Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 230
Filter
1.
Viruses ; 14(7)2022 07 13.
Article in English | MEDLINE | ID: mdl-35891505

ABSTRACT

Herpesvirus saimiri (HVS) is discussed as a possible vector in gene therapy. In order to create a self-repairing HVS vector, the F plasmid vector moiety of the bacterial artificial chromosome (BAC) was transposed via Red recombination into the virus genes ORF22 or ORF29b, both important for virus replication. Repetitive sequences were additionally inserted, allowing the removal of the F-derived sequences from the viral DNA genome upon reconstitution in permissive epithelial cells. Moreover, these self-repair-enabled BACs were used to generate deletion variants of the transforming strain C488 in order to minimalize the virus genome. Using the en passant mutagenesis with two subsequent homologous recombination steps, the BAC was seamlessly manipulated. To ensure the replication capacity in permissive monkey cells, replication kinetics for all generated virus variants were documented. HVS variants with increased insert capacity reached the self-repair within two to three passages in permissive epithelial cells. The seamless deletion of ORFs 3/21, 12-14, 16 or 71 did not abolish replication competence. Apoptosis induction did not seem to be altered in human T cells transformed with deletion variants lacking ORF16 or ORF71. These virus variants form an important step towards creating a potential minimal virus vector for gene therapy, for example, in human T cells.


Subject(s)
Herpesvirus 2, Saimiriine , Chromosomes, Artificial, Bacterial/genetics , Genes, Viral , Genetic Vectors , Genome, Viral , Herpesvirus 2, Saimiriine/genetics , Humans
2.
Nucleic Acids Res ; 50(11): 6511-6520, 2022 06 24.
Article in English | MEDLINE | ID: mdl-35648438

ABSTRACT

HSUR1 and HSUR2, two noncoding RNAs expressed by the oncogenic Herpesvirus saimiri, bind host microRNAs miR-142-3p, miR-16, and miR-27 with different purposes. While binding of miR-27 to HSUR1 triggers the degradation of the microRNA, miR-16 is tethered by HSUR2 to target host mRNAs to repress their expression. Here we show that the interaction with miR-142-3p is required for the activity of both HSURs. Coimmunoprecipitation experiments revealed that miR-142-3p allosterically regulates the binding of miR-27 and miR-16 to HSUR1 and HSUR2, respectively. The binding of two different miRNAs to each HSUR is not cooperative. HSURs can be engineered to be regulated by other miRNAs, indicating that the identity of the binding miRNA is not important for HSUR regulation. Our results uncover a mechanism for allosteric regulation of noncoding RNA function and a previously unappreciated way in which microRNAs can regulate gene expression.


Subject(s)
Allosteric Regulation , Herpesvirus 2, Saimiriine , MicroRNAs , RNA, Untranslated , Herpesviridae Infections/metabolism , Herpesvirus 2, Saimiriine/genetics , MicroRNAs/genetics , MicroRNAs/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Untranslated/genetics , RNA, Untranslated/metabolism
3.
Mamm Genome ; 33(2): 412-420, 2022 06.
Article in English | MEDLINE | ID: mdl-34491378

ABSTRACT

Insights into interactions between viral factors and the cellular machinery usually lead to discoveries concerning host cell biology. Thus, the gene expression field has historically relied on viral model systems to discover mechanisms underlying different cellular processes. In recent years, the functional characterization of the small nuclear noncoding RNAs expressed by the oncogenic Herpesvirus saimiri, called HSURs, resulted in the discovery of two mechanisms for the regulation of gene expression. HSUR1 and HSUR2 associate with host microRNAs, which are small noncoding RNAs that broadly regulate gene expression by binding to messenger RNAs. HSUR1 provided the first example of a process known as target-directed miRNA degradation that operates in cells to regulate miRNA populations. HSUR2 functions as a miRNA adaptor, uncovering an entirely new, indirect mechanism by which miRNAs can inhibit mRNA function. Here, I review the path that led to these discoveries and their implications and postulate new exciting questions about the functions of these fascinating viral noncoding RNAs.


