Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 108
Filter
Add more filters

Country/Region as subject
Publication year range
1.
J Biol Chem ; 300(6): 107318, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38677513

ABSTRACT

Sidney Altman's discovery of the processing of one RNA by another RNA that acts like an enzyme was revolutionary in biology and the basis for his sharing the 1989 Nobel Prize in Chemistry with Thomas Cech. These breakthrough findings support the key role of RNA in molecular evolution, where replicating RNAs (and similar chemical derivatives) either with or without peptides functioned in protocells during the early stages of life on Earth, an era referred to as the RNA world. Here, we cover the historical background highlighting the work of Altman and his colleagues and the subsequent efforts of other researchers to understand the biological function of RNase P and its catalytic RNA subunit and to employ it as a tool to downregulate gene expression. We primarily discuss bacterial RNase P-related studies but acknowledge that many groups have significantly contributed to our understanding of archaeal and eukaryotic RNase P, as reviewed in this special issue and elsewhere.


Subject(s)
RNA, Catalytic , Ribonuclease P , Ribonuclease P/metabolism , Ribonuclease P/chemistry , Ribonuclease P/genetics , History, 20th Century , RNA, Catalytic/metabolism , RNA, Catalytic/chemistry , RNA, Catalytic/genetics , History, 21st Century , Humans
2.
RNA ; 29(3): 300-307, 2023 03.
Article in English | MEDLINE | ID: mdl-36549864

ABSTRACT

The seminal discovery of ribonuclease P (RNase P) and its catalytic RNA by Sidney Altman has not only revolutionized our understanding of life, but also opened new fields for scientific exploration and investigation. This review focuses on human RNase P and its use as a gene-targeting tool, two topics initiated in Altman's laboratory. We outline early works on human RNase P as a tRNA processing enzyme and comment on its expanding nonconventional functions in molecular networks of transcription, chromatin remodeling, homology-directed repair, and innate immunity. The important implications and insights from these discoveries on the potential use of RNase P as a gene-targeting tool are presented. This multifunctionality calls to a modified structure-function partitioning of domains in human RNase P, as well as its relative ribonucleoprotein, RNase MRP. The role of these two catalysts in innate immunity is of particular interest in molecular evolution, as this dynamic molecular network could have originated and evolved from primordial enzymes and sensors of RNA, including predecessors of these two ribonucleoproteins.


Subject(s)
RNA, Catalytic , Ribonuclease P , Humans , Ribonuclease P/genetics , Ribonuclease P/metabolism , RNA/genetics , RNA Processing, Post-Transcriptional , RNA, Catalytic/metabolism
3.
Molecules ; 29(9)2024 Apr 29.
Article in English | MEDLINE | ID: mdl-38731543

ABSTRACT

Ribonuclease P (RNase P) complexed with an external guide sequence (EGS) represents a promising nucleic acid-based gene targeting approach for gene expression knock-down and modulation. The RNase P-EGS strategy is unique as an EGS can be designed to basepair any mRNA sequence and recruit intracellular RNase P for hydrolysis of the target mRNA. In this study, we provide the first direct evidence that the RNase P-based approach effectively blocks the gene expression and replication of herpes simplex virus 2 (HSV-2), the causative agent of genital herpes. We constructed EGSs to target the mRNA encoding HSV-2 single-stranded DNA binding protein ICP8, which is essential for viral DNA genome replication and growth. In HSV-2 infected cells expressing a functional EGS, ICP8 levels were reduced by 85%, and viral growth decreased by 3000 folds. On the contrary, ICP8 expression and viral growth exhibited no substantial differences between cells expressing no EGS and those expressing a disabled EGS with mutations precluding RNase P recognition. The anti-ICP8 EGS is specific in targeting ICP8 because it only affects ICP8 expression but does not affect the expression of the other viral immediate-early and early genes examined. This study shows the effective and specific anti-HSV-2 activity of the RNase P-EGS approach and demonstrates the potential of EGS RNAs for anti-HSV-2 applications.


Subject(s)
Gene Expression Regulation, Viral , Herpesvirus 2, Human , Virus Replication , Herpesvirus 2, Human/genetics , Herpesvirus 2, Human/physiology , Humans , Ribonuclease P/metabolism , Ribonuclease P/genetics , Animals , Viral Proteins/genetics , Viral Proteins/metabolism , Chlorocebus aethiops , RNA, Messenger/genetics , RNA, Messenger/metabolism , Vero Cells , Immediate-Early Proteins/genetics , Immediate-Early Proteins/metabolism , DNA-Binding Proteins
4.
Molecules ; 28(8)2023 Apr 21.
Article in English | MEDLINE | ID: mdl-37110852

