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.
Proc Natl Acad Sci U S A ; 121(12): e2312290121, 2024 Mar 19.
Article in English | MEDLINE | ID: mdl-38483999

ABSTRACT

Human cytomegalovirus (HCMV) infection of monocytes is essential for viral dissemination and persistence. We previously identified that HCMV entry/internalization and subsequent productive infection of this clinically relevant cell type is distinct when compared to other infected cells. We showed that internalization and productive infection required activation of epidermal growth factor receptor (EGFR) and integrin/c-Src, via binding of viral glycoprotein B to EGFR, and the pentamer complex to ß1/ß3 integrins. To understand how virus attachment drives entry, we compared infection of monocytes with viruses containing the pentamer vs. those without the pentamer and then used a phosphoproteomic screen to identify potential phosphorylated proteins that influence HCMV entry and trafficking. The screen revealed that the most prominent pentamer-biased phosphorylated protein was the lipid- and protein-phosphatase phosphatase and tensin homolog (PTEN). PTEN knockdown with siRNA or PTEN inhibition with a PTEN inhibitor decreased pentamer-mediated HCMV entry, without affecting trimer-mediated entry. Inhibition of PTEN activity affected lipid metabolism and interfered with the onset of the endocytic processes required for HCMV entry. PTEN inactivation was sufficient to rescue pentamer-null HCMV from lysosomal degradation. We next examined dephosphorylation of a PTEN substrate Rab7, a regulator of endosomal maturation. Inhibition of PTEN activity prevented dephosphorylation of Rab7. Phosphorylated Rab7, in turn, blocked early endosome to late endosome maturation and promoted nuclear localization of the virus and productive infection.


Subject(s)
Monocytes , Virus Internalization , Humans , Cells, Cultured , Monocytes/metabolism , Cytomegalovirus/physiology , ErbB Receptors/metabolism , Phosphoric Monoester Hydrolases/metabolism , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism
2.
J Virol ; 96(14): e0081922, 2022 07 27.
Article in English | MEDLINE | ID: mdl-35862696

ABSTRACT

Human cytomegalovirus (HCMV) exhibits a complex host-pathogen interaction with peripheral blood monocytes. We have identified a unique, cell-type specific retrograde-like intracellular trafficking pattern that HCMV utilizes to gain access to the monocyte nucleus and for productive infection. We show that infection of primary human monocytes, epithelial cells, and fibroblasts leads to an increase in the amount of the trafficking protein Syntaxin 6 (Stx6). However, only knockdown (KD) of Stx6 in monocytes inhibited viral trafficking to the trans-Golgi network (TGN), a requisite step for nuclear translocation in monocytes. Conversely, KD of Stx6 in epithelial cells and fibroblasts did not change the kinetics of nuclear translocation and productive infection. Stx6 predominantly functions at the level of the TGN where it facilitates retrograde transport, a trafficking pathway used by only a few cellular proteins and seldom by pathogens. We also newly identify that in monocytes, Stx6 exhibits an irregular vesicular localization rather than being concentrated at the TGN as seen in other cell-types. Lastly, we implicate that viral particles that associate with both Stx6 and EEA1 early in infection are the viral population that successfully traffics to the TGN at later time points and undergo nuclear translocation. Additionally, we show for the first time that HCMV enters the TGN, and that lack of Stx6 prevents viral trafficking to this organelle. We argue that we have identified an essential cell-type specific regulator that controls early steps in efficient productive infection of a cell-type required for viral persistence and disease. IMPORTANCE Human cytomegalovirus (HCMV) infection causes severe and often fatal disease in the immunocompromised. It is one of the leading infectious causes of birth defects and causes severe complications in transplant recipients. By uncovering the unique pathways used by the virus to infect key cells, such as monocytes, responsible for dissemination and persistence, we provide new potential targets for therapeutic intervention.


Subject(s)
Cytomegalovirus , Monocytes , Qa-SNARE Proteins , Cytomegalovirus/pathogenicity , Humans , Monocytes/virology , Qa-SNARE Proteins/genetics , Qa-SNARE Proteins/metabolism , Signal Transduction , trans-Golgi Network/metabolism
3.
Methods Mol Biol ; 2244: 1-18, 2021.
Article in English | MEDLINE | ID: mdl-33555579

ABSTRACT

Human cytomegalovirus (HCMV) is a betaherpesvirus with a global seroprevalence of 60-90%. HCMV is the leading cause of congenital infections and poses a great health risk to immunocompromised individuals. Although HCMV infection is typically asymptomatic in the immunocompetent population, infection can result in mononucleosis and has also been associated with the development of certain cancers, as well as chronic inflammatory diseases such as various cardiovascular diseases. In immunocompromised patients, including AIDS patients, transplant recipients, and developing fetuses, HCMV infection is associated with increased rates of morbidity and mortality. Currently there is no vaccine for HCMV and there is a need for new pharmacological treatments. Ongoing research seeks to further define the complex aspects of HCMV pathogenesis, which could potentially lead to the generation of new therapeutics to mitigate the disease states associated with HCMV infection. The following chapter reviews the advancements in our understanding of HCMV pathogenesis in the immunocompetent and immunocompromised hosts.


Subject(s)
Cytomegalovirus Infections/immunology , Cytomegalovirus/metabolism , Cytomegalovirus/physiology , Cytomegalovirus/genetics , Cytomegalovirus/pathogenicity , DNA, Viral/genetics , Humans , Immunocompromised Host/immunology , Seroepidemiologic Studies
4.
Methods Mol Biol ; 2244: 103-113, 2021.
Article in English | MEDLINE | ID: mdl-33555584

ABSTRACT

Human cytomegalovirus (HCMV) can cause severe disease in the immunocompromised. One of the hallmarks of HCMV infection of a human host is the targeted infection of peripheral blood monocytes (but not other leukocyte populations) that in turn serve as the key cell type for hematogenous dissemination and the establishment of persistence following primary infection. Monocytes are also a key cell type associated with viral reactivation and spread following viral reactivation. Because of their importance in the HCMV-host infection cycle and lifelong infection, it is critical to be able to study their infection in controlled in vitro systems in the laboratory. In this chapter, we discuss a viable protocol for harvesting fresh ex vivo blood monocytes from human donors that are pure and unactivated cells and that can be used in a research setting.


Subject(s)
Centrifugation, Density Gradient/methods , Cytomegalovirus/metabolism , Primary Cell Culture/methods , Cell Differentiation , Cytomegalovirus Infections/virology , Host-Pathogen Interactions , Humans , Models, Biological , Monocytes/cytology , Monocytes/metabolism , Signal Transduction , Viral Proteins , Virus Activation/genetics , Virus Activation/physiology , Virus Internalization , Virus Latency/physiology
5.
Proc Natl Acad Sci U S A ; 117(32): 19507-19516, 2020 08 11.
Article in English | MEDLINE | ID: mdl-32723814

ABSTRACT

Previous analysis of postentry events revealed that human cytomegalovirus (HCMV) displays a unique, extended nuclear translocation pattern in monocytes. We determined that c-Src signaling through pentamer engagement of integrins is required upon HCMV entry to avoid sorting of the virus into late endosomes and subsequent degradation. To follow up on this previous study, we designed experiments to investigate how HCMV-induced signaling through the other major axis-the epidermal growth factor receptor (EGFR) kinase-regulates viral postentry events. Here we show that HCMV induces chronic and functional EGFR signaling that is distinct to the virus as compared to the natural EGFR ligand: EGF. This chronic EGFR kinase activity in infected monocytes is required for the proper subcellular localization of the viral particle during trafficking events, as well as for promoting translocation of viral DNA into the host nucleus. Our data indicate that HCMV glycoprotein B (gB) binds to EGFR at the monocyte surface, the virus and EGFR are internalized together, and gB remains bound to EGFR throughout viral postentry events until de-envelopment to promote the chronic EGFR kinase activity required for viral trafficking and nuclear translocation. These data highlight how initial EGFR signaling via viral binding is necessary for entry, but not sufficient to promote each viral trafficking event. HCMV appears to manipulate the EGFR kinase postentry, via gB-EGFR interaction, to be active at the critical points throughout the trafficking process that leads to nuclear translocation and productive infection of peripheral blood monocytes.


Subject(s)
Cell Nucleus/metabolism , Cytomegalovirus/physiology , Monocytes/virology , Viral Envelope Proteins/metabolism , Cell Nucleus/virology , Cells, Cultured , DNA, Viral/metabolism , Endosomes/metabolism , Endosomes/virology , Epidermal Growth Factor/metabolism , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/metabolism , Host-Pathogen Interactions , Humans , Monocytes/metabolism , Protein Binding , Signal Transduction , trans-Golgi Network/metabolism , trans-Golgi Network/virology
6.
Viruses ; 10(10)2018 09 29.
Article in English | MEDLINE | ID: mdl-30274264

ABSTRACT

Human cytomegalovirus (HCMV) infection of peripheral blood monocytes plays a key role in the hematogenous dissemination of the virus to multiple organ systems following primary infection or reactivation of latent virus in the bone marrow. Monocytes have a short life span of 1⁻3 days in circulation; thus, HCMV must alter their survival and differentiation to utilize these cells and their differentiated counterparts-macrophages-for dissemination and long term viral persistence. Because monocytes are not initially permissive for viral gene expression and replication, HCMV must control host-derived factors early during infection to prevent apoptosis or programmed cell death prior to viral induced differentiation into naturally long-lived macrophages. This review provides a short overview of HCMV infection of monocytes and describes how HCMV has evolved to utilize host cell anti-apoptotic pathways to allow infected monocytes to bridge the 48⁻72 h viability gate so that differentiation into a long term stable mature cell can occur. Because viral gene expression is delayed in monocytes following initial infection and only occurs (begins around two to three weeks post infection in our model) following what appears to be complete differentiation into mature macrophages or dendritic cells, or both; virally-encoded anti-apoptotic gene products cannot initially control long term infected cell survival. Anti-apoptotic viral genes are discussed in the second section of this review and we argue they would play an important role in long term macrophage or dendritic cell survival following infection-induced differentiation.


Subject(s)
Apoptosis , Cytomegalovirus Infections/virology , Cytomegalovirus/immunology , Gene Expression Regulation, Viral/genetics , Monocytes/virology , Viral Proteins/immunology , Cell Differentiation , Cell Survival , Cytomegalovirus/genetics , Humans , Macrophages/immunology , Macrophages/virology , Monocytes/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...