Subject(s)
Herpesvirus 2, Saimiriine , MicroRNAs , Viruses , Herpesvirus 2, Saimiriine/genetics , Herpesvirus 2, Saimiriine/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Untranslated/genetics , RNA, Untranslated/metabolism , Viruses/genetics , Viruses/metabolism
4.
Elife ; 82019 09 20.
Article in English | MEDLINE | ID: mdl-31538617

ABSTRACT

HSUR2 is a viral non-coding RNA (ncRNA) that functions as a microRNA (miRNA) adaptor. HSUR2 inhibits apoptosis in infected cells by recruiting host miRNAs miR-142-3p and miR-16 to mRNAs encoding apoptotic factors. HSUR2's target recognition mechanism is not understood. It is also unknown why HSUR2 utilizes miR-16 to downregulate only a subset of transcripts. We developed a general method for individual-nucleotide resolution RNA-RNA interaction identification by crosslinking and capture (iRICC) to identify sequences mediating interactions between HSUR2 and target mRNAs in vivo. Mutational analyses confirmed identified HSUR2-mRNA interactions and validated iRICC as a method that confidently determines sequences mediating RNA-RNA interactions in vivo. We show that HSUR2 does not display a 'seed' region to base-pair with most target mRNAs, but instead uses different regions to interact with different transcripts. We further demonstrate that this versatile mode of interaction via variable base-pairing provides HSUR2 with a mechanism for differential miRNA recruitment.


Subject(s)
Base Pairing , Herpesvirus 2, Saimiriine/genetics , Host-Pathogen Interactions , MicroRNAs/genetics , RNA, Messenger/genetics , RNA, Viral/genetics , Animals , Cell Line , DNA Mutational Analysis , Herpesvirus 2, Saimiriine/growth & development , Humans , MicroRNAs/metabolism , RNA, Messenger/metabolism , RNA, Viral/metabolism
5.
Nucleic Acids Res ; 47(4): 1987-2001, 2019 02 28.
Article in English | MEDLINE | ID: mdl-30462297

ABSTRACT

Kaposi's sarcoma-associated herpesvirus (KSHV) transcribes a long noncoding polyadenylated nuclear (PAN) RNA, which promotes the latent to lytic transition by repressing host genes involved in antiviral responses as well as viral proteins that support the latent state. KSHV also expresses several early proteins including ORF57 (Mta), a member of the conserved multifunctional ICP27 protein family, which is essential for productive replication. ORF57/Mta interacts with PAN RNA via a region termed the Mta responsive element (MRE), stabilizing the transcript and supporting nuclear accumulation. Here, using a close homolog of KSHV ORF57 from herpesvirus saimiri (HVS), we determined the crystal structure of the globular domain in complex with a PAN RNA MRE, revealing a uracil specific binding site that is also conserved in KSHV. Using solution NMR, RNA binding was also mapped within the disordered N-terminal domain of KSHV ORF57, and showed specificity for an RNA fragment containing a GAAGRG motif previously known to bind a homologous region in HVS ORF57. Together these data located novel differential RNA recognition sites within neighboring domains of herpesvirus ORF57 homologs, and revealed high-resolution details of their interactions with PAN RNA, thus providing insight into interactions crucial to viral function.


Subject(s)
Herpesvirus 8, Human/genetics , RNA, Long Noncoding/genetics , RNA-Binding Proteins/genetics , Viral Regulatory and Accessory Proteins/genetics , Binding Sites/genetics , Gene Expression Regulation, Viral , Herpesvirus 2, Saimiriine/genetics , Humans , Immediate-Early Proteins/genetics , Nucleotide Motifs/genetics , RNA, Messenger/genetics
6.
RNA Biol ; 15(7): 856-862, 2018.
Article in English | MEDLINE | ID: mdl-29895222

ABSTRACT

Viruses masterfully regulate host gene expression during infection. Many do so, in part, by expressing non-coding RNAs. Recent work has shown that HSUR 2, a viral non-coding RNA expressed by the oncogenic Herpesvirus saimiri, regulates mRNA expression through a novel mechanism. HSUR 2 base pairs with both target mRNAs and host miRNAs in infected cells. This results in HSUR 2-dependent recruitment of host miRNAs and associated Ago proteins to target mRNAs, and the subsequent destabilization of target mRNAs. Using this mechanism, this virus regulates key cellular pathways during viral infection. Here I discuss the evolution of our thinking about HSUR function and explore the implications of recent findings in relation to the current views on the functions of interactions between miRNAs and other classes of non-coding RNAs, the potential advantages of this mechanism of regulation of gene expression, and the evolutionary origin of HSUR 2.


Subject(s)
Gene Expression Regulation, Viral , Herpesviridae Infections/metabolism , Herpesvirus 2, Saimiriine/genetics , RNA, Untranslated/metabolism , RNA, Viral/metabolism , Argonaute Proteins/genetics , Argonaute Proteins/metabolism , Base Sequence , Herpesviridae Infections/virology , Host-Pathogen Interactions , Humans , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Untranslated/genetics , RNA, Viral/genetics
7.
Nature ; 550(7675): 275-279, 2017 10 12.
Article in English | MEDLINE | ID: mdl-28976967

ABSTRACT

Viruses express several classes of non-coding RNAs; the functions and mechanisms by which most of these act are unknown. Herpesvirus saimiri, a γ-herpesvirus that establishes latency in the T cells of New World primates and has the ability to cause aggressive leukaemias and lymphomas in non-natural hosts, expresses seven small nuclear uracil-rich non-coding RNAs (called HSURs) in latently infected cells. These HSURs associate with Sm proteins, and share biogenesis and structural features with cellular Sm-class small nuclear RNAs. One of these HSURs (HSUR2) base-pairs with two host cellular microRNAs (miR-142-3p and miR-16) but does not affect their abundance or activity, which suggests that its interactions with them perform alternative functions. Here we show that HSUR2 also base-pairs with mRNAs in infected cells. We combined in vivo psoralen-mediated RNA-RNA crosslinking and high-throughput sequencing to identify the mRNAs targeted by HSUR2, which include mRNAs that encode retinoblastoma and factors involved in p53 signalling and apoptosis. We show that HSUR2 represses the expression of target mRNAs and that base-pairing between HSUR2 and miR-142-3p and miR-16 is essential for this repression, suggesting that HSUR2 recruits these two cellular microRNAs to its target mRNAs. Furthermore, we show that HSUR2 uses this mechanism to inhibit apoptosis. Our results uncover a role for this viral Sm-class RNA as a microRNA adaptor in the regulation of gene expression that follows precursor mRNA processing.


Subject(s)
Apoptosis/genetics , Base Pairing , Herpesvirus 2, Saimiriine/genetics , MicroRNAs/metabolism , RNA, Messenger/genetics , RNA, Viral/genetics , Animals , Callithrix , Gene Expression Regulation , MicroRNAs/genetics , Models, Biological , Protein Biosynthesis , RNA Stability , RNA, Messenger/biosynthesis , RNA, Messenger/chemistry , RNA, Messenger/metabolism , RNA, Viral/chemistry , T-Lymphocytes/metabolism , T-Lymphocytes/virology
8.
Biochim Biophys Acta Mol Cell Res ; 1864(1): 1-11, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27751885

ABSTRACT

Members of the herpesviral family use multiple strategies to hijack infected host cells and exploit cellular signaling for their pathogenesis and latent infection. Among the most intriguing weapons in the arsenal of pathogenic herpesviruses are the constitutively active virally-encoded G protein-coupled receptors (vGPCRs). Even though vGPCRs contribute to viral pathogenesis such as immune evasion and proliferative disorders, the molecular details of how vGPCRs continuously activate cellular signaling are largely unknown. Here, we report that the vGPCR of Herpesvirus saimiri (HVS), an oncogenic γ2-herpesvirus, constitutively activates T cells via a heteromeric interaction with cellular CXCR4. Constitutive T cell activation also occurs with expression of the vGPCR of Kaposi's sarcoma-associated herpesvirus (KSHV), but not the vGPCR of Epstein-Barr virus. Expression of HVS vGPCR down-regulated the surface expression of CXCR4 but did not induce the degradation of the chemokine receptor, suggesting that vGPCR/CXCR4 signaling continues in cytosolic compartments. The physical association of vGPCR with CXCR4 was demonstrated by proximity ligation assay as well as immunoprecipitation. Interestingly, the constitutive activation of T cells by HVS vGPCR is independent of proximal T cell receptor (TCR) signaling molecules, such as TCRß, Lck, and ZAP70, whereas CXCR4 silencing by shRNA abolished T cell activation by vGPCRs of HVS and KSHV. Furthermore, previously identified inactive vGPCR mutants failed to interact with CXCR4. These findings on the positive cooperativity of vGPCR with cellular CXCR4 in T cell activation extend our current understanding of the molecular mechanisms of vGPCR function and highlight the importance of heteromerization for GPCR activity.


Subject(s)
Herpesvirus 2, Saimiriine/metabolism , Herpesvirus 8, Human/metabolism , Receptors, CXCR4/genetics , Receptors, Chemokine/genetics , T-Lymphocytes/virology , Gene Expression Regulation , HEK293 Cells , Herpesvirus 2, Saimiriine/genetics , Herpesvirus 2, Saimiriine/growth & development , Herpesvirus 4, Human/genetics , Herpesvirus 4, Human/growth & development , Herpesvirus 4, Human/metabolism , Herpesvirus 8, Human/genetics , Herpesvirus 8, Human/growth & development , Host-Pathogen Interactions , Humans , Lymphocyte Activation , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/genetics , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/immunology , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/metabolism , Primary Cell Culture , Protein Binding , Protein Multimerization , Receptors, Antigen, T-Cell, alpha-beta/genetics , Receptors, Antigen, T-Cell, alpha-beta/immunology , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Receptors, CXCR4/immunology , Receptors, CXCR4/metabolism , Receptors, Chemokine/immunology , Receptors, Chemokine/metabolism , Signal Transduction , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , ZAP-70 Protein-Tyrosine Kinase/genetics , ZAP-70 Protein-Tyrosine Kinase/immunology , ZAP-70 Protein-Tyrosine Kinase/metabolism
9.
Curr Protoc Immunol ; 115: 7.21C.1-7.21C.12, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27801513