ABSTRACT

Kaposi's sarcoma, an AIDS-defining illness, is caused by Kaposi's sarcoma-associated herpesvirus (KSHV), an oncogenic virus. In this study, we engineered ribozymes derived from ribonuclease P (RNase P) catalytic RNA with targeting against the mRNA encoding KSHV immediate early replication and transcription activator (RTA), which is vital for KSHV gene expression. The functional ribozyme F-RTA efficiently sliced the RTA mRNA sequence in vitro. In cells, KSHV production was suppressed with ribozyme F-RTA expression by 250-fold, and RTA expression was suppressed by 92-94%. In contrast, expression of control ribozymes hardly affected RTA expression or viral production. Further studies revealed both overall KSHV early and late gene expression and viral growth decreased because of F-RTA-facilitated suppression of RTA expression. Our results indicate the first instance of RNase P ribozymes having potential for use in anti-KSHV therapy.


Subject(s)
Herpesvirus 8, Human , Immediate-Early Proteins , RNA, Catalytic , Herpesvirus 8, Human/genetics , Herpesvirus 8, Human/metabolism , RNA, Catalytic/genetics , RNA, Catalytic/metabolism , Ribonuclease P/genetics , Ribonuclease P/metabolism , Immediate-Early Proteins/metabolism , Virus Replication/genetics , Virus Latency , Trans-Activators/genetics , RNA, Messenger/genetics , Gene Expression , Gene Expression Regulation, Viral
5.
RNA ; 25(5): 645-655, 2019 05.
Article in English | MEDLINE | ID: mdl-30803999

ABSTRACT

External guide sequences (EGSs) signify the short RNAs that induce ribonuclease P (RNase P), an enzyme responsible for processing the 5' termini of tRNA, to specifically cleave a target mRNA by forming a precursor tRNA-like complex. Hence, the EGS technology may serve as a potential strategy for gene-targeting therapy. Our previous studies have revealed that engineered EGS variants induced RNase P to efficiently hydrolyze target mRNAs. In the present research, an EGS variant was designed to be complementary to the mRNA coding for human cytomegalovirus (HCMV) major capsid protein (MCP), which is vital to form the viral capsid. In vitro, the EGS variant was about 80-fold more efficient in inducing human RNase P-mediated cleavage of the target mRNA than a natural tRNA-derived EGS. Moreover, the expressed variant and natural tRNA-originated EGSs led to a decrease of MCP expression by 98% and 73%-74% and a decrease of viral growth by about 10,000- and 200-fold in cells infected with HCMV, respectively. These results reveal direct evidence that the engineered EGS variant has higher efficiency in blocking the expression of HCMV genes and viral growth than the natural tRNA-originated EGS. Therefore, our findings imply that the EGS variant can be a potent candidate agent for the treatment of infections caused by HCMV.


Subject(s)
Capsid Proteins/genetics , Cytomegalovirus/genetics , RNA, Guide, Kinetoplastida/genetics , RNA, Messenger/genetics , RNA, Transfer, Ser/genetics , RNA, Viral/genetics , Ribonuclease P/metabolism , Base Pairing , Capsid Proteins/biosynthesis , Cell Line, Transformed , Cell Line, Tumor , Cytomegalovirus/metabolism , Fibroblasts/metabolism , Fibroblasts/virology , Gene Expression Regulation, Viral , Gene Targeting/methods , Genetic Engineering/methods , Host-Pathogen Interactions/genetics , Humans , Molecular Targeted Therapy , Neuroglia/metabolism , Neuroglia/virology , Nucleic Acid Conformation , Primary Cell Culture , RNA Cleavage , RNA, Guide, Kinetoplastida/chemistry , RNA, Guide, Kinetoplastida/metabolism , RNA, Messenger/chemistry , RNA, Messenger/metabolism , RNA, Transfer, Ser/chemistry , RNA, Transfer, Ser/metabolism , RNA, Viral/chemistry , RNA, Viral/metabolism , Ribonuclease P/chemistry , Ribonuclease P/genetics , Virus Replication/physiology
6.
PLoS Pathog ; 15(2): e1007615, 2019 02.
Article in English | MEDLINE | ID: mdl-30779794

