Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Nat Commun ; 14(1): 3087, 2023 05 29.
Article in English | MEDLINE | ID: mdl-37248241

ABSTRACT

To date, no herpesvirus has been shown to latently persist in fibroblastic cells. Here, we show that murine cytomegalovirus, a ß-herpesvirus, persists for the long term and across organs in PDGFRα-positive fibroblastic cells, with similar or higher genome loads than in the previously known sites of murine cytomegalovirus latency. Whereas murine cytomegalovirus gene transcription in PDGFRα-positive fibroblastic cells is almost completely silenced at 5 months post-infection, these cells give rise to reactivated virus ex vivo, arguing that they support latent murine cytomegalovirus infection. Notably, PDGFRα-positive fibroblastic cells also support productive virus replication during primary murine cytomegalovirus infection. Mechanistically, Stat1-deficiency promotes lytic infection but abolishes latent persistence of murine cytomegalovirus in PDGFRα-positive fibroblastic cells in vivo. In sum, fibroblastic cells have a dual role as a site of lytic murine cytomegalovirus replication and a reservoir of latent murine cytomegalovirus in vivo and STAT1 is required for murine cytomegalovirus latent persistence in vivo.


Subject(s)
Cytomegalovirus Infections , Muromegalovirus , Animals , Mice , Cytomegalovirus/genetics , Virus Latency/genetics , Receptor, Platelet-Derived Growth Factor alpha , Virus Replication , Fibroblasts , STAT1 Transcription Factor/genetics
2.
Nucleic Acids Res ; 48(20): 11799-11811, 2020 11 18.
Article in English | MEDLINE | ID: mdl-33137201

ABSTRACT

Mammalian first line of defense against viruses is accomplished by the interferon (IFN) system. Viruses have evolved numerous mechanisms to reduce the IFN action allowing them to invade the host and/or to establish latency. We generated an IFN responsive intracellular hub by integrating the synthetic transactivator tTA into the chromosomal Mx2 locus for IFN-based activation of tTA dependent expression modules. The additional implementation of a synthetic amplifier module with positive feedback even allowed for monitoring and reacting to infections of viruses that can antagonize the IFN system. Low and transient IFN amounts are sufficient to trigger these amplifier cells. This gives rise to higher and sustained-but optionally de-activatable-expression even when the initial stimulus has faded out. Amplification of the IFN response induced by IFN suppressing viruses is sufficient to protect cells from infection. Together, this interfaced sensor/actuator system provides a toolbox for robust sensing and counteracting viral infections.


Subject(s)
Interferon Type I/metabolism , Virus Physiological Phenomena , Animals , Cells, Cultured , Feedback, Physiological , Luciferases/analysis , Mice , Newcastle disease virus/physiology
3.
Front Cell Infect Microbiol ; 10: 536150, 2020.
Article in English | MEDLINE | ID: mdl-33489928

ABSTRACT

Human Cytomegalovirus (HCMV) infection may result in severe outcomes in immunocompromised individuals such as AIDS patients, transplant recipients, and neonates. To date, no vaccines are available and there are only few drugs for anti-HCMV therapy. Adverse effects and the continuous emergence of drug-resistance strains require the identification of new drug candidates in the near future. Identification and characterization of such compounds and biological factors requires sensitive and reliable detection techniques of HCMV infection, gene expression and spread. In this work, we present and validate a novel concept for multi-reporter herpesviruses, identified through iterative testing of minimally invasive mutations. We integrated up to three fluorescence reporter genes into replication-competent HCMV strains, generating reporter HCMVs that allow the visualization of replication cycle stages of HCMV, namely the immediate early (IE), early (E), and late (L) phase. Fluorescent proteins with clearly distinguishable emission spectra were linked by 2A peptides to essential viral genes, allowing bicistronic expression of the viral and the fluorescent protein without major effects on viral fitness. By using this triple color reporter HCMV, we monitored gene expression dynamics of the IE, E, and L genes by measuring the fluorescent signal of the viral gene-associated fluorophores within infected cell populations and at high temporal resolution. We demonstrate distinct inhibitory profiles of foscarnet, fomivirsen, phosphonoacetic acid, ganciclovir, and letermovir reflecting their mode-of-action. In conclusion, our data argues that this experimental approach allows the identification and characterization of new drug candidates in a single step.


Subject(s)
Herpesviridae , Pharmaceutical Preparations , Antiviral Agents/pharmacology , Cytomegalovirus/genetics , Gene Expression , Humans , Infant, Newborn , Virus Replication
4.
J Virol ; 92(1)2018 01 01.
Article in English | MEDLINE | ID: mdl-29046460

ABSTRACT

Cytomegalovirus (CMV) is a betaherpesvirus that latently infects most adult humans worldwide and is a major cause of morbidity and mortality in immunocompromised hosts. Latent human CMV (HCMV) is believed to reside in precursors of myeloid-lineage leukocytes and monocytes, which give rise to macrophages and dendritic cells (DC). We report here that human monocyte-derived DC (mo-DC) suppress HCMV infection in coculture with infected fibroblast target cells in a manner dependent on the effector-to-target ratio. Intriguingly, optimal activation of mo-DC was achieved under coculture conditions and not by direct infection with HCMV, implying that mo-DC may recognize unique molecular patterns on, or within, infected fibroblasts. We show that HCMV is controlled by secreted factors that act by priming defenses in target cells rather than by direct viral neutralization, but we excluded a role for interferons (IFNs) in this control. The expression of lytic viral genes in infected cells and the progression of infection were significantly slowed, but this effect was reversible, indicating that the control of infection depended on the transient induction of antiviral effector molecules in target cells. Using immediate early or late-phase reporter HCMVs, we show that soluble factors secreted in the cocultures suppress HCMV replication at both stages of the infection and that their antiviral effects are robust and comparable in numerous batches of mo-DC as well as in primary fibroblasts and stromal cells.IMPORTANCE Human cytomegalovirus is a widespread opportunistic pathogen that can cause severe disease and complications in vulnerable individuals. This includes newborn children, HIV AIDS patients, and transplant recipients. Although the majority of healthy humans carry this virus throughout their lives without symptoms, it is not exactly clear which tissues in the body are the main reservoirs of latent virus infection or how the delicate balance between the virus and the immune system is maintained over an individual's lifetime. Here, for the first time, we provide evidence for a novel mechanism of direct virus control by a subset of human innate immune cells called dendritic cells, which are regarded as a major site of virus latency and reactivation. Our findings may have important implications in HCMV disease prevention as well as in development of novel therapeutic approaches.


Subject(s)
Antiviral Agents/metabolism , Cytomegalovirus/genetics , Dendritic Cells/immunology , Dendritic Cells/virology , Fibroblasts/virology , Gene Expression , Antiviral Agents/chemistry , Antiviral Agents/immunology , Coculture Techniques , Cytomegalovirus/physiology , Dendritic Cells/physiology , Genes, Viral , Humans , Immunity, Innate , Interferons/immunology , Microscopy, Video , Myeloid Cells/immunology , Myeloid Cells/virology , Solubility , Virus Activation , Virus Latency
5.
Sci Rep ; 7(1): 15588, 2017 Nov 14.
Article in English | MEDLINE | ID: mdl-29138436

ABSTRACT

Cell rounding is a hallmark of the cytopathic effect induced by cytomegaloviruses. By screening a panel of deletion mutants of mouse cytomegalovirus (MCMV) a mutant was identified that did not elicit cell rounding and lacked the ability to form typical plaques. Altered cell morphology was assigned to the viral M25 gene. We detected an early 2.8 kb M25 mRNA directing the synthesis of a 105 kDa M25 protein, and confirmed that a late 3.1 kb mRNA encodes a 130 kDa M25 tegument protein. Virions lacking the M25 tegument protein were of smaller size because the tegument layer between capsid and viral envelope was reduced. The ΔM25 mutant did not provoke the rearrangement of the actin cytoskeleton observed after wild-type MCMV infection, and isolated expression of the M25 proteins led to cell size reduction, confirming that they contribute to the morphological changes. Yields of progeny virus and cell-to-cell spread of the ΔM25 mutant in vitro were diminished and replication in vivo was impaired. The identification of an MCMV gene involved in cell rounding provides the basis for investigating the role of this cytopathic effect in CMV pathogenesis.


Subject(s)
Herpesviridae Infections/genetics , Muromegalovirus/genetics , Viral Envelope Proteins/genetics , Animals , Herpesviridae Infections/virology , Mice , Muromegalovirus/pathogenicity , Sequence Deletion/genetics , Virion/genetics , Virion/growth & development
6.
Sci Rep ; 7(1): 8661, 2017 08 17.
Article in English | MEDLINE | ID: mdl-28819195

ABSTRACT

Human cytomegalovirus (HCMV) is a major human pathogen, causing serious diseases in immunocompromised populations and congenially infected neonates. One of the main immune cells acting against the virus are Natural Killer (NK) cells. Killing by NK cells is mediated by a small family of activating receptors such as NKp30 that interact with the cellular ligand B7-H6. The outcome of B7-H6-NKp30 interaction was, so far, mainly studied with regard to NK recognition and killing of tumors. Here, we demonstrated that the expression of B7-H6 is upregulated following HCMV infection and that HCMV uses two of its genes: US18 and US20, to interfere with B7-H6 surface expression, in a mechanism involving endosomal degradation, in order to evade NK cell recognition.


Subject(s)
B7 Antigens/genetics , Cytomegalovirus Infections/immunology , Cytomegalovirus Infections/virology , Cytomegalovirus/physiology , Host-Pathogen Interactions , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Viral Proteins/genetics , B7 Antigens/metabolism , Cell Line , Cytomegalovirus Infections/metabolism , Cytotoxicity, Immunologic , Gene Expression Regulation , Gene Order , Genome, Viral , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Humans , Lysosomes/metabolism , Natural Cytotoxicity Triggering Receptor 3/genetics , Natural Cytotoxicity Triggering Receptor 3/metabolism , Virulence/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...