ABSTRACT

Human T cells can be transformed and expanded with herpesvirus saimiri (HVS). HVS-transformed T cells from patients have facilitated the study of a broad range of primary immunodeficiencies (PID) in which T-cell development or function is altered. However, the utility of HVS-transformed T cells for genetic studies has been limited by technical challenges in the expression of exogenous genes, including wild-type or mutant alleles. A novel, gamma retrovirus-based method for the simple and reliable transduction, purification, and study of HVS-transformed T cells is described. © 2016 by John Wiley & Sons, Inc.


Subject(s)
Genetic Vectors/genetics , Herpesvirus 2, Saimiriine/genetics , T-Lymphocytes/metabolism , Transduction, Genetic/methods , Transgenes , Animals , Cell Line, Transformed , Gene Order , Gene Transfer Techniques , Humans , Plasmids/genetics , Retroviridae/genetics
10.
RNA ; 22(8): 1181-9, 2016 08.
Article in English | MEDLINE | ID: mdl-27335146

ABSTRACT

Herpesvirus saimiri, an oncogenic herpesvirus, during latency produces seven small nuclear RNAs, called the Herpesvirus saimiri U RNAs (HSUR1-7). HSUR1 mediates degradation of the host microRNA, miR-27, via a process that requires imperfect base-pairing. The decreased levels of miR-27 lead to prolonged T-cell activation and likely contribute to oncogenesis. To gain insight into HSUR1-mediated degradation of miR-27, we probed the in vivo secondary structure of HSUR1 and coupled this with bioinformatic structural analyses. The results suggest that HSUR1 adopts a conformation different than previously believed and that the region complementary to miR-27 lacks stable structure. To determine whether HSUR1 structural flexibility is important for its ability to mediate miR-27 degradation, we performed structurally informative mutagenic analyses of HSUR1. HSUR1 mutants in which the miR-27 binding site sequence is preserved, but sequestered in predicted helices, lose their ability to decrease miR-27 levels. These results indicate that the HSUR1 miR27-binding region must be available in a conformationally flexible segment for noncoding RNA function.


Subject(s)
Genes, Viral , Herpesvirus 2, Saimiriine/metabolism , MicroRNAs/metabolism , RNA, Small Nuclear/metabolism , Animals , Callithrix , Herpesvirus 2, Saimiriine/genetics
11.
J Immunol Res ; 2015: 395371, 2015.
Article in English | MEDLINE | ID: mdl-26539553

ABSTRACT

The Lck interacting protein Tip of Herpesvirus saimiri is responsible for T-cell transformation both in vitro and in vivo. Here we designed the chimeric peptide hTip-CSKH, comprising the Lck specific interacting motif CSKH of Tip and its hydrophobic transmembrane sequence (hTip), the latter as a vector targeting lipid rafts. We found that hTip-CSKH can induce a fivefold increase in proliferation of human and Aotus sp. T-cells. Costimulation with PMA did not enhance this proliferation rate, suggesting that hTip-CSKH is sufficient and independent of further PKC stimulation. We also found that human Lck phosphorylation was increased earlier after stimulation when T-cells were incubated previously with hTip-CSKH, supporting a strong signalling and proliferative effect of the chimeric peptide. Additionally, Lck downstream signalling was evident with hTip-CSKH but not with control peptides. Importantly, hTip-CSKH could be identified in heavy lipid rafts membrane fractions, a compartment where important T-cell signalling molecules (LAT, Ras, and Lck) are present during T-cell activation. Interestingly, hTip-CSKH was inhibitory to Jurkat cells, in total agreement with the different signalling pathways and activation requirements of this leukemic cell line. These results provide the basis for the development of new compounds capable of modulating therapeutic targets present in lipid rafts.


Subject(s)
Herpesvirus 2, Saimiriine/chemistry , Lymphocyte Activation , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/metabolism , Peptides/genetics , Phosphoproteins/chemistry , Phosphoproteins/metabolism , T-Lymphocytes/immunology , Viral Proteins/chemistry , Viral Proteins/metabolism , Amino Acid Motifs , Animals , Aotidae , Herpesvirus 2, Saimiriine/genetics , Humans , Jurkat Cells , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/genetics , Membrane Microdomains/metabolism , Peptides/chemistry , Phosphoproteins/immunology , Phosphorylation , Phytohemagglutinins/immunology , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/immunology , Signal Transduction , T-Lymphocytes/metabolism , Viral Proteins/immunology
12.
J Virol ; 89(21): 10901-11, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26292323

ABSTRACT

UNLABELLED: In latently infected marmoset T cells, Herpesvirus saimiri (HVS) expresses six microRNAs (known as miR-HSURs [H. saimiri U-rich RNAs]). The viral miR-HSURs are processed from chimeric primary transcripts, each containing a noncoding U-rich RNA (HSUR) and a pre-miRNA hairpin. To uncover the functions of miR-HSURs, we identified mRNA targets in infected cells using high-throughput sequencing of RNA isolated by cross-linking immunoprecipitation (HITS-CLIP). HITS-CLIP revealed hundreds of robust Argonaute (Ago) binding sites mediated by miR-HSURs that map to the host genome but few in the HVS genome. Gene ontology analysis showed that several pathways regulating the cell cycle are enriched among cellular targets of miR-HSURs. Interestingly, miR-HSUR4-3p represses expression of the p300 transcriptional coactivator by binding the open reading frame of its mRNA. miR-HSUR5-3p directly regulates BiP, an endoplasmic reticulum (ER)-localized chaperone facilitating maturation of major histocompatibility complex class I (MHC-I) and the antiviral response. miR-HSUR5-3p also robustly downregulates WEE1, a key negative regulator of cell cycle progression, leading to reduced phosphorylation of its substrate, cyclin-dependent kinase (Cdk1). Consistently, inhibition of miR-HSUR5-3p in HVS-infected cells decreases their proliferation. Together, our results shed light on the roles of viral miRNAs in cellular transformation and viral latency. IMPORTANCE: Viruses express miRNAs during various stages of infection, suggesting that viral miRNAs play critical roles in the viral life cycle. Compared to protein-coding genes, the functions of viral miRNAs are not well understood. This is because it has been challenging to identify their mRNA targets. Here, we focused on the functions of the recently discovered HVS miRNAs, called miR-HSURs. HVS is an oncogenic gammaherpesvirus that causes acute T-cell lymphomas and leukemias in New World primates and transforms human T cells. A better understanding of HVS biology will help advance our knowledge of virus-induced oncogenesis. Because numerous cellular miRNAs play crucial roles in cancer, viral miRNAs from the highly oncogenic HVS might also be important for transformation. Here, we found that the miR-HSURs preferentially modulate expression of host cell cycle regulators, as well as antiviral response factors. Our work provides further insight into the functions of herpesviral miRNAs in virus-induced oncogenesis and latency.


Subject(s)
Cell Cycle Proteins/metabolism , Herpesvirus 2, Saimiriine/genetics , MicroRNAs/metabolism , RNA, Messenger/metabolism , T-Lymphocytes/metabolism , Animals , Binding Sites/genetics , Blotting, Western , CDC2 Protein Kinase/metabolism , Callithrix , Endoplasmic Reticulum Chaperone BiP , HEK293 Cells , Heat-Shock Proteins/metabolism , High-Throughput Nucleotide Sequencing , Humans , Immunoprecipitation , Luciferases , MicroRNAs/genetics , Phosphorylation , RNA, Messenger/genetics
13.
Genes Dev ; 29(14): 1552-64, 2015 Jul 15.
Article in English | MEDLINE | ID: mdl-26220997

ABSTRACT

Herpesvirus saimiri (HVS) is an oncogenic γ-herpesvirus that produces microRNAs (miRNAs) by cotranscription of precursor miRNA (pre-miRNA) hairpins immediately downstream from viral small nuclear RNAs (snRNA). The host cell Integrator complex, which recognizes the snRNA 3' end processing signal (3' box), generates the 5' ends of HVS pre-miRNA hairpins. Here, we identify a novel 3' box-like sequence (miRNA 3' box) downstream from HVS pre-miRNAs that is essential for miRNA biogenesis. In vivo knockdown and rescue experiments confirmed that the 3' end processing of HVS pre-miRNAs also depends on Integrator activity. Interaction between Integrator and HVS primary miRNA (pri-miRNA) substrates that contain only the miRNA 3' box was confirmed by coimmunoprecipitation and an in situ proximity ligation assay (PLA) that we developed to localize specific transient RNA-protein interactions inside cells. Surprisingly, in contrast to snRNA 3' end processing, HVS pre-miRNA 3' end processing by Integrator can be uncoupled from transcription, enabling new approaches to study Integrator enzymology.


Subject(s)
Herpesvirus 2, Saimiriine/genetics , MicroRNAs/metabolism , RNA 3' End Processing/physiology , Gene Knockdown Techniques , HEK293 Cells , HeLa Cells , Herpesvirus 2, Saimiriine/metabolism , Host-Pathogen Interactions/genetics , Humans , Immunoprecipitation , MicroRNAs/chemistry , MicroRNAs/genetics , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , RNA 3' End Processing/genetics , RNA Precursors/genetics , RNA Precursors/metabolism , RNA, Small Nuclear/metabolism , Transcription, Genetic
14.
Mol Cell ; 54(1): 67-79, 2014 Apr 10.
Article in English | MEDLINE | ID: mdl-24725595

ABSTRACT

In marmoset T cells transformed by Herpesvirus saimiri (HVS), a viral U-rich noncoding (nc) RNA, HSUR 1, specifically mediates degradation of host microRNA-27 (miR-27). High-throughput sequencing of RNA after crosslinking immunoprecipitation (HITS-CLIP) identified mRNAs targeted by miR-27 as enriched in the T cell receptor (TCR) signaling pathway, including GRB2. Accordingly, transfection of miR-27 into human T cells attenuates TCR-induced activation of mitogen-activated protein kinases (MAPKs) and induction of CD69. MiR-27 also robustly regulates SEMA7A and IFN-γ, key modulators and effectors of T cell function. Knockdown or ectopic expression of HSUR 1 alters levels of these proteins in virally transformed cells. Two other T-lymphotropic γ-herpesviruses, AlHV-1 and OvHV-2, do not produce a noncoding RNA to downregulate miR-27 but instead encode homologs of miR-27 target genes. Thus, oncogenic γ-herpesviruses have evolved diverse strategies to converge on common targets in host T cells.


Subject(s)
Herpesvirus 2, Saimiriine/metabolism , Lymphocyte Activation , MicroRNAs/metabolism , RNA, Untranslated/metabolism , RNA, Viral/metabolism , T-Lymphocytes/metabolism , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Antigens, Differentiation, T-Lymphocyte/metabolism , Base Sequence , Callithrix , Enzyme Activation , GPI-Linked Proteins/genetics , GPI-Linked Proteins/metabolism , GRB2 Adaptor Protein/genetics , GRB2 Adaptor Protein/metabolism , Gene Expression Regulation , HEK293 Cells , Herpesvirus 2, Saimiriine/genetics , Herpesvirus 2, Saimiriine/pathogenicity , High-Throughput Nucleotide Sequencing , Host-Pathogen Interactions , Humans , Immunoprecipitation , Interferon-gamma/genetics , Interferon-gamma/metabolism , Jurkat Cells , Lectins, C-Type/metabolism , MicroRNAs/genetics , Mitogen-Activated Protein Kinases/metabolism , Molecular Sequence Data , RNA Stability , RNA, Untranslated/genetics , RNA, Viral/genetics , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , Semaphorins/genetics , Semaphorins/metabolism , Sequence Analysis, RNA , Signal Transduction , T-Lymphocytes/immunology , T-Lymphocytes/virology , Time Factors , Transfection
15.
BMC Med Imaging ; 13: 35, 2013 Nov 12.
Article in English | MEDLINE | ID: mdl-24219244

ABSTRACT

BACKGROUND: Non-invasive imaging of the biodistribution of novel therapeutics including gene therapy vectors in animal models is essential. METHODS: This study assessed the utility of high-frequency ultrasound (HF-US) combined with biofluoresence imaging (BFI) to determine the longitudinal impact of a Herpesvirus saimiri amplicon on human colorectal cancer xenograft growth. RESULTS: HF-US imaging of xenografts resulted in an accurate and informative xenograft volume in a longitudinal study. The volumes correlated better with final ex vivo volume than mechanical callipers (R2 = 0.7993, p = 0.0002 vs. R2 = 0.7867, p = 0.0014). HF-US showed that the amplicon caused lobe formation. BFI demonstrated retention and expression of the amplicon in the xenografts and quantitation of the fluorescence levels also correlated with tumour volumes. CONCLUSIONS: The use of multi-modal imaging provided useful and enhanced insights into the behaviour of gene therapy vectors in vivo in real-time. These relatively inexpensive technologies are easy to incorporate into pre-clinical studies.


Subject(s)
Colorectal Neoplasms/pathology , Colorectal Neoplasms/therapy , Genetic Therapy , Genetic Vectors , Herpesvirus 2, Saimiriine/genetics , Optical Imaging/methods , Ultrasonography/methods , Animals , Green Fluorescent Proteins , HCT116 Cells , Humans , Longitudinal Studies , Mice , Mice, Nude , Multimodal Imaging , Tumor Burden , Xenograft Model Antitumor Assays
16.
J Virol ; 87(12): 7127-39, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23596304

ABSTRACT

Herpesvirus saimiri (HVS) infects a range of human cell types with high efficiency. Upon infection, the viral genome can persist as high-copy-number, circular, nonintegrated episomes that segregate to progeny cells upon division. This allows HVS-based vectors to stably transduce a dividing cell population and provide sustained transgene expression in vitro and in vivo. Moreover, the HVS episome is able to persist and provide prolonged transgene expression during in vitro differentiation of mouse and human hemopoietic progenitor cells. Together, these properties are advantageous for induced pluripotent stem cell (iPSC) technology, whereby stem cell-like cells are generated from adult somatic cells by exogenous expression of specific reprogramming factors. Here we assess the potential of HVS-based vectors for the generation of induced pluripotent cancer stem-like cells (iPCs). We demonstrate that HVS-based exogenous delivery of Oct4, Nanog, and Lin28 can reprogram the Ewing's sarcoma family tumor cell line A673 to produce stem cell-like colonies that can grow under feeder-free stem cell culture conditions. Further analysis of the HVS-derived putative iPCs showed some degree of reprogramming into a stem cell-like state. Specifically, the putative iPCs had a number of embryonic stem cell characteristics, staining positive for alkaline phosphatase and SSEA4, in addition to expressing elevated levels of pluripotent marker genes involved in proliferation and self-renewal. However, differentiation trials suggest that although the HVS-derived putative iPCs are capable of differentiation toward the ectodermal lineage, they do not exhibit pluripotency. Therefore, they are hereby termed induced multipotent cancer cells.


Subject(s)
Cellular Reprogramming/genetics , Genetic Vectors , Herpesvirus 2, Saimiriine/physiology , Induced Pluripotent Stem Cells , Kidney/virology , Sarcoma, Ewing/virology , Transgenes , Animals , Cell Culture Techniques , Cell Differentiation , Cell Line, Tumor , HEK293 Cells , Herpesvirus 2, Saimiriine/genetics , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/physiology , Kidney/cytology , Mice , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Transgenes/genetics , Transgenes/physiology
17.
J Virol ; 87(1): 282-95, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23077301

ABSTRACT

Herpesvirus saimiri is known to encode a homolog of human complement regulators named complement control protein homolog (CCPH). We have previously reported that this virally encoded inhibitor effectively inactivates complement by supporting factor I-mediated inactivation of complement proteins C3b and C4b (termed cofactor activity), as well as by accelerating the irreversible decay of the classical/lectin and alternative pathway C3 convertases (termed decay-accelerating activity). To fine map its functional sites, in the present study, we have generated a homology model of CCPH and performed substitution mutagenesis of its conserved residues. Functional analyses of 24 substitution mutants of CCPH indicated that (i) amino acids R118 and F144 play a critical role in imparting C3b and C4b cofactor activities, (ii) amino acids R35, K142, and K191 are required for efficient decay of the C3 convertases, (iii) positively charged amino acids of the linker regions, which are dubbed to be critical for functioning in other complement regulators, are not crucial for its function, and (iv) S100K and G110D mutations substantially enhance its decay-accelerating activities without affecting the cofactor activities. Overall, our data point out that ionic interactions form a major component of the binding interface between CCPH and its interacting partners.


Subject(s)
Complement Factor I/genetics , Complement Factor I/metabolism , Herpesvirus 2, Saimiriine/immunology , Viral Proteins/genetics , Viral Proteins/metabolism , Amino Acid Substitution , DNA Mutational Analysis , Herpesvirus 2, Saimiriine/genetics , Mutant Proteins/genetics , Mutant Proteins/metabolism , Protein Binding
18.
RNA ; 18(12): 2166-73, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23097423

ABSTRACT

RNase III enzymes are fundamental to the biogenesis of microRNAs (miRNAs) and small interfering RNAs (siRNAs) in all species studied. Although alternative miRNA pathways independent of Drosha or Dicer exist, each still requires one RNase III-type enzyme. Here, we describe two strategies that marry either RNase Z or the Integrator complex with the slicing activity of Argonaute2 to generate highly functional mature miRNAs. We provide stringent validation of their RNase III independence by demonstrating efficient miRNA biogenesis and activity in Drosha and Dicer knockout cells. These data provide proof-of-principle evidence for additional mechanistic possibilities for efficient generation of small regulatory RNAs, and represent novel silencing triggers that may be exploited for technical purposes.


Subject(s)
MicroRNAs/biosynthesis , Ribonuclease III/metabolism , Animals , Argonaute Proteins/deficiency , Argonaute Proteins/genetics , Argonaute Proteins/metabolism , Base Sequence , Cell Line , DEAD-box RNA Helicases/deficiency , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/metabolism , Endoribonucleases/deficiency , Endoribonucleases/genetics , Endoribonucleases/metabolism , Gene Knockout Techniques , HEK293 Cells , HeLa Cells , Herpesvirus 2, Saimiriine/genetics , Herpesvirus 2, Saimiriine/metabolism , Humans , Mice , MicroRNAs/genetics , RNA, Small Nuclear/genetics , RNA, Small Nuclear/metabolism , RNA, Viral/genetics , RNA, Viral/metabolism , Ribonuclease III/deficiency , Ribonuclease III/genetics
19.
Int J Oncol ; 40(6): 2081-9, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22396105

ABSTRACT

Herpesvirus saimiri (HVS) is capable of infecting a range of human carcinoma cell types with high efficiency and the viral genome persists as high copy number, circular, non-integrated episomes which segregate to progeny upon cell division. This allows HVS-based vectors to stably transduce a dividing cell population and provide sustained transgene expression for an extended period of time both in vitro and in vivo. Moreover, the insertion of a bacterial artificial chromosome cassette into the HVS genome simplifies the incorporation of large amounts of heterologous DNA for gene delivery. Herein we have produced a recombinant HVS-based vector containing full-length human TRAIL under the control of the α-survivin promoter, and subsequently challenged a variety of cancer cell lines with this vector. The TRAIL transgene was expressed in infected colorectal SW480 cells, causing considerable apoptosis induction. Apoptosis was also observed when several other cancer cell lines derived from different tissues were infected. Moreover, co-treatment with Jak inhibitor AG490 led to the disruption of spheroid cultures grown from the melanoma Mel888 line. These data suggest that an HVS gene therapy vector expressing TRAIL could be an effective treatment against cancer.


Subject(s)
Apoptosis , Herpesvirus 2, Saimiriine/genetics , Neoplasms/therapy , TNF-Related Apoptosis-Inducing Ligand/biosynthesis , Antineoplastic Agents/pharmacology , Caspases/metabolism , Cell Line, Tumor , Cloning, Molecular , Genetic Therapy , Genetic Vectors , HEK293 Cells , Humans , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Sequence Deletion , Spheroids, Cellular , TNF-Related Apoptosis-Inducing Ligand/genetics , Transcription, Genetic , Tyrphostins/pharmacology
20.
J Virol ; 86(7): 3541-53, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22278248

ABSTRACT

In recent studies, the nuclear domain 10 (ND10) components PML, Sp100, human Daxx (hDaxx), and ATRX were identified to be cellular restriction factors that are able to inhibit the replication of several herpesviruses. The antiviral function of ND10, however, is antagonized by viral effector proteins by a variety of strategies, including degradation of PML or relocalization of ND10 proteins. In this study, we analyzed the interplay between infection with herpesvirus saimiri (HVS), the prototypic rhadinovirus, and cellular defense by ND10. In contrast to other herpesviruses, we found that HVS specifically degraded the cellular ND10 component Sp100, whereas other factors like PML or hDaxx remained intact. We could further identify the ORF3 tegument protein of HVS, which shares homology with the cellular formylglycinamide ribotide amidotransferase (FGARAT) enzyme, to be the viral factor that induces the proteasomal degradation of Sp100. Interestingly, recent studies showed that the ORF3-homologous proteins ORF75c of murine gammaherpesvirus 68 and BNRF-1 of Epstein-Barr virus modulate the ND10 proteins PML and ATRX, respectively, suggesting that the ND10 targets of viral FGARAT-homologous proteins diversified during evolution. Furthermore, a virus with the ORF3 deletion was efficiently complemented in Sp100-depleted cells, indicating that Sp100 is able to inhibit HVS in the absence of antagonistic mechanisms. In contrast, we observed that PML, which was neither degraded nor redistributed after HVS infection, strongly restricted both wild-type HVS and virus with the ORF3 deletion. Thus, HVS may lack a factor that efficiently counteracts the repressive function of PML, which may foster latency as the outcome of infection.


Subject(s)
Antigens, Nuclear/metabolism , Autoantigens/metabolism , Herpesviridae Infections/metabolism , Herpesvirus 2, Saimiriine/metabolism , Nuclear Proteins/immunology , Open Reading Frames , Viral Proteins/metabolism , Antigens, Nuclear/genetics , Autoantigens/genetics , Cell Line , Gene Expression Regulation, Viral , Herpesviridae Infections/genetics , Herpesviridae Infections/immunology , Herpesviridae Infections/virology , Herpesvirus 2, Saimiriine/genetics , Humans , Immunity , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/genetics , Protein Binding , Viral Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL