Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
bioRxiv ; 2023 Nov 02.
Article in English | MEDLINE | ID: mdl-37961113

ABSTRACT

Replication of the complex retrovirus mouse mammary tumor virus (MMTV) is antagonized by murine Apobec3 (mA3), a member of the Apobec family of cytidine deaminases. We have shown that MMTV-encoded Rem protein inhibits proviral mutagenesis by the Apobec enzyme, activation-induced cytidine deaminase (AID) during viral replication in BALB/c mice. To further study the role of Rem in vivo , we have infected C57BL/6 (B6) mice with a superantigen-independent lymphomagenic strain of MMTV (TBLV-WT) or a mutant strain (TBLV-SD) that is defective in Rem and its cleavage product Rem-CT. Unlike MMTV, TBLV induced T-cell tumors in µMT mice, indicating that mature B cells, which express the highest AID levels, are not required for TBLV replication. Compared to BALB/c, B6 mice were more susceptible to TBLV infection and tumorigenesis. The lack of Rem expression accelerated B6 tumorigenesis at limiting doses compared to TBLV-WT in either wild-type B6 or AID-deficient mice. However, unlike proviruses from BALB/c mice, high-throughput sequencing indicated that proviral G-to-A or C-to-T changes did not significantly differ in the presence and absence of Rem expression. Ex vivo stimulation showed higher levels of mA3 relative to AID in B6 compared to BALB/c splenocytes, but effects of agonists differed in the two strains. RNA-Seq revealed increased transcripts related to growth factor and cytokine signaling in TBLV-SD-induced tumors relative to those from TBLV-WT, consistent with a third Rem function. Thus, Rem-mediated effects on tumorigenesis in B6 mice are independent of Apobec-mediated proviral hypermutation.

2.
Microorganisms ; 8(12)2020 Nov 30.
Article in English | MEDLINE | ID: mdl-33266042

ABSTRACT

Apolipoprotein B mRNA-editing enzyme catalytic polypeptide-like (APOBEC) proteins are a diverse and evolutionarily conserved family of cytidine deaminases that provide a variety of functions from tissue-specific gene expression and immunoglobulin diversity to control of viruses and retrotransposons. APOBEC family expansion has been documented among mammalian species, suggesting a powerful selection for their activity. Enzymes with a duplicated zinc-binding domain often have catalytically active and inactive domains, yet both have antiviral function. Although APOBEC antiviral function was discovered through hypermutation of HIV-1 genomes lacking an active Vif protein, much evidence indicates that APOBECs also inhibit virus replication through mechanisms other than mutagenesis. Multiple steps of the viral replication cycle may be affected, although nucleic acid replication is a primary target. Packaging of APOBECs into virions was first noted with HIV-1, yet is not a prerequisite for viral inhibition. APOBEC antagonism may occur in viral producer and recipient cells. Signatures of APOBEC activity include G-to-A and C-to-T mutations in a particular sequence context. The importance of APOBEC activity for viral inhibition is reflected in the identification of numerous viral factors, including HIV-1 Vif, which are dedicated to antagonism of these deaminases. Such viral antagonists often are only partially successful, leading to APOBEC selection for viral variants that enhance replication or avoid immune elimination.

3.
mBio ; 10(4)2019 08 13.
Article in English | MEDLINE | ID: mdl-31409681

ABSTRACT

Complex human-pathogenic retroviruses cause high morbidity and mortality worldwide, but resist antiviral drugs and vaccine development due to evasion of the immune response. A complex retrovirus, mouse mammary tumor virus (MMTV), requires replication in B and T lymphocytes for mammary gland transmission and is antagonized by the innate immune restriction factor murine Apobec3 (mA3). To determine whether the regulatory/accessory protein Rem affects innate responses to MMTV, a splice-donor mutant (MMTV-SD) lacking Rem expression was injected into BALB/c mice. Mammary tumors induced by MMTV-SD had a lower proviral load, lower incidence, and longer latency than mammary tumors induced by wild-type MMTV (MMTV-WT). MMTV-SD proviruses had many G-to-A mutations on the proviral plus strand, but also C-to-T transitions within WRC motifs. Similarly, a lymphomagenic MMTV variant lacking Rem expression showed decreased proviral loads and increased WRC motif mutations relative to those in wild-type-virus-induced tumors, consistent with activation-induced cytidine deaminase (AID) mutagenesis in lymphoid cells. These mutations are typical of the Apobec family member AID, a B-cell-specific mutagenic protein involved in antibody variable region hypermutation. In contrast, mutations in WRC motifs and proviral loads were similar in MMTV-WT and MMTV-SD proviruses from tumors in AID-insufficient mice. AID was not packaged in MMTV virions. Rem coexpression in transfection experiments led to AID proteasomal degradation. Our data suggest that rem specifies a human-pathogenic immunodeficiency virus type 1 (HIV-1) Vif-like protein that inhibits AID and antagonizes innate immunity during MMTV replication in lymphocytes.IMPORTANCE Complex retroviruses, such as human-pathogenic immunodeficiency virus type 1 (HIV-1), cause many human deaths. These retroviruses produce lifelong infections through viral proteins that interfere with host immunity. The complex retrovirus mouse mammary tumor virus (MMTV) allows for studies of host-pathogen interactions not possible in humans. A mutation preventing expression of the MMTV Rem protein in two different MMTV strains decreased proviral loads in tumors and increased viral genome mutations typical of an evolutionarily ancient enzyme, AID. Although the presence of AID generally improves antibody-based immunity, it may contribute to human cancer progression. We observed that coexpression of MMTV Rem and AID led to AID destruction. Our results suggest that Rem is the first known protein inhibitor of AID and that further experiments could lead to new disease treatments.


Subject(s)
Cytidine Deaminase/antagonists & inhibitors , Mammary Tumor Virus, Mouse/genetics , Proviruses/genetics , Viral Regulatory and Accessory Proteins/genetics , Animals , Cytidine Deaminase/genetics , Cytidine Deaminase/metabolism , Female , Immunity, Innate , Male , Mammary Neoplasms, Experimental/immunology , Mammary Neoplasms, Experimental/virology , Mammary Tumor Virus, Mouse/physiology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Mutant Strains , Mutation , Proviruses/physiology , Retroviridae Infections/immunology , Retroviridae Infections/virology , Tumor Virus Infections/immunology , Tumor Virus Infections/virology , Viral Load/genetics , Viral Regulatory and Accessory Proteins/metabolism , Virus Replication
4.
mBio ; 8(2)2017 03 28.
Article in English | MEDLINE | ID: mdl-28351922

ABSTRACT

Multiple pathogens, including viruses and bacteria, manipulate endoplasmic reticulum-associated degradation (ERAD) to avoid the host immune response and promote their replication. The betaretrovirus mouse mammary tumor virus (MMTV) encodes Rem, which is a precursor protein that is cleaved into a 98-amino-acid signal peptide (SP) and a C-terminal protein (Rem-CT). SP uses retrotranslocation for ER membrane extraction and yet avoids ERAD by an unknown mechanism to enter the nucleus and function as a Rev-like protein. To determine how SP escapes ERAD, we used a ubiquitin-activated interaction trap (UBAIT) screen to trap and identify transient protein interactions with SP, including the ERAD-associated p97 ATPase, but not E3 ligases or Derlin proteins linked to retrotranslocation, polyubiquitylation, and proteasomal degradation of extracted proteins. A dominant negative p97 ATPase inhibited both Rem and SP function. Immunoprecipitation experiments indicated that Rem, but not SP, is polyubiquitylated. Using both yeast and mammalian expression systems, linkage of a ubiquitin-like domain (UbL) to SP or Rem induced degradation by the proteasome, whereas SP was stable in the absence of the UbL. ERAD-associated Derlin proteins were not required for SP activity. Together, these results suggested that Rem uses a novel p97-dependent, Derlin-independent retrotranslocation mechanism distinct from other pathogens to avoid SP ubiquitylation and proteasomal degradation.IMPORTANCE Bacterial and viral infections produce pathogen-specific proteins that interfere with host functions, including the immune response. Mouse mammary tumor virus (MMTV) is a model system for studies of human complex retroviruses, such as HIV-1, as well as cancer induction. We have shown that MMTV encodes a regulatory protein, Rem, which is cleaved into an N-terminal signal peptide (SP) and a C-terminal protein (Rem-CT) within the endoplasmic reticulum (ER) membrane. SP function requires ER membrane extraction by retrotranslocation, which is part of a protein quality control system known as ER-associated degradation (ERAD) that is essential to cellular health. Through poorly understood mechanisms, certain pathogen-derived proteins are retrotranslocated but not degraded. We demonstrate here that MMTV SP retrotranslocation from the ER membrane avoids degradation through a unique process involving interaction with cellular p97 ATPase and failure to acquire cellular proteasome-targeting sequences.


Subject(s)
Adenosine Triphosphatases/metabolism , Immune Evasion , Mammary Tumor Virus, Mouse/immunology , Mammary Tumor Virus, Mouse/physiology , Nuclear Proteins/metabolism , Proteasome Endopeptidase Complex/metabolism , Protein Sorting Signals , Viral Proteins/metabolism , Cell Line , Humans , Membrane Proteins/metabolism , Protein Transport , Proteolysis
5.
Proc Natl Acad Sci U S A ; 112(11): 3505-10, 2015 Mar 17.
Article in English | MEDLINE | ID: mdl-25737543

ABSTRACT

Retroviruses cause immunodeficiency and cancer but also are used as vectors for the expression of heterologous genes. Nevertheless, optimal translation of introduced genes often is not achieved. Here we show that transfection into mammalian cells of lentiviral or gammaretroviral vectors, including those with specific shRNAs, increased expression of a cotransfected gene relative to standard plasmid vectors. Levels of most endogenous cellular proteins were unchanged. Transfer of lentiviral vector sequences into a standard plasmid conferred the ability to give increased expression of cotransfected genes (superinduction). Superinduction by the retroviral vector was not dependent on the cell type or species, the type of reporter gene, or the method of transfection. No differences were detected in the IFN, unfolded protein, or stress responses in the presence of retroviral vectors. RT-PCRs revealed that RNA levels of cotransfected genes were unchanged during superinduction, yet Western blotting, pulse labeling, and the use of bicistronic vectors showed increased cap-dependent translation of cointroduced genes. Expression of the mammalian target of rapamycin (mTOR) kinase target 4E-BP1, but not the mTOR inhibitor Torin 1, preferentially inhibited superinduction relative to basal protein expression. Furthermore, transcription of lentiviral vector sequences from a doxycycline-inducible promoter eliminated superinduction, consistent with a DNA-triggered event. Thus, retroviral DNA increased translation of cointroduced genes in trans by an mTOR-independent signaling mechanism. Our experiments have broad applications for the design of retroviral vectors for transfections, DNA vaccines, and gene therapy.


Subject(s)
Genetic Vectors/metabolism , Lentivirus/metabolism , Protein Biosynthesis , RNA Caps/metabolism , DNA/metabolism , Eukaryotic Initiation Factor-4E/biosynthesis , Gene Expression , Interferons/metabolism , RNA, Small Interfering/metabolism , Stress, Physiological , TOR Serine-Threonine Kinases/metabolism , Transcription, Genetic , Transfection
6.
Front Microbiol ; 5: 330, 2014.
Article in English | MEDLINE | ID: mdl-25071743

ABSTRACT

Endoplasmic reticulum (ER)-associated degradation (ERAD) is a universally important process among eukaryotic cells. ERAD is necessary to preserve cell integrity since the accumulation of defective proteins results in diseases associated with neurological dysfunction, cancer, and infections. This process involves recognition of misfolded or misassembled proteins that have been translated in association with ER membranes. Recognition of ERAD substrates leads to their extraction through the ER membrane (retrotranslocation or dislocation), ubiquitination, and destruction by cytosolic proteasomes. This review focuses on ERAD and its components as well as how viruses use this process to promote their replication and to avoid the immune response.

7.
J Virol ; 86(1): 214-25, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22072771

ABSTRACT

Mouse mammary tumor virus (MMTV) encodes a Rev-like protein, Rem, which is involved in the nuclear export and expression of viral RNA. Previous data have shown that all Rev-like functions are localized to the 98-amino-acid signal peptide (SP) at the N terminus of MMTV Rem or envelope proteins. MMTV-SP uses endoplasmic reticulum-associated degradation (ERAD) for protein trafficking. Rem cleavage by signal peptidase in the ER is necessary for MMTV-SP function in a reporter assay, but many requirements for trafficking are not known. To allow detection and localization of both MMTV-SP and the C-terminal cleavage product, we prepared plasmids expressing green fluorescent protein (GFP) tags. N-terminal Rem tagging led to protein accumulation relative to untagged Rem and allowed signal peptidase cleavage but reduced its specific activity. C-terminal tagging also led to Rem accumulation yet dramatically reduced cleavage, GFP fluorescence, and activity relative to N-terminally tagged Rem (GFPRem). Substitutions of an invariant leucine at position 71 between the known RNA-binding and nuclear export sequences interfered with GFPRem accumulation and activity but not cleavage. Similarly, deletion of 100 or 150 C-terminal amino acids from GFPRem dramatically reduced both Rem and MMTV-SP levels and function. Removal of the entire C terminus (203 amino acids) restored both protein levels and activity of MMTV-SP. Only C-terminal GFP tagging, and not other modifications, appeared to trap Rem in the ER membrane. Thus, Rem conformation in both the ER lumen and cytoplasm determines cleavage, retrotranslocation, and MMTV-SP function. These mutants further characterize intermediates in Rem trafficking and have implications for all proteins affected by ERAD.


Subject(s)
Mammary Tumor Virus, Mouse/metabolism , Protein Processing, Post-Translational , Protein Sorting Signals , Retroviridae Infections/virology , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/metabolism , Amino Acid Motifs , Animals , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/virology , Endoplasmic Reticulum-Associated Degradation , Humans , Mammary Tumor Virus, Mouse/chemistry , Mammary Tumor Virus, Mouse/genetics , Mice , Retroviridae Infections/metabolism , Viral Envelope Proteins/genetics
8.
Proc Natl Acad Sci U S A ; 107(27): 12287-92, 2010 Jul 06.
Article in English | MEDLINE | ID: mdl-20566871

ABSTRACT

Mouse mammary tumor virus (MMTV) is a complex murine retrovirus that encodes an HIV Rev-like export protein, Rem, from a doubly spliced version of envelope (Env) mRNA. Previously, the N-terminal 98-amino acid sequence of Rem, which is identical to Env signal peptide (SP), and full-length Rem were shown to be functional in a reporter assay that measures a postexport function. Here we show that MMTV-infected cells or cells transfected with rem or env cDNAs express SP, which is the active component in the reporter assay. Uncleaved Rem was partially glycosylated, but mutations in both glycosylation sites within the C terminus prevented Rem function. Mutations that reduced Rem or Env cleavage by signal peptidase greatly reduced SP levels and functional activity in the reporter assay and allowed accumulation of the uncleaved protein. Fluorescence microscopy revealed that GFP-tagged cleavage-site mutants are unstable and lack fluorescence compared with wild-type Rem, suggesting improper folding. Proteasome inhibitors allowed accumulation of uncleaved Rem relative to SP and increased reporter activity, consistent with SP retrotranslocation and proteasome escape before nuclear entry. Expression of a dominant-negative p97 ATPase did not alter levels of unprocessed Rem and SP but decreased reporter activity, suggesting p97-facilitated retrotranslocation of SP. Our results provide an example of a SP that is processed by signal peptidase and retrotranslocated to allow nuclear localization and function.


Subject(s)
Cell Nucleus/metabolism , Mammary Tumor Virus, Mouse/metabolism , Membrane Proteins/metabolism , Serine Endopeptidases/metabolism , Viral Envelope Proteins/metabolism , Active Transport, Cell Nucleus , Alternative Splicing , Animals , Binding Sites/genetics , Blotting, Western , Cell Line , Glycosylation , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Jurkat Cells , Mammary Tumor Virus, Mouse/genetics , Mice , Microscopy, Fluorescence , Mutation , Protein Processing, Post-Translational , Protein Sorting Signals/genetics , Viral Envelope Proteins/genetics
9.
J Biol Chem ; 284(38): 25642-52, 2009 Sep 18.
Article in English | MEDLINE | ID: mdl-19632991

ABSTRACT

Mouse mammary tumor virus (MMTV) is a complex retrovirus that encodes at least three regulatory and accessory proteins, including Rem. Rem is required for nuclear export of unspliced viral RNA and efficient expression of viral proteins. Our previous data indicated that sequences at the envelope-3' long terminal repeat junction are required for proper export of viral RNA. To further map the Rem-responsive element (RmRE), reporter vectors containing various portions of the viral envelope gene and the 3' long terminal repeat were tested in the presence and absence of Rem in transient transfection assays. A 476-bp fragment that spans the envelope-long terminal repeat junction had activity equivalent to the entire 3'-end of the mouse mammary tumor virus genome, but further deletions at the 5'- or 3'-ends reduced Rem responsiveness. RNase structure mapping of the full-length RmRE and a 3'-truncation suggested multiple domains with local base pairing and intervening single-stranded segments. A secondary structure model constrained by these data is reminiscent of the RNA response elements of other complex retroviruses, with numerous local stem-loops and long-range base pairs near the 5'- and 3'-boundaries, and differs substantially from an earlier model generated without experimental constraints. Covariation analysis provides limited support for basic features of our model. Reporter assays in human and mouse cell lines revealed similar boundaries, suggesting that the RmRE does not require cell type-specific proteins to form a functional structure.


Subject(s)
Genome, Viral/physiology , Mammary Tumor Virus, Mouse/metabolism , Monomeric GTP-Binding Proteins/metabolism , RNA, Viral/metabolism , Response Elements/physiology , Terminal Repeat Sequences/physiology , Animals , Cell Line , Chromosome Mapping/methods , Humans , Mammary Tumor Virus, Mouse/genetics , Mice , Monomeric GTP-Binding Proteins/genetics , RNA, Viral/genetics , Rats , Viral Envelope Proteins/genetics , Viral Envelope Proteins/metabolism
10.
Mol Cells ; 14(2): 185-91, 2002 Oct 31.
Article in English | MEDLINE | ID: mdl-12442889

ABSTRACT

Kaposi's sarcoma (KS)-associated herpesvirus (KSHV) belongs to the human gammaherpesvirus family that undergoes both lytic and latent life cycles in host cells. Open reading frame (ORF) 50 is the most important protein in reactivation to lytic phase and functions as a strong transcriptional activator of the early and late genes of KSHV. Since transactivation of promoters by ORF50 is achieved via response elements, we have attempted to identify ORF50 response elements in K8 and ORF57 promoters of KSHV by transient transfection assays with deletion mutants. Our data reveal that specific regions within the K8 (74661-74760) and ORF57 (81851-81931) promoters contain ORF50 response elements, which are heterogeneous, unlike those of Epstein-Barr virus and Herpesvirus saimiri. We additionally identify an AP-1 binding site at the ORF57 promoter between 81882 and 81889, and show that AP-1 participates in ORF57 promoter activation by ORF50. Our findings collectively indicate that ORF50 activates various viral proteins through both direct binding and cellular transcriptional factor-mediated mechanisms.


Subject(s)
Carrier Proteins/metabolism , Herpesvirus 8, Human/metabolism , Immediate-Early Proteins/metabolism , Promoter Regions, Genetic , Trans-Activators/metabolism , Viral Proteins/metabolism , Basic-Leucine Zipper Transcription Factors , Binding Sites/genetics , Carrier Proteins/genetics , Herpesvirus 8, Human/genetics , Humans , Immediate-Early Proteins/genetics , Repressor Proteins , Trans-Activators/genetics , Transcription Factor AP-1/metabolism , Viral Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...