERINC3/5 in vitro We recently developed SERINC3 and SERINC5 knockout mice to examine the in vivo function of these genes. We found that SERINC5 restriction is dependent on the absence of glycosylated Gag and the expression of a specific viral envelope glycoprotein. On the other hand, SERINC3 had no antiviral function. Our findings have implications for the development of therapeutics that target SERINC5 during retrovirus infection.


Subject(s)
Host-Pathogen Interactions , Leukemia, Experimental/virology , Membrane Proteins/genetics , Retroviridae Infections/virology , Tumor Virus Infections/virology , Animals , Female , Glycosylation , Leukemia Virus, Murine/pathogenicity , Male , Membrane Glycoproteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout
18.
J Exp Med ; 217(12)2020 12 07.
Article in English | MEDLINE | ID: mdl-32870257

ABSTRACT

The APOBEC3 family of antiviral DNA cytosine deaminases is implicated as the second largest source of mutation in cancer. This mutational process may be a causal driver or inconsequential passenger to the overall tumor phenotype. We show that human APOBEC3A expression in murine colon and liver tissues increases tumorigenesis. All other APOBEC3 family members, including APOBEC3B, fail to promote liver tumor formation. Tumor DNA sequences from APOBEC3A-expressing animals display hallmark APOBEC signature mutations in TCA/T motifs. Bioinformatic comparisons of the observed APOBEC3A mutation signature in murine tumors, previously reported APOBEC3A and APOBEC3B mutation signatures in yeast, and reanalyzed APOBEC mutation signatures in human tumor datasets support cause-and-effect relationships for APOBEC3A-catalyzed deamination and mutagenesis in driving multiple human cancers.


Subject(s)
Biocatalysis , Carcinogenesis/genetics , Cytidine Deaminase/genetics , Mutation/genetics , Proteins/genetics , Adenomatous Polyposis Coli/pathology , Animals , Base Sequence , Carcinogenesis/pathology , DNA Transposable Elements/genetics , Humans , Hydrolases/genetics , Intestinal Neoplasms/pathology , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Liver Regeneration , Loss of Heterozygosity/genetics , Mice, Transgenic , Polyps/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism
19.
J Virol ; 82(12): 5879-86, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18400855

ABSTRACT

The DA strain of Theiler's murine encephalomyelitis virus (TMEV) causes a persistent central nervous system (CNS) infection of mice with a restricted virus gene expression and induces an inflammatory demyelinating disease that is thought to be immune mediated and a model of multiple sclerosis (MS). The relative contribution of virus vis-à-vis the immune system in the pathogenesis of DA-induced white matter disease remains unclear, as is also true in MS. To clarify the pathogenesis of DA-induced demyelination, we used Cre/loxP technology to generate a transgenic mouse that has tamoxifen (Tm)-inducible expression of a subgenomic segment of DA RNA in oligodendrocytes and Schwann cells. Tm-treated young transgenic mice developed progressive weakness leading to death, with abnormalities of oligodendrocytes and Schwann cells and demyelination, but without inflammation, demonstrating that DA virus can play a direct pathogenic role in demyelination. Tm treatment of mice at a later age resulted in milder disease, with evidence of peripheral nerve remyelination and focal fur depigmentation; surviving weak mice had persistent expression of the recombined transgene in the CNS, suggesting that the DA subgenomic segment can cause cellular dysfunction but not death, possibly similar to the situation seen during DA virus persistence. These studies demonstrate that DA RNA or a DA protein(s) is toxic to myelin-synthesizing cells. This Cre/loxP transgenic system allows for spatially and temporally controlled expression of the viral transgene and is valuable for clarifying nonimmune (and immune) mechanisms of demyelination induced by TMEV as well as other viruses.


Subject(s)
Demyelinating Diseases/etiology , RNA, Viral/analysis , Theilovirus/pathogenicity , Animals , Brain/metabolism , Brain/ultrastructure , Brain/virology , Cell Line , Cricetinae , Demyelinating Diseases/pathology , Disease Models, Animal , Green Fluorescent Proteins/metabolism , Immunohistochemistry , Kidney/cytology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Recombination, Genetic , Sciatic Nerve/metabolism , Sciatic Nerve/ultrastructure , Sciatic Nerve/virology , Spinal Cord/metabolism , Spinal Cord/ultrastructure , Spinal Cord/virology
20.
mBio ; 9(3)2018 06 05.
Article in English | MEDLINE | ID: mdl-29871919

ABSTRACT

Host recognition of viral nucleic acids generated during infection leads to the activation of innate immune responses essential for early control of virus. Retrovirus reverse transcription creates numerous potential ligands for cytosolic host sensors that recognize foreign nucleic acids, including single-stranded RNA (ssRNA), RNA/DNA hybrids, and double-stranded DNA (dsDNA). We and others recently showed that the sensors cyclic GMP-AMP synthase (cGAS), DEAD-box helicase 41 (DDX41), and members of the Aim2-like receptor (ALR) family participate in the recognition of retroviral reverse transcripts. However, why multiple sensors might be required and their relative importance in in vivo control of retroviral infection are not known. Here, we show that DDX41 primarily senses the DNA/RNA hybrid generated at the first step of reverse transcription, while cGAS recognizes dsDNA generated at the next step. We also show that both DDX41 and cGAS are needed for the antiretroviral innate immune response to murine leukemia virus (MLV) and HIV in primary mouse macrophages and dendritic cells (DCs). Using mice with cell type-specific knockout of the Ddx41 gene, we show that DDX41 sensing in DCs but not macrophages was critical for controlling in vivo MLV infection. This suggests that DCs are essential in vivo targets for infection, as well as for initiating the antiviral response. Our work demonstrates that the innate immune response to retrovirus infection depends on multiple host nucleic acid sensors that recognize different reverse transcription intermediates.IMPORTANCE Viruses are detected by many different host sensors of nucleic acid, which in turn trigger innate immune responses, such as type I interferon (IFN) production, required to control infection. We show here that at least two sensors are needed to initiate a highly effective innate immune response to retroviruses-DDX41, which preferentially senses the RNA/DNA hybrid generated at the first step of retrovirus replication, and cGAS, which recognizes double-stranded DNA generated at the second step. Importantly, we demonstrate using mice lacking DDX41 or cGAS that both sensors are needed for the full antiviral response needed to control in vivo MLV infection. These findings underscore the need for multiple host factors to counteract retroviral infection.


Subject(s)
DEAD-box RNA Helicases/metabolism , DNA, Viral/metabolism , Leukemia Virus, Murine/genetics , RNA, Viral/metabolism , Retroviridae Infections/enzymology , Retroviridae Infections/virology , Animals , DEAD-box RNA Helicases/genetics , DNA, Viral/chemistry , DNA, Viral/genetics , Dendritic Cells/enzymology , Dendritic Cells/virology , Host-Pathogen Interactions , Humans , Leukemia Virus, Murine/physiology , Macrophages/enzymology , Macrophages/virology , Male , Mice , Mice, Inbred C57BL , Nucleotidyltransferases/genetics , Nucleotidyltransferases/metabolism , RNA, Viral/chemistry , RNA, Viral/genetics , Retroviridae Infections/genetics
SELECTION OF CITATIONS
SEARCH DETAIL