1
|
Lankina A, Raposo M, Hargreaves A, Atkinson C, Griffiths P, Reeves MB. Developing a Vaccine Against Human Cytomegalovirus: Identifying and Targeting HCMV's Immunological Achilles' Heel. Vaccines (Basel) 2025; 13:435. [PMID: 40432047 PMCID: PMC12115399 DOI: 10.3390/vaccines13050435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Revised: 04/10/2025] [Accepted: 04/18/2025] [Indexed: 05/29/2025] Open
Abstract
Human cytomegalovirus (HCMV) is a critical pathogen in immunocompromised populations, such as organ transplant recipients as well as congenitally infected neonates with immature immune systems. Despite decades of research and the growing financial burden associated with the management of HCMV, there is no licensed vaccine to date. In this review, we aim to outline the complexity of HCMV and the antigens it presents and the journey and challenges of developing an effective HCMV vaccine, as well as further highlight the recent analyses of the most successful vaccine candidate so far-gB/MF59.
Collapse
Affiliation(s)
- Anastasia Lankina
- Institute of Immunity and Transplantation, Division of Infection and Immunity, University College London, London NW3 2PP, UK; (A.L.); (M.R.); (A.H.); (P.G.)
| | - Marta Raposo
- Institute of Immunity and Transplantation, Division of Infection and Immunity, University College London, London NW3 2PP, UK; (A.L.); (M.R.); (A.H.); (P.G.)
| | - Alexander Hargreaves
- Institute of Immunity and Transplantation, Division of Infection and Immunity, University College London, London NW3 2PP, UK; (A.L.); (M.R.); (A.H.); (P.G.)
| | - Claire Atkinson
- School of Applied and Health Sciences, London South Bank University, London SE1 0AA, UK;
| | - Paul Griffiths
- Institute of Immunity and Transplantation, Division of Infection and Immunity, University College London, London NW3 2PP, UK; (A.L.); (M.R.); (A.H.); (P.G.)
| | - Matthew B. Reeves
- Institute of Immunity and Transplantation, Division of Infection and Immunity, University College London, London NW3 2PP, UK; (A.L.); (M.R.); (A.H.); (P.G.)
| |
Collapse
|
2
|
Lee BJ, Min CK, Hancock M, Streblow DN, Caposio P, Goodrum FD, Yurochko AD. Human Cytomegalovirus Host Interactions: EGFR and Host Cell Signaling Is a Point of Convergence Between Viral Infection and Functional Changes in Infected Cells. Front Microbiol 2021; 12:660901. [PMID: 34025614 PMCID: PMC8138183 DOI: 10.3389/fmicb.2021.660901] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 04/07/2021] [Indexed: 12/22/2022] Open
Abstract
Viruses have evolved diverse strategies to manipulate cellular signaling pathways in order to promote infection and/or persistence. Human cytomegalovirus (HCMV) possesses a number of unique properties that allow the virus to alter cellular events required for infection of a diverse array of host cell types and long-term persistence. Of specific importance is infection of bone marrow derived and myeloid lineage cells, such as peripheral blood monocytes and CD34+ hematopoietic progenitor cells (HPCs) because of their essential role in dissemination of the virus and for the establishment of latency. Viral induced signaling through the Epidermal Growth Factor Receptor (EGFR) and other receptors such as integrins are key control points for viral-induced cellular changes and productive and latent infection in host organ systems. This review will explore the current understanding of HCMV strategies utilized to hijack cellular signaling pathways, such as EGFR, to promote the wide-spread dissemination and the classic life-long herpesvirus persistence.
Collapse
Affiliation(s)
- Byeong-Jae Lee
- Department of Microbiology & Immunology, Center for Molecular and Tumor Virology, Louisiana State University Health Sciences Center Shreveport, Shreveport, LA, United States.,Center for Applied Immunology and Pathological Processes, Louisiana State University Health Sciences Center Shreveport, Shreveport, LA, United States.,Center of Excellence for Emerging Viral Threats, Louisiana State University Health Sciences Center Shreveport, Shreveport, LA, United States
| | - Chan-Ki Min
- Department of Microbiology & Immunology, Center for Molecular and Tumor Virology, Louisiana State University Health Sciences Center Shreveport, Shreveport, LA, United States.,Center for Applied Immunology and Pathological Processes, Louisiana State University Health Sciences Center Shreveport, Shreveport, LA, United States.,Center of Excellence for Emerging Viral Threats, Louisiana State University Health Sciences Center Shreveport, Shreveport, LA, United States
| | - Meaghan Hancock
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR, United States
| | - Daniel N Streblow
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR, United States
| | - Patrizia Caposio
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR, United States
| | | | - Andrew D Yurochko
- Department of Microbiology & Immunology, Center for Molecular and Tumor Virology, Louisiana State University Health Sciences Center Shreveport, Shreveport, LA, United States.,Feist-Weiller Cancer Center, Louisiana State University Health Sciences Center Shreveport, Shreveport, LA, United States.,Center for Cardiovascular Diseases and Sciences, Louisiana State University Health Sciences Center Shreveport, Shreveport, LA, United States.,Center of Excellence in Arthritis and Rheumatology, Louisiana State University Health Sciences Center Shreveport, Shreveport, LA, United States
| |
Collapse
|
3
|
A Luciferase Gene Driven by an Alphaherpesviral Promoter Also Responds to Immediate Early Antigens of the Betaherpesvirus HCMV, Allowing Comparative Analyses of Different Human Herpesviruses in One Reporter Cell Line. PLoS One 2017; 12:e0169580. [PMID: 28060895 PMCID: PMC5217978 DOI: 10.1371/journal.pone.0169580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Accepted: 12/19/2016] [Indexed: 11/19/2022] Open
Abstract
Widely used methods for quantification of human cytomegalovirus (HCMV) infection in cell culture such as immunoblotting or plaque reduction assays are generally restricted to low throughput and require time-consuming evaluation. Up to now, only few HCMV reporter cell lines have been generated to overcome these restrictions and they are afflicted with other limitations because permanently expandable cell lines are normally not fully permissive to HCMV. In this work, a previously existing epithelial cell line hosting a luciferase gene under control of a Varicella-zoster virus promoter was adopted to investigate HCMV infection. The cells were susceptible to different HCMV strains at infection efficiencies that corresponded to their respective degree of epithelial cell tropism. Expression of early and late viral antigens, formation of nuclear inclusions, release of infectious virus progeny, and focal growth indicated productive viral replication. However, viral release and spread occurred at lower levels than in primary cell lines which appears to be due to a malfunction of virion morphogenesis during the nuclear stage. Expression of the luciferase reporter gene was specifically induced in HCMV infected cultures as a function of the virus dose and dependent on viral immediate early gene expression. The level of reporter activity accurately reflected infection efficiencies as determined by viral antigen immunostaining, and hence could discriminate the cell tropism of the tested virus strains. As proof-of-principle, we demonstrate that this cell line is applicable to evaluate drug resistance of clinical HCMV isolates and the neutralization capacity of human sera, and that it allows comparative and simultaneous analysis of HCMV and human herpes simplex virus type 1. In summary, the permanent epithelial reporter cell line allows robust, rapid and objective quantitation of HCMV infection and it will be particularly useful in higher throughput analyses as well as in comparative analyses of different human herpesviruses.
Collapse
|
4
|
Krömmelbein N, Wiebusch L, Schiedner G, Büscher N, Sauer C, Florin L, Sehn E, Wolfrum U, Plachter B. Adenovirus E1A/E1B Transformed Amniotic Fluid Cells Support Human Cytomegalovirus Replication. Viruses 2016; 8:v8020037. [PMID: 26848680 PMCID: PMC4776192 DOI: 10.3390/v8020037] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Revised: 01/22/2016] [Accepted: 01/28/2016] [Indexed: 11/30/2022] Open
Abstract
The human cytomegalovirus (HCMV) replicates to high titers in primary human fibroblast cell cultures. A variety of primary human cells and some tumor-derived cell lines do also support permissive HCMV replication, yet at low levels. Cell lines established by transfection of the transforming functions of adenoviruses have been notoriously resistant to HCMV replication and progeny production. Here, we provide first-time evidence that a permanent cell line immortalized by adenovirus type 5 E1A and E1B (CAP) is supporting the full HCMV replication cycle and is releasing infectious progeny. The CAP cell line had previously been established from amniotic fluid cells which were likely derived from membranes of the developing fetus. These cells can be grown under serum-free conditions. HCMV efficiently penetrated CAP cells, expressed its immediate-early proteins and dispersed restrictive PML-bodies. Viral DNA replication was initiated and viral progeny became detectable by electron microscopy in CAP cells. Furthermore, infectious virus was released from CAP cells, yet to lower levels compared to fibroblasts. Subviral dense bodies were also secreted from CAP cells. The results show that E1A/E1B expression in transformed cells is not generally repressive to HCMV replication and that CAP cells may be a good substrate for dense body based vaccine production.
Collapse
Affiliation(s)
- Natascha Krömmelbein
- Institute for Virology, University Medical Center of the Johannes Gutenberg-University Mainz, D-55131 Mainz, Germany.
| | - Lüder Wiebusch
- Department of Pediatric Molecular Biology, Charité University Medical Centre Berlin, D-10117 Berlin, Germany.
| | | | - Nicole Büscher
- Institute for Virology, University Medical Center of the Johannes Gutenberg-University Mainz, D-55131 Mainz, Germany.
| | - Caroline Sauer
- Institute for Virology, University Medical Center of the Johannes Gutenberg-University Mainz, D-55131 Mainz, Germany.
| | - Luise Florin
- Institute for Medical Microbiology and Hygiene, University Medical Center of the Johannes Gutenberg-University Mainz, D-55131 Mainz, Germany.
| | - Elisabeth Sehn
- Institute for Zoology, Johannes Gutenberg-University Mainz, D-55099 Mainz, Germany.
| | - Uwe Wolfrum
- Institute for Zoology, Johannes Gutenberg-University Mainz, D-55099 Mainz, Germany.
| | - Bodo Plachter
- Institute for Virology, University Medical Center of the Johannes Gutenberg-University Mainz, D-55131 Mainz, Germany.
- Research Center for Immunotherapy, University Medical Center of the Johannes Gutenberg-University Mainz, D-55131 Mainz, Germany.
| |
Collapse
|
5
|
Muntasell A, Costa-Garcia M, Vera A, Marina-Garcia N, Kirschning CJ, López-Botet M. Priming of NK cell anti-viral effector mechanisms by direct recognition of human cytomegalovirus. Front Immunol 2013; 4:40. [PMID: 23440148 PMCID: PMC3578278 DOI: 10.3389/fimmu.2013.00040] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2012] [Accepted: 02/04/2013] [Indexed: 12/24/2022] Open
Abstract
Natural killer (NK) cells play an important role in the defense against viral infections. Activation of resting NK cells is tightly controlled by the balance of surface inhibitory and activating receptors and aided by cytokines released by accessory cells along the anti-viral response. On the other hand, NK cells express functional pattern recognition receptors (PRRs) whose function has been mostly addressed by the use of synthetic agonists. The present study was undertaken to investigate whether NK cells could directly recognize a complex pathogen such as Human Cytomegalovirus (HCMV). Exposure of primary human NK cells to HCMV (TB40/E strain) induced the expression of CD69, promoted IFNγ secretion, and increased their cytotoxic activity against HCMV-infected autologous monocyte-derived dendritic cells. The divergent response induced by infective and UV-inactivated virions indicated the involvement of different NK cell sensors in the recognition of HCMV. The fact that NK cell activation could be partially prevented by blocking mAb specific for IFNAR and TLR2, together with the induction of IFNβ mRNA, supported the involvement of IFNβ and TLR2 in the response to HCMV. Thus, our data indicate that simultaneous activation of several PRRs leads to the autonomous priming of NK cell effector functions and could be a previously unappreciated mechanism presumably contributing to the control of HCMV infection.
Collapse
Affiliation(s)
- Aura Muntasell
- Hospital del Mar Medical Research Institute Barcelona, Spain
| | | | | | | | | | | |
Collapse
|
6
|
Manley K, Anderson J, Yang F, Szustakowski J, Oakeley EJ, Compton T, Feire AL. Human cytomegalovirus escapes a naturally occurring neutralizing antibody by incorporating it into assembling virions. Cell Host Microbe 2012; 10:197-209. [PMID: 21925108 DOI: 10.1016/j.chom.2011.07.010] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2011] [Revised: 06/01/2011] [Accepted: 07/20/2011] [Indexed: 01/31/2023]
Abstract
Human cytomegalovirus (CMV) is a common but difficult to treat infection of immunocompromised patients. MSL-109 is a human monoclonal IgG isolated from a CMV seropositive individual that recognizes the viral glycoprotein H (gH) surface antigen complexes that mediate entry. Although MSL-109 blocks CMV infection in vitro, it lacked sufficient efficacy in human trials, and CMV isolated from treated patients suggested the evolution of MSL-109 resistance. To understand how CMV escapes MSL-109, we characterized a MSL-109-resistant CMV strain. Our results elucidate a nongenetic escape mechanism in which the antibody is selectively taken up by infected cells and incorporated into assembling virions in a dose-dependent manner. The resistant virus then utilizes the Fc domain of the incorporated antibody to infect naive nonimmune cells. This resistance mechanism may explain the clinical failure of MSL-109, illustrate a general mechanism of viral antibody escape, and inform antiviral vaccine and therapeutic development.
Collapse
Affiliation(s)
- Kate Manley
- Novartis Institutes for Biomedical Research, Cambridge, MA 02139, USA
| | | | | | | | | | | | | |
Collapse
|
7
|
Mercorelli B, Lembo D, Palù G, Loregian A. Early inhibitors of human cytomegalovirus: state-of-art and therapeutic perspectives. Pharmacol Ther 2011; 131:309-29. [PMID: 21570424 PMCID: PMC7112563 DOI: 10.1016/j.pharmthera.2011.04.007] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2011] [Accepted: 04/05/2011] [Indexed: 12/31/2022]
Abstract
Human cytomegalovirus (HCMV) infection is associated with severe morbidity and mortality in immunocompromised individuals, mainly transplant recipients and AIDS patients, and is the most frequent cause of congenital malformations in newborn children. To date, few drugs are licensed for the treatment of HCMV infections, most of which target the viral DNA polymerase and suffer from many drawbacks, including long-term toxicity, low potency, and poor bioavailability. In addition, the emergence of drug-resistant viral strains is becoming an increasing problem for disease management. Finally, none of the current anti-HCMV drugs have been approved for the treatment of congenital infections. For all these reasons, there is still a strong need for new anti-HCMV drugs with novel mechanisms of action. The first events of the virus replication cycle, including attachment, entry, immediate-early gene expression, and immediate-early functions—in particular that of Immediate-Early 2 protein—represent attractive targets for the development of novel antiviral compounds. Such inhibitors would block not only the expression of viral immediate-early proteins, which play a key role in the pathogenesis of HCMV infection, but also the host immunomodulation and the changes to cell physiology induced by the first events of virus infection. This review describes the current knowledge on the initial phases of HCMV replication, their validation as potential novel antiviral targets, and the development of compounds that block such processes.
Collapse
Affiliation(s)
- Beatrice Mercorelli
- Department of Histology, Microbiology and Medical Biotechnologies, University of Padova, 35121 Padova, Italy
| | | | | | | |
Collapse
|
8
|
Ibig-Rehm Y, Götte M, Gabriel D, Woodhall D, Shea A, Brown NE, Compton T, Feire AL. High-content screening to distinguish between attachment and post-attachment steps of human cytomegalovirus entry into fibroblasts and epithelial cells. Antiviral Res 2011; 89:246-56. [PMID: 21277329 DOI: 10.1016/j.antiviral.2011.01.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2010] [Revised: 01/13/2011] [Accepted: 01/21/2011] [Indexed: 11/17/2022]
Abstract
Human cytomegalovirus (HCMV) enters cells through a complex pathway involving the interaction of multiple viral glycoproteins and cellular receptors. While HCMV clinical isolates enter a wide range of cell types, entry has historically been studied using a laboratory strain of virus that can only infect fibroblasts. Herein, we have constructed a HCMV reporter strain that contains GFP fused to the abundant tegument protein pp65 to allow for the direct visualization of virus attachment and entry. Furthermore, the UL131 gene of this strain was restored to clinical isolate sequence to expand our studies of entry into physiologically relevant epithelial cell types. Using the HCMV-GFP reporter virus, we developed an image-based assay and screened a library containing 65,000 compounds for the inhibition of virus entry into fibroblasts. In addition to assessing the effect on virus entry, automated image analysis provided information on compound toxicity and whether the compounds acted as attachment or post-attachment inhibitors. To identify therapeutically viable inhibitors capable of blocking entry in multiple cell types, the inhibitors were screened further for their ability to inhibit virus entry into epithelial cells. Compounds were identified that were able to inhibit virus entry into both cell types at either attachment or post-attachment steps.
Collapse
Affiliation(s)
- Yvonne Ibig-Rehm
- Lead Finding Platform, Novartis Institutes for Biomedical Research, Novartis Campus, Basle, Switzerland
| | | | | | | | | | | | | | | |
Collapse
|
9
|
Real-time analysis of antibody interactions with whole enveloped human cytomegalovirus using surface plasmon resonance. Anal Biochem 2010; 411:58-63. [PMID: 21167121 DOI: 10.1016/j.ab.2010.12.017] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2010] [Revised: 12/04/2010] [Accepted: 12/08/2010] [Indexed: 11/24/2022]
Abstract
Human cytomegalovirus (CMV) is a large enveloped virus that encodes multiple glycoproteins required for virus-cell binding and fusion. To assess the binding properties of antibodies with target glycoprotein in a natural context of infection, we investigated the feasibility of using the surface plasmon resonance (SPR) technique for studying the direct binding of antibodies with CMV virions. Direct immobilization of whole virions to sensor surface and a surface regeneration procedure allowed for quantitative and reproducible measurements of binding affinity and binding kinetics of antibody-whole virion interactions. The conformational and functional integrity of viral particles was not compromised by the regeneration condition as evaluated with antibodies recognizing conformational epitopes and by electron microscopy. Binding of an irrelevant antibody was not observed, indicating the high specificity of the method. A panel of anti-gB antibodies was measured and the binding affinities correlated fairly well with those determined by ELISA. These data demonstrated that the interaction of anti-gB antibody with whole virion of large enveloped CMV can be quantitatively studied using SPR. This method has been successfully applied for screening and selection of anti-CMV antibodies and can be potentially extended to study antibody-glycoprotein interactions of other related herpesviruses.
Collapse
|
10
|
The glycoprotein B disintegrin-like domain binds beta 1 integrin to mediate cytomegalovirus entry. J Virol 2010; 84:10026-37. [PMID: 20660204 DOI: 10.1128/jvi.00710-10] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Cellular integrins were identified as human cytomegalovirus (HCMV) entry receptors and signaling mediators in both fibroblasts and endothelial cells. The goal of these studies was to determine the mechanism by which HCMV binds to cellular integrins to mediate virus entry. HCMV envelope glycoprotein B (gB) has sequence similarity to the integrin-binding disintegrin-like domain found in the ADAM (a disintegrin and metalloprotease) family of proteins. To test the ability of this region to bind to cellular integrins, we generated a recombinant soluble version of the gB disintegrin-like domain (gB-DLD). The gB-DLD protein bound to human fibroblasts in a specific, dose-dependent and saturable manner that required the expression of an intact beta1 integrin ectodomain. Furthermore, a physical association between gB-DLD and beta1 integrin was demonstrated through in vitro pull-down assays. The function of this interaction was shown by the ability of cell-bound gB-DLD to efficiently block HCMV entry and the infectivity of multiple in vivo target cells. Additionally, rabbit polyclonal antibodies raised against gB-DLD neutralized HCMV infection. Mimicry of the ADAM family disintegrin-like domain by HCMV gB represents a novel mechanism for integrin engagement by a virus and reveals a unique therapeutic target for HCMV neutralization. The strong conservation of the DLD across beta- and gammaherpesviruses suggests that integrin recognition and utilization may be a more broadly conserved feature throughout the Herpesviridae.
Collapse
|
11
|
Ueno T, Ogawa-Goto K. Use of a GFP-PML-expressing cell line as a biosensor for human cytomegalovirus infection. Methods Mol Biol 2009; 515:33-44. [PMID: 19378119 DOI: 10.1007/978-1-59745-559-6_3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
Human cytomegalovirus (HCMV) infection has a marked effect on promyelocytic leukemia (PML) bodies. Here, we describe a novel real-time monitoring system for HCMV-infected cells in vitro using a newly established cell line that stably expresses GFP-PML protein. Upon infection, HCMV causes specific dispersion of GFP-PML bodies, thereby allowing the infected cells to be monitored by fluorescence microscopy without immunostaining. Quantitative protocols using either an NPB fluorescence assay or a GFP-PML imaging assay are also described. The NPB fluorescence assay is rapid, sensitive, and sufficiently simple for screening of inhibitory reagents, while the GFP-PML imaging assay is highly sensitive and applicable to drug susceptibility testing of low-titer clinical isolates.
Collapse
Affiliation(s)
- Tomonori Ueno
- Nippi Research Institute of Biomatrix, Toride, Ibaraki, Japan
| | | |
Collapse
|
12
|
Abstract
Human cytomegalovirus (HCMV) exhibits an exceptionally broad cellular tropism as it is capable of infecting most major organ systems and cell types. Definitive proof of an essential role for a cellular molecule that serves as an entry receptor has proven very challenging. It is widely hypothesized that receptor utilization, envelope glycoprotein requirements and entry pathways may all vary according to cell type, which is partially supported by the data. What has clearly emerged in recent years is that virus entry is not going undetected by the host. Robust and rapid induction of innate immune response is intimately associated with entry-related events. Here we review the state of knowledge on HCMV cellular entry mediators confronting the scientific challenges by accruing a definitive data set. We also review the roles of pattern recognition receptors such as Toll-like receptors in activation of specific innate immune response and discuss how entry events are tightly coordinated with innate immune initiation steps.
Collapse
|
13
|
Isaacson MK, Feire AL, Compton T. Epidermal growth factor receptor is not required for human cytomegalovirus entry or signaling. J Virol 2007; 81:6241-7. [PMID: 17428848 PMCID: PMC1900073 DOI: 10.1128/jvi.00169-07] [Citation(s) in RCA: 91] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Human cytomegalovirus (HCMV) can bind, fuse, and initiate gene expression in a diverse range of vertebrate cell types. This broad cellular tropism suggests that multiple receptors and/or universally distributed receptors mediate HCMV entry. Our laboratory has recently discovered that certain beta1 and beta3 integrin heterodimers are critical mediators of HCMV entry into permissive fibroblasts (A. L. Feire, H. Koss, and T. Compton, Proc. Natl. Acad. Sci. USA 101:15470-15475, 2004). It has also been reported that epidermal growth factor receptor (EGFR) is necessary for HCMV-mediated signaling and entry (X. Wang, S. M. Huong, M. L. Chiu, N. Raab-Traub, and E. E. Huang, Nature 424:456-461, 2003). Integrins are known to signal synergistically with growth factor receptors, and this coordination was recently reported for EGFR and beta3 integrins in the context of HCMV entry (X. Wang, D. Y. Huang, S. M. Huong, and E. S. Huang, Nat. Med. 11:515-521, 2005). However, EGFR-negative cell lines, such as hematopoietic cells, are known to be infected by HCMV. Therefore, we wished to confirm a role for EGFR in HCMV entry and then examine any interaction between beta1 integrins and EGFR during the entry process. Surprisingly, we were unable to detect any role for EGFR in the process of HCMV entry into fibroblast, epithelial, or endothelial cell lines. Additionally, HCMV did not activate the EGFR kinase in fibroblast cell lines. We first examined HCMV entry into two EGFR-positive or -negative cell lines but observed no increase in entry when EGFR was expressed to high levels. Physically blocking EGFR with a neutralizing antibody in fibroblast, epithelial, or endothelial cell lines or blocking EGFR kinase signaling with a chemical inhibitor in fibroblast cells did not inhibit virus entry. Lastly, we were unable to detect phosphorylation of EGFR in fibroblasts cells in response to HCMV stimulation. Our findings demonstrate that EGFR does not play a significant role in HCMV entry or signaling. These results suggest that specific integrin heterodimers either act alone as the primary entry receptors or interact in conjunction with an additional receptor(s), other than EGFR, to facilitate virus entry.
Collapse
Affiliation(s)
- Marisa K Isaacson
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison Medical School 53706, USA
| | | | | |
Collapse
|
14
|
Derry MC, Sutherland MR, Restall CM, Waisman DM, Pryzdial ELG. Annexin 2-mediated enhancement of cytomegalovirus infection opposes inhibition by annexin 1 or annexin 5. J Gen Virol 2007; 88:19-27. [PMID: 17170432 DOI: 10.1099/vir.0.82294-0] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Biochemical studies have suggested that annexin 2 (A2) may participate in cytomegalovirus (CMV) infection. In the current work, effects of A2 monomer (p36) and heterotetramer (A2t; p36(2)p11(2)) were investigated. Demonstrating a role for endogenous A2, the four stages of infection that were followed were each inhibited by anti-p36 or anti-p11 at 37 degrees C. Immuno-inhibition was attenuated when the virus and cells were pre-incubated at 4 degrees C to coordinate virus entry initiated afterwards at 37 degrees C, reconciling controversy in the literature. As an explanation, CMV-induced phosphorylation of p36 was prevented by the 4 degrees C treatment. Supporting these immuno-inhibition data, purified A2t or p11 increased CMV infectious-progeny generation and CMV gene expression. A specific role for A2t was indicated by purified p36 having no effect. Unlike other steps, primary plaque formation was not enhanced by purified A2t or p11, possibly because of undetectable phosphorylation. As annexins 1 (A1) and 5 (A5) interact with A2, their effect on CMV was also tested. Both purified proteins inhibited CMV infection. In each experiment, the concentration of A1 required for half-maximal inhibition was five- to 10-fold lower than that of A5. Addition of A2 opposed A1- or A5-mediated inhibition of CMV, as did certain A2-specific antibodies that had no effect in the absence of added A1 or A5. Transfection of the p36-deficient cell line HepG2 increased CMV infection and was required for inhibition by the other annexins. These data suggest that CMV exploits A2t at physiological temperature to oppose the protection of cells conferred by A1 or A5.
Collapse
Affiliation(s)
- Mélanie C Derry
- Department of Pathology and Laboratory Medicine, University of British Columbia/Centre for Blood Research, 2350 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada
- Research and Development Department, Canadian Blood Services, Ottawa, ON, Canada
| | - Michael R Sutherland
- Department of Pathology and Laboratory Medicine, University of British Columbia/Centre for Blood Research, 2350 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada
- Research and Development Department, Canadian Blood Services, Ottawa, ON, Canada
| | - Christina M Restall
- Research and Development Department, Canadian Blood Services, Ottawa, ON, Canada
| | - David M Waisman
- Department of Medical Biochemistry, University of Calgary, Calgary, AB, Canada
| | - Edward L G Pryzdial
- Department of Pathology and Laboratory Medicine, University of British Columbia/Centre for Blood Research, 2350 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada
- Research and Development Department, Canadian Blood Services, Ottawa, ON, Canada
| |
Collapse
|
15
|
Ueno T, Eizuru Y, Katano H, Kurata T, Sata T, Irie S, Ogawa-Goto K. Novel real-time monitoring system for human cytomegalovirus-infected cells in vitro that uses a green fluorescent protein-PML-expressing cell line. Antimicrob Agents Chemother 2006; 50:2806-13. [PMID: 16870775 PMCID: PMC1538688 DOI: 10.1128/aac.01641-05] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Promyelocytic leukemia (PML) bodies are discrete nuclear foci that are intimately associated with many DNA viruses. In human cytomegalovirus (HCMV) infection, the IE1 (for "immediate-early 1") protein has a marked effect on PML bodies via de-SUMOylation of PML protein. Here, we report a novel real-time monitoring system for HCMV-infected cells using a newly established cell line (SE/15) that stably expresses green fluorescent protein (GFP)-PML protein. In SE/15 cells, HCMV infection causes specific and efficient dispersion of GFP-PML bodies in an IE1-dependent manner, allowing the infected cells to be monitored by fluorescence microscopy without immunostaining. Since a specific change in the detergent solubility of GFP-PML occurs upon infection, the infected cells can be quantified by GFP fluorescence measurement after extraction. With this assay, the inhibitory effects of heparin and neutralizing antibodies were determined in small-scale cultures, indicating its usefulness for screening inhibitory reagents for laboratory virus strains. Furthermore, we established a sensitive imaging assay by counting the number of nuclei containing dispersed GFP-PML, which is applicable for titration of slow-growing clinical isolates. In all strains tested, the virus titers estimated by the GFP-PML imaging assay were well correlated with the plaque-forming cell numbers determined in human embryonic lung cells. Coculture of SE/15 cells and HCMV-infected fibroblasts permitted a rapid and reliable method for estimating the 50% inhibitory concentration values of drugs for clinical isolates in susceptibility testing. Taken together, these results demonstrate the development of a rapid, sensitive, quantitative, and specific detection system for HCMV-infected cells involving a simple procedure that can be used for titration of low-titer clinical isolates.
Collapse
Affiliation(s)
- T Ueno
- Nippi Research Institute of Biomatrix, Adachi, Tokyo 120-8601, Japan
| | | | | | | | | | | | | |
Collapse
|
16
|
Feire AL, Koss H, Compton T. Cellular integrins function as entry receptors for human cytomegalovirus via a highly conserved disintegrin-like domain. Proc Natl Acad Sci U S A 2004; 101:15470-5. [PMID: 15494436 PMCID: PMC524452 DOI: 10.1073/pnas.0406821101] [Citation(s) in RCA: 251] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Human cytomegalovirus (HCMV) is capable of manifesting disease in nearly every organ system in immunocompromised patients. This broad pathogenic tropism correlates with the ability of the virus to infect all tested vertebrate cell types in vitro, a characteristic that has made receptor identification extremely difficult. During virus entry, HCMV induces cellular morphological changes and signaling cascades consistent with engagement of cellular integrins; however, HCMV structural proteins do not possess the widely used RGD integrin-binding motif. We identified an integrin-binding disintegrin-like domain within HCMV envelope glycoprotein B, a protein required for virus entry and fusion throughout the Herpesviridae. Accepted receptor criteria are met through the use of function-blocking integrin Abs, beta1 integrin knockout mouse fibroblasts, and glycoprotein B disintegrin-like peptides, all of which support a critical role for alpha2beta1, alpha6beta1, and alphaVbeta3 integrins as HCMV entry receptors and signaling mediators acting during the penetration stage of the entry pathway. Strikingly, the glycoprotein B disintegrin-like domain is conserved in many human and animal herpesviruses, suggesting that integrins may support entry across this medically important virus family.
Collapse
Affiliation(s)
- Adam L Feire
- McArdle Laboratory for Cancer Research, University of Wisconsin, Madison, WI 53706, USA
| | | | | |
Collapse
|
17
|
Abstract
The ability of human cytomegalovirus (HCMV) to infect an extensive range of cell types has complicated efforts to identify cellular receptors for this significant pathogen. Recent findings demonstrate that epidermal growth factor receptor (EGFR) serves also as a receptor for HCMV. Additional evidence has shown that HCMV entry occurs in concert with immune detection through toll-like receptors. Here, the implications of EGFR activation, the existence of other receptors and the coordination of entry with the innate sensing are discussed.
Collapse
Affiliation(s)
- Teresa Compton
- McArdle Laboratory for Cancer Research, 1400 University Avenue, Madison, WI 53706, USA.
| |
Collapse
|
18
|
Kawasaki H, Tsutsui Y. Brain slice culture for analysis of developmental brain disorders with special reference to congenital cytomegalovirus infection. Congenit Anom (Kyoto) 2003; 43:105-13. [PMID: 12893969 DOI: 10.1111/j.1741-4520.2003.tb01034.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Cytomegalovirus (CMV) is the most significant infectious cause of congenital abnormalities of the central nervous system (CNS) with variation from the fatal cytomegalic inclusion disease to functional brain disorder. The phenotype and degree of the brain disorder depends on infection time during the developing stage, virulence, route of infection and the viral susceptibility of the cells. The pathogenesis of the CMV infection to the CNS seems to be strongly related to neural migration, neural death, cellular compositions and the immune system of the brain. To understand the complex mechanism of this disorder, we used organotypic brain slice cultures. In the brain slice culture system, migration of CMV-infected neuronal cells was observed, which reflects infectious dynamics in vivo. Neural progenitor cells or glial immature cells in the subventricular zone and marginal area are most susceptible to murine cytomegalovirus (MCMV) infection in this system. The susceptibility declined as the number of immature glial cells decreased with age. The immature glial cells proliferated in brain slice cultures during prolonged incubation, and the susceptibility to MCMV infection also increased in association with the proliferation of these cells. The brain slice from an immunocompromised mouse (Beige-SCID mouse) unexpectedly showed lower susceptibility than that of an immunocompetent mouse during any prolonged incubation. These results suggest that the number of immature glial cells might determine the susceptibility of CMV infection to the brain, independent of the immune system. We reviewed recent findings of CMV infection to the brain from the perspective of brain slice cultures and the possibility that this system could be a useful method to investigate mechanisms of congenital anomaly of the brain.
Collapse
Affiliation(s)
- Hideya Kawasaki
- Second Department of Pathology, Hamamatsu University School of Medicine, Hamamatsu 431-3192, Japan.
| | | |
Collapse
|
19
|
Kawasaki H, Kosugi I, Arai Y, Tsutsui Y. The amount of immature glial cells in organotypic brain slices determines the susceptibility to murine cytomegalovirus infection. J Transl Med 2002; 82:1347-58. [PMID: 12379769 DOI: 10.1097/01.lab.0000032376.58688.d4] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Cytomegalovirus (CMV) is the most common infectious cause of congenital anomalies of the brain and also causes brain damage in immunocompromised individuals. We investigated the effects of murine cytomegalovirus (MCMV) infection on the developing mouse brain in terms of susceptible cells and age-related resistance to MCMV in brain slice cultures. Brain slices from BALB/c mice at different developmental stages were infected with recombinant MCMV in which the lacZ gene was inserted into a late gene. The subventricular zone and cortical marginal region were the sites most susceptible to MCMV infection, and the susceptibility declined with the development of the brain. Immunohistochemical staining showed that the virus-susceptible cells were positive for GFAP, nestin, and Musashi-1, and that most of the infected cells were positive for the proliferative cell nuclear antigen and labeled with bromodeoxyuridine. These results suggest that the susceptible cells in the subventricular zone are immature glial cells, including neural progenitor cells. Immature glial cells proliferated when the brain slices were cultured for a prolonged time and furthermore, they showed themselves to be susceptible to virus infection even under serum-free conditions. These results suggest that the amount of immature glial cells, which include neural progenitor cells, in the developing brain or in the damaged brain with neural proliferation may be closely associated with the susceptibility of the brain to CMV infection in humans.
Collapse
Affiliation(s)
- Hideya Kawasaki
- Second Department of Pathology, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | | | | | | |
Collapse
|
20
|
Huber MT, Tomazin R, Wisner T, Boname J, Johnson DC. Human cytomegalovirus US7, US8, US9, and US10 are cytoplasmic glycoproteins, not found at cell surfaces, and US9 does not mediate cell-to-cell spread. J Virol 2002; 76:5748-58. [PMID: 11992003 PMCID: PMC137039 DOI: 10.1128/jvi.76.11.5748-5758.2002] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Human cytomegalovirus (HCMV) expresses a large number of membrane proteins with unknown functions. One class of these membrane proteins apparently acts to allow HCMV to escape detection by the immune system. The best characterized of these are the glycoproteins encoded within the US2 to US11 region of the HCMV genome that mediate resistance to CD8(+) and CD4(+) T cells. US2, US3, US6, and US11 block various aspects of the major histocompatibility complex (MHC) class I and class II antigen presentation pathways, functioning in cytoplasmic membranes to cause retention, degradation, or mislocalization of MHC proteins. Distantly homologous genes in this region, US7, US8, US9, and US10, are not well characterized. Here, we report expression of the glycoproteins encoded by US7 to US10 by using replication-defective adenovirus (Ad) vectors. US7, US9, and US10 remained sensitive to endoglycosidase H and were exclusively or largely present in the endoplasmic reticulum (ER) as determined by confocal microscopy. US8 reached the Golgi apparatus and trans-Golgi network and was more quickly degraded. Previous studies suggested that US9 could localize to cell junctions and mediate cell-to-cell spread in ARPE-19 retinal epithelial cells. We found no evidence of US9 at cell junctions of HEC-1A epithelial cells. HCMV recombinants lacking US9 produced smaller plaques on ARPE-19 cell monolayers but also exhibited defects in virus replication compared with wild-type HCMV in these cells. Other HCMV recombinants constructed in a similar fashion that were able to express US9 also produced small plaques and some of these exhibited defects in production of infectious progeny in ARPE-19 cells. Thus, there was no correlation between defects in cell-to-cell spread (plaque size) and loss of expression of US9, and it is possible that US9(-) mutants produce smaller plaques because they produce fewer progeny. Together, our results do not support the hypothesis that US9 plays a direct role in HCMV cell-to-cell spread.
Collapse
Affiliation(s)
- Mary T Huber
- Department of Molecular Microbiology and Immunology, Oregon Health Sciences University, Portland, Oregon 97201, USA
| | | | | | | | | |
Collapse
|
21
|
Ogawa-Goto K, Irie S, Omori A, Miura Y, Katano H, Hasegawa H, Kurata T, Sata T, Arao Y. An endoplasmic reticulum protein, p180, is highly expressed in human cytomegalovirus-permissive cells and interacts with the tegument protein encoded by UL48. J Virol 2002; 76:2350-62. [PMID: 11836413 PMCID: PMC153829 DOI: 10.1128/jvi.76.5.2350-2362.2002] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
We have used a virus overlay assay to detect cellular proteins associated with human cytomegalovirus (HCMV) particles. The radiolabeled HCMV particles specifically bound to two host proteins with molecular sizes of 150 and 180 kDa. By a micro-amino-acid sequencing technique, the 180-kDa protein was identified as a human homologue of the ES130/p180 ribosome receptor (p180), which is an integral endoplasmic reticulum (ER) membrane protein possessing a very unique tandem repeat domain at its N-terminal region. The virus overlay assay using truncated p180 polypeptides revealed that HCMV binding to human p180 occurred through the N-terminal region. In HCMV-permissive cells the high level of expression of the human p180 protein was clearly observed regardless of cell type. Furthermore, we showed that p180 binds to the UL48 gene product, which is one of the predominant tegument proteins of HCMV and which is considered to be tightly associated with the capsid. The interaction between the two proteins was assumed to be specific and was observed both in vitro and in vivo. During the late phase of infection, the unique relocation of human p180 was observed, that is, to the juxtanuclear region, which appeared to be in the vicinity of the area where naked virions were frequently observed in an electron-microscopic study. Thus our data suggest that p180 interacts with the HCMV tegument, at least through pUL48, during the HCMV replication process. We discuss the possible role of the interaction between p180 and pUL48 in the intracellular transport of HCMV virions.
Collapse
Affiliation(s)
- K Ogawa-Goto
- Department of Pathology, National Institute of Infectious Diseases, Toyama 1-23-1, Shinjuku, Tokyo 162-8640, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Theiler RN, Compton T. Characterization of the signal peptide processing and membrane association of human cytomegalovirus glycoprotein O. J Biol Chem 2001; 276:39226-31. [PMID: 11504733 DOI: 10.1074/jbc.m106300200] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Human cytomegalovirus (HCMV) has a structurally complex envelope that contains multiple glycoproteins. These glycoproteins are involved in virus entry, virus maturation, and cell-cell spread of infection. Glycoprotein H (gH), glycoprotein L (gL), and glycoprotein O (gO) associate covalently to form a unique disulfide-bonded tripartite complex. Glycoprotein O was recently discovered, and its basic structure, as well as that of the tripartite complex, remains uncharacterized. Based on hydropathy analysis, we hypothesized that gO could adopt a type II transmembrane orientation. The data presented here, however, reveal that the single hydrophobic domain of gO functions as a cleavable signal peptide that is absent from the mature molecule. Although it lacks a membrane anchor, glycoprotein O is associated with the membranes of HCMV-infected cells. The sophisticated organization of the gH.gL.gO complex reflects the intricate nature of the multicomponent entry and fusion machinery encoded by HCMV.
Collapse
Affiliation(s)
- R N Theiler
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison Medical School, Madison, Wisconsin 53706, USA
| | | |
Collapse
|
23
|
Esclatine A, Bellon A, Michelson S, Servin AL, Quéro AM, Géniteau-Legendre M. Differentiation-dependent redistribution of heparan sulfate in epithelial intestinal Caco-2 cells leads to basolateral entry of cytomegalovirus. Virology 2001; 289:23-33. [PMID: 11601914 DOI: 10.1006/viro.2001.1122] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Human cytomegalovirus (HCMV) causes a broad spectrum of clinical manifestations in immunocompromised patients, including infection of the gastrointestinal tract. To investigate the role of epithelial cells in the gastrointestinal HCMV disease, we used the intestinal epithelial cell line Caco-2, which is permissive for HCMV replication. In differentiated Caco-2 cells, we showed previously that HCMV infection proceeds preferentially from the basolateral membrane, suggesting that receptors for HCMV may be contained predominantly in the basolateral membrane (A. Esclatine et al., 2000, J. Virol. 74, 513-517). Therefore, we examined expression and localization in Caco-2 cells of heparan sulfate (HS) proteoglycan and annexin II, previously implicated in initial events of HCMV infection. We observed that annexin II is expressed in Caco-2 cells, but is not essential for entry of HCMV. We showed that, during the differentiation process, HS, initially present on the entire surface of the membrane of undifferentiated cells, ultimately became sequestered at the basolateral cell surface of fully differentiated cells. We established by biochemical assays that membrane-associated HS proteoglycan mediates both viral attachment to, and subsequent infection of, Caco-2 cells, regardless of the cell differentiation state. Thus, the redistribution of HS is implicated in the basolateral entry of HCMV into differentiated Caco-2 cells.
Collapse
Affiliation(s)
- A Esclatine
- Institut National de la Santé et de la Recherche Médicale, Unité 510, Pathogènes et Fonctions des Cellules Epithéliales Polarisées, Faculté de Pharmacie, Université Paris XI, 92296 Châtenay-Malabry Cedex, France.
| | | | | | | | | | | |
Collapse
|
24
|
García-Ramírez JJ, Ruchti F, Huang H, Simmen K, Angulo A, Ghazal P. Dominance of virus over host factors in cross-species activation of human cytomegalovirus early gene expression. J Virol 2001; 75:26-35. [PMID: 11119570 PMCID: PMC113894 DOI: 10.1128/jvi.75.1.26-35.2001] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Human cytomegalovirus (HCMV) exhibits a highly restricted host range. In this study, we sought to examine the relative significance of host and viral factors in activating early gene expression of the HCMV UL54 (DNA polymerase) promoter in murine cells. Appropriate activation of the UL54 promoter at early times is essential for viral DNA replication. To study how the HCMV UL54 promoter is activated in murine cells, a transgenesis system based on yeast artificial chromosomes (YACs) was established for HCMV. A 178-kb YAC, containing a subgenomic fragment of HCMV encompassing the majority of the unique long (UL) region, was constructed by homologous recombination in yeast. This HCMV YAC backbone is defective for viral growth and lacks the major immediate-early (IE) gene region, thus permitting the analysis of essential cis-acting sequences when complemented in trans. To quantitatively measure the level of gene expression, we generated HCMV YACs containing a luciferase reporter gene inserted downstream of either the UL54 promoter or, as a control for late gene expression, the UL86 promoter, which directs expression of the major capsid protein. To determine the early gene activation pathway, point mutations were introduced into the inverted repeat 1 (IR1) element of the UL54 promoter of the HCMV YAC. In the transgenesis experiments, HCMV YACs and derivatives generated in yeast were introduced into NIH 3T3 murine cells by polyethylene glycol-mediated fusion. We found that infection of YAC, but not plasmid, transgenic lines with HCMV was sufficient to fully recapitulate the UL54 expression program at early times of infection, indicating the importance of remote regulatory elements in influencing regulation of the UL54 promoter. Moreover, YACs containing a mutant IR1 in the UL54 promoter led to reduced ( approximately 30-fold) reporter gene expression levels, indicating that HCMV major IE gene activation of the UL54 promoter is fully permissive in murine cells. In comparison with HCMV, infection of YAC transgenic NIH 3T3 lines with murine cytomegalovirus (MCMV) resulted in lower (more than one order of magnitude) efficiency in activating UL54 early gene expression. MCMV is therefore not able to fully activate HCMV early gene expression, indicating the significance of virus over host determinants in the cross-species activation of key early gene promoters. Finally, these studies show that YAC transgenesis can be a useful tool in functional analysis of viral proteins and control of gene expression for large viral genomes.
Collapse
Affiliation(s)
- J J García-Ramírez
- Departments of Immunology and Molecular Biology, Division of Virology, The Scripps Research Institute, La Jolla, California 92037, USA
| | | | | | | | | | | |
Collapse
|
25
|
Raynor CM, Wright JF, Waisman DM, Pryzdial EL. Annexin II enhances cytomegalovirus binding and fusion to phospholipid membranes. Biochemistry 1999; 38:5089-95. [PMID: 10213612 DOI: 10.1021/bi982095b] [Citation(s) in RCA: 61] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
A number of studies have suggested that the anionic phospholipid (anPL)-binding protein annexin II may play a role in cytomegalovirus (CMV) infection. Since annexin II has been shown to mediate aggregation and fusion of certain membranes, we investigated whether these properties could be exploited by CMV directly. The experiments showed that purified annexin II, but not the homologous protein annexin V (AnV), can mediate the binding of 35S-CMV (strain AD169) to anPL-coated microtiter wells. This association required Ca2+, could be titrated by varying either annexin II (apparent Kd = 4 x 10(-)8 M) or 35S-CMV, was inhibited by unlabeled CMV, and was observed for the heterotetrameric or monomeric form of annexin II. In experiments utilizing the fluorescence dequenching of octadecyl rhodamine incorporated into the CMV envelope, annexin II was furthermore found to enhance the rate of virus-anPL vesicle fusion. The observed fusion was dependent on the concentration of annexin II, Ca2+, and anPL and was mediated principally by the heterotetramer. Interestingly, AnV was observed to inhibit the effects of annexin II on CMV fusion but not binding to anPL, which indicates that annexin II enhances these processes by distinct mechanisms. The results presented here provide the first direct evidence that annexin II has the capacity to bridge CMV to a phospholipid membrane and to enhance virus-membrane fusion. These observations furthermore suggest that AnV may regulate the fusogenic function of annexin II.
Collapse
Affiliation(s)
- C M Raynor
- Canadian Blood Services, R&D Department, University of Ottawa, Ontario, Canada
| | | | | | | |
Collapse
|
26
|
Kledal TN, Rosenkilde MM, Schwartz TW. Selective recognition of the membrane-bound CX3C chemokine, fractalkine, by the human cytomegalovirus-encoded broad-spectrum receptor US28. FEBS Lett 1998; 441:209-14. [PMID: 9883886 DOI: 10.1016/s0014-5793(98)01551-8] [Citation(s) in RCA: 159] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The 7TM receptor, US28, encoded by human cytomegalovirus binds a broad spectrum of endogenous CC chemokines with sub-nanomolar affinity as determined in homologous competition binding assays. We here find that US28 also recognizes the membrane-associated CX3C chemokine, fractalkine, with sub-nanomolar affinity (IC50=0.42+/-0.09 nM). Importantly, although fractalkine could compete with high affinity against the binding of CC chemokines, the secreted CC chemokines were only able to compete for binding against radioactive fractalkine with very low affinity. It is concluded that US28, which is known to enhance cell-cell fusion processes through interaction with an as yet unidentified, human cell-specific factor, has been optimized by cytomegalovirus to selectively recognize the membrane-associated fractalkine. It is suggested that US28 expressed on the surface of infected cells and possibly on the envelope of the virion is involved in transfer of the virus from cell to cell.
Collapse
Affiliation(s)
- T N Kledal
- Department of Pharmacology, The Panum Institute 18.6, Copenhagen, Denmark
| | | | | |
Collapse
|
27
|
Slobbe-van Drunen ME, Hendrickx AT, Vossen RC, Speel EJ, van Dam-Mieras MC, Bruggeman CA. Nuclear import as a barrier to infection of human umbilical vein endothelial cells by human cytomegalovirus strain AD169. Virus Res 1998; 56:149-56. [PMID: 9783463 DOI: 10.1016/s0168-1702(98)00064-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Human embryonal fibroblasts (HEF) are fully permissive for infection by human cytomegalovirus (HCMV) strain AD169, whereas human umbilical vein endothelial cells (HUVEC) seem to form an almost complete barrier to infection with this virus. To investigate this difference in permissiveness, HCMV infection of both cell types was studied using in situ hybridisation (ISH) as well as immunocytochemistry to detect viral DNA and viral proteins. At 2 h post-infection (p.i.), viral DNA was detected dispersed throughout the cytoplasm in both HEF and HUVEC, indicating that HCMV enters all cells of both cell types. At 4 h p.i., the viral DNA was found in the nucleus in HEF, and at the same time expression of immediate early (IE) antigen was found. In contrast, in HUVEC the expression of the IE proteins occurred in a limited number of cells at 8 h p.i., while in most HUVEC an accumulation of viral DNA around the nuclei was observed at this time point. In HUVEC, the nuclear localisation of viral DNA was detected 16 h p.i. in a minority of cells, indicating that transport of HCMV DNA into the nucleus is considerably slower in HUVEC than in HEF. Furthermore, the number of HUVEC containing HCMV DNA decreased about six-fold between 8 and 48 h p.i., indicating that HCMV DNA is either transported into the nucleus or eliminated. Apparently, the lower permissiveness of HUVEC for the HCMV strain AD169 relative to HEF is due to inefficient transport of HCMV DNA into the nuclei of infected HUVEC.
Collapse
|
28
|
Boyle KA, Compton T. Receptor-binding properties of a soluble form of human cytomegalovirus glycoprotein B. J Virol 1998; 72:1826-33. [PMID: 9499033 PMCID: PMC109472 DOI: 10.1128/jvi.72.3.1826-1833.1998] [Citation(s) in RCA: 84] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/1997] [Accepted: 12/10/1997] [Indexed: 02/06/2023] Open
Abstract
The human cytomegalovirus (HCMV) glycoprotein B (gB) (also known as gpUL55) homolog is an important mediator of virus entry and cell-to-cell dissemination of infection. To examine the potential ligand-binding properties of gB, a soluble form of gB (gB-S) was radiolabeled, purified, and tested in cell-binding experiments. Binding of gB-S to human fibroblast cells was found to occur in a dose-dependent, saturable, and specific manner. Scatchard analysis demonstrated a biphasic plot with the following estimated dissociation constants (Kd): Kd1, 4.96 x 10(-6) M; Kd2, 3.07 x 10(-7) M. Cell surface heparan sulfate proteoglycans (HSPGs) were determined to serve as one class of receptors able to facilitate gB-S binding. Both HSPG-deficient Chinese hamster ovary (CHO) cells and fibroblast cells with enzymatically removed HSPGs had 40% reductions in gB-S binding, whereas removal of chondroitin sulfate had no effect. However, a significant proportion of gB-S was able to associate with the cell surface in the absence of HSPGs via an undefined nonheparin component. Binding affinity analysis of gB-S binding to wild-type CHO-K1 cells demonstrated biphasic binding kinetics (Kd1, 9.85 x 10(-6) M; Kd2, 4.03 x 10(-8) M), whereas gB-S binding to HSPG-deficient CHO-677 cells exhibited single-component binding kinetics (Kd, 7.46 x 10(-6) M). Together, these data suggest that gB-S associates with two classes of cellular receptors. The interaction of gB with its receptors is physiologically relevant, as evidenced by an inhibitory effect on HCMV entry when cells were pretreated with purified gB-S. This inhibition was determined to be manifested at the level of virus attachment. We conclude that gB is a ligand for HCMV that mediates an interaction with a cellular receptor(s) during HCMV infection.
Collapse
Affiliation(s)
- K A Boyle
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, 53706-1532, USA
| | | |
Collapse
|
29
|
Pietropaolo RL, Compton T. Direct interaction between human cytomegalovirus glycoprotein B and cellular annexin II. J Virol 1997; 71:9803-7. [PMID: 9371650 PMCID: PMC230294 DOI: 10.1128/jvi.71.12.9803-9807.1997] [Citation(s) in RCA: 48] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Cellular annexin II has been shown to specifically bind human cytomegalovirus (HCMV) and be a component of highly purified virions. In this report, we characterize the interaction of annexin II with HCMV. We found that the binding of annexin II to the HCMV envelope occurs partially through the calcium-dependent phospholipid-binding ability of annexin II since some annexin II was dissociated from virions with chelating agents. However, a substantial proportion of virion-associated annexin II was resistant to chelation, which suggested a calcium-independent interaction between annexin II and an HCMV envelope component. The search for a nonphospholipid component to account for this binding led to the discovery that HCMV glycoprotein B (gpUL55) (gB) can physically interact with annexin II. We present three lines of evidence to support the conclusion that HCMV gB can bind host cell annexin II.
Collapse
Affiliation(s)
- R L Pietropaolo
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison Medical School, 53706-1532, USA
| | | |
Collapse
|
30
|
Yurochko AD, Hwang ES, Rasmussen L, Keay S, Pereira L, Huang ES. The human cytomegalovirus UL55 (gB) and UL75 (gH) glycoprotein ligands initiate the rapid activation of Sp1 and NF-kappaB during infection. J Virol 1997; 71:5051-9. [PMID: 9188570 PMCID: PMC191738 DOI: 10.1128/jvi.71.7.5051-5059.1997] [Citation(s) in RCA: 171] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The cellular transcription factors Sp1 and NF-kappaB were upregulated shortly after the binding of purified live or UV-inactivated human cytomegalovirus (HCMV) to the cell surface. The rapid time frame of transcription factor induction is similar to that seen in other systems in which cellular factors are induced following receptor-ligand engagement. This similarity suggested that a cellular receptor-viral ligand interaction might be involved in Sp1 and NF-kappaB activation during the earliest stages of HCMV infection. To focus on the possible role viral ligands play in initiating cellular events following infection, we first used purified viral membrane extracts to demonstrate that constituents on the membrane are responsible for cellular activation. Additionally, these studies showed, through the use of neutralizing antibodies, that the viral membrane mediators of this activation are the major envelope glycoproteins gB (UL55) and gH (UL75). To confirm these results, neutralizing anti-gB and -gH antibodies were used to block the interactions of these glycoproteins on whole purified virus with their cell surface receptors. In so doing, we found that Sp1 and NF-kappaB induction was inhibited. Lastly, through the use of purified viral gB protein and an anti-idiotypic antibody that mimics the image of the viral gH protein, it was found that the engagement of individual viral ligands with their appropriate cell surface receptors was sufficient to activate cellular Sp1 and NF-kappaB. These results support our hypothesis that HCMV glycoproteins mediate an initial signal transduction pathway which leads to the upregulation of host cell transcription factors and suggests a model wherein the orderly sequence of virus-mediated changes in cellular activation initiates with viral binding via envelope glycoproteins to the cognate cellular receptor(s).
Collapse
Affiliation(s)
- A D Yurochko
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, 27599-7295, USA.
| | | | | | | | | | | |
Collapse
|
31
|
Huber MT, Compton T. Characterization of a novel third member of the human cytomegalovirus glycoprotein H-glycoprotein L complex. J Virol 1997; 71:5391-8. [PMID: 9188610 PMCID: PMC191778 DOI: 10.1128/jvi.71.7.5391-5398.1997] [Citation(s) in RCA: 61] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
A prerequisite for understanding the molecular function of the human cytomegalovirus (HCMV) gH (UL75)-gL (UL115) complex is a detailed knowledge of the structure of this complex in its functional form, as it is present in mature virions. The gH protein is known to be a component of a 240-kDa envelope complex designated as gCIII (D. R. Gretch, B. Kari, L. Rasmussen, R. C. Gehrz, and M. F. Stinski, J. Virol. 62:875-881, 1988). However, the exact composition of the gCIII complex remains unknown. In this report, we attempted reconstitution of the gCIII complex by coexpression of gH and gL in the baculovirus expression system. Formation of recombinant gH-gL complexes of approximately 115 kDa was demonstrated; however, no higher-molecular-mass (approximately 240-kDa) recombinant gH-gL complexes were detected, suggesting that the presence of gH and gL alone is not sufficient for reconstitution of the gCIII complex. To identify other mammalian and/or HCMV factors which may be necessary for gCIII formation, immunoprecipitates of gH and gL from HCMV-infected fibroblasts and purified HCMV virions were examined. This analysis did reveal a number of coprecipitating proteins which associate either transiently or integrally with gH and gL. One coprecipitating protein of 145 kDa was shown to be an integral component of gCIII, along with gH and gL. Characterization of the 145-kDa protein demonstrates that it is structurally and antigenically unrelated to gH and gL and that it appears to be virally encoded. Together, these data indicate that the 145-kDa protein is a third novel component of the mature HCMV gH-gL complex.
Collapse
Affiliation(s)
- M T Huber
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, 53706-1532, USA
| | | |
Collapse
|
32
|
Glasgow BJ. Evidence for breaches of the retinal vasculature in acquired immune deficiency syndrome angiopathy. A fluorescent microsphere study. Ophthalmology 1997; 104:753-60. [PMID: 9160019 DOI: 10.1016/s0161-6420(97)30237-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
PURPOSE The author studied the retinal vasculature in patients with acquired immune deficiency syndrome (AIDS) by infusion of microspheres impregnated with fluorescent dye. METHODS Experimental study of the structural integrity of the retinal microvasculature in 14 autopsy patients with AIDS was compared with age- and gender-matched control retinas. MATERIALS Fourteen autopsy eyes from patients with AIDS, eight autopsy eyes from immunosuppressed control patients, and four autopsy eyes from nonimmunosuppressed control patients were studied. EXPERIMENTAL METHOD The central retinal arteries of autopsy eyes were perfused with fluorescent microspheres of 10 and 200 nm in diameter. The retinas were dissected from the eyes and viewed by fluorescence microscopy. RESULTS Vascular breaches permeable to 200-nm microspheres were discovered in eyes from patients with AIDS. Ruptured microaneurysms were identified at the center of retinal hemorrhages in 7 of 14 eyes from patients with AIDS and 5 of 8 immunosuppressed control eyes. Leakage around microaneurysms occurred even in the absence of hemorrhage and were more frequent in eyes from patients with AIDS (11/14) than in control eyes (3/12). Cotton wool patches were surrounded by tortuous retinal vessels and microaneurysms. CONCLUSIONS Ruptured microaneurysms are a frequent cause of retinal hemorrhage in immunosuppressed patients. Breaches in microaneurysms occur even in the absence of hemorrhage. These breaches are often at least 200 nm in diameter, a size that is permissive to capsids and virions of cytomegalovirus (CMV). Ruptured and/or leaky microaneurysms are potential sources of CMV permeation of the blood-retinal barrier. Breaches of the retinal microvasculature are not specific to patients with AIDS and occur frequently in other immunosuppressed patients.
Collapse
Affiliation(s)
- B J Glasgow
- Department of Ophthalmology, University of California, Los Angeles School of Medicine, USA
| |
Collapse
|
33
|
Ludwig GV, Kondig JP, Smith JF. A putative receptor for Venezuelan equine encephalitis virus from mosquito cells. J Virol 1996; 70:5592-9. [PMID: 8764073 PMCID: PMC190519 DOI: 10.1128/jvi.70.8.5592-5599.1996] [Citation(s) in RCA: 103] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
We have identified a cellular protein from a continuous mosquito cell line (C6/36) that appears to play a significant role in the attachment of Venezuelan equine encephalitis (VEE) virus to these cells. VEE virus bound to a 32-kDa polypeptide present in the C6/36 plasma membrane fraction, and binding to this polypeptide was dose dependent and saturable and competed with homologous and heterologous alphaviruses. These observations suggest that this polypeptide binds virus via a receptor-ligand interaction. The 32-kDa polypeptide was expressed on the surfaces of C6/36 cells, and monoclonal antibodies directed against either this cell polypeptide or the VEE virus E2 glycoprotein, which is thought to be the viral attachment protein, interfered with virus attachment. Collectively, these data provide evidence suggesting that the 32-kDa polypeptide serves as a receptor for VEE virus infection of cells. We have characterized this cell polypeptide as a laminin-binding protein on the basis of its ability to interact directly with laminin as well as its immunologic cross-reactivity with the high-affinity human laminin receptor.
Collapse
Affiliation(s)
- G V Ludwig
- Virology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, Maryland 21702-5011, USA
| | | | | |
Collapse
|
34
|
Baskar JF, Smith PP, Ciment GS, Hoffmann S, Tucker C, Tenney DJ, Colberg-Poley AM, Nelson JA, Ghazal P. Developmental analysis of the cytomegalovirus enhancer in transgenic animals. J Virol 1996; 70:3215-26. [PMID: 8627802 PMCID: PMC190185 DOI: 10.1128/jvi.70.5.3215-3226.1996] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
The major immediate-early promoter (MIEP) of human, cytomegalovirus (HCMV) constitutes a primary genetic switch for viral activation. In this study, regulation of the enhancer-containing segment (nucleotides -670 to +54) of the HCMV MIEP attached to the 1acZ reporter gene was examined in the developing embryos of transgenic mice to identify temporal and tissue-specific expression. We find that the transgene reporter is first detected as a dorsal stripe of expression in the neural folds of embryos at day 8.5 postcoitum (p.c.). A broad expression pattern is exhibited in embryos at day 9.5 p.c. This pattern becomes more restricted by day 10.5 p.c. as organogenesis progresses. By day 14.5 p.c., prominent expression is observed in a subpopulation of central nervous system cells and spinal ganglia, endothelial cells, muscle, skin, thyroid, parathyroid, kidney, lung, liver, and gut cells, and the pancreas and submandibular and pituitary glands. This distribution pattern is discussed in relation to human congenital HCMV infection. These results suggest that the transcriptional activity of the HCMV MIEP may determine in part, the ability of the virus to specifically target developing fetal tissues in utero.
Collapse
Affiliation(s)
- J F Baskar
- Department of Immunology, Division of Virology R307B, Scripps Research Institute, La Jolla, California 92037, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Shanley JD, Biegel D, Pachter JS. A rapid and sensitive radioimmunoassay for the detection of human cytomegalovirus binding and infection of human fibroblasts. J Virol Methods 1996; 58:121-9. [PMID: 8783157 DOI: 10.1016/0166-0934(95)02000-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
A rapid and sensitive radioimmunoassay for the quantitation of HCMV binding and infection of human fibroblasts (HFF) was developed. The protocol involves the use of a monoclonal antibody (27-156) reactive with HCMV gB (alpha-gB), followed by an 125I-labeled second antibody to mouse IgG. Antibody to gB bound specifically to HFF inoculated with HCMV when compared to sham inoculated cells or cells inoculated with HSV (strain KOS). Antibody to gB also bound to HFF infected with HCMV 48 h prior to assay. The binding of antibody to HFF inoculated with HCMV was found to be dependent on antibody concentration and to demonstrate saturable kinetics. Moreover, antibody binding was directly dependent on the concentration of the virus inoculum, using either conventional viral preparations or gradient purified HCMV. The binding of antibody to HFF inoculated with HCMV at 4 degrees C was found to be dependent on antibody concentration and to demonstrate saturable kinetics. Displacement of HCMV binding to HFF with the proteoglycan heparin sulfate could be detected, thus allowing for competitive binding studies. This binding assay allows for the relative quantitation of HCMV binding to cells and will be useful for examining the early events of cell-viral interactions.
Collapse
Affiliation(s)
- J D Shanley
- Department of Medicine, University of Connecticut Health Center, Farmington 06030, USA
| | | | | |
Collapse
|
36
|
Abstract
Anti-idiotypic antibodies (anti-ids) have been used successfully in studies on bovine viral diarrhea virus (BVDV) receptor(s) in our laboratory. The anti-ids specifically bound to cultured cells and identified a 50 kDa cellular membrane protein, which is thought to be a specific receptor for BVDV. In this study, flow cytometric analyses demonstrated that the anti-ids also specifically bound to different cell types, namely MDBK, EBK, BT, PK15, MA1O4, and Vero. Experiments on virus attachment and replication showed that BVDV adsorbed to all cells and replicated in them except monkey kidney cells MA 104 and Vero (non-permissive). Results from plaque reduction assays indicated that cellular membrane proteins from all cell lines competitively inhibited BVDV attachment to cultured MDBK cells, suggesting the presence of BVDV receptor on all cells. Immunoblotting of cell membrane proteins with the anti-ids revealed a 50 kDa protein in both permissive and nonpermissive cells. Subcloned or synchronized MDBK cells demonstrated no significant difference of binding with anti-ids as compared to normal cultured cells.
Collapse
Affiliation(s)
- W Xue
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan 66506, USA
| | | |
Collapse
|
37
|
Plachter B, Sinzger C, Jahn G. Cell types involved in replication and distribution of human cytomegalovirus. Adv Virus Res 1996; 46:195-261. [PMID: 8824701 DOI: 10.1016/s0065-3527(08)60073-1] [Citation(s) in RCA: 122] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
As the number of patients suffering from severe HCMV infections has steadily increased, there is a growing need to understand the molecular mechanisms by which the virus causes disease. The factors that control infection at one time and the events leading to virus multiplication at another time are only beginning to be understood. The interaction of HCMV with different host cells is one key for elucidating these processes. Through modern techniques, much has been learned about the biology of HCMV infections in culture systems. In addition to endothelial cells, epithelial cells, and smooth muscle cells, fibroblasts are one cell population preferentially infected in solid tissues in vivo. From these sites of multiplication, the virus may be carried by peripheral monocytes and circulating endothelial cells to reach distant sites of the body. This would explain the multiorgan involvement in acute HCMV infection and the modes of viral transmission. From what has been learned mainly from human fibroblast culture systems, future studies will focus on how HCMV regulates the expression of its putative 200 genes in different host cells at different stages of cell differentiation and activation to result in viral latency and pathogenesis.
Collapse
Affiliation(s)
- B Plachter
- Institut für Klinische und Molekulare Virologie, Universität Erlangen-Nürnberg, Germany
| | | | | |
Collapse
|
38
|
Lobert PE, Hober D, Dewilde A, Wattré P. Cell membrane bound N-acetylneuraminic acid is involved in the infection of fibroblasts and phorbol-ester differentiated monocyte-like cells with human cytomegalovirus (HCMV). Arch Virol 1995; 140:1357-71. [PMID: 7661690 DOI: 10.1007/bf01322663] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
We focused on the role of membrane bound sugar residues in the infection of fibroblasts and monocyte-like cells with human cytomegalovirus (HCMV). Treatment of phorbol 12-myristate 13-acetate (PMA) differentiated monocyte-like cells THP-1 or human fibroblasts MRC-5 with lectins specific for N-acetylneuraminic acid (NeuAc) blocked infection with HCMV. HCMV failed to infect sialidase-treated differentiated THP-1 cells or MRC-5 cells. By using NeuAc, N-glycolylneuraminic acid (NeuGl) and alpha 2-3, but not alpha 2-6, sialyl-oligosaccharide, the infection of cells was less efficient. NeuAc was more potent inhibitor than NeuGl. These observations suggest that the sialic acid specificity and the nature of the interglycosidic linkage at the end of the complex carbohydrates may play an important role. Analogous experiments indicated that HCMV binds to N-acetylglucosamine (GlcNAc) in addition to NeuAc. Human cytomegalovirus infection in differentiated THP-1 cells and in human fibroblasts was inhibited by incubation of the virus with 20 micrograms/ml of heparin before and during the adsorption period. Treatment of the cells with heparinase or heparitinase inhibited infection with HCMV. We emphasized the role of NeuAc and GlcNAc and heparan sulfate proteoglycans at the surface of the cells, in the early steps of infection of both human fibroblasts and PMA differentiated monocyte-like cells with HCMV.
Collapse
Affiliation(s)
- P E Lobert
- Laboratoire de Virologie, Centre Hospitalier, Lille, France
| | | | | | | |
Collapse
|
39
|
Price P, Allcock RJ, Coombe DR, Shellam GR, McCluskey J. MHC proteins and heparan sulphate proteoglycans regulate murine cytomegalovirus infection. Immunol Cell Biol 1995; 73:308-15. [PMID: 7493766 DOI: 10.1038/icb.1995.47] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Factors influencing MCMV infection mediated by MHC class 1 molecules were analysed further as previous studies showed that the effects of the MHC genotype on sensitivity to this virus are important in vivo. Here we show that H-2d, H-2b, H-2r and H-2v macrophages are highly sensitive to MCMV. Moreover, transfection of H-2k L-cells with Kb or Dd conferred sensitivity to MCMV. This was not affected by amino acid substitutions in Kb alpha 1 or alpha 2, although previous studies demonstrated that exchange of the alpha 1 domain of Dd with Ld alpha 1 compromised sensitivity. Here replacement of Kb alpha 3 with Ld alpha 3 reduced susceptibility to low doses of MCMV. In addition, extracellular beta 2-microglobulin (beta 2m) promoted infection of beta 2m-negative RIE/TL8X.1 cells transfected with Db with or without a beta 2m gene. Hence MCMV infection can involve beta 2m and the alpha 1 and alpha 3 domains of MHC heavy chains. MCMV infection of L-cells expressing Dd or Kb was also inhibited by heparin, but infection of the parental L-cell line was not reproducibly affected. A role for heparan sulphate proteoglycan in MHC-mediated MCMV infection was confirmed using cells pre-treated with heparinase I or III, or propagated in chlorate to inhibit the sulphation of the glycosaminoglycan chains.
Collapse
Affiliation(s)
- P Price
- Department of Microbiology, University of Western Australia, Nedlands, Australia
| | | | | | | | | |
Collapse
|
40
|
Wright JF, Kurosky A, Pryzdial EL, Wasi S. Host cellular annexin II is associated with cytomegalovirus particles isolated from cultured human fibroblasts. J Virol 1995; 69:4784-91. [PMID: 7609045 PMCID: PMC189289 DOI: 10.1128/jvi.69.8.4784-4791.1995] [Citation(s) in RCA: 71] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
A significant amount of host cellular annexin II was found to be associated with human cytomegalovirus isolated from cultured human fibroblasts (approximately 1,160 molecules per virion). This composition was established by four different analytical approaches that included (i) Western blot (immunoblot) analysis of gradient-purified virions with a monoclonal antibody specific for annexin II, (ii) peptide mapping and sequence analysis of virus-associated proteins and proteins dissociated from virus following EDTA treatment, (iii) electron microscopic immunocytochemistry of gradient-purified virions, and (iv) labeling of virus-associated proteins by lactoperoxidase-catalyzed radioiodination. These results indicated that annexin II was primarily localized to the viral surface, where it bound in a divalent cation-dependent manner. In functional experiments, a rabbit antiserum raised against annexin II inhibited cytomegalovirus plaque formation in human foreskin fibroblast monolayers in a concentration-dependent manner. Cumulatively, these studies demonstrate an association of host annexin II with cytomegalovirus particles and provide evidence for the involvement of this cellular protein in virus infectivity.
Collapse
Affiliation(s)
- J F Wright
- Canadian Red Cross Society, Toronto Centre
| | | | | | | |
Collapse
|
41
|
Norkin LC. Virus receptors: implications for pathogenesis and the design of antiviral agents. Clin Microbiol Rev 1995; 8:293-315. [PMID: 7621403 PMCID: PMC172860 DOI: 10.1128/cmr.8.2.293] [Citation(s) in RCA: 54] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
A virus initiates infection by attaching to its specific receptor on the surface of a susceptible host cell. This prepares the way for the virus to enter the cell. Consequently, the expression of the receptor on specific cells and tissues of the host is a major determinant of the route of entry of the virus into the host and of the patterns of virus spread and pathogenesis in the host. This review emphasizes the virus-receptor interactions of human immunodeficiency virus, the rhinoviruses, the herpesviruses, and the coronaviruses. These interactions are often found to be complex and dynamic, involving multiple sites or factors on both the virus and the host cell. Also, the receptor may play an important role in virus entry per se in addition to its role in virus binding. In the cases of human immunodeficiency virus and the rhinoviruses, ingenious approaches to therapeutic strategies based on inhibiting virus attachment and entry are under development and in clinical trials.
Collapse
Affiliation(s)
- L C Norkin
- Department of Microbiology, University of Massachusetts, Amherst 01003, USA
| |
Collapse
|
42
|
Holberg-Petersen M, Bukholm G, Rollag H, Degré M. Infection with human cytomegalovirus enhances bacterial adhesiveness and invasiveness in permissive and semipermissive cells. APMIS 1994; 102:703-10. [PMID: 7946274 DOI: 10.1111/j.1699-0463.1994.tb05223.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
The effect of human cytomegalovirus (HCMV) infection on adhesiveness and invasiveness of Salmonella typhimurium was examined in cells permissive (human embryo fibroblasts (HE)), semipermissive (A549) and nonpermissive (HEp-2) for the virus. Preinfection of the cells with HCMV induced enhanced adhesiveness and invasiveness of bacteria in the permissive HE cells. In the semipermissive A549 cells, where HCMV immediate-early (IE) mRNA transcripts and IE proteins were detected, a significant effect on the initial phase of invasiveness, the adherence phase, was demonstrated. HCMV had no effect on invasiveness of S. typhimurium in nonpermissive HEp-2 cells. Neither HCMV IE transcripts nor IE proteins could be detected in these cells.
Collapse
Affiliation(s)
- M Holberg-Petersen
- Kaptein W. Wilhelmsen og Frues Bakteriologiske Institutt, University of Oslo, Rikshospitalet, Norway
| | | | | | | |
Collapse
|
43
|
Lipson SM, Ciamician Z, Kaplan MH. Comparison of cytomegalovirus antigenemia assay with shell vial-indirect immunofluorescence assay for rapid detection of cytomegalovirus in blood. J Clin Microbiol 1994; 32:1619-20. [PMID: 8077419 PMCID: PMC264055 DOI: 10.1128/jcm.32.6.1619-1620.1994] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
|
44
|
Kreutz LC, Seal BS, Mengeling WL. Early interaction of feline calicivirus with cells in culture. Arch Virol 1994; 136:19-34. [PMID: 8002786 DOI: 10.1007/bf01538814] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
The kinetics and biochemical properties of feline calicivirus (FCV) attachment to Crandell-Reese feline kidney cells were determined. Maximum binding was observed at pH 6.5. Cells in suspension at 4 degrees C bound virus more efficiently than cells in monolayers at 4 degrees C or 37 degrees C. High initial binding rate was observed in monolayers or cells in suspension and proceeded to a maximum at 90 min, although half maximal binding was observed as early as 15 min. Binding was specific and competitively blocked by serotypically homologous or heterologous FCV as well as by San Miguel sea lion virus. Treatment of cells with proteases increased FCV binding, whereas phospholipase had no effect on virus attachment. Conversely, cells treated with neuraminidase followed by O-glycanase treatment showed a decreased binding ability. Cells of feline origin bound FCV very efficiently, and non-permissive cells showed a poor binding ability. Following transfection of viral RNA, infectious virus could be recovered from all non-permissive cells, except from Madin-Darby canine kidney cells. These results suggest that FCV binds to a receptor in which carbohydrates may be an important component and that FCV replication in non-permissive cells is primarily restricted by the absence of appropriate receptors on the cell surface.
Collapse
Affiliation(s)
- L C Kreutz
- Virology Swine Research Unit, National Animal Disease Center, USDA, Ames, Iowa
| | | | | |
Collapse
|
45
|
Geoffroy F, Ogier G, Chantepie J, Quash G. Inactivation of human cytomegalovirus by sodium periodate oxidation. Arch Virol 1994; 135:61-74. [PMID: 8198450 DOI: 10.1007/bf01309765] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Human cytomegalovirus (HCMV), oxidized by sodium periodate (NaIO4), is incapable of giving rise to viral progeny in cell culture. At a NaIO4 concentration as low as 5 mM, there is a loss of at least 6 logs of viral infectivity which occurs very rapidly (less than 5 min). Further, the inactivation is a first-order reaction depending on the periodate concentration. Adsorption to the cell surface, penetration into cells, and penetration of the viral DNA into cell nuclei were found to occur identically in mock oxidized and oxidized HCMV. Since the carbohydrate moiety of viral glycoproteins was the target of periodate attack, these observations strongly suggest that the structural integrity of the sugar residues is not a prerequisite for adsorption and penetration. Nevertheless, no evidence for viral DNA or protein synthesis was detected in cells infected with oxidized virus, and even after 3 weeks in culture, no cytopathic effect was observed.
Collapse
Affiliation(s)
- F Geoffroy
- Laboratoire d'Immunochimie INSERM-C.J.F. 89-05, Faculté de Médecine Lyon-Sud, Oullins, France
| | | | | | | |
Collapse
|
46
|
Ouwehand AJ, Balk AH, Baan CC, Vaessen LM, Jutte NH, Bos E, Claas FH, Weimar W. Cytomegalovirus infection and allospecific cytotoxic activity of graft-infiltrating cells after heart transplantation. J Med Virol 1994; 42:175-81. [PMID: 8158113 DOI: 10.1002/jmv.1890420214] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
We have investigated whether cytomegalovirus (CMV) infection has an effect on donor directed cytotoxicity of graft-infiltrating cells in human heart transplants. Our study group consisted of 89 heart transplant recipients. Thirty-eight (43%) showed signs of CMV infection; in 28 of them, cytolytic activity of biopsy derived cultures could be tested during the infection. Eight patients had a primary and 20 a secondary infection. We found that during CMV infection, both primary and secondary, a significantly higher proportion of the biopsy-derived cultures showed cytotoxicity against donor HLA antigens (chi 2 test; P < 0.01 in comparison with 51 patients without infection). This was most evident in patients with both infection and acute rejection episodes when compared to patients with only one of these complications. This suggests that one process amplifies the other with regard to the up-regulation of alloreactivity within the transplanted heart. In secondary infections, only an increase of donor class I-directed cytotoxicity was found, while in primary infections cytotoxicity against donor class I and II antigens was increased (P < 0.005 vs. secondary infection).
Collapse
Affiliation(s)
- A J Ouwehand
- Department of Thoracic Surgery, Erasmus University, Rotterdam, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Söderberg C, Giugni TD, Zaia JA, Larsson S, Wahlberg JM, Möller E. CD13 (human aminopeptidase N) mediates human cytomegalovirus infection. J Virol 1993; 67:6576-85. [PMID: 8105105 PMCID: PMC238095 DOI: 10.1128/jvi.67.11.6576-6585.1993] [Citation(s) in RCA: 97] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Human cytomegalovirus (HCMV) infects cells by a series of processes including attachment, penetration via fusion of the envelope with the plasma membrane, and transport of the viral DNA to the nucleus. The details of the early events of HCMV infection are poorly understood. We have recently reported that CD13, human aminopeptidase N, a metalloprotease, is present on blood cells susceptible in vitro to HCMV infection (C. Söderberg, S. Larsson, S. Bergstedt-Lindqvist, and E. Möller, J. Virol. 67:3166-3175, 1993). Here we report that human CD13 is involved in HCMV infection. Antibodies directed against human CD13 not only inhibit infection but also block binding of HCMV virions to susceptible cells. Compounds known to inhibit aminopeptidase activity block HCMV infection. HCMV-resistant murine fibroblasts have heightened susceptibility to HCMV infection after transfection with complementary DNA encoding human CD13. A significant increase in binding of HCMV was observed in the CD13-expressing transfectants compared with neomycin-resistant control mouse cells. However, murine fibroblasts transfected with mutant CD13, lacking a portion of the aminopeptidase active site, remained susceptible to HCMV infection. Thus, human CD13 appears to mediate HCMV infection by a process that increases binding, but its enzymatic domain is not necessary for infection.
Collapse
Affiliation(s)
- C Söderberg
- Department of Clinical Immunology, NOVUM, Karolinska Institute at Huddinge Hospital, Stockholm, Sweden
| | | | | | | | | | | |
Collapse
|
48
|
|
49
|
Wykes MN, Shellam GR, McCluskey J, Kast WM, Dallas PB, Price P. Murine cytomegalovirus interacts with major histocompatibility complex class I molecules to establish cellular infection. J Virol 1993; 67:4182-9. [PMID: 8389923 PMCID: PMC237787 DOI: 10.1128/jvi.67.7.4182-4189.1993] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
The expression of stable, correctly folded major histocompatibility complex class I molecules conferred susceptibility to murine cytomegalovirus (MCMV) in cells which were previously resistant to infection, demonstrating that these molecules interact critically with MCMV to initiate infection. All class I molecules could potentiate MCMV infection but H-2Dd and Kb molecules were most efficient. Monoclonal antibodies specific for the alpha 1 and/or alpha 2 domains of Dd and Kb inhibited infection. Infection of L cells transfected with hybrid major histocompatibility complex class I molecules demonstrated that allelic control of susceptibility to MCMV mapped to the alpha 1 domain of Dd when in correct configuration with the alpha 2 and alpha 3 domains. In MCMV-resistant RMA-S cells, an improvement in the conformation of class I molecules introduced susceptibility to infection.
Collapse
Affiliation(s)
- M N Wykes
- Department of Microbiology, University of Western Australia, Nedlands
| | | | | | | | | | | |
Collapse
|
50
|
Söderberg C, Larsson S, Bergstedt-Lindqvist S, Möller E. Definition of a subset of human peripheral blood mononuclear cells that are permissive to human cytomegalovirus infection. J Virol 1993; 67:3166-75. [PMID: 7684461 PMCID: PMC237655 DOI: 10.1128/jvi.67.6.3166-3175.1993] [Citation(s) in RCA: 99] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
The identity of cells responsible for transmission of human cytomegalovirus (HCMV) in blood products or bone marrow transplants is unknown. We have tested the capacity of HCMV to in vitro infect human peripheral blood mononuclear cells (PBMC) from healthy donors and found that certain PBMC are permissive to HCMV infection. In vitro-infected viable cells were double stained for surface expression of different HMCV proteins and for cell-type-specific antigens to allow the identification of sensitive cells. All analysis were performed on viable cells, using HCMV-specific monoclonal antibodies and automated flow cytofluorimetry. PBMC were infected either with the laboratory-adapted HCMV strain AD169 or with a virus isolate obtained from a viremic patient. Up to 25% of all PBMC could express the major immediate-early antigen as well as the pp65 antigen, known at the lower matrix protein. Infected cells were mainly CD14+ monocytes, but also a small population of large CD8+ cells were susceptible to HCMV infection. CD19+ B lymphocytes were resistant to HCMV infection. Different populations of infected cells were enriched by using Dynabeads coated with cell-type-specific antibodies, and the presence of infectious virus was demonstrated by incubating the selected and sonicated cell material on human fibroblasts. Only material from infected monocytes and from CD3+ CD8+ cells gave rise to HCMV-specific plaques. The presence of HCMV mRNA as a sign of active viral transcription of the major immediate-early and late pp150 genes in infected cells was demonstrated by using nested reversed polymerase chain reaction. A common denominator was found for all cells that could be infected with HCMV. The CD13 antigen, a 130- to 150-kDa integral membrane protein identical to the enzyme aminopeptidase N, was expressed on all HCMV-permissive cells.
Collapse
MESH Headings
- Antigens, CD/immunology
- Antigens, CD19
- Antigens, Differentiation, B-Lymphocyte/immunology
- Antigens, Differentiation, Myelomonocytic/immunology
- B-Lymphocyte Subsets/immunology
- B-Lymphocyte Subsets/microbiology
- Base Sequence
- CD13 Antigens
- CD8 Antigens/immunology
- Cell Separation
- Cytomegalovirus/growth & development
- Cytomegalovirus Infections/microbiology
- Genetic Variation
- Humans
- Immunity, Innate
- Leukocytes, Mononuclear/immunology
- Leukocytes, Mononuclear/microbiology
- Lipopolysaccharide Receptors
- Molecular Sequence Data
- Monocytes/immunology
- Monocytes/microbiology
- Receptors, Virus
- T-Lymphocyte Subsets/immunology
- T-Lymphocyte Subsets/microbiology
- Transcription, Genetic
Collapse
Affiliation(s)
- C Söderberg
- Department of Clinical Immunology, NOVUM, Karolinska Institute, Huddinge Hospital, Stockholm, Sweden
| | | | | | | |
Collapse
|