Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
Add more filters










Publication year range
1.
Adv Virus Res ; 118: 1-75, 2024.
Article in English | MEDLINE | ID: mdl-38461029

ABSTRACT

G protein coupled receptors (GPCRs) are seven-transmembrane domain proteins that modulate cellular processes in response to external stimuli. These receptors represent the largest family of membrane proteins, and in mammals, their signaling regulates important physiological functions, such as vision, taste, and olfaction. Many organisms, including yeast, slime molds, and viruses encode GPCRs. Cytomegaloviruses (CMVs) are large, betaherpesviruses, that encode viral GPCRs (vGPCRs). Human CMV (HCMV) encodes four vGPCRs, including UL33, UL78, US27, and US28. Each of these vGPCRs, as well as their rodent and primate orthologues, have been investigated for their contributions to viral infection and disease. Herein, we discuss how the CMV vGPCRs function during lytic and latent infection, as well as our understanding of how they impact viral pathogenesis.


Subject(s)
Cytomegalovirus Infections , Receptors, G-Protein-Coupled , Humans , Animals , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Cytomegalovirus/genetics , Cytomegalovirus Infections/metabolism , Mammals/metabolism
2.
Proc Natl Acad Sci U S A ; 121(10): e2315860121, 2024 Mar 05.
Article in English | MEDLINE | ID: mdl-38408244

ABSTRACT

Human cytomegalovirus (HCMV) is a prevalent pathogen that establishes life-long latent infection in hematopoietic cells. While this infection is usually asymptomatic, immune dysregulation leads to viral reactivation, which can cause significant morbidity and mortality. However, the mechanisms underpinning reactivation remain incompletely understood. The HCMV major immediate early promoter (MIEP)/enhancer is a key factor in this process, as its transactivation from a repressed to active state helps drive viral gene transcription necessary for reactivation from latency. Numerous host transcription factors bind the MIE locus and recruit repressive chromatin modifiers, thus impeding virus reactivation. One such factor is CCCTC-binding protein (CTCF), a highly conserved host zinc finger protein that mediates chromatin conformation and nuclear architecture. However, the mechanisms by which CTCF contributes to HCMV latency were previously unexplored. Here, we confirm that CTCF binds two convergent sites within the MIE locus during latency in primary CD14+ monocytes, and following cellular differentiation, CTCF association is lost as the virus reactivates. While mutation of the MIE enhancer CTCF binding site does not impact viral lytic growth in fibroblasts, this mutant virus fails to maintain latency in myeloid cells. Furthermore, we show the two convergent CTCF binding sites allow looping to occur across the MIEP, supporting transcriptional repression during latency. Indeed, looping between the two sites diminishes during virus reactivation, concurrent with activation of MIE transcription. Taken together, our data reveal that three-dimensional chromatin looping aids in the regulation of HCMV latency and provides insight into promoter/enhancer regulation that may prove broadly applicable across biological systems.


Subject(s)
Cytomegalovirus Infections , Cytomegalovirus , Humans , Chromatin/genetics , Cytomegalovirus/genetics , Cytomegalovirus Infections/genetics , Gene Expression Regulation, Viral , Promoter Regions, Genetic , Virus Activation/genetics , Virus Latency/genetics
3.
bioRxiv ; 2023 Sep 05.
Article in English | MEDLINE | ID: mdl-37732204

ABSTRACT

Establishing a non-productive quiescent/silent infection within monocytes is essential for spread of human cytomegalovirus (HCMV). Yet, how HCMV establishes a quiescent infection in monocytes remains unclear. US28 is a viral G protein-coupled receptor (GPCR) essential for silent infections within cells of the myeloid lineage. We found virion-associated US28 was rapidly delivered to monocytes, while de novo synthesized US28 was delayed for several days. A recombinant mutant virus lacking US28 (US28Δ) was unable to establish a quiescent infection, resulting in a fully productive lytic replication cycle. Mechanistically, viral entry of US28Δ phosphorylated Akt at both serine 473 (S473) and threonine 308 (T308), which contrasted with the site-specific phosphorylation of Akt at S473 following WT infection. Preventing Akt bi-phosphorylation prevented lytic replication of US28Δ, and ectopic expression of a constitutively phosphorylated Akt variant triggered lytic replication of WT infection. Our data demonstrate that virion-delivered US28 fine-tunes Akt activity to permit HCMV infection to enter a quiescent state following primary infection of monocytes.

4.
mBio ; 14(4): e0032623, 2023 08 31.
Article in English | MEDLINE | ID: mdl-37439556

ABSTRACT

Human cytomegalovirus (HCMV) is a betaherpesvirus that establishes lifelong infection in its host and can cause severe comorbidities in individuals with suppressed or compromised immune systems. The lifecycle of HCMV consists of lytic and latent phases, largely dependent upon the cell type infected and whether transcription from the major immediate early locus can ensue. Control of this locus, which acts as a critical "switch" region from where the lytic gene expression cascade originates, as well as regulation of the additional ~235 kilobases of virus genome, occurs through chromatinization with cellular histone proteins after infection. Upon infection of a host cell, an initial intrinsic antiviral response represses gene expression from the incoming genome, which is relieved in permissive cells by viral and host factors in concert. Latency is established in a subset of hematopoietic cells, during which viral transcription is largely repressed while the genome is maintained. As these latently infected cells differentiate, the cellular milieu and epigenetic modifications change, giving rise to the initial stages of virus reactivation from latency. Thus, throughout the cycle of infection, chromatinization, chromatin modifiers, and the recruitment of specific transcription factors influence the expression of genes from the HCMV genome. In this review, we discuss epigenetic regulation of the HCMV genome during the different phases of infection, with an emphasis on recent reports that add to our current perspective.


Subject(s)
Chromatin , Cytomegalovirus Infections , Humans , Epigenesis, Genetic , Virus Latency/genetics , Histones/metabolism , Cytomegalovirus/physiology , Gene Expression Regulation, Viral
5.
Sci Adv ; 8(43): eadd1168, 2022 Oct 28.
Article in English | MEDLINE | ID: mdl-36288299

ABSTRACT

Cytomegalovirus (CMV) reactivation from latency following immune dysregulation remains a serious risk for patients, often causing substantial morbidity and mortality. Here, we demonstrate the CMV-encoded G protein-coupled receptor, US28, in coordination with cellular Ephrin receptor A2, attenuates mitogen-activated protein kinase signaling, thereby limiting viral replication in latently infected primary monocytes. Furthermore, treatment of latently infected primary monocytes with dasatinib, a Food and Drug Association-approved kinase inhibitor used to treat a subset of leukemias, results in CMV reactivation. These ex vivo data correlate with our retrospective analyses of the Explorys electronic health record database, where we find dasatinib treatment is associated with a significant risk of CMV-associated disease (odds ratio 1.58, P = 0.0004). Collectively, our findings elucidate a signaling pathway that plays a central role in the balance between CMV latency and reactivation and identifies a common therapeutic cancer treatment that elevates the risk of CMV-associated disease.

6.
Viruses ; 14(6)2022 06 12.
Article in English | MEDLINE | ID: mdl-35746751

ABSTRACT

Human cytomegalovirus (CMV) is a ubiquitous pathogen that latently resides in hematopoietic cells. Latently infected individuals with dysfunctional immune systems often experience CMV reactivation, which can cause devastating disease and mortality. While factors dictating the balance between latency and reactivation are not completely understood, CMV US28 is required for maintaining latent infection, and viral mutants that alter US28 function result in a lytic-like, rather than latent, infection in hematopoietic cells. In turn, viral lytic factors alter the host cell, making it challenging to characterize the US28-specific changes in the cellular milieu. To circumvent this, we generated a temperature-sensitive TB40/E recombinant virus, TB40/EgfpC510G (tsC510G), into which we engineered an amino acid change at position 510 (C510G) of IE2, as previously described in the CMV Towne strain. Using tsC510G, we then deleted the US28 ORF, termed tsC510G-US28Δ. Consistent with previous findings, tsC510G-US28Δ fails to undergo latency in Kasumi-3 cells at the permissive temperature. However, parallel cultures maintained at the non-permissive temperature showed a significant reduction in infectious center frequency, as measured by limiting dilution assay. Thus, we generated a new US28 mutant virus for use as a tool to study US28-specific changes in latently infected hematopoietic cells in the absence of induced lytic replication.


Subject(s)
Cytomegalovirus Infections , Cytomegalovirus , Cytomegalovirus/physiology , Humans , Temperature , Viral Proteins/genetics , Viral Proteins/metabolism , Virus Latency , Virus Replication
7.
Virol J ; 18(1): 207, 2021 Oct 18.
Article in English | MEDLINE | ID: mdl-34663377

ABSTRACT

BACKGROUND: Human cytomegalovirus (HCMV) resides latently in cells of the myeloid compartment, including CD34+ hematopoietic progenitor cells and circulating monocytes. Healthy hosts maintain the virus latently, and this infection is, for the most part, asymptomatic. However, given the proper external cues, HCMV reactivates from latency, at which point the virus disseminates, causing disease. The viral and cellular factors dictating the balance between these phases of infection are incompletely understood, though a large body of literature support a role for viral-mediated manipulation of host cell signaling. MAIN BODY: To establish and maintain latency, HCMV has evolved various means by which it usurps host cell factors to alter the cellular environment to its own advantage, including altering host cell signaling cascades. As early as virus entry into myeloid cells, HCMV usurps cellular signaling to change the cellular milieu, and this regulation includes upregulation, as well as downregulation, of different signaling cascades. Indeed, given proper reactivation cues, this signaling is again altered to allow for transactivation of viral lytic genes. CONCLUSIONS: HCMV modulation of host cell signaling is not binary, and many of the cellular pathways altered are finely regulated, wherein the slightest modification imparts profound changes to the cellular milieu. It is also evident that viral-mediated cell signaling differs not only between these phases of infection, but also is myeloid cell type specific. Nonetheless, understanding the exact pathways and the means by which HCMV mediates them will undoubtedly provide novel targets for therapeutic intervention.


Subject(s)
Cytomegalovirus , Virus Latency , Cells, Cultured , Cytomegalovirus/genetics , Host-Pathogen Interactions , Signal Transduction , Virus Latency/genetics
8.
Cells ; 10(8)2021 08 18.
Article in English | MEDLINE | ID: mdl-34440891

ABSTRACT

Infection of a host cell by an invading viral pathogen triggers a multifaceted antiviral response. One of the most potent defense mechanisms host cells possess is the interferon (IFN) system, which initiates a targeted, coordinated attack against various stages of viral infection. This immediate innate immune response provides the most proximal defense and includes the accumulation of antiviral proteins, such as IFN-stimulated genes (ISGs), as well as a variety of protective cytokines. However, viruses have co-evolved with their hosts, and as such, have devised distinct mechanisms to undermine host innate responses. As large, double-stranded DNA viruses, herpesviruses rely on a multitude of means by which to counter the antiviral attack. Herein, we review the various approaches the human herpesviruses employ as countermeasures to the host innate immune response.


Subject(s)
Herpesviridae Infections/immunology , Immunity, Innate/physiology , Animals , Herpesviridae Infections/metabolism , Humans , Immunity, Innate/genetics , Signal Transduction/physiology , Virus Replication/physiology
9.
Pathogens ; 10(3)2021 Mar 15.
Article in English | MEDLINE | ID: mdl-33804183

ABSTRACT

Cytomegalovirus (CMV) is a herpesvirus that infects a majority of the human population worldwide [...].

10.
Methods Mol Biol ; 2244: 159-197, 2021.
Article in English | MEDLINE | ID: mdl-33555587

ABSTRACT

All of the cytomegaloviruses discovered to date encode two or more genes with significant homology to G protein-coupled receptors (GPCRs). The functions of these cytomegalovirus GPCRs continue to be actively studied and it is clear that they exhibit numerous interesting functions in vitro and in vivo. In this chapter, we review the various methodologies that can be used to examine biochemical aspects of viral GPCR signaling in vitro, as well as examine the biological activity of these viral GPCRs in vitro and in vivo in virus infected cells using recombinant cytomegaloviruses.


Subject(s)
Cell Culture Techniques/methods , Cytomegalovirus/genetics , Receptors, G-Protein-Coupled/genetics , Animals , Cell Line/virology , Cytomegalovirus/metabolism , Humans , Primary Cell Culture/methods , Signal Transduction/genetics , Signal Transduction/physiology
11.
J Cell Sci ; 134(5)2021 01 25.
Article in English | MEDLINE | ID: mdl-33199520

ABSTRACT

Human cytomegalovirus (HCMV) establishes life-long latent infection in hematopoietic progenitor cells and circulating monocytes in infected individuals. Myeloid differentiation coupled with immune dysregulation leads to viral reactivation, which can cause severe disease and mortality. Reactivation of latent virus requires chromatin reorganization and the removal of transcriptional repressors in exchange for transcriptional activators. While some factors involved in these processes are identified, a complete characterization of the viral and cellular factors involved in their upstream regulation remains elusive. Herein, we show the HCMV-encoded G protein-coupled receptor (GPCR), UL33, is expressed during latency. Although this viral GPCR is not required to maintain latent infection, our data reveal UL33-mediated signaling is important for efficient viral reactivation. Additionally, UL33 signaling induces cellular cyclic AMP response element binding protein (CREB1, referred to here as CREB) phosphorylation, a transcription factor that promotes reactivation when recruited to the major immediate early (MIE) enhancer/promoter. Finally, targeted pharmacological inhibition of CREB activity reverses the reactivation phenotype of the UL33 signaling-deficient mutant. In sum, our data reveal UL33-mediated signaling functions to activate CREB, resulting in successful viral reactivation.


Subject(s)
Cyclic AMP Response Element-Binding Protein , Cytomegalovirus Infections , Cytomegalovirus , Receptors, G-Protein-Coupled , Virus Activation , Cyclic AMP Response Element-Binding Protein/genetics , Cyclic AMP Response Element-Binding Protein/metabolism , Cytomegalovirus/genetics , Cytomegalovirus/metabolism , Cytomegalovirus Infections/genetics , Humans , Signal Transduction
12.
Pathogens ; 9(11)2020 Oct 23.
Article in English | MEDLINE | ID: mdl-33113934

ABSTRACT

Human cytomegalovirus (HCMV) is a ubiquitous herpesviral pathogen that results in life-long infection. HCMV maintains a latent or quiescent infection in hematopoietic cells, which is broadly defined by transcriptional silencing and the absence of de novo virion production. However, upon cell differentiation coupled with immune dysfunction, the virus can reactivate, which leads to lytic replication in a variety of cell and tissue types. One of the mechanisms controlling the balance between latency and reactivation/lytic replication is the regulation of the major immediate-early (MIE) locus. This enhancer/promoter region is complex, and it is regulated by chromatinization and associated factors, as well as a variety of transcription factors. Herein, we discuss these factors and how they influence the MIE locus, which ultimately impacts the phase of HCMV infection.

13.
Virology ; 548: 49-58, 2020 09.
Article in English | MEDLINE | ID: mdl-32838946

ABSTRACT

Human cytomegalovirus (HCMV) is a widespread herpesvirus that establishes latency in myeloid cells and persists by manipulating immune signaling. Chemokine receptor CXCR4 and its ligand CXCL12 regulate movement of myeloid progenitors into bone marrow and out into peripheral tissues. HCMV amplifies CXCL12-CXCR4 signaling through viral chemokine receptor US27 and cmvIL-10, a viral cytokine that binds the cellular IL-10 receptor (IL-10R), but precisely how these viral proteins influence CXCR4 is unknown. We used the proximity ligation assay (PLA) to examine association of CXCR4, IL-10R, and US27 in both transfected and HCMV-infected cells. CXCR4 and IL-10R colocalized to discrete clusters, and treatment with CXCL12 and cmvIL-10 dramatically increased receptor clustering and calcium flux. US27 was associated with CXCR4 and IL-10R in PLA clusters and further enhanced cluster formation and calcium signaling. These results indicate that CXCR4, IL-10R, and US27 form a novel virus-host signaling complex that enhances CXCL12 signaling during HCMV infection.


Subject(s)
Cytomegalovirus Infections/metabolism , Cytomegalovirus/metabolism , Receptors, CXCR4/metabolism , Receptors, Chemokine/metabolism , Receptors, Interleukin-10/metabolism , Viral Proteins/metabolism , Chemokine CXCL12/genetics , Chemokine CXCL12/metabolism , Cytomegalovirus/genetics , Cytomegalovirus Infections/genetics , Cytomegalovirus Infections/virology , Host-Pathogen Interactions , Humans , Protein Binding , Receptors, CXCR4/genetics , Receptors, Chemokine/genetics , Receptors, Interleukin-10/genetics , Signal Transduction , Viral Proteins/genetics
14.
Proc Natl Acad Sci U S A ; 117(34): 20860-20867, 2020 08 25.
Article in English | MEDLINE | ID: mdl-32788362

ABSTRACT

Human cytomegalovirus (HCMV) is a ubiquitous pathogen that latently infects hematopoietic cells and has the ability to reactivate when triggered by immunological stress. This reactivation causes significant morbidity and mortality in immune-deficient patients, who are unable to control viral dissemination. While a competent immune system helps prevent clinically detectable viremia, a portrait of the factors that induce reactivation following the proper cues remains incomplete. Our understanding of the complex molecular mechanisms underlying latency and reactivation continues to evolve. We previously showed the HCMV-encoded G protein-coupled receptor US28 is expressed during latency and facilitates latent infection by attenuating the activator protein-1 (AP-1) transcription factor subunit, c-fos, expression and activity. We now show AP-1 is a critical component for HCMV reactivation. Pharmacological inhibition of c-fos significantly attenuates viral reactivation. In agreement, infection with a virus in which we disrupted the proximal AP-1 binding site in the major immediate early (MIE) enhancer results in inefficient reactivation compared to WT. Concomitantly, AP-1 recruitment to the MIE enhancer is significantly decreased following reactivation of the mutant virus. Furthermore, AP-1 is critical for derepression of MIE-driven transcripts and downstream early and late genes, while immediate early genes from other loci remain unaffected. Our data also reveal MIE transcripts driven from the MIE promoter, the distal promoter, and the internal promoter, iP2, are dependent upon AP-1 recruitment, while iP1-driven transcripts are AP-1-independent. Collectively, our data demonstrate AP-1 binding to and activation of the MIE enhancer is a key molecular process controlling reactivation from latency.


Subject(s)
Cytomegalovirus/genetics , Transcription Factor AP-1/metabolism , Virus Activation/genetics , Cytomegalovirus/metabolism , Cytomegalovirus/physiology , Cytomegalovirus Infections/virology , Genes, Immediate-Early/genetics , Humans , Immediate-Early Proteins/genetics , Promoter Regions, Genetic/genetics , Signal Transduction/genetics , Transcription Factor AP-1/genetics , Transcriptional Activation/genetics , Virus Latency/genetics
15.
Article in English | MEDLINE | ID: mdl-32411622

ABSTRACT

The ability to establish a latent infection with periodic reactivation events ensures herpesviruses, like human cytomegalovirus (HCMV), lifelong infection, and serial passage. The host-pathogen relationship throughout HCMV latency is complex, though both cellular and viral factors influence the equilibrium between latent and lytic infection. We and others have shown one of the viral-encoded G protein-coupled receptors, US28, is required for HCMV latency. US28 potentiates signals both constitutively and in response to ligand binding, and we previously showed deletion of the ligand binding domain or mutation of the G protein-coupling domain results in the failure to maintain latency similar to deletion of the entire US28 open reading frame (ORF). Interestingly, a recent publication detailed an altered phenotype from that previously reported, showing US28 is required for viral reactivation rather than latency, suggesting the US28 ORF deletion impacts transcription of the surrounding genes. Here, we show an independently generated US28-stop mutant, like the US28 ORF deletion mutant, fails to maintain latency in hematopoietic cells. Further, we found US27 and US29 transcription in each of these mutants was comparable to their expression during wild type infection, suggesting neither US28 mutant alters mRNA levels of the surrounding genes. Finally, infection with a US28 ORF deletion virus expressed US27 protein comparable to its expression following wild type infection. In sum, our new data strongly support previous findings from our lab and others, detailing a requirement for US28 during HCMV latent infection.


Subject(s)
Cytomegalovirus , Signal Transduction , Virus Latency , Cytomegalovirus/genetics , Gene Expression , Humans , Receptors, Chemokine , Viral Proteins/genetics
16.
mBio ; 10(6)2019 12 03.
Article in English | MEDLINE | ID: mdl-31796538

ABSTRACT

Human cytomegalovirus (HCMV) latency is an active process which remodels the latently infected cell to optimize latent carriage and reactivation. This is achieved, in part, through the expression of viral genes, including the G-protein-coupled receptor US28. Here, we use an unbiased proteomic screen to assess changes in host proteins induced by US28, revealing that interferon-inducible genes are downregulated by US28. We validate that major histocompatibility complex (MHC) class II and two pyrin and HIN domain (PYHIN) proteins, myeloid cell nuclear differentiation antigen (MNDA) and IFI16, are downregulated during experimental latency in primary human CD14+ monocytes. We find that IFI16 is targeted rapidly during the establishment of latency in a US28-dependent manner but only in undifferentiated myeloid cells, a natural site of latent carriage. Finally, by overexpressing IFI16, we show that IFI16 can activate the viral major immediate early promoter and immediate early gene expression during latency via NF-κB, a function which explains why downregulation of IFI16 during latency is advantageous for the virus.IMPORTANCE Human cytomegalovirus (HCMV) is a ubiquitous herpesvirus which infects 50 to 100% of humans worldwide. HCMV causes a lifelong subclinical infection in immunocompetent individuals but is a serious cause of mortality and morbidity in the immunocompromised and neonates. In particular, reactivation of HCMV in the transplant setting is a major cause of transplant failure and related disease. Therefore, a molecular understanding of HCMV latency and reactivation could provide insights into potential ways to target the latent viral reservoir in at-risk patient populations.


Subject(s)
Cytomegalovirus Infections/genetics , Cytomegalovirus/immunology , Interferons/genetics , Virus Latency/genetics , Cell Differentiation/genetics , Cell Differentiation/immunology , Cell Line , Cytomegalovirus Infections/immunology , Cytomegalovirus Infections/virology , Down-Regulation/genetics , Down-Regulation/immunology , Gene Expression/genetics , Gene Expression/immunology , Gene Expression Regulation, Viral/genetics , Gene Expression Regulation, Viral/immunology , HEK293 Cells , Humans , Interferons/immunology , Monocytes/immunology , Monocytes/virology , Myeloid Cells/immunology , Myeloid Cells/virology , NF-kappa B/genetics , NF-kappa B/immunology , Promoter Regions, Genetic/genetics , Promoter Regions, Genetic/immunology , Proteomics/methods , Receptors, G-Protein-Coupled/immunology , THP-1 Cells , Viral Proteins/genetics , Viral Proteins/immunology , Virus Activation/genetics , Virus Activation/immunology , Virus Latency/immunology
17.
Proc Natl Acad Sci U S A ; 116(5): 1755-1764, 2019 01 29.
Article in English | MEDLINE | ID: mdl-30647114

ABSTRACT

Human cytomegalovirus (HCMV) is a ubiquitous pathogen that undergoes latency in cells of the hematopoietic compartment, although the mechanisms underlying establishment and maintenance of latency remain elusive. We previously reported that the HCMV-encoded G protein-coupled receptor (GPCR) homolog US28 is required for successful latent infection. We now show that US28 protein (pUS28) provided in trans complements the US28Δ lytic phenotype in myeloid cells, suggesting that sustained US28 expression is necessary for long-term latency. Furthermore, expression of pUS28 at the time of infection represses transcription from the major immediate early promoter (MIEP) within 24 h. However, this repression is only maintained in the presence of continual pUS28 expression provided in trans Our data also reveal that pUS28-mediated signaling attenuates both expression and phosphorylation of cellular fos (c-fos), an AP-1 transcription factor subunit, to repress MIEP-driven transcription. AP-1 binds to the MIEP and promotes lytic replication, and in line with this we find that US28Δ infection results in an increase in AP-1 binding to the MIEP, compared with WT latent infection. Pharmacological inhibition of c-fos represses the MIEP during US28Δ infection to levels similar to those we observe during WT latent infection. Together, our data reveal that US28 is required for both establishment and long-term maintenance of HCMV latency, which is modulated, at least in part, by repressing functional AP-1 binding to the MIEP.


Subject(s)
Cytomegalovirus Infections/virology , Cytomegalovirus/genetics , Receptors, Chemokine/genetics , Receptors, G-Protein-Coupled/genetics , Viral Proteins/genetics , Virus Latency/genetics , Cell Line , Gene Expression Regulation, Viral/genetics , HEK293 Cells , Humans , Promoter Regions, Genetic/genetics , Signal Transduction/genetics , Transcription Factor AP-1/genetics , Virus Replication/genetics
18.
Viruses ; 10(11)2018 11 07.
Article in English | MEDLINE | ID: mdl-30405048

ABSTRACT

Human cytomegalovirus (HCMV) is a ubiquitous herpesvirus for which there is no vaccine or cure. This viral infection, once acquired, is life-long, residing latently in hematopoietic cells. However, latently infected individuals with weakened immune systems often undergo HCMV reactivation, which can cause serious complications in immunosuppressed and immunocompromised patients. Current anti-viral therapies target late stages of viral replication, and are often met with therapeutic resistance, necessitating the development of novel therapeutics. In this current study, we identified a naturally-occurring flavonoid compound, deguelin, which inhibits HCMV lytic replication. Our findings reveal that nanomolar concentrations of deguelin significantly suppress the production of the infectious virus. Further, we show that deguelin inhibits the lytic cycle during the phase of the replication cycle consistent with early (E) gene and protein expression. Importantly, our data reveal that deguelin inhibits replication of a ganciclovir-resistant strain of HCMV. Together, our findings identify a novel, naturally occurring compound that may prove useful in the treatment of HCMV replication.


Subject(s)
Cytomegalovirus/drug effects , Cytomegalovirus/physiology , Fibroblasts/drug effects , Fibroblasts/virology , Flavonoids/pharmacology , Rotenone/analogs & derivatives , Virus Replication/drug effects , Cell Survival/drug effects , Drug Resistance, Viral , Flavonoids/chemistry , Gene Expression Regulation, Viral/drug effects , Humans , Inhibitory Concentration 50 , Rotenone/chemistry , Rotenone/pharmacology , Virus Internalization/drug effects
19.
J Virol ; 92(23)2018 12 01.
Article in English | MEDLINE | ID: mdl-30209167

ABSTRACT

Human cytomegalovirus (HCMV) is a widespread pathogen that modulates host chemokine signaling during persistent infection in the host. HCMV encodes four proteins with homology to the chemokine receptor family of G protein-coupled receptors (GPCRs): US27, US28, UL33, and UL78. Each of the four receptors modulates host CXCR4 signaling. US28, UL33, and UL78 impair CXCR4 signaling outcomes, while US27 enhances signaling, as evidenced by increased calcium mobilization and cell migration to CXCL12. To investigate the effects of US27 on CXCR4 during virus infection, fibroblasts were infected with bacterial artificial chromosome-derived clinical strain HCMV TB40/E-mCherry (wild type [WT]), mutants lacking US27 (TB40/E-mCherry-US27Δ [US27Δ]) or all four GPCRs (TB40 E-mCherry-allΔ), or mutants expressing only US27 but not US28, UL33, or UL78 (TB40/E-mCherry-US27wt [US27wt]). CXCR4 gene expression was significantly higher in WT- and US27wt-infected fibroblasts. This effect was evident at 3 h postinfection, suggesting that US27 derived from the parental virion enhanced CXCR4 expression. Reporter gene assays demonstrated that US27 increased transcriptional activity regulated by the antioxidant response element (ARE), and small interfering RNA treatment indicated that this effect was mediated by NRF-1, the primary transcription factor for CXCR4. Increased translocation of NRF-1 into the nucleus of WT-infected cells compared to mock- or US27Δ-infected cells was confirmed by immunofluorescence microscopy. Chemical inhibitors targeting Gßγ and phosphoinositide 3-kinase (PI3K) ablated the increase in ARE-driven transcription, implicating these proteins as mediators of US27-stimulated gene transcription. This work identifies the first signaling pathway activated by HCMV US27 and may reveal a novel regulatory function for this orphan viral receptor in stimulating stress response genes during infection.IMPORTANCE Human cytomegalovirus (HCMV) is the most common congenital infection worldwide, causing deafness, blindness, and other serious birth defects. CXCR4 is a human chemokine receptor that is crucial for both fetal development and immune responses. We found that the HCMV protein US27 stimulates increased expression of CXCR4 through activation of the transcription factor nuclear respiratory factor 1 (NRF-1). NRF-1 regulates stress response genes that contain the antioxidant response element (ARE), and HCMV infection is associated with increased expression of many stress response genes when US27 is present. Our results show that the US27 protein activates the NRF-1/ARE pathway, stimulating higher expression of CXCR4 and other stress response genes, which is likely to be beneficial for virus replication and/or immune evasion.


Subject(s)
Antioxidant Response Elements , GTP-Binding Protein beta Subunits/metabolism , GTP-Binding Protein gamma Subunits/metabolism , Gene Expression Regulation , Nuclear Respiratory Factor 1/metabolism , Phosphatidylinositol 3-Kinase/metabolism , Receptors, CXCR4/genetics , Receptors, Chemokine/metabolism , Viral Proteins/metabolism , Cell Movement , Cytomegalovirus/physiology , Cytomegalovirus Infections/genetics , Cytomegalovirus Infections/metabolism , Cytomegalovirus Infections/virology , GTP-Binding Protein beta Subunits/genetics , GTP-Binding Protein gamma Subunits/genetics , HEK293 Cells , Humans , Nuclear Respiratory Factor 1/genetics , Phosphatidylinositol 3-Kinase/genetics , Promoter Regions, Genetic , Protein Binding , Receptors, CXCR4/metabolism , Receptors, Chemokine/genetics , Signal Transduction , Viral Proteins/genetics , Virus Replication
20.
Viruses ; 10(8)2018 08 20.
Article in English | MEDLINE | ID: mdl-30127279

ABSTRACT

US28 is one of four G protein coupled receptors (GPCRs) encoded by human cytomegalovirus (HCMV). The US28 protein (pUS28) is a potent signaling molecule that alters a variety of cellular pathways that ultimately alter the host cell environment. This viral GPCR is expressed not only in the context of lytic replication but also during viral latency, highlighting its multifunctional properties. pUS28 is a functional GPCR, and its manipulation of multiple signaling pathways likely impacts HCMV pathogenesis. Herein, we will discuss the impact of pUS28 on both lytic and latent infection, pUS28-mediated signaling and its downstream consequences, and the influence this viral GPCR may have on disease states, including cardiovascular disease and cancer. We will also discuss the potential for and progress towards exploiting pUS28 as a novel therapeutic to combat HCMV.


Subject(s)
Cardiovascular Diseases/virology , Cytomegalovirus Infections/virology , Cytomegalovirus/pathogenicity , Gene Expression Regulation, Viral , Host-Pathogen Interactions , Neoplasms/virology , Receptors, Chemokine/genetics , Viral Proteins/genetics , Cardiovascular Diseases/pathology , Cytomegalovirus/genetics , Cytomegalovirus/metabolism , Cytomegalovirus Infections/drug therapy , Cytomegalovirus Infections/pathology , Humans , Models, Molecular , Neoplasms/pathology , Protein Structure, Secondary , Receptors, Chemokine/therapeutic use , Signal Transduction , Viral Proteins/therapeutic use , Virus Latency/genetics , Virus Replication/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...