ABSTRACT

Cytomegalovirus (CMV) infection causes birth defects and life-threatening complications in immunosuppressed patients. Lack of vaccine and need for more effective drugs have driven widespread ongoing therapeutic development efforts against human CMV (HCMV), mostly using murine CMV (MCMV) as the model system for preclinical animal tests. The recent publication (Yu et al., 2017, DOI: 10.1126/science.aam6892) of an atomic model for HCMV capsid with associated tegument protein pp150 has infused impetus for rational design of novel vaccines and drugs, but the absence of high-resolution structural data on MCMV remains a significant knowledge gap in such development efforts. Here, by cryoEM with sub-particle reconstruction method, we have obtained the first atomic structure of MCMV capsid with associated pp150. Surprisingly, the capsid-binding patterns of pp150 differ between HCMV and MCMV despite their highly similar capsid structures. In MCMV, pp150 is absent on triplex Tc and exists as a "Λ"-shaped dimer on other triplexes, leading to only 260 groups of two pp150 subunits per capsid in contrast to 320 groups of three pp150 subunits each in a "Δ"-shaped fortifying configuration. Many more amino acids contribute to pp150-pp150 interactions in MCMV than in HCMV, making MCMV pp150 dimer inflexible thus incompatible to instigate triplex Tc-binding as observed in HCMV. While pp150 is essential in HCMV, our pp150-deletion mutant of MCMV remained viable though with attenuated infectivity and exhibiting defects in retaining viral genome. These results thus invalidate targeting pp150, but lend support to targeting capsid proteins, when using MCMV as a model for HCMV pathogenesis and therapeutic studies.


Subject(s)
Capsid Proteins/ultrastructure , Phosphoproteins/metabolism , Phosphoproteins/physiology , Viral Matrix Proteins/metabolism , Viral Matrix Proteins/physiology , Animals , Capsid , Capsid Proteins/metabolism , Cryoelectron Microscopy/methods , Cytomegalovirus/genetics , Cytomegalovirus/metabolism , Cytomegalovirus/pathogenicity , Cytomegalovirus Infections/metabolism , Genome, Viral/genetics , Humans , Mice , Muromegalovirus/metabolism , Muromegalovirus/pathogenicity , Phosphoproteins/ultrastructure , Sequence Deletion/genetics , Viral Matrix Proteins/ultrastructure , Virion , Virus Assembly
7.
PLoS Pathog ; 14(1): e1006867, 2018 01.
Article in English | MEDLINE | ID: mdl-29377960

ABSTRACT

Interferon-γ (IFN-γ) represents one of the most important innate immunity responses in a host to combat infections of many human viruses including human herpesviruses. Human N-myc interactor (Nmi) protein, which has been shown to interact with signal transducer and activator of transcription (STAT) proteins including STAT1, is important for the activation of IFN-γ induced STAT1-dependent transcription of many genes responsible for IFN-γ immune responses. However, no proteins encoded by herpesviruses have been reported to interact with Nmi and inhibit Nmi-mediated activation of IFN-γ immune responses to achieve immune evasion from IFN-γ responses. In this study, we show strong evidence that the UL23 protein of human cytomegalovirus (HCMV), a human herpesvirus, specifically interacts with Nmi. This interaction was identified through a yeast two-hybrid screen and co-immunoprecipitation in human cells. We observed that Nmi, when bound to UL23, was not associated with STAT1, suggesting that UL23 binding of Nmi disrupts the interaction of Nmi with STAT1. In cells overexpressing UL23, we observed (a) significantly reduced levels of Nmi and STAT1 in the nuclei, the sites where these proteins act to induce transcription of IFN-γ stimulated genes, and (b) decreased levels of the induction of the transcription of IFN-γ stimulated genes. UL23-deficient HCMV mutants induced higher transcription of IFN-γ stimulated genes and exhibited lower titers than parental and control revertant viruses expressing functional UL23 in IFN-γ treated cells. Thus, UL23 appears to interact directly with Nmi and inhibit nuclear translocation of Nmi and its associated protein STAT1, leading to a decrease of IFN-γ induced responses and an increase of viral resistance to IFN-γ. Our results further highlight the roles of UL23-Nmi interactions in facilitating viral immune escape from IFN-γ responses and enhancing viral resistance to IFN antiviral effects.


Subject(s)
Cytomegalovirus/physiology , Immune Evasion , Immunity, Innate/drug effects , Interferon-gamma/pharmacology , Intracellular Signaling Peptides and Proteins/metabolism , Viral Matrix Proteins/physiology , Cells, Cultured , Cytomegalovirus/immunology , Gene Expression Regulation/immunology , HEK293 Cells , Humans , Immune Evasion/drug effects , Immune Evasion/genetics , Immunity, Innate/genetics , Protein Binding , Signal Transduction/genetics , Signal Transduction/immunology
8.
FASEB J ; 33(3): 4287-4299, 2019 03.
Article in English | MEDLINE | ID: mdl-30571312

ABSTRACT

Antiviral drug development against respiratory syncytial virus (RSV) is urgently needed due to the public health significance of the viral infection. Here, we report the anti-RSV activity of a small molecule, (1S,3R,4R,5R)-3,4- bis{[(E)-3-(3,4-dihydroxyphenyl)prop-2-enoyl]oxy}-1,5-dihydroxycyclohexane-1-carboxylic methyl ester (3,4-DCQAME) or 3,4- O-Dicaffeoylquinic acid methyl ester, which can be isolated from several plants of traditional Chinese medicine. We showed for the first time that compound 3,4-DCQAME potently inhibits RSV entry and infection. In vitro, 3,4-DCQAME can interact with F(ecto), the ectodomain of RSV fusion (F) protein. In cultured cells, the compound can block the interaction of F(ecto) protein with the cellular membrane and inhibit viral fusion during RSV entry, leading to inhibition of viral gene expression and infection. In RSV-infected mice that were treated with 3,4-DCQAME, we observed a reduction of RSV-induced pathologic changes and substantial inhibition of viral infection and growth in the lung tissues. Our results provide the first direct evidence of the anti-RSV activity of 3,4-DCQAME. Furthermore, these results suggest that 3,4-DCQAME represents a promising lead compound for anti-RSV therapeutic development.-Tang, W., Li, M., Liu, Y., Liang, N., Yang, Z., Zhao, Y., Wu, S., Lu, S., Li, Y., Liu, F. Small molecule inhibits respiratory syncytial virus entry and infection by blocking the interaction of the viral fusion protein with the cell membrane.


Subject(s)
Antiviral Agents/pharmacology , Cell Membrane/drug effects , Respiratory Syncytial Virus Infections/drug therapy , Respiratory Syncytial Virus, Human/drug effects , Small Molecule Libraries/pharmacology , Viral Fusion Proteins/metabolism , Animals , Cell Line , Cell Membrane/metabolism , Cell Membrane/virology , Gene Expression/drug effects , Lung/metabolism , Lung/virology , Male , Medicine, Chinese Traditional/methods , Mice , Mice, Inbred BALB C , Respiratory Syncytial Virus Infections/metabolism , Respiratory Syncytial Virus Infections/virology
10.
Blood ; 129(12): 1680-1684, 2017 03 23.
Article in English | MEDLINE | ID: mdl-27979823

ABSTRACT

It is widely suspected, yet controversial, that infection plays an etiologic role in the development of acute lymphoblastic leukemia (ALL), the most common childhood cancer and a disease with a confirmed prenatal origin in most cases. We investigated infections at diagnosis and then assessed the timing of infection at birth in children with ALL and age, gender, and ethnicity matched controls to identify potential causal initiating infections. Comprehensive untargeted virome and bacterial analyses of pretreatment bone marrow specimens (n = 127 ALL in comparison with 38 acute myeloid leukemia cases in a comparison group) revealed prevalent cytomegalovirus (CMV) infection at diagnosis in childhood ALL, demonstrating active viral transcription in leukemia blasts as well as intact virions in serum. Screening of newborn blood samples revealed a significantly higher prevalence of in utero CMV infection in ALL cases (n = 268) than healthy controls (n = 270) (odds ratio [OR], 3.71, confidence interval [CI], 1.56-7.92, P = .0016). Risk was more pronounced in Hispanics (OR=5.90, CI=1.89-25.96) than in non-Hispanic whites (OR=2.10 CI= 0.69-7.13). This is the first study to suggest that congenital CMV infection is a risk factor for childhood ALL and is more prominent in Hispanic children. Further investigation of CMV as an etiologic agent for ALL is warranted.


Subject(s)
Cytomegalovirus Infections/complications , Neonatal Screening/methods , Precursor Cell Lymphoblastic Leukemia-Lymphoma/virology , Bone Marrow Examination , Case-Control Studies , Cytomegalovirus Infections/congenital , Cytomegalovirus Infections/ethnology , Hispanic or Latino , Humans , Infant, Newborn , Precursor Cell Lymphoblastic Leukemia-Lymphoma/etiology , Prevalence , White People
11.
Chemistry ; 25(6): 1446-1450, 2019 Jan 28.
Article in English | MEDLINE | ID: mdl-30479034

ABSTRACT

This work demonstrates single-molecule imaging of metal-ion induced double-stranded DNA formation in DNA nanostructures. The formation of the metal ion-mediated base pairing in a DNA origami frame was examined using C-Ag-C and T-Hg-T metallo-base pairs. The target DNA strands containing consecutive C or T were incorporated into the DNA frame, and the binding was controlled by the addition of metal ions. Double-stranded DNA formation was monitored by observing the structural changes in the incorporated DNA strands using high-speed atomic force microscopy (AFM). Using the T-Hg-T base pair, the dynamic formation of unique dsDNA and its dissociation were observed. The formation of an unusual shape of dsDNA with consecutive T-Hg-T base pairs was visualized in the designed nanoscale structure.


Subject(s)
DNA/chemistry , Metals/chemistry , Nanostructures/chemistry , Base Pairing , DNA/metabolism , Ions/chemistry , Microscopy, Atomic Force , Nanotechnology
12.
Anal Biochem ; 567: 85-89, 2019 02 15.
Article in English | MEDLINE | ID: mdl-30157446

ABSTRACT

Terminal deoxynucleotidyl transferase (TdT) is a unique template-free polymerase that randomly adds multiple deoxyribonucleoside triphosphates (dNTPs) to the 3'-OH terminus of ssDNA. This characteristic makes TdT a versatile enzymatic tool in many fields. Moreover, aberrant TdT expression is a well-recognized biomarker of several leukemic diseases and is related to carcinogenesis. In this study, we developed a facile, rapid, label-free, and convenient assay for TdT detection. TdT-generated poly A tails formed a fluorescent enhancement complex in the presence of coralyne. To achieve a better signal-to-noise ratio, we used potassium thiocyanate (KSCN), instead of other halogen anions (KCl, KBr, KI, NaI) as the quenching agent of dissociate coralyne. Our results demonstrate that this assay is extremely facile, rapid, and label-free; at levels as low as 0.025 U/mL, TdT was distinctly detected within 55 min. And the determination of TdT activity in RBL-2H3 and Reh cells lysates exhibited a good sensing performance, demonstrating its potential applications in biochemical research and clinical diagnosis.


Subject(s)
Adenosine/chemistry , Berberine Alkaloids/chemistry , Biosensing Techniques/methods , DNA Nucleotidylexotransferase/analysis , Polymers/chemistry , DNA Nucleotidylexotransferase/metabolism , DNA, Single-Stranded/chemistry , Fluorescent Dyes/chemistry
13.
PLoS Pathog ; 12(11): e1006007, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27824944

ABSTRACT

The mechanisms underlying human cytomegalovirus (HCMV) latency remain incompletely understood. Here, we showed that a HCMV-encoded miRNA, miR-UL148D, robustly accumulates during late stages of experimental latent HCMV infection in host cells and promotes HCMV latency by modulating the immediate early response gene 5 (IER5)-cell division cycle 25B (CDC25B) axis in host cells. miR-UL148D inhibited IER5 expression by directly targeting the three-prime untranslated region(3'UTR) of IER5 mRNA and thus rescued CDC25B expression during the establishment of viral latency. Infection with NR-1ΔmiR-UL148D, a derivative of the HCMV clinical strain NR-1 with a miR-UL148D knockout mutation, resulted in sustained induction of IER5 expression but decreased CDC25B expression in host cells. Mechanistically, we further showed that CDC25B plays an important role in suppressing HCMV IE1 and lytic gene transcription by activating cyclin-dependent kinase 1 (CDK-1). Both gain-of-function and lose-of-function assays demonstrated that miR-UL148D promotes HCMV latency by helping maintain CDC25B activity in host cells. These results provide a novel mechanism through which a HCMV miRNA regulates viral latency.


Subject(s)
Cytomegalovirus/physiology , Immediate-Early Proteins/metabolism , Nuclear Proteins/metabolism , Virus Latency/physiology , cdc25 Phosphatases/metabolism , Blotting, Western , Cell Line , Enzyme-Linked Immunosorbent Assay , Fluorescent Antibody Technique , Gene Expression Regulation, Viral , Host-Pathogen Interactions/physiology , Humans , MicroRNAs/genetics , RNA, Viral/genetics , Transfection
14.
J Immunol ; 196(2): 792-802, 2016 Jan 15.
Article in English | MEDLINE | ID: mdl-26673132

ABSTRACT

Effector proteins encoded by Salmonella pathogenicity islands play a key role in promoting bacterial intracellular survival, colonization, and pathogenesis. In this study, we investigated the function of the virulence-associated effector SrfA (SsrAB regulated factor) both in macrophages in vitro and in infected mice in vivo. SrfA was secreted into the cytoplasm during S. Typhimurium infection and disassociated IL-1R-associated kinase-1 (IRAK-1) from the IRAK-1-Toll interacting protein (Tollip) complex by interacting with Tollip. The released IRAK-1 was phosphorylated and subsequently activated the NF-κB signaling pathway, which enhanced the LPS-induced expression of inflammatory cytokines, such as IL-8, IL-1ß, and TNF-α. The coupling of ubiquitin to endoplasmic reticulum degradation aa 183-219 domain of Tollip is the binding region for SrfA, and both the MDaa207-226 and CTaa357-377 regions of SrfA mediate binding to Tollip and NF-κB signaling activation. Deletion of SrfA in S. Typhimurium had no notable effects on its replication but impaired the induction of NF-κB activation in infected macrophages. The mice infected with srfA-deficient bacteria exhibited a decreased inflammatory response and an increased survival rate compared with those infected with wild-type S. Typhimurium. We conclude that SrfA is a novel Salmonella virulence effector that helps modulate host inflammatory responses by promoting NF-κB signaling activation.


Subject(s)
NF-kappa B/immunology , Salmonella Infections, Animal/immunology , Signal Transduction/immunology , Virulence Factors/immunology , Animals , Blotting, Western , Enzyme-Linked Immunosorbent Assay , Female , Fluorescent Antibody Technique , HEK293 Cells , Humans , Immunoprecipitation , Inflammation/immunology , Inflammation/microbiology , Macrophages/immunology , Macrophages/microbiology , Mice , Mice, Inbred BALB C , Real-Time Polymerase Chain Reaction , Salmonella typhimurium/immunology , Two-Hybrid System Techniques
15.
J Virol ; 88(6): 3411-22, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24403583

ABSTRACT

Infection with the human gammaherpesviruses, Epstein-Barr virus (EBV) and Kaposi's sarcoma-associated herpesvirus (KSHV), is associated with several cancers. During lytic replication of herpesviruses, viral genes are expressed in an ordered cascade. However, the mechanism by which late gene expression is regulated has not been well characterized in gammaherpesviruses. In this study, we have investigated the cis element that mediates late gene expression during de novo lytic infection with murine gammaherpesvirus 68 (MHV-68). A reporter system was established and used to assess the activity of viral late gene promoters upon infection with MHV-68. It was found that the viral origin of lytic replication, orilyt, must be on the reporter plasmid to support activation of the late gene promoter. Furthermore, the DNA sequence required for the activation of late gene promoters was mapped to a core element containing a distinct TATT box and its neighboring sequences. The critical nucleotides of the TATT box region were determined by systematic mutagenesis in the reporter system, and the significance of these nucleotides was confirmed in the context of the viral genome. In addition, EBV and KSHV late gene core promoters could be activated by MHV-68 lytic replication, indicating that the mechanisms controlling late gene expression are conserved among gammaherpesviruses. Therefore, our results on MHV-68 establish a solid foundation for mechanistic studies of late gene regulation.


Subject(s)
Herpesviridae Infections/veterinary , Promoter Regions, Genetic , Rhadinovirus/genetics , Rodent Diseases/virology , Transcription, Genetic , Viral Proteins/genetics , Animals , Base Sequence , Gene Expression Regulation, Viral , Herpesviridae Infections/virology , Mice , Molecular Sequence Data , Replication Origin , Rhadinovirus/physiology , TATA Box , Viral Proteins/metabolism , Virus Replication
16.
PLoS Pathog ; 9(8): e1003525, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23966856

ABSTRACT

Human cytomegalovirus (HCMV) is a ubiquitous herpesvirus that causes birth defects in newborns and life-threatening complications in immunocompromised individuals. Among all human herpesviruses, HCMV contains a much larger dsDNA genome within a similarly-sized capsid compared to the others, and it was proposed to require pp150, a tegument protein only found in cytomegaloviruses, to stabilize its genome-containing capsid. However, little is known about how pp150 interacts with the underlying capsid. Moreover, the smallest capsid protein (SCP), while dispensable in herpes simplex virus type 1, was shown to play essential, yet undefined, role in HCMV infection. Here, by cryo electron microscopy (cryoEM), we determine three-dimensional structures of HCMV capsid (no pp150) and virion (with pp150) at sub-nanometer resolution. Comparison of these two structures reveals that each pp150 tegument density is composed of two helix bundles connected by a long central helix. Correlation between the resolved helices and sequence-based secondary structure prediction maps the tegument density to the N-terminal half of pp150. The structures also show that SCP mediates interactions between the capsid and pp150 at the upper helix bundle of pp150. Consistent with this structural observation, ribozyme inhibition of SCP expression in HCMV-infected cells impairs the formation of DNA-containing viral particles and reduces viral yield by 10,000 fold. By cryoEM reconstruction of the resulting "SCP-deficient" viral particles, we further demonstrate that SCP is required for pp150 functionally binding to the capsid. Together, our structural and biochemical results point to a mechanism whereby SCP recruits pp150 to stabilize genome-containing capsid for the production of infectious HCMV virion.


Subject(s)
Capsid Proteins/metabolism , Capsid/chemistry , Cytomegalovirus/physiology , DNA, Viral/genetics , Phosphoproteins/metabolism , Viral Matrix Proteins/metabolism , Virion/metabolism , Virus Assembly , Amino Acid Sequence , Blotting, Northern , Blotting, Western , Capsid/metabolism , Capsid/ultrastructure , Capsid Proteins/chemistry , Cells, Cultured , Cryoelectron Microscopy , Cytomegalovirus Infections/genetics , Cytomegalovirus Infections/metabolism , Cytomegalovirus Infections/virology , Fibroblasts/cytology , Fibroblasts/metabolism , Fibroblasts/virology , Humans , Molecular Sequence Data , Phosphoproteins/chemistry , Protein Conformation , RNA, Catalytic/metabolism , Viral Matrix Proteins/chemistry , Virion/chemistry
17.
Proc Natl Acad Sci U S A ; 109(32): 13070-5, 2012 Aug 07.
Article in English | MEDLINE | ID: mdl-22826233

ABSTRACT

Ribonuclease P complexed with external guide sequence (EGS) bound to mRNA represents a unique nucleic acid-based gene interference approach for modulation of gene expression. Compared with other strategies, such as RNA interference, the EGS-based technology is unique because a custom-designed EGS molecule can hybridize with any mRNA and recruit intracellular ribonuclease P for specific degradation of the target mRNA. It has not been reported whether the EGS-based technology can modulate gene expression in mice. In this study, a functional EGS was constructed to target the mRNA encoding the protease (mPR) of murine cytomegalovirus (MCMV), which is essential for viral replication. Furthermore, a unique attenuated strain of Salmonella was generated for gene delivery of EGS in cultured cells and in mice. Efficient expression of EGS was observed in cultured cells treated with the generated Salmonella vector carrying constructs with the EGS expression cassette. Moreover, a significant reduction in mPR expression and viral growth was found in MCMV-infected cells treated with Salmonella carrying the construct with the functional EGS sequence. When MCMV-infected mice were orally treated with Salmonella carrying EGS expression cassettes, viral gene expression and growth in various organs of these animals were reduced and animal survival improved. Our study suggests that EGS RNAs, when expressed following Salmonella-mediated gene transfer, effectively inhibit viral gene expression and infection in mice. Furthermore, these results demonstrate the feasibility of developing Salmonella-mediated delivery of EGS as a unique approach for treatment that reduces viral diseases in vivo.


Subject(s)
Cytomegalovirus Infections/prevention & control , Gene Expression Regulation, Viral/genetics , Muromegalovirus/genetics , RNA, Messenger/metabolism , Ribonuclease P/metabolism , Animals , Blotting, Northern , Blotting, Western , Cytomegalovirus Infections/genetics , DNA Primers/genetics , Gene Transfer Techniques , Genetic Therapy/methods , Mice , RNA, Messenger/genetics , Ribonuclease P/genetics , Salmonella , RNA, Small Untranslated
18.
J Infect Dis ; 209(12): 2000-11, 2014 Jun 15.
Article in English | MEDLINE | ID: mdl-24415783

ABSTRACT

BACKGROUND: The mechanism underlying the ability of virulent Salmonella organisms to escape clearance by macrophages is incompletely understood. Here, we report a novel mechanism by which Salmonella escapes macrophages. METHODS: Microarray and quantitative real-time polymerase chain reaction analyses were used to screen key microRNAs regulating Salmonella-host cell interactions. Target gene was tested using luciferase reporter and Western blot assays. The role of microRNA 128 (miR-128) was assayed using intestinal epithelial cells and a mouse infection model. RESULTS: The miR-128 level in human intestinal epithelial HT29 cells was strongly increased by infection with strain SE2472, and the elevation in miR-128 levels in mouse intestine and colon tissues correlated with the level of Salmonella infection in mice. Macrophage colony-stimulating factor (M-CSF) was identified as a target of miR-128, and increased miR-128 levels in epithelial cells due to infection with strain SE2472 significantly decreased the level of cell-secreted M-CSF, leading to impaired M-CSF-mediated macrophage recruitment. The secreted proteins from Salmonella were identified as possible effectors to induce miR-128 expression via the p53 signaling pathway. Moreover, intragastric delivery of anti-miR-128 antagomir into mice significantly increased M-CSF-mediated macrophage recruitment and suppressed Salmonella infection. CONCLUSIONS: Salmonella can upregulate intestinal epithelial miR-128 expression, which, in turn, decreases levels of epithelial cell-secreted M-CSF and M-CSF-induced macrophage recruitment.


Subject(s)
Intestines/immunology , Macrophage Colony-Stimulating Factor/metabolism , Macrophages/immunology , MicroRNAs/metabolism , Salmonella enteritidis/isolation & purification , Animals , Epithelial Cells/immunology , Epithelial Cells/microbiology , Female , HT29 Cells , Humans , Intestines/cytology , Intestines/microbiology , Macrophage Colony-Stimulating Factor/genetics , Mice , Mice, Inbred BALB C , MicroRNAs/genetics , Salmonella Infections/prevention & control , Salmonella enteritidis/growth & development , Signal Transduction , Tumor Suppressor Protein p53/metabolism , Up-Regulation
19.
J Virol ; 87(19): 10628-40, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23885069

ABSTRACT

Controlled regulation of genomic DNA synthesis is a universally conserved process for all herpesviruses, including human cytomegalovirus (HCMV), and plays a key role in viral pathogenesis, such as persistent infections. HCMV UL105 is believed to encode the helicase of the DNA replication machinery that needs to localize in the nuclei, the site of viral DNA synthesis. No host factors that interact with UL105 have been identified. In this study, we show that UL105 specifically interacts with Snapin, a human protein that is predominantly localized in the cytoplasm and associated with cellular vesicles. UL105 was found to interact with Snapin in both the yeast two-hybrid screen and coimmunoprecipitation experiments in HCMV-infected cells. The nuclear and cytoplasmic levels of UL105 were decreased and increased in cells overexpressing Snapin, respectively, while the levels of UL105 in the nuclei and cytoplasm were increased and decreased in cells in which the expression of Snapin was downregulated with anti-Snapin small interfering RNA (siRNA) molecules, respectively. Furthermore, viral DNA synthesis and progeny production were decreased in cells overexpressing Snapin and increased in the anti-Snapin siRNA-treated cells, respectively. Our results provide the first direct evidence to suggest that Snapin interacts with UL105 and alters its cellular distribution, leading to modulation of viral DNA synthesis and progeny production. Our study further suggests that modulation of the cellular distribution of viral helicase by Snapin may represent a possible mechanism for regulating HCMV genomic DNA synthesis, a key step during herpesvirus lytic and persistent infections.


Subject(s)
Cytomegalovirus Infections/metabolism , Cytomegalovirus/physiology , Cytoplasm/metabolism , DNA Helicases/metabolism , Neoplasms/metabolism , Vesicular Transport Proteins/metabolism , Virus Replication , Blotting, Western , Cytomegalovirus Infections/virology , DNA Replication , DNA, Viral/genetics , Humans , Immunoprecipitation , Microscopy, Fluorescence , Neoplasms/genetics , Neoplasms/virology , RNA, Small Interfering/genetics , Tumor Cells, Cultured , Two-Hybrid System Techniques , Vesicular Transport Proteins/antagonists & inhibitors , Vesicular Transport Proteins/genetics
20.
PLoS Pathog ; 8(11): e1002968, 2012.
Article in English | MEDLINE | ID: mdl-23133382

ABSTRACT

Genomic DNA replication is a universal and essential process for all herpesvirus including human cytomegalovirus (HCMV). HCMV UL70 protein, which is believed to encode the primase activity of the viral DNA replication machinery and is highly conserved among herpesviruses, needs to be localized in the nucleus, the site of viral DNA synthesis. No host factors that facilitate the nuclear import of UL70 have been reported. In this study, we provided the first direct evidence that UL70 specifically interacts with a highly conserved and ubiquitously expressed member of the heat shock protein Hsp40/DNAJ family, DNAJB6, which is expressed as two isoforms, a and b, as a result of alternative splicing. The interaction of UL70 with a common region of DNAJB6a and b was identified by both a two hybrid screen in yeast and coimmunoprecipitation in human cells. In transfected cells, UL70 was primarily co-localized with DNAJB6a in the nuclei and with DNAJB6b in the cytoplasm, respectively. The nuclear import of UL70 was increased in cells in which DNAJB6a was up-regulated or DNAJB6b was down-regulated, and was reduced in cells in which DNAJB6a was down-regulated or DNAJB6b was up-regulated. Furthermore, the level of viral DNA synthesis and progeny production was increased in cells in which DNAJB6a was up-regulated or DNAJB6b was down-regulated, and was reduced in cells in which DNAJB6a was down-regulated or DNAJB6b was up-regulated. Thus, DNAJB6a and b appear to enhance the nuclear import and cytoplasmic accumulation of UL70, respectively. Our results also suggest that the relative expression levels of DNAJB6 isoforms may play a key role in regulating the cellular localization of UL70, leading to modulation of HCMV DNA synthesis and lytic infection.


Subject(s)
Cell Nucleus/metabolism , Cytomegalovirus/enzymology , DNA Primase/metabolism , DNA, Viral/biosynthesis , HSP40 Heat-Shock Proteins/metabolism , Molecular Chaperones/metabolism , Nerve Tissue Proteins/metabolism , Viral Proteins/metabolism , Alternative Splicing/genetics , Animals , Cell Nucleus/genetics , Cell Nucleus/virology , Cytomegalovirus/genetics , Cytomegalovirus Infections/genetics , Cytomegalovirus Infections/metabolism , DNA Primase/genetics , DNA, Viral/genetics , HSP40 Heat-Shock Proteins/genetics , HeLa Cells , Humans , Mice , Mice, Inbred BALB C , Molecular Chaperones/genetics , Nerve Tissue Proteins/genetics , Protein Binding , Viral